Health Literacy in Dermatology Patients: How to Level the Playing Field

Article Type
Changed
Thu, 09/08/2022 - 14:04
Display Headline
Health Literacy in Dermatology Patients: How to Level the Playing Field

Health literacy is a multifaceted construct that encompasses the knowledge of health and health systems, utilization of information related to health, and ability to maintain health.1 Low health literacy impairs health outcomes, disproportionately affecting socioeconomically disadvantaged populations, including racial minorities and the older population. Consistently, it is associated with fewer vaccinations and screenings, higher health care utilization, and poorer ability to take medications or interpret health information.2

With growing utilization of the Internet for health information,3 much patient education now occurs outside the clinic. Differential utilization of the Internet can exacerbate disparities in health outcomes: people with a lower family income more frequently engage in health information and dialogue online.3 Despite opportunities to improve literacy and narrow gaps in care, a lack of awareness, advocacy, and funding limit patient- and community-based initiatives. Herein, we discuss health literacy challenges in dermatology, offer potential solutions, and propose ways that stakeholders can prioritize health literacy advocacy to improve outcomes.

The Importance of Health Literacy in Dermatology

Dermatology patients often face challenges that demand greater health literacy. Active participation in health promotion, protection, and maintenance can remarkably improve outcomes. When patients understand disease pathogenesis and the rationale behind treatment choices, adherence to a treatment regimen might improve.

However, understanding dermatologic diseases and disorders can be challenging. First, many are chronic inflammatory conditions that require intricate treatment regimens. Second, the complexity of those diseases and disorders continues to grow in the era of new research and unprecedented expansion of treatment options.

For chronic conditions that require ongoing complex management, researchers have developed advanced patient tools. For instance, the eczema action plan helps atopic dermatitis patients manage conditions from home.4 However, patients with greater literacy and the ability to participate will better utilize such tools and have fewer uncontrolled flares. Patient tools meant to improve outcomes might, instead, widen gaps in care. Even with nonchronic conditions, such as nonmelanoma skin cancer, continued awareness and the need for preventive care, timely diagnosis, and appropriate intervention remain critical.

Limited Accessibility of Patient Education Materials

Patient education in dermatology occurs through several formats. Because online health resources are more readily available to those with less access to health care, the potential for such resources to narrow health disparities is immense. However, online resources have not adequately taken advantage of the opportunity to make health information openly accessible to its users. The readability of online patient education materials on a large expanse of dermatologic conditions is far too advanced.5 The readability level of some resources is as high as 17th grade (graduate school), which is much higher than the American Medical Association recommendation6 that patient education materials be presented at a 6th-grade level or less. Furthermore, the quality and comprehensiveness of content is highly variable. Rather than serving as an equalizer, the Internet may widen the gap as low health literacy continues to impair the accessibility of health information.

Solutions to Level the Playing Field

What can be done to increase the readability of patient education materials? Leveling the playing field begins with creating materials at an appropriate readability level, including online content, printed handouts, and after-visit summaries in the clinic. Writers of patient education materials should be cognizant of their choice of language and routinely use a free readability checker (https://readabilityformulas.com). Patient education materials should reflect the American Medical Association’s recommended 6th-grade level. Creators should maintain a high standard of quality and comprehensiveness; prior studies note no inverse correlation between readability and quality.5 In the age of multimedia presentation, non–print-based materials can be explored, such as audio or video for online content, podcasts, and webinars. Providers also should take the opportunity to be mindful of health literacy in clinic. Beyond assessing the readability of written resources for a patient, assessing that patient’s health literacy and tailoring one’s language will maximize engagement.

Systemic Change Is Needed

Ultimately, systemic change is needed to address the root causes of health literacy disparity, requiring advocacy for social welfare, public health, and public policy initiatives. In recognizing existing efforts, such as community outreach teams and hospital committees to evaluate health literacy materials, numerous barriers remain. Despite the notable impact of health literacy on health outcomes, there is a lack of advocacy and funds to conduct health literacy–related work.7 Because dermatologists provide holistic care and remain mindful of patients’ health literacy in the clinic, they should continue to advocate for increased awareness, improved funding, and support for local and federal initiatives.

Final Thoughts

With more opportunities to narrow gaps in care, it is more pertinent than ever to acknowledge the impact of health literacy on dermatology outcomes. Leveling the playing field begins with (1) an awareness of health literacy and (2) creating readable and comprehensible patient education content. Greater advocacy from community and professional organizations; increased funding from nonprofit organizations, industry, and federal institutions; and increased involvement by dermatologists in bringing greater attention to health literacy will improve outcomes in dermatology.

References
  1. Liu C, Wang D, Liu C, et al. What is the meaning of health literacy? a systematic review and qualitative synthesis. Fam Med Community Health. 2020;8:e000351. doi:10.1136/fmch-2020-000351
  2. Berkman ND, Sheridan SL, Donahue KE, et al. Low health literacy and health outcomes: an updated systematic review. Ann Intern Med. 2011;155:97-107. doi:10.7326/0003-4819-155-2-201107190-00005
  3. Rice RE. Influences, usage, and outcomes of Internet health information searching: multivariate results from the Pew surveys. Int J Med Inform. 2006;75:8-28. doi:10.1016/j.ijmedinf.2005.07.032
  4. Brown J, Weitz NW, Liang A, et al. Does an eczema action plan improve atopic dermatitis? a single-site randomized controlled trial. Clin Pediatr (Phila). 2018;57:1624-1629. doi:10.1177/0009922818795906
  5. De DR, Shih T, Katta R, et al. Readability, quality, and timeliness of patient online health resources for contact dermatitis and patch testing. Dermatitis. 2022;33:155-160. doi:10.1097/DER.0000000000000789
  6. Weiss BD. Health Literacy: A Manual for Clinicians. American Medical Association, American Medical Foundation; 2003.
  7. Nutbeam D, McGill B, Premkumar P. Improving health literacy in community populations: a review of progress. Health Promot Int. 2018;33:901-911. doi:10.1093/heapro/dax015
Article PDF
Author and Disclosure Information

Ms. Shih is from the David Geffen School of Medicine, University of California, Los Angeles. Ms. De is from the Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, New York. Drs. Tran and Shi are from the Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock. Dr. Hsiao is from the Department of Dermatology, University of Southern California, Los Angeles.

Ms. Shih, Ms. De, and Dr. Tran report no conflict of interest. Dr. Hsiao is on the board of directors for the HS Foundation; has served as a consultant for Boehringer Ingelheim, Novartis, and UCB; and has served as a consultant and speaker for AbbVie. Dr. Shi is on the board of directors for the HS Foundation and is a shareholder of LearnHealth. She also has served as an advisory board member, investigator, and/or speaker for, and/or received research funding from AbbVie; Altus Labs; Alumis; Aristea Therapeutics; Boehringer Ingelheim; Burt’s Bees; Dermira; Eli Lilly and Company; Galderma; Genzyme, a Sanofi company; gpskin; Incyte; Kiniksa Pharmaceuticals; LEO Pharma; MyOr; Novartis; Pfizer; Polyfins; Regeneron; Skin Actives Scientific; SUN Pharma Industries; TARGET PharmaSolutions; UCB; and VYNE Therapeutics.

Correspondence: Vivian Y. Shi, MD, Department of Dermatology, University of Arkansas for Medical Sciences, 4301 W Markham St, #576, Little Rock, AR 72205-7199 (vivian.shi.publications@gmail.com).

Issue
Cutis - 110(3)
Publications
Topics
Page Number
119-120
Sections
Author and Disclosure Information

Ms. Shih is from the David Geffen School of Medicine, University of California, Los Angeles. Ms. De is from the Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, New York. Drs. Tran and Shi are from the Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock. Dr. Hsiao is from the Department of Dermatology, University of Southern California, Los Angeles.

Ms. Shih, Ms. De, and Dr. Tran report no conflict of interest. Dr. Hsiao is on the board of directors for the HS Foundation; has served as a consultant for Boehringer Ingelheim, Novartis, and UCB; and has served as a consultant and speaker for AbbVie. Dr. Shi is on the board of directors for the HS Foundation and is a shareholder of LearnHealth. She also has served as an advisory board member, investigator, and/or speaker for, and/or received research funding from AbbVie; Altus Labs; Alumis; Aristea Therapeutics; Boehringer Ingelheim; Burt’s Bees; Dermira; Eli Lilly and Company; Galderma; Genzyme, a Sanofi company; gpskin; Incyte; Kiniksa Pharmaceuticals; LEO Pharma; MyOr; Novartis; Pfizer; Polyfins; Regeneron; Skin Actives Scientific; SUN Pharma Industries; TARGET PharmaSolutions; UCB; and VYNE Therapeutics.

Correspondence: Vivian Y. Shi, MD, Department of Dermatology, University of Arkansas for Medical Sciences, 4301 W Markham St, #576, Little Rock, AR 72205-7199 (vivian.shi.publications@gmail.com).

Author and Disclosure Information

Ms. Shih is from the David Geffen School of Medicine, University of California, Los Angeles. Ms. De is from the Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, New York. Drs. Tran and Shi are from the Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock. Dr. Hsiao is from the Department of Dermatology, University of Southern California, Los Angeles.

Ms. Shih, Ms. De, and Dr. Tran report no conflict of interest. Dr. Hsiao is on the board of directors for the HS Foundation; has served as a consultant for Boehringer Ingelheim, Novartis, and UCB; and has served as a consultant and speaker for AbbVie. Dr. Shi is on the board of directors for the HS Foundation and is a shareholder of LearnHealth. She also has served as an advisory board member, investigator, and/or speaker for, and/or received research funding from AbbVie; Altus Labs; Alumis; Aristea Therapeutics; Boehringer Ingelheim; Burt’s Bees; Dermira; Eli Lilly and Company; Galderma; Genzyme, a Sanofi company; gpskin; Incyte; Kiniksa Pharmaceuticals; LEO Pharma; MyOr; Novartis; Pfizer; Polyfins; Regeneron; Skin Actives Scientific; SUN Pharma Industries; TARGET PharmaSolutions; UCB; and VYNE Therapeutics.

Correspondence: Vivian Y. Shi, MD, Department of Dermatology, University of Arkansas for Medical Sciences, 4301 W Markham St, #576, Little Rock, AR 72205-7199 (vivian.shi.publications@gmail.com).

Article PDF
Article PDF

Health literacy is a multifaceted construct that encompasses the knowledge of health and health systems, utilization of information related to health, and ability to maintain health.1 Low health literacy impairs health outcomes, disproportionately affecting socioeconomically disadvantaged populations, including racial minorities and the older population. Consistently, it is associated with fewer vaccinations and screenings, higher health care utilization, and poorer ability to take medications or interpret health information.2

With growing utilization of the Internet for health information,3 much patient education now occurs outside the clinic. Differential utilization of the Internet can exacerbate disparities in health outcomes: people with a lower family income more frequently engage in health information and dialogue online.3 Despite opportunities to improve literacy and narrow gaps in care, a lack of awareness, advocacy, and funding limit patient- and community-based initiatives. Herein, we discuss health literacy challenges in dermatology, offer potential solutions, and propose ways that stakeholders can prioritize health literacy advocacy to improve outcomes.

The Importance of Health Literacy in Dermatology

Dermatology patients often face challenges that demand greater health literacy. Active participation in health promotion, protection, and maintenance can remarkably improve outcomes. When patients understand disease pathogenesis and the rationale behind treatment choices, adherence to a treatment regimen might improve.

However, understanding dermatologic diseases and disorders can be challenging. First, many are chronic inflammatory conditions that require intricate treatment regimens. Second, the complexity of those diseases and disorders continues to grow in the era of new research and unprecedented expansion of treatment options.

For chronic conditions that require ongoing complex management, researchers have developed advanced patient tools. For instance, the eczema action plan helps atopic dermatitis patients manage conditions from home.4 However, patients with greater literacy and the ability to participate will better utilize such tools and have fewer uncontrolled flares. Patient tools meant to improve outcomes might, instead, widen gaps in care. Even with nonchronic conditions, such as nonmelanoma skin cancer, continued awareness and the need for preventive care, timely diagnosis, and appropriate intervention remain critical.

Limited Accessibility of Patient Education Materials

Patient education in dermatology occurs through several formats. Because online health resources are more readily available to those with less access to health care, the potential for such resources to narrow health disparities is immense. However, online resources have not adequately taken advantage of the opportunity to make health information openly accessible to its users. The readability of online patient education materials on a large expanse of dermatologic conditions is far too advanced.5 The readability level of some resources is as high as 17th grade (graduate school), which is much higher than the American Medical Association recommendation6 that patient education materials be presented at a 6th-grade level or less. Furthermore, the quality and comprehensiveness of content is highly variable. Rather than serving as an equalizer, the Internet may widen the gap as low health literacy continues to impair the accessibility of health information.

Solutions to Level the Playing Field

What can be done to increase the readability of patient education materials? Leveling the playing field begins with creating materials at an appropriate readability level, including online content, printed handouts, and after-visit summaries in the clinic. Writers of patient education materials should be cognizant of their choice of language and routinely use a free readability checker (https://readabilityformulas.com). Patient education materials should reflect the American Medical Association’s recommended 6th-grade level. Creators should maintain a high standard of quality and comprehensiveness; prior studies note no inverse correlation between readability and quality.5 In the age of multimedia presentation, non–print-based materials can be explored, such as audio or video for online content, podcasts, and webinars. Providers also should take the opportunity to be mindful of health literacy in clinic. Beyond assessing the readability of written resources for a patient, assessing that patient’s health literacy and tailoring one’s language will maximize engagement.

Systemic Change Is Needed

Ultimately, systemic change is needed to address the root causes of health literacy disparity, requiring advocacy for social welfare, public health, and public policy initiatives. In recognizing existing efforts, such as community outreach teams and hospital committees to evaluate health literacy materials, numerous barriers remain. Despite the notable impact of health literacy on health outcomes, there is a lack of advocacy and funds to conduct health literacy–related work.7 Because dermatologists provide holistic care and remain mindful of patients’ health literacy in the clinic, they should continue to advocate for increased awareness, improved funding, and support for local and federal initiatives.

Final Thoughts

With more opportunities to narrow gaps in care, it is more pertinent than ever to acknowledge the impact of health literacy on dermatology outcomes. Leveling the playing field begins with (1) an awareness of health literacy and (2) creating readable and comprehensible patient education content. Greater advocacy from community and professional organizations; increased funding from nonprofit organizations, industry, and federal institutions; and increased involvement by dermatologists in bringing greater attention to health literacy will improve outcomes in dermatology.

Health literacy is a multifaceted construct that encompasses the knowledge of health and health systems, utilization of information related to health, and ability to maintain health.1 Low health literacy impairs health outcomes, disproportionately affecting socioeconomically disadvantaged populations, including racial minorities and the older population. Consistently, it is associated with fewer vaccinations and screenings, higher health care utilization, and poorer ability to take medications or interpret health information.2

With growing utilization of the Internet for health information,3 much patient education now occurs outside the clinic. Differential utilization of the Internet can exacerbate disparities in health outcomes: people with a lower family income more frequently engage in health information and dialogue online.3 Despite opportunities to improve literacy and narrow gaps in care, a lack of awareness, advocacy, and funding limit patient- and community-based initiatives. Herein, we discuss health literacy challenges in dermatology, offer potential solutions, and propose ways that stakeholders can prioritize health literacy advocacy to improve outcomes.

The Importance of Health Literacy in Dermatology

Dermatology patients often face challenges that demand greater health literacy. Active participation in health promotion, protection, and maintenance can remarkably improve outcomes. When patients understand disease pathogenesis and the rationale behind treatment choices, adherence to a treatment regimen might improve.

However, understanding dermatologic diseases and disorders can be challenging. First, many are chronic inflammatory conditions that require intricate treatment regimens. Second, the complexity of those diseases and disorders continues to grow in the era of new research and unprecedented expansion of treatment options.

For chronic conditions that require ongoing complex management, researchers have developed advanced patient tools. For instance, the eczema action plan helps atopic dermatitis patients manage conditions from home.4 However, patients with greater literacy and the ability to participate will better utilize such tools and have fewer uncontrolled flares. Patient tools meant to improve outcomes might, instead, widen gaps in care. Even with nonchronic conditions, such as nonmelanoma skin cancer, continued awareness and the need for preventive care, timely diagnosis, and appropriate intervention remain critical.

Limited Accessibility of Patient Education Materials

Patient education in dermatology occurs through several formats. Because online health resources are more readily available to those with less access to health care, the potential for such resources to narrow health disparities is immense. However, online resources have not adequately taken advantage of the opportunity to make health information openly accessible to its users. The readability of online patient education materials on a large expanse of dermatologic conditions is far too advanced.5 The readability level of some resources is as high as 17th grade (graduate school), which is much higher than the American Medical Association recommendation6 that patient education materials be presented at a 6th-grade level or less. Furthermore, the quality and comprehensiveness of content is highly variable. Rather than serving as an equalizer, the Internet may widen the gap as low health literacy continues to impair the accessibility of health information.

Solutions to Level the Playing Field

What can be done to increase the readability of patient education materials? Leveling the playing field begins with creating materials at an appropriate readability level, including online content, printed handouts, and after-visit summaries in the clinic. Writers of patient education materials should be cognizant of their choice of language and routinely use a free readability checker (https://readabilityformulas.com). Patient education materials should reflect the American Medical Association’s recommended 6th-grade level. Creators should maintain a high standard of quality and comprehensiveness; prior studies note no inverse correlation between readability and quality.5 In the age of multimedia presentation, non–print-based materials can be explored, such as audio or video for online content, podcasts, and webinars. Providers also should take the opportunity to be mindful of health literacy in clinic. Beyond assessing the readability of written resources for a patient, assessing that patient’s health literacy and tailoring one’s language will maximize engagement.

Systemic Change Is Needed

Ultimately, systemic change is needed to address the root causes of health literacy disparity, requiring advocacy for social welfare, public health, and public policy initiatives. In recognizing existing efforts, such as community outreach teams and hospital committees to evaluate health literacy materials, numerous barriers remain. Despite the notable impact of health literacy on health outcomes, there is a lack of advocacy and funds to conduct health literacy–related work.7 Because dermatologists provide holistic care and remain mindful of patients’ health literacy in the clinic, they should continue to advocate for increased awareness, improved funding, and support for local and federal initiatives.

Final Thoughts

With more opportunities to narrow gaps in care, it is more pertinent than ever to acknowledge the impact of health literacy on dermatology outcomes. Leveling the playing field begins with (1) an awareness of health literacy and (2) creating readable and comprehensible patient education content. Greater advocacy from community and professional organizations; increased funding from nonprofit organizations, industry, and federal institutions; and increased involvement by dermatologists in bringing greater attention to health literacy will improve outcomes in dermatology.

References
  1. Liu C, Wang D, Liu C, et al. What is the meaning of health literacy? a systematic review and qualitative synthesis. Fam Med Community Health. 2020;8:e000351. doi:10.1136/fmch-2020-000351
  2. Berkman ND, Sheridan SL, Donahue KE, et al. Low health literacy and health outcomes: an updated systematic review. Ann Intern Med. 2011;155:97-107. doi:10.7326/0003-4819-155-2-201107190-00005
  3. Rice RE. Influences, usage, and outcomes of Internet health information searching: multivariate results from the Pew surveys. Int J Med Inform. 2006;75:8-28. doi:10.1016/j.ijmedinf.2005.07.032
  4. Brown J, Weitz NW, Liang A, et al. Does an eczema action plan improve atopic dermatitis? a single-site randomized controlled trial. Clin Pediatr (Phila). 2018;57:1624-1629. doi:10.1177/0009922818795906
  5. De DR, Shih T, Katta R, et al. Readability, quality, and timeliness of patient online health resources for contact dermatitis and patch testing. Dermatitis. 2022;33:155-160. doi:10.1097/DER.0000000000000789
  6. Weiss BD. Health Literacy: A Manual for Clinicians. American Medical Association, American Medical Foundation; 2003.
  7. Nutbeam D, McGill B, Premkumar P. Improving health literacy in community populations: a review of progress. Health Promot Int. 2018;33:901-911. doi:10.1093/heapro/dax015
References
  1. Liu C, Wang D, Liu C, et al. What is the meaning of health literacy? a systematic review and qualitative synthesis. Fam Med Community Health. 2020;8:e000351. doi:10.1136/fmch-2020-000351
  2. Berkman ND, Sheridan SL, Donahue KE, et al. Low health literacy and health outcomes: an updated systematic review. Ann Intern Med. 2011;155:97-107. doi:10.7326/0003-4819-155-2-201107190-00005
  3. Rice RE. Influences, usage, and outcomes of Internet health information searching: multivariate results from the Pew surveys. Int J Med Inform. 2006;75:8-28. doi:10.1016/j.ijmedinf.2005.07.032
  4. Brown J, Weitz NW, Liang A, et al. Does an eczema action plan improve atopic dermatitis? a single-site randomized controlled trial. Clin Pediatr (Phila). 2018;57:1624-1629. doi:10.1177/0009922818795906
  5. De DR, Shih T, Katta R, et al. Readability, quality, and timeliness of patient online health resources for contact dermatitis and patch testing. Dermatitis. 2022;33:155-160. doi:10.1097/DER.0000000000000789
  6. Weiss BD. Health Literacy: A Manual for Clinicians. American Medical Association, American Medical Foundation; 2003.
  7. Nutbeam D, McGill B, Premkumar P. Improving health literacy in community populations: a review of progress. Health Promot Int. 2018;33:901-911. doi:10.1093/heapro/dax015
Issue
Cutis - 110(3)
Issue
Cutis - 110(3)
Page Number
119-120
Page Number
119-120
Publications
Publications
Topics
Article Type
Display Headline
Health Literacy in Dermatology Patients: How to Level the Playing Field
Display Headline
Health Literacy in Dermatology Patients: How to Level the Playing Field
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>Shih Leveling</fileName> <TBEID>0C02A8C6.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02A8C6</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Shih Leveling</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20220906T141951</firstPublished> <LastPublished>20220906T141951</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20220906T141950</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Terri Shih, BS; Devea R. De, BS; Khiem A. Tran, MD</byline> <bylineText>Terri Shih, BS; Devea R. De, BS; Khiem A. Tran, MD; Jennifer L. Hsiao, MD; Vivian Y. Shi, MD</bylineText> <bylineFull>Terri Shih, BS; Devea R. De, BS; Khiem A. Tran, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>119-120</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Health literacy is a multifaceted construct that encompasses the knowledge of health and health systems, utilization of information related to health, and abili</metaDescription> <articlePDF>289197</articlePDF> <teaserImage/> <title>Health Literacy in Dermatology Patients: How to Level the Playing Field</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2022</pubPubdateYear> <pubPubdateMonth>September</pubPubdateMonth> <pubPubdateDay/> <pubVolume>110</pubVolume> <pubNumber>3</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2159</CMSID> </CMSIDs> <keywords> <keyword>diversity in medicine</keyword> <keyword> health literacy</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>September 2022</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">52</term> </sections> <topics> <term canonical="true">66772</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/180021f9.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Health Literacy in Dermatology Patients: How to Level the Playing Field</title> <deck/> </itemMeta> <itemContent> <p>Health literacy is a multifaceted construct that encompasses the knowledge of health and health systems, utilization of information related to health, and ability to maintain health.<sup>1</sup> Low health literacy impairs health outcomes, disproportionately affecting socioeconomically disadvantaged populations, including racial minorities and the older population. Consistently, it is associated with fewer vaccinations and screenings, higher health care utilization, and poorer ability to take medications or interpret health information.<sup>2</sup> </p> <p>With growing utilization of the Internet for health information,<sup>3</sup> much patient education now occurs outside the clinic. Differential utilization of the Internet can exacerbate disparities in health outcomes: people with a lower family income more frequently engage in health information and dialogue online.<sup>3</sup> Despite opportunities to improve literacy and narrow gaps in care, a lack of awareness, advocacy, and funding limit patient- and community-based initiatives. Herein, we discuss health literacy challenges in dermatology, offer potential solutions, and propose ways that stakeholders can prioritize health literacy advocacy to improve outcomes.</p> <h3>The Importance of Health Literacy in Dermatology</h3> <p>Dermatology patients often face challenges that demand greater health literacy. Active participation in health promotion, protection, and maintenance can remarkably improve outcomes. When patients understand disease pathogenesis and the rationale behind treatment choices, adherence to a treatment regimen might improve. </p> <p>However, understanding dermatologic diseases and disorders can be challenging. First, many are chronic inflammatory conditions that require intricate treatment regimens. Second, the complexity of those diseases and disorders continues to grow in the era of new research and unprecedented expansion of treatment options. <br/><br/>For chronic conditions that require ongoing complex management, researchers have developed advanced patient tools. For instance, the eczema action plan helps atopic dermatitis patients manage conditions from home.<sup>4</sup> However, patients with greater literacy and the ability to participate will better utilize such tools and have fewer uncontrolled flares. Patient tools meant to improve outcomes might, instead, widen gaps in care. Even with nonchronic conditions, such as nonmelanoma skin cancer, continued awareness and the need for preventive care, timely diagnosis, and appropriate intervention remain critical.</p> <h3>Limited Accessibility of Patient Education Materials</h3> <p>Patient education in dermatology occurs through several formats. Because online health resources are more readily available to those with less access to health care, the potential for such resources to narrow health disparities is immense. However, online resources have not adequately taken advantage of the opportunity to make health information openly accessible to its users. The readability of online patient education materials on a large expanse of dermatologic conditions is far too advanced.<sup>5</sup> The readability level of some resources is as high as 17th grade (graduate school), which is much higher than the American Medical Association recommendation<sup>6</sup> that patient education materials be presented at a 6th-grade level or less. Furthermore, the quality and comprehensiveness of content is highly variable. Rather than serving as an equalizer, the Internet may widen the gap as low health literacy continues to impair the accessibility of health information.</p> <h3>Solutions to Level the Playing Field</h3> <p>What can be done to increase the readability of patient education materials? Leveling the playing field begins with creating materials at an appropriate readability level, including online content, printed handouts, and after-visit summaries in the clinic. Writers of patient education materials should be cognizant of their choice of language and routinely use a<i> </i>free readability checker (https://readabilityformulas.com). Patient education materials should reflect the American Medical Association’s<i> </i>recommended 6th-grade level. Creators should maintain a high standard of quality and comprehensiveness; prior studies note no inverse correlation between readability and quality.<sup>5</sup> In the age of multimedia presentation, non–print-based materials can be explored, such as audio or video for online content, podcasts, and webinars. Providers also should take the opportunity to be mindful of health literacy in clinic. Beyond assessing the readability of written resources for a patient, assessing that patient’s health literacy and tailoring one’s language will maximize engagement.</p> <h3>Systemic Change Is Needed</h3> <p>Ultimately, systemic change is needed to address the root causes of health literacy disparity, requiring advocacy for social welfare, public health, and public policy initiatives. In recognizing existing efforts, such as community outreach teams and hospital committees to evaluate health literacy materials, numerous barriers remain. Despite the notable impact of health literacy on health outcomes, there is a lack of advocacy and funds to conduct health literacy–related work.<sup>7</sup> Because dermatologists provide holistic care and remain mindful of patients’ health literacy in the clinic, they should continue to advocate for increased awareness, improved funding, and support for local and federal initiatives.</p> <h3>Final Thoughts</h3> <p>With more opportunities to narrow gaps in care, it is more pertinent than ever to acknowledge the impact of health literacy on dermatology outcomes. Leveling the playing field begins with (1) an awareness of health literacy and (2) creating readable and comprehensible patient education content. Greater advocacy from community and professional organizations; increased funding from nonprofit organizations, industry, and federal institutions; and increased involvement by dermatologists in bringing greater attention to health literacy will improve outcomes in dermatology.</p> <h2>References</h2> <p class="reference"> 1. Liu C, Wang D, Liu C, et al. What is the meaning of health literacy? a systematic review and qualitative synthesis. <i>Fam Med Community Health</i>. 2020;8:e000351. doi:10.1136/fmch-2020-000351</p> <p class="reference"> 2. Berkman ND, Sheridan SL, Donahue KE, et al. Low health literacy and health outcomes: an updated systematic review. <i>Ann Intern Med</i>. 2011;155:97-107. doi:10.7326/0003-4819-155-2-201107190-00005<br/><br/> 3. Rice RE. Influences, usage, and outcomes of Internet health information searching: multivariate results from the Pew surveys. <i>Int J Med Inform</i>. 2006;75:8-28. doi:10.1016/j.ijmedinf.2005.07.032<br/><br/> 4. Brown J, Weitz NW, Liang A, et al. Does an eczema action plan improve atopic dermatitis? a single-site randomized controlled trial. <i>Clin Pediatr (Phila).</i> 2018;57:1624-1629. doi:10.1177/0009922818795906<br/><br/> 5. De DR, Shih T, Katta R, et al. Readability, quality, and timeliness of patient online health resources for contact dermatitis and patch testing. <i>Dermatitis</i>. 2022;33:155-160. doi:10.1097/DER.0000000000000789<br/><br/> 6. Weiss BD. <i>Health Literacy: A Manual for Clinicians</i>. American Medical Association, American Medical Foundation; 2003.<br/><br/> 7. Nutbeam D, McGill B, Premkumar P. Improving health literacy in community populations: a review of progress. <i>Health Promot Int</i>. 2018;33:901-911. doi:10.1093/heapro/dax015</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Ms. Shih is from the David Geffen School of Medicine, University of California, Los Angeles. Ms. De is from the Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, New York. Drs. Tran and Shi are from the Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock. Dr. Hsiao is from the Department of Dermatology, University of Southern California, Los Angeles.</p> <p class="disclosure">Ms. Shih, Ms. De, and Dr. Tran report no conflict of interest. Dr. Hsiao is on the board of directors for the HS Foundation; has served as a consultant for Boehringer Ingelheim, Novartis, and UCB; and has served as a consultant and speaker for AbbVie. Dr. Shi is on the board of directors for the HS Foundation and is a shareholder of LearnHealth. She also has served as an advisory board member, investigator, and/or speaker for, and/or received research funding from AbbVie; Altus Labs; Alumis; Aristea Therapeutics; Boehringer Ingelheim; Burt’s Bees; Dermira; Eli Lilly and Company; Galderma; Genzyme, a Sanofi company; gpskin; Incyte; Kiniksa Pharmaceuticals; LEO Pharma; MyOr; Novartis; Pfizer; Polyfins; Regeneron; Skin Actives Scientific; SUN Pharma Industries; TARGET PharmaSolutions; UCB; and VYNE Therapeutics. <br/><br/>Correspondence: Vivian Y. Shi, MD, Department of Dermatology, University of Arkansas for Medical Sciences, 4301 W Markham St, #576, Little Rock, AR 72205-7199 (vivian.shi.publications@gmail.com).<br/><br/>doi:10.12788/cutis.0608</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
Article PDF Media
Image
Teambase ID
180021F9.SIG
Disable zoom
Off

Treatment Stacking: Optimizing Therapeutic Regimens for Hidradenitis Suppurativa

Article Type
Changed
Mon, 11/15/2021 - 09:56
Display Headline
Treatment Stacking: Optimizing Therapeutic Regimens for Hidradenitis Suppurativa

Hidradenitis suppurativa (HS) is a debilitating chronic condition that often is recalcitrant to first-line treatments, and mechanisms underlying its pathology remain unclear. Existing data suggest a multifactorial etiology with different pathophysiologic contributors, including genetic, hormonal, and immune dysregulation factors. At this time, only one medication (adalimumab) is US Food and Drug Administration approved for HS, but multiple medical and procedural therapies are available.1 Herein, we discuss the concept of treatment stacking, or the combination of unique therapeutic modalities—an approach we believe is key to optimizing management of HS patients.

Stacking Treatments for HS

Unlike psoriasis, in which a single biologic agent may provide 100% clearance (psoriasis area and severity index 100 [PASI 100]) without adjuvant treatment,2,3 the field of HS currently lacks medications that are efficacious to that degree of success as monotherapy. In HS, the benchmark for a positive treatment outcome is Hidradenitis Suppurativa Clinical Response 50 (HiSCR50),4 a 50% reduction in inflammatory lesion count—a far less stringent marker for disease improvement. Thus, providers should design HS treatment regimens with a model of combining therapies and shift away from monotherapy. Targeting different pathophysiologic pathways by stacking multiple treatments may provide synergistic benefits for HS patients. Treatment stacking is a familiar concept in acne; for instance, patients who benefit tremendously from isotretinoin may still require a hormone-modulating treatment (eg, spironolactone) to attain optimal results.

Adherence to a rigid treatment algorithm based on disease severity limits the potential to create comprehensive regimens that account for unique patient characteristics and clinical manifestations. When evaluating an HS patient, providers should systematically consider each pathophysiologic factor and target the ones that appear to be most involved in that particular patient. The North American HS guidelines illustrate this point by supporting use of several treatments across different Hurley stages, such as recommending hormonal treatment in patients with Hurley stages 1, 2, or 3.1 Of note, treatment stacking also includes procedural therapies. Surgeons typically prefer a patient’s disease management to be optimized prior to surgery, including reduced drainage and inflammation. In addition, even after surgery, patients often still require medical management to prevent continued disease worsening.

Treatment Pathways for HS

A multimodal approach with treatment stacking (Figure) can be useful to all HS patients, from those with the mildest to the most severe disease. Modifiable pathophysiologic factors and examples of their targeted treatments include (1) follicular occlusion (eg, oral retinoids), (2) metabolic dysfunction (eg, metformin), (3) hormones (eg, oral contraceptive pills, spironolactone, finasteride), (4) dysbiosis (eg, antibiotics such as clindamycin and rifampin combination therapy), (5) immune dysregulation (eg, biologic agents), and (6) friction/irritation (eg, weight loss, clothing recommendations).

CT108004176_figure.JPG
%3Cp%3ETargeted%20treatments%20for%20modifiable%20pathophysiologic%20arms%20of%20hidradenitis%20suppurativa%20(HS).%20Surgical%20and%20laser%20excisions%20(not%20shown)%20remove%20persistent%20inflamed%20and%20diseased%20tissue.%20Asterisk%20indicates%20mixed%20data%20in%20literature%3B%20should%20be%20considered%20in%20patients%20with%20severe%20acne.%20Dagger%20indicates%20exclusive%20usage%20in%20female%20HS%20patients.%20Double%20dagger%20indicates%20biologics%20including%20anti%E2%80%93tumor%20necrosis%20factor%20%CE%B1%2C%20IL-1%2C%20IL-17%2C%20IL-12%2F23%2C%20and%20IL-23.%3C%2Fp%3E

Combining treatments from different pathways enables potentiation of individual treatment efficacies. A female patient with only a few HS nodules that flare with menses may be well controlled with spironolactone as her only systemic agent; however, she still may benefit from use of an antiseptic wash, topical clindamycin, and lifestyle changes such as weight loss and reduction of mechanical irritation. A patient with severe recalcitrant HS could notably benefit from concomitant biologic, systemic antibiotic, and hormonal/metabolic treatments. If disease control is still inadequate, agents within the same class can be switched (eg, choosing a different biologic) or other disease-modifying agents such as colchicine also can be added. The goal is to create an effective treatment toolbox with therapies targeting different pathophysiologic arms of HS and working together in synergy. Each tool can be refined by modifying dosing frequency and duration of use to strive for optimal response. At this time, the literature on HS combination therapy is sparse. A retrospective study of 31 patients reported promising combinations, including isotretinoin with spironolactone for mild disease, isotretinoin or doxycycline with adalimumab for moderate disease, and cyclosporine with adalimumab for severe disease.5 Larger prospective studies on clinical response to different combination regimens are warranted.

Optimizing Therapy for HS and Its Comorbidities

Additional considerations may further optimize treatment plans. Some therapies benefit all patients; for example, providers should counsel all HS patients on healthy weight management, optimized clothing choices,6 and friction reduction in the intertriginous folds. Providers also may consider adding therapies with faster onset of efficacy as a bridge to long-term, slower-onset therapies. For instance, female HS patients with menstrual flares who are prescribed spironolactone also may benefit from a course of systemic antibiotics, which typically provides more prompt relief. Treatment regimens also can concomitantly treat HS and its comorbidities.7 For example, metformin serves a dual purpose in HS patients with diabetes mellitus, and adalimumab in patients with both HS and inflammatory bowel disease.

Final Thoughts

The last decade has seen tremendous growth in HS research8 coupled with a remarkable expansion in the therapeutic pipeline.9 However, currently no single therapy for HS can guarantee satisfactory disease remission or durability of remission. The contrast between clinical trials and real-world practice should be acknowledged; the former often is restrictive in design with monotherapy and allowance of very limited concomitant treatments, such as topical or oral antibiotics. This limits our ability to draw conclusions regarding the additive synergistic potential of different therapeutics in combination. In clinical practice, we are not restricted by monotherapy trial protocols. As we await new tools, treatment stacking allows for creating a framework to best utilize the tools that are available to us.

Although HS has continued to affect the lives of many patients, improved understanding of underlying pathophysiology and a well-placed sense of urgency from all stakeholders (ie, patients, clinicians, researchers, industry partners) has pushed this field forward. Until our therapeutic armamentarium has expanded to include highly efficacious monotherapy options, providers should consider treatment stacking for every HS patient.

References
  1. Alikhan A, Sayed C, Alavi A, et al. North American clinical management guidelines for hidradenitis suppurativa: a publication from the United States and Canadian Hidradenitis Suppurativa Foundations: part II: topical, intralesional, and systemic medical management. J Am Acad Dermatol. 2019;81:91-101. doi:10.1016/j.jaad.2019.02.068
  2. Reich K, Warren RB, Lebwohl M, et al. Bimekizumab versus secukinumab in plaque psoriasis. N Engl J Med. 2021;385:142-152. doi:10.1056/NEJMoa2102383
  3. Imafuku S, Nakagawa H, Igarashi A, et al. Long-term efficacy and safety of tildrakizumab in Japanese patients with moderate to severe plaque psoriasis: results from a 5-year extension of a phase 3 study (reSURFACE 1). J Dermatol. 2021;48:844-852. doi:10.1111/1346-8138.15763
  4. Kimball AB, Okun MM, Williams DA, et al. Two phase 3 trials of adalimumab for hidradenitis suppurativa. N Engl J Med. 2016;375:422-434. doi:10.1056/NEJMoa1504370
  5. McPhie ML, Bridgman AC, Kirchhof MG. Combination therapies for hidradenitis suppurativa: a retrospective chart review of 31 patients. J Cutan Med Surg. 2019;23:270-276. doi:10.1177/1203475418823529
  6. Loh TY, Hendricks AJ, Hsiao JL, et al. Undergarment and fabric selection in the management of hidradenitis suppurativa. Dermatol Basel Switz. 2021;237:119-124. doi:10.1159/000501611
  7. Garg A, Malviya N, Strunk A, et al. Comorbidity screening in hidradenitis suppurativa: evidence-based recommendations from the US and Canadian Hidradenitis Suppurativa Foundations [published online January 23, 2021]. J Am Acad Dermatol. doi:10.1016/j.jaad.2021.01.059
  8. Savage KT, Brant EG, Flood KS, et al. Publication trends in hidradenitis suppurativa from 2008 to 2018. J Eur Acad Dermatol Venereol. 2020;34:1885-1889. doi:10.1111/jdv.16213
  9. van Straalen KR, Schneider-Burrus S, Prens EP. Current and future treatment of hidradenitis suppurativa. Br J Dermatol. 2020;183:E178-E187. doi:10.1111/bjd.16768
Article PDF
Author and Disclosure Information

Ms. Shih and Dr. Hsiao are from the University of California, Los Angeles. Ms. Shih is from the David Geffen School of Medicine, and Dr. Hsiao is from the Division of Dermatology. Dr. Shi is from the Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock.

Ms. Shih reports no conflict of interest. Dr. Shi is on the Board of Directors for the Hidradenitis Suppurativa Foundation and is a stock shareholder for Learn Health. Dr. Shi also has served as an advisory board member, investigator, or speaker and/or has received research funding from AbbVie; Boehringer Ingelheim; Burt’s Bees, Inc; CQuell/Altus Lab; Dermira, Inc; Eli Lilly and Company; Galderma; Gpskin; Incyte Corporation; Kiniksa Pharmaceuticals; LEO Pharma; Menlo Therapeutics; MyOR; Novartis; Pfizer; Polyfins Technology; Regeneron Pharmaceuticals; Sanofi Genzyme; Skin Actives Scientific; Sun Pharmaceutical Industries Ltd; TARGET PHARMASOLUTIONS; and UCB. Dr. Hsiao is on the Board of Directors for the Hidradenitis Suppurativa Foundation, a speaker for AbbVie, and consultant for Novartis.

Correspondence: Jennifer L. Hsiao, MD, Division of Dermatology, UCLA, 2020 Santa Monica Blvd, Ste 510, Santa Monica, CA 9040 (j.hsiao.publications@gmail.com).

Issue
Cutis - 108(4)
Publications
Topics
Page Number
176-178
Sections
Author and Disclosure Information

Ms. Shih and Dr. Hsiao are from the University of California, Los Angeles. Ms. Shih is from the David Geffen School of Medicine, and Dr. Hsiao is from the Division of Dermatology. Dr. Shi is from the Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock.

Ms. Shih reports no conflict of interest. Dr. Shi is on the Board of Directors for the Hidradenitis Suppurativa Foundation and is a stock shareholder for Learn Health. Dr. Shi also has served as an advisory board member, investigator, or speaker and/or has received research funding from AbbVie; Boehringer Ingelheim; Burt’s Bees, Inc; CQuell/Altus Lab; Dermira, Inc; Eli Lilly and Company; Galderma; Gpskin; Incyte Corporation; Kiniksa Pharmaceuticals; LEO Pharma; Menlo Therapeutics; MyOR; Novartis; Pfizer; Polyfins Technology; Regeneron Pharmaceuticals; Sanofi Genzyme; Skin Actives Scientific; Sun Pharmaceutical Industries Ltd; TARGET PHARMASOLUTIONS; and UCB. Dr. Hsiao is on the Board of Directors for the Hidradenitis Suppurativa Foundation, a speaker for AbbVie, and consultant for Novartis.

Correspondence: Jennifer L. Hsiao, MD, Division of Dermatology, UCLA, 2020 Santa Monica Blvd, Ste 510, Santa Monica, CA 9040 (j.hsiao.publications@gmail.com).

Author and Disclosure Information

Ms. Shih and Dr. Hsiao are from the University of California, Los Angeles. Ms. Shih is from the David Geffen School of Medicine, and Dr. Hsiao is from the Division of Dermatology. Dr. Shi is from the Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock.

Ms. Shih reports no conflict of interest. Dr. Shi is on the Board of Directors for the Hidradenitis Suppurativa Foundation and is a stock shareholder for Learn Health. Dr. Shi also has served as an advisory board member, investigator, or speaker and/or has received research funding from AbbVie; Boehringer Ingelheim; Burt’s Bees, Inc; CQuell/Altus Lab; Dermira, Inc; Eli Lilly and Company; Galderma; Gpskin; Incyte Corporation; Kiniksa Pharmaceuticals; LEO Pharma; Menlo Therapeutics; MyOR; Novartis; Pfizer; Polyfins Technology; Regeneron Pharmaceuticals; Sanofi Genzyme; Skin Actives Scientific; Sun Pharmaceutical Industries Ltd; TARGET PHARMASOLUTIONS; and UCB. Dr. Hsiao is on the Board of Directors for the Hidradenitis Suppurativa Foundation, a speaker for AbbVie, and consultant for Novartis.

Correspondence: Jennifer L. Hsiao, MD, Division of Dermatology, UCLA, 2020 Santa Monica Blvd, Ste 510, Santa Monica, CA 9040 (j.hsiao.publications@gmail.com).

Article PDF
Article PDF

Hidradenitis suppurativa (HS) is a debilitating chronic condition that often is recalcitrant to first-line treatments, and mechanisms underlying its pathology remain unclear. Existing data suggest a multifactorial etiology with different pathophysiologic contributors, including genetic, hormonal, and immune dysregulation factors. At this time, only one medication (adalimumab) is US Food and Drug Administration approved for HS, but multiple medical and procedural therapies are available.1 Herein, we discuss the concept of treatment stacking, or the combination of unique therapeutic modalities—an approach we believe is key to optimizing management of HS patients.

Stacking Treatments for HS

Unlike psoriasis, in which a single biologic agent may provide 100% clearance (psoriasis area and severity index 100 [PASI 100]) without adjuvant treatment,2,3 the field of HS currently lacks medications that are efficacious to that degree of success as monotherapy. In HS, the benchmark for a positive treatment outcome is Hidradenitis Suppurativa Clinical Response 50 (HiSCR50),4 a 50% reduction in inflammatory lesion count—a far less stringent marker for disease improvement. Thus, providers should design HS treatment regimens with a model of combining therapies and shift away from monotherapy. Targeting different pathophysiologic pathways by stacking multiple treatments may provide synergistic benefits for HS patients. Treatment stacking is a familiar concept in acne; for instance, patients who benefit tremendously from isotretinoin may still require a hormone-modulating treatment (eg, spironolactone) to attain optimal results.

Adherence to a rigid treatment algorithm based on disease severity limits the potential to create comprehensive regimens that account for unique patient characteristics and clinical manifestations. When evaluating an HS patient, providers should systematically consider each pathophysiologic factor and target the ones that appear to be most involved in that particular patient. The North American HS guidelines illustrate this point by supporting use of several treatments across different Hurley stages, such as recommending hormonal treatment in patients with Hurley stages 1, 2, or 3.1 Of note, treatment stacking also includes procedural therapies. Surgeons typically prefer a patient’s disease management to be optimized prior to surgery, including reduced drainage and inflammation. In addition, even after surgery, patients often still require medical management to prevent continued disease worsening.

Treatment Pathways for HS

A multimodal approach with treatment stacking (Figure) can be useful to all HS patients, from those with the mildest to the most severe disease. Modifiable pathophysiologic factors and examples of their targeted treatments include (1) follicular occlusion (eg, oral retinoids), (2) metabolic dysfunction (eg, metformin), (3) hormones (eg, oral contraceptive pills, spironolactone, finasteride), (4) dysbiosis (eg, antibiotics such as clindamycin and rifampin combination therapy), (5) immune dysregulation (eg, biologic agents), and (6) friction/irritation (eg, weight loss, clothing recommendations).

CT108004176_figure.JPG
%3Cp%3ETargeted%20treatments%20for%20modifiable%20pathophysiologic%20arms%20of%20hidradenitis%20suppurativa%20(HS).%20Surgical%20and%20laser%20excisions%20(not%20shown)%20remove%20persistent%20inflamed%20and%20diseased%20tissue.%20Asterisk%20indicates%20mixed%20data%20in%20literature%3B%20should%20be%20considered%20in%20patients%20with%20severe%20acne.%20Dagger%20indicates%20exclusive%20usage%20in%20female%20HS%20patients.%20Double%20dagger%20indicates%20biologics%20including%20anti%E2%80%93tumor%20necrosis%20factor%20%CE%B1%2C%20IL-1%2C%20IL-17%2C%20IL-12%2F23%2C%20and%20IL-23.%3C%2Fp%3E

Combining treatments from different pathways enables potentiation of individual treatment efficacies. A female patient with only a few HS nodules that flare with menses may be well controlled with spironolactone as her only systemic agent; however, she still may benefit from use of an antiseptic wash, topical clindamycin, and lifestyle changes such as weight loss and reduction of mechanical irritation. A patient with severe recalcitrant HS could notably benefit from concomitant biologic, systemic antibiotic, and hormonal/metabolic treatments. If disease control is still inadequate, agents within the same class can be switched (eg, choosing a different biologic) or other disease-modifying agents such as colchicine also can be added. The goal is to create an effective treatment toolbox with therapies targeting different pathophysiologic arms of HS and working together in synergy. Each tool can be refined by modifying dosing frequency and duration of use to strive for optimal response. At this time, the literature on HS combination therapy is sparse. A retrospective study of 31 patients reported promising combinations, including isotretinoin with spironolactone for mild disease, isotretinoin or doxycycline with adalimumab for moderate disease, and cyclosporine with adalimumab for severe disease.5 Larger prospective studies on clinical response to different combination regimens are warranted.

Optimizing Therapy for HS and Its Comorbidities

Additional considerations may further optimize treatment plans. Some therapies benefit all patients; for example, providers should counsel all HS patients on healthy weight management, optimized clothing choices,6 and friction reduction in the intertriginous folds. Providers also may consider adding therapies with faster onset of efficacy as a bridge to long-term, slower-onset therapies. For instance, female HS patients with menstrual flares who are prescribed spironolactone also may benefit from a course of systemic antibiotics, which typically provides more prompt relief. Treatment regimens also can concomitantly treat HS and its comorbidities.7 For example, metformin serves a dual purpose in HS patients with diabetes mellitus, and adalimumab in patients with both HS and inflammatory bowel disease.

Final Thoughts

The last decade has seen tremendous growth in HS research8 coupled with a remarkable expansion in the therapeutic pipeline.9 However, currently no single therapy for HS can guarantee satisfactory disease remission or durability of remission. The contrast between clinical trials and real-world practice should be acknowledged; the former often is restrictive in design with monotherapy and allowance of very limited concomitant treatments, such as topical or oral antibiotics. This limits our ability to draw conclusions regarding the additive synergistic potential of different therapeutics in combination. In clinical practice, we are not restricted by monotherapy trial protocols. As we await new tools, treatment stacking allows for creating a framework to best utilize the tools that are available to us.

Although HS has continued to affect the lives of many patients, improved understanding of underlying pathophysiology and a well-placed sense of urgency from all stakeholders (ie, patients, clinicians, researchers, industry partners) has pushed this field forward. Until our therapeutic armamentarium has expanded to include highly efficacious monotherapy options, providers should consider treatment stacking for every HS patient.

Hidradenitis suppurativa (HS) is a debilitating chronic condition that often is recalcitrant to first-line treatments, and mechanisms underlying its pathology remain unclear. Existing data suggest a multifactorial etiology with different pathophysiologic contributors, including genetic, hormonal, and immune dysregulation factors. At this time, only one medication (adalimumab) is US Food and Drug Administration approved for HS, but multiple medical and procedural therapies are available.1 Herein, we discuss the concept of treatment stacking, or the combination of unique therapeutic modalities—an approach we believe is key to optimizing management of HS patients.

Stacking Treatments for HS

Unlike psoriasis, in which a single biologic agent may provide 100% clearance (psoriasis area and severity index 100 [PASI 100]) without adjuvant treatment,2,3 the field of HS currently lacks medications that are efficacious to that degree of success as monotherapy. In HS, the benchmark for a positive treatment outcome is Hidradenitis Suppurativa Clinical Response 50 (HiSCR50),4 a 50% reduction in inflammatory lesion count—a far less stringent marker for disease improvement. Thus, providers should design HS treatment regimens with a model of combining therapies and shift away from monotherapy. Targeting different pathophysiologic pathways by stacking multiple treatments may provide synergistic benefits for HS patients. Treatment stacking is a familiar concept in acne; for instance, patients who benefit tremendously from isotretinoin may still require a hormone-modulating treatment (eg, spironolactone) to attain optimal results.

Adherence to a rigid treatment algorithm based on disease severity limits the potential to create comprehensive regimens that account for unique patient characteristics and clinical manifestations. When evaluating an HS patient, providers should systematically consider each pathophysiologic factor and target the ones that appear to be most involved in that particular patient. The North American HS guidelines illustrate this point by supporting use of several treatments across different Hurley stages, such as recommending hormonal treatment in patients with Hurley stages 1, 2, or 3.1 Of note, treatment stacking also includes procedural therapies. Surgeons typically prefer a patient’s disease management to be optimized prior to surgery, including reduced drainage and inflammation. In addition, even after surgery, patients often still require medical management to prevent continued disease worsening.

Treatment Pathways for HS

A multimodal approach with treatment stacking (Figure) can be useful to all HS patients, from those with the mildest to the most severe disease. Modifiable pathophysiologic factors and examples of their targeted treatments include (1) follicular occlusion (eg, oral retinoids), (2) metabolic dysfunction (eg, metformin), (3) hormones (eg, oral contraceptive pills, spironolactone, finasteride), (4) dysbiosis (eg, antibiotics such as clindamycin and rifampin combination therapy), (5) immune dysregulation (eg, biologic agents), and (6) friction/irritation (eg, weight loss, clothing recommendations).

CT108004176_figure.JPG
%3Cp%3ETargeted%20treatments%20for%20modifiable%20pathophysiologic%20arms%20of%20hidradenitis%20suppurativa%20(HS).%20Surgical%20and%20laser%20excisions%20(not%20shown)%20remove%20persistent%20inflamed%20and%20diseased%20tissue.%20Asterisk%20indicates%20mixed%20data%20in%20literature%3B%20should%20be%20considered%20in%20patients%20with%20severe%20acne.%20Dagger%20indicates%20exclusive%20usage%20in%20female%20HS%20patients.%20Double%20dagger%20indicates%20biologics%20including%20anti%E2%80%93tumor%20necrosis%20factor%20%CE%B1%2C%20IL-1%2C%20IL-17%2C%20IL-12%2F23%2C%20and%20IL-23.%3C%2Fp%3E

Combining treatments from different pathways enables potentiation of individual treatment efficacies. A female patient with only a few HS nodules that flare with menses may be well controlled with spironolactone as her only systemic agent; however, she still may benefit from use of an antiseptic wash, topical clindamycin, and lifestyle changes such as weight loss and reduction of mechanical irritation. A patient with severe recalcitrant HS could notably benefit from concomitant biologic, systemic antibiotic, and hormonal/metabolic treatments. If disease control is still inadequate, agents within the same class can be switched (eg, choosing a different biologic) or other disease-modifying agents such as colchicine also can be added. The goal is to create an effective treatment toolbox with therapies targeting different pathophysiologic arms of HS and working together in synergy. Each tool can be refined by modifying dosing frequency and duration of use to strive for optimal response. At this time, the literature on HS combination therapy is sparse. A retrospective study of 31 patients reported promising combinations, including isotretinoin with spironolactone for mild disease, isotretinoin or doxycycline with adalimumab for moderate disease, and cyclosporine with adalimumab for severe disease.5 Larger prospective studies on clinical response to different combination regimens are warranted.

Optimizing Therapy for HS and Its Comorbidities

Additional considerations may further optimize treatment plans. Some therapies benefit all patients; for example, providers should counsel all HS patients on healthy weight management, optimized clothing choices,6 and friction reduction in the intertriginous folds. Providers also may consider adding therapies with faster onset of efficacy as a bridge to long-term, slower-onset therapies. For instance, female HS patients with menstrual flares who are prescribed spironolactone also may benefit from a course of systemic antibiotics, which typically provides more prompt relief. Treatment regimens also can concomitantly treat HS and its comorbidities.7 For example, metformin serves a dual purpose in HS patients with diabetes mellitus, and adalimumab in patients with both HS and inflammatory bowel disease.

Final Thoughts

The last decade has seen tremendous growth in HS research8 coupled with a remarkable expansion in the therapeutic pipeline.9 However, currently no single therapy for HS can guarantee satisfactory disease remission or durability of remission. The contrast between clinical trials and real-world practice should be acknowledged; the former often is restrictive in design with monotherapy and allowance of very limited concomitant treatments, such as topical or oral antibiotics. This limits our ability to draw conclusions regarding the additive synergistic potential of different therapeutics in combination. In clinical practice, we are not restricted by monotherapy trial protocols. As we await new tools, treatment stacking allows for creating a framework to best utilize the tools that are available to us.

Although HS has continued to affect the lives of many patients, improved understanding of underlying pathophysiology and a well-placed sense of urgency from all stakeholders (ie, patients, clinicians, researchers, industry partners) has pushed this field forward. Until our therapeutic armamentarium has expanded to include highly efficacious monotherapy options, providers should consider treatment stacking for every HS patient.

References
  1. Alikhan A, Sayed C, Alavi A, et al. North American clinical management guidelines for hidradenitis suppurativa: a publication from the United States and Canadian Hidradenitis Suppurativa Foundations: part II: topical, intralesional, and systemic medical management. J Am Acad Dermatol. 2019;81:91-101. doi:10.1016/j.jaad.2019.02.068
  2. Reich K, Warren RB, Lebwohl M, et al. Bimekizumab versus secukinumab in plaque psoriasis. N Engl J Med. 2021;385:142-152. doi:10.1056/NEJMoa2102383
  3. Imafuku S, Nakagawa H, Igarashi A, et al. Long-term efficacy and safety of tildrakizumab in Japanese patients with moderate to severe plaque psoriasis: results from a 5-year extension of a phase 3 study (reSURFACE 1). J Dermatol. 2021;48:844-852. doi:10.1111/1346-8138.15763
  4. Kimball AB, Okun MM, Williams DA, et al. Two phase 3 trials of adalimumab for hidradenitis suppurativa. N Engl J Med. 2016;375:422-434. doi:10.1056/NEJMoa1504370
  5. McPhie ML, Bridgman AC, Kirchhof MG. Combination therapies for hidradenitis suppurativa: a retrospective chart review of 31 patients. J Cutan Med Surg. 2019;23:270-276. doi:10.1177/1203475418823529
  6. Loh TY, Hendricks AJ, Hsiao JL, et al. Undergarment and fabric selection in the management of hidradenitis suppurativa. Dermatol Basel Switz. 2021;237:119-124. doi:10.1159/000501611
  7. Garg A, Malviya N, Strunk A, et al. Comorbidity screening in hidradenitis suppurativa: evidence-based recommendations from the US and Canadian Hidradenitis Suppurativa Foundations [published online January 23, 2021]. J Am Acad Dermatol. doi:10.1016/j.jaad.2021.01.059
  8. Savage KT, Brant EG, Flood KS, et al. Publication trends in hidradenitis suppurativa from 2008 to 2018. J Eur Acad Dermatol Venereol. 2020;34:1885-1889. doi:10.1111/jdv.16213
  9. van Straalen KR, Schneider-Burrus S, Prens EP. Current and future treatment of hidradenitis suppurativa. Br J Dermatol. 2020;183:E178-E187. doi:10.1111/bjd.16768
References
  1. Alikhan A, Sayed C, Alavi A, et al. North American clinical management guidelines for hidradenitis suppurativa: a publication from the United States and Canadian Hidradenitis Suppurativa Foundations: part II: topical, intralesional, and systemic medical management. J Am Acad Dermatol. 2019;81:91-101. doi:10.1016/j.jaad.2019.02.068
  2. Reich K, Warren RB, Lebwohl M, et al. Bimekizumab versus secukinumab in plaque psoriasis. N Engl J Med. 2021;385:142-152. doi:10.1056/NEJMoa2102383
  3. Imafuku S, Nakagawa H, Igarashi A, et al. Long-term efficacy and safety of tildrakizumab in Japanese patients with moderate to severe plaque psoriasis: results from a 5-year extension of a phase 3 study (reSURFACE 1). J Dermatol. 2021;48:844-852. doi:10.1111/1346-8138.15763
  4. Kimball AB, Okun MM, Williams DA, et al. Two phase 3 trials of adalimumab for hidradenitis suppurativa. N Engl J Med. 2016;375:422-434. doi:10.1056/NEJMoa1504370
  5. McPhie ML, Bridgman AC, Kirchhof MG. Combination therapies for hidradenitis suppurativa: a retrospective chart review of 31 patients. J Cutan Med Surg. 2019;23:270-276. doi:10.1177/1203475418823529
  6. Loh TY, Hendricks AJ, Hsiao JL, et al. Undergarment and fabric selection in the management of hidradenitis suppurativa. Dermatol Basel Switz. 2021;237:119-124. doi:10.1159/000501611
  7. Garg A, Malviya N, Strunk A, et al. Comorbidity screening in hidradenitis suppurativa: evidence-based recommendations from the US and Canadian Hidradenitis Suppurativa Foundations [published online January 23, 2021]. J Am Acad Dermatol. doi:10.1016/j.jaad.2021.01.059
  8. Savage KT, Brant EG, Flood KS, et al. Publication trends in hidradenitis suppurativa from 2008 to 2018. J Eur Acad Dermatol Venereol. 2020;34:1885-1889. doi:10.1111/jdv.16213
  9. van Straalen KR, Schneider-Burrus S, Prens EP. Current and future treatment of hidradenitis suppurativa. Br J Dermatol. 2020;183:E178-E187. doi:10.1111/bjd.16768
Issue
Cutis - 108(4)
Issue
Cutis - 108(4)
Page Number
176-178
Page Number
176-178
Publications
Publications
Topics
Article Type
Display Headline
Treatment Stacking: Optimizing Therapeutic Regimens for Hidradenitis Suppurativa
Display Headline
Treatment Stacking: Optimizing Therapeutic Regimens for Hidradenitis Suppurativa
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Asymptomatic Transient Lingual Hyperpigmentation

Article Type
Changed
Mon, 12/07/2020 - 15:18
Display Headline
Asymptomatic Transient Lingual Hyperpigmentation

The Diagnosis: Pseudo-Black Hairy Tongue

Pseudo-black hairy tongue is a benign and painless disorder characterized by transient hyperpigmentation of the tongue with a substance that can be easily scraped off. In this case, the patient's lingual discoloration was secondary to the ingestion of bismuth salicylate. The phenomenon is thought to occur due to a reaction between bismuth and sulfur-containing compounds in the saliva, resulting in the characteristic black substance on the surface of the tongue that nestles between the lingual papillae.1 An associated feature may include black stools. Other etiologic factors involved in pseudo-black hairy tongue include food coloring, tobacco, and other drugs such antibiotics and antidepressants.2  

The differential diagnosis of lingual hyperpigmentation includes lingua villosa nigra (also known as black hairy tongue), pigmented fungiform papillae of the tongue, acanthosis nigricans, and oral hairy leukoplakia. Lingua villosa nigra is a similar condition in which individuals present with a black tongue; however, the tongue also appears hairy. The tongue may appear as other colors such as brown, yellow, or green. Patients additionally may have symptoms of burning, dysgeusia, halitosis, or gagging. Poor oral hygiene, xerostomia, use of tobacco or alcohol, and different medications including antibiotics and antipsychotic medications increase the risk for developing lingua villosa nigra.2,3 This condition is distinguished from pseudo-black hairy tongue by proliferation and elongation of the filiform papillae.3 Pigmented fungiform papillae of the tongue is a normal variant of tongue morphology, is more common in individuals with darker skin types, and primarily affects the lateral aspect and apex of the tongue.4 Acanthosis nigricans can appear in the oral cavity as multiple pigmented papillary lesions on the dorsal and lateral regions of the tongue and frequently involves the lips; this condition may be associated with metabolic disorders or underlying malignancy.2,3 Oral hairy leukoplakia is caused by Epstein-Barr virus infection and typically presents as white plaques on the dorsal and ventral surfaces of the tongue; this condition largely is found in immunocompromised patients.5

In our patient there was an acute onset of tongue discoloration associated with ingestion of bismuth salicylate, no hypertrophy or lengthening of the lingual papillae, and no involvement of the patient's lips, which was consistent with the diagnosis of pseudo-black hairy tongue. Pseudo-black hairy tongue is transient and treated by discontinuation of offending agents and proper hygiene practices.

References
  1. Bradley B, Singleton M, Lin Wan Po A. Bismuth toxicity--a reassessment. J Clin Pharm Ther. 1989;14:423-441.
  2. Gurvits GE, Tan A. Black hairy tongue syndrome. World J Gastroenterol. 2014;20:10845-10850.
  3. Schlager E, St Claire C, Ashack K, et al. Black hairy tongue: predisposing factors, diagnosis, and treatment. Am J Clin Dermatol. 2017;18:563-569.  
  4. Mangold AR, Torgerson RR, Rogers RS. Diseases of the tongue. Clin Dermatol. 2016;34:458-469.
  5. Husak R, Garbe C, Orfanos CE. Oral hairy leukoplakia in 71 HIV-seropositive patients: clinical symptoms, relation to immunologic status, and prognostic significance. J Am Acad Dermatol. 1996;35:928-934.
Article PDF
Author and Disclosure Information

Ms. Patel and Dr. Hsiao are from the Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles. Dr. Hsiao also is from the Division of Dermatology. Dr. Harview is from Harbor-UCLA Medical Center Graduate Medical Education, Torrance, California.

The authors report no conflict of interest.  

Correspondence: Jennifer L. Hsiao, MD, 2020 Santa Monica Blvd, Ste 510, Santa Monica, CA 90404 (Jhsiao@mednet.ucla.edu).

Issue
Cutis - 105(6)
Publications
Topics
Page Number
288, 293
Sections
Author and Disclosure Information

Ms. Patel and Dr. Hsiao are from the Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles. Dr. Hsiao also is from the Division of Dermatology. Dr. Harview is from Harbor-UCLA Medical Center Graduate Medical Education, Torrance, California.

The authors report no conflict of interest.  

Correspondence: Jennifer L. Hsiao, MD, 2020 Santa Monica Blvd, Ste 510, Santa Monica, CA 90404 (Jhsiao@mednet.ucla.edu).

Author and Disclosure Information

Ms. Patel and Dr. Hsiao are from the Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles. Dr. Hsiao also is from the Division of Dermatology. Dr. Harview is from Harbor-UCLA Medical Center Graduate Medical Education, Torrance, California.

The authors report no conflict of interest.  

Correspondence: Jennifer L. Hsiao, MD, 2020 Santa Monica Blvd, Ste 510, Santa Monica, CA 90404 (Jhsiao@mednet.ucla.edu).

Article PDF
Article PDF
Related Articles

The Diagnosis: Pseudo-Black Hairy Tongue

Pseudo-black hairy tongue is a benign and painless disorder characterized by transient hyperpigmentation of the tongue with a substance that can be easily scraped off. In this case, the patient's lingual discoloration was secondary to the ingestion of bismuth salicylate. The phenomenon is thought to occur due to a reaction between bismuth and sulfur-containing compounds in the saliva, resulting in the characteristic black substance on the surface of the tongue that nestles between the lingual papillae.1 An associated feature may include black stools. Other etiologic factors involved in pseudo-black hairy tongue include food coloring, tobacco, and other drugs such antibiotics and antidepressants.2  

The differential diagnosis of lingual hyperpigmentation includes lingua villosa nigra (also known as black hairy tongue), pigmented fungiform papillae of the tongue, acanthosis nigricans, and oral hairy leukoplakia. Lingua villosa nigra is a similar condition in which individuals present with a black tongue; however, the tongue also appears hairy. The tongue may appear as other colors such as brown, yellow, or green. Patients additionally may have symptoms of burning, dysgeusia, halitosis, or gagging. Poor oral hygiene, xerostomia, use of tobacco or alcohol, and different medications including antibiotics and antipsychotic medications increase the risk for developing lingua villosa nigra.2,3 This condition is distinguished from pseudo-black hairy tongue by proliferation and elongation of the filiform papillae.3 Pigmented fungiform papillae of the tongue is a normal variant of tongue morphology, is more common in individuals with darker skin types, and primarily affects the lateral aspect and apex of the tongue.4 Acanthosis nigricans can appear in the oral cavity as multiple pigmented papillary lesions on the dorsal and lateral regions of the tongue and frequently involves the lips; this condition may be associated with metabolic disorders or underlying malignancy.2,3 Oral hairy leukoplakia is caused by Epstein-Barr virus infection and typically presents as white plaques on the dorsal and ventral surfaces of the tongue; this condition largely is found in immunocompromised patients.5

In our patient there was an acute onset of tongue discoloration associated with ingestion of bismuth salicylate, no hypertrophy or lengthening of the lingual papillae, and no involvement of the patient's lips, which was consistent with the diagnosis of pseudo-black hairy tongue. Pseudo-black hairy tongue is transient and treated by discontinuation of offending agents and proper hygiene practices.

The Diagnosis: Pseudo-Black Hairy Tongue

Pseudo-black hairy tongue is a benign and painless disorder characterized by transient hyperpigmentation of the tongue with a substance that can be easily scraped off. In this case, the patient's lingual discoloration was secondary to the ingestion of bismuth salicylate. The phenomenon is thought to occur due to a reaction between bismuth and sulfur-containing compounds in the saliva, resulting in the characteristic black substance on the surface of the tongue that nestles between the lingual papillae.1 An associated feature may include black stools. Other etiologic factors involved in pseudo-black hairy tongue include food coloring, tobacco, and other drugs such antibiotics and antidepressants.2  

The differential diagnosis of lingual hyperpigmentation includes lingua villosa nigra (also known as black hairy tongue), pigmented fungiform papillae of the tongue, acanthosis nigricans, and oral hairy leukoplakia. Lingua villosa nigra is a similar condition in which individuals present with a black tongue; however, the tongue also appears hairy. The tongue may appear as other colors such as brown, yellow, or green. Patients additionally may have symptoms of burning, dysgeusia, halitosis, or gagging. Poor oral hygiene, xerostomia, use of tobacco or alcohol, and different medications including antibiotics and antipsychotic medications increase the risk for developing lingua villosa nigra.2,3 This condition is distinguished from pseudo-black hairy tongue by proliferation and elongation of the filiform papillae.3 Pigmented fungiform papillae of the tongue is a normal variant of tongue morphology, is more common in individuals with darker skin types, and primarily affects the lateral aspect and apex of the tongue.4 Acanthosis nigricans can appear in the oral cavity as multiple pigmented papillary lesions on the dorsal and lateral regions of the tongue and frequently involves the lips; this condition may be associated with metabolic disorders or underlying malignancy.2,3 Oral hairy leukoplakia is caused by Epstein-Barr virus infection and typically presents as white plaques on the dorsal and ventral surfaces of the tongue; this condition largely is found in immunocompromised patients.5

In our patient there was an acute onset of tongue discoloration associated with ingestion of bismuth salicylate, no hypertrophy or lengthening of the lingual papillae, and no involvement of the patient's lips, which was consistent with the diagnosis of pseudo-black hairy tongue. Pseudo-black hairy tongue is transient and treated by discontinuation of offending agents and proper hygiene practices.

References
  1. Bradley B, Singleton M, Lin Wan Po A. Bismuth toxicity--a reassessment. J Clin Pharm Ther. 1989;14:423-441.
  2. Gurvits GE, Tan A. Black hairy tongue syndrome. World J Gastroenterol. 2014;20:10845-10850.
  3. Schlager E, St Claire C, Ashack K, et al. Black hairy tongue: predisposing factors, diagnosis, and treatment. Am J Clin Dermatol. 2017;18:563-569.  
  4. Mangold AR, Torgerson RR, Rogers RS. Diseases of the tongue. Clin Dermatol. 2016;34:458-469.
  5. Husak R, Garbe C, Orfanos CE. Oral hairy leukoplakia in 71 HIV-seropositive patients: clinical symptoms, relation to immunologic status, and prognostic significance. J Am Acad Dermatol. 1996;35:928-934.
References
  1. Bradley B, Singleton M, Lin Wan Po A. Bismuth toxicity--a reassessment. J Clin Pharm Ther. 1989;14:423-441.
  2. Gurvits GE, Tan A. Black hairy tongue syndrome. World J Gastroenterol. 2014;20:10845-10850.
  3. Schlager E, St Claire C, Ashack K, et al. Black hairy tongue: predisposing factors, diagnosis, and treatment. Am J Clin Dermatol. 2017;18:563-569.  
  4. Mangold AR, Torgerson RR, Rogers RS. Diseases of the tongue. Clin Dermatol. 2016;34:458-469.
  5. Husak R, Garbe C, Orfanos CE. Oral hairy leukoplakia in 71 HIV-seropositive patients: clinical symptoms, relation to immunologic status, and prognostic significance. J Am Acad Dermatol. 1996;35:928-934.
Issue
Cutis - 105(6)
Issue
Cutis - 105(6)
Page Number
288, 293
Page Number
288, 293
Publications
Publications
Topics
Article Type
Display Headline
Asymptomatic Transient Lingual Hyperpigmentation
Display Headline
Asymptomatic Transient Lingual Hyperpigmentation
Sections
Questionnaire Body

patel_tongue_quiz.jpg

A 77-year-old woman incidentally was noted to have black discoloration of the tongue during a routine dermatologic examination. The patient was unaware of the tongue discoloration and reported that her tongue appeared normal the day prior. The tongue was asymptomatic. Clinical examination revealed black hyperpigmentation on the dorsal aspect of the tongue without appreciable hypertrophy or hyperkeratosis of the filiform papillae. The patient had a half-pack daily smoking habit for many years but had abstained from any smoking or tobacco use for the last 15 years. The patient endorsed good oral hygiene. Upon further questioning, the patient revealed that she had ingested 1 tablet of bismuth salicylate the prior night to relieve postprandial dyspepsia. A cotton-tipped applicator was rubbed gently against the affected area and removed some of the black pigment. 

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Mon, 06/01/2020 - 16:00
Un-Gate On Date
Mon, 06/01/2020 - 16:00
Use ProPublica
CFC Schedule Remove Status
Mon, 06/01/2020 - 16:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Article PDF Media

Rapidly growing lesions on the forehead

Article Type
Changed
Wed, 07/10/2019 - 12:25
Display Headline
Rapidly growing lesions on the forehead

A 97-year-old woman with a history of atrial fibrillation and nonmelanoma skin cancer presented to our clinic from an assisted living facility with a several-month history of rapidly growing forehead lesions. She denied symptoms, other than some bleeding and crusting, but was concerned about their appearance. She reported a notable history of sun exposure.

The patient had 3 confluent, but distinct, lesions on her forehead: an erythematous crateriform nodule with overlying hyperkeratotic scale (FIGURE, Lesion A); a nodular hyperpigmented plaque with irregular color and borders (Lesion B); and a pearly well-vascularized erythematous nodule with surrounding hemorrhagic crust (Lesion C).

JFP06806293_f1.JPG

She also had scattered, thin, gritty pink papules and plaques on the face that were thought to be actinic keratosis and nonmelanoma skin cancers based on clinical morphology; however, the patient deferred workup and treatment of these lesions to focus on the forehead lesions. The decision was made to biopsy all 3 clinical morphologies seen. The risks and benefits of biopsy were reviewed with the patient and her daughter, and they opted to proceed. The areas were anesthetized with an injection of 1% lidocaine and epinephrine 1:100,000; 3 shave biopsies were performed. Hemostasis was obtained with electrodesiccation.

WHAT IS YOUR DIAGNOSIS?
HOW WOULD YOU TREAT THIS PATIENT?

 

 

Diagnosis: Skin cancer

A histopathology report revealed that Lesion A was squamous cell carcinoma (SCC), Lesion B was a melanoma with a Breslow depth of at least 1.2 mm, and Lesion C was basal cell carcinoma (BCC). It is unusual to have a patient present with BCC, SCC, and melanoma concurrently in the same anatomic region.

Two of the lesions were nonmelanoma skin cancers (NMSC). BCC is the most common NMSC in the United States, affecting more than 3.3 million people per year.1 Although there are several subtypes of BCC with varying clinical presentations, the most classic appearance is a pearly papule with or without surface telangiectasias.2

The findings in this case serve as an important reminder to biopsy lesions with varying morphologies even when they are in close proximity to one another.

SCC has an incidence of 200,000 to 400,000 cases per year in the United States and the lifetime risk is 9% to 14% in men and 4% to 9% in women.3 SCC most commonly presents as a hyperkeratotic papule or plaque.2 Lesions suspicious for SCC and BCC should be biopsied and the diagnosis confirmed by histopathologic analysis. These NMSCs are locally destructive, but rarely metastatic with a generally good prognosis. The standard treatment for both is surgical excision with consideration for other treatment modalities, such as topical therapies, chemotherapy, and radiation, depending on tumor characteristics as well as whether the patient is a good surgical candidate.1,3

 

Melanoma is rising in incidence each year, with nearly 100,000 new cases expected in the United States this year.4 It is the leading cause of skin cancer related mortality.5 The most common suspicious lesions are variably pigmented macules with irregular borders. Biopsy and subsequent histopathologic analysis will confirm the diagnosis.

When a lesion is clinically suspicious for melanoma, it is particularly important to consider an excisional biopsy to allow for proper staging.5 Examples of appropriate excisional biopsies include elliptical excisions, punch biopsies, and deep shave biopsies.5 Definitive treatment involves a wider and deeper excision with histologically confirmed clear margins.5

Continue to: This case required a multidisciplinary team

 

 

This case required a multidisciplinary team

The patient underwent magnetic resonance imaging and positron emission tomography/computed tomography; the scans revealed no metastatic disease. She was evaluated by a multidisciplinary head and neck cancer team, and various treatment options were explored. Resection typically is the definitive treatment for localized cutaneous melanoma; however, given the configuration of the lesions, it was deemed impractical to resect this patient’s melanoma and not the other lesions. Radiotherapy can be effective for BCC and SCC, but it is traditionally not as effective for melanoma.6 The options presented to the patient were radiotherapy or surgical resection to all 3 lesions, and she decided to pursue resection.

The patient was cleared for surgery; however, after the patient held her warfarin in preparation for the resection, she suffered a left frontal operculum infarction. At this point, she was re-evaluated by her head and neck physician, cardiologist, and anesthesiologist. Consensus was reached that the patient was at high perioperative risk for morbidity and mortality, and surgical intervention was no longer considered a viable option.

The patient then opted for palliative radiation therapy to all 3 lesions, with the understanding that the local control offered by radiotherapy would be inferior to what resection would provide for the melanoma lesion. Although not curative, radiotherapy was expected to provide local symptom relief for the melanoma, consistent with the patient’s palliative goals of care. In the past, melanoma was thought to be resistant to radiation, but recent evidence suggests that it may be at least partially susceptible to hypofractionated courses of radiation.6

Radiation oncology recommended a 6 to 15 fraction regimen and she had a good clinical response with > 50% decrease in the size of all 3 lesions along with cessation of bleeding.

The take-home lesson. The findings in this case serve as an important reminder to biopsy lesions with varying morphologies—even when they are in close proximity to one another. Foregoing any of the biopsies in this case would have led to a missed diagnosis, which has implications for optimal management and treatment.

CORRESPONDENCE
Jennifer L. Hsiao, MD, 2020 Santa Monica Boulevard, Suite 510, Santa Monica, CA 90404; jhsiao@mednet.ucla.edu

References

1. Kim JYS, Kozlow JH, Mittal B, et al. Guidelines of care for the management of basal cell carcinoma. J Am Acad Dermatol. 2018;78:540-559.

2. Firnhaber JM. Diagnosis and treatment of basal cell and squamous cell carcinoma. Am Fam Physician. 2012;86:161-168.

3. Kim JYS, Kozlow JH, Mittal B, et al. Guidelines of care for the management of cutaneous squamous cell carcinoma. J Am Acad Dermatol. 2018;78:560-578.

4. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69:7-34.

5. Swetter SM, Tsao H, Bichakjian CK, et al. Guidelines of care for the management of primary cutaneous melanoma. J Am Acad Dermatol. 2019;80:208-250.

6. Vuong W, Lin J, Wei RL. Palliative radiotherapy for skin malignancies. Ann Palliat Med. 2017;6:165-172.

Article PDF
Author and Disclosure Information

Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles
jhsiao@mednet.ucla.edu

DEPARTMENT EDITOR
Richard P. Usatine, MD

University of Texas Health at San Antonio

The authors reported no potential conflict of interest relevant to this article.

Issue
The Journal of Family Practice - 68(5)
Publications
Topics
Page Number
293-295
Sections
Author and Disclosure Information

Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles
jhsiao@mednet.ucla.edu

DEPARTMENT EDITOR
Richard P. Usatine, MD

University of Texas Health at San Antonio

The authors reported no potential conflict of interest relevant to this article.

Author and Disclosure Information

Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles
jhsiao@mednet.ucla.edu

DEPARTMENT EDITOR
Richard P. Usatine, MD

University of Texas Health at San Antonio

The authors reported no potential conflict of interest relevant to this article.

Article PDF
Article PDF

A 97-year-old woman with a history of atrial fibrillation and nonmelanoma skin cancer presented to our clinic from an assisted living facility with a several-month history of rapidly growing forehead lesions. She denied symptoms, other than some bleeding and crusting, but was concerned about their appearance. She reported a notable history of sun exposure.

The patient had 3 confluent, but distinct, lesions on her forehead: an erythematous crateriform nodule with overlying hyperkeratotic scale (FIGURE, Lesion A); a nodular hyperpigmented plaque with irregular color and borders (Lesion B); and a pearly well-vascularized erythematous nodule with surrounding hemorrhagic crust (Lesion C).

JFP06806293_f1.JPG

She also had scattered, thin, gritty pink papules and plaques on the face that were thought to be actinic keratosis and nonmelanoma skin cancers based on clinical morphology; however, the patient deferred workup and treatment of these lesions to focus on the forehead lesions. The decision was made to biopsy all 3 clinical morphologies seen. The risks and benefits of biopsy were reviewed with the patient and her daughter, and they opted to proceed. The areas were anesthetized with an injection of 1% lidocaine and epinephrine 1:100,000; 3 shave biopsies were performed. Hemostasis was obtained with electrodesiccation.

WHAT IS YOUR DIAGNOSIS?
HOW WOULD YOU TREAT THIS PATIENT?

 

 

Diagnosis: Skin cancer

A histopathology report revealed that Lesion A was squamous cell carcinoma (SCC), Lesion B was a melanoma with a Breslow depth of at least 1.2 mm, and Lesion C was basal cell carcinoma (BCC). It is unusual to have a patient present with BCC, SCC, and melanoma concurrently in the same anatomic region.

Two of the lesions were nonmelanoma skin cancers (NMSC). BCC is the most common NMSC in the United States, affecting more than 3.3 million people per year.1 Although there are several subtypes of BCC with varying clinical presentations, the most classic appearance is a pearly papule with or without surface telangiectasias.2

The findings in this case serve as an important reminder to biopsy lesions with varying morphologies even when they are in close proximity to one another.

SCC has an incidence of 200,000 to 400,000 cases per year in the United States and the lifetime risk is 9% to 14% in men and 4% to 9% in women.3 SCC most commonly presents as a hyperkeratotic papule or plaque.2 Lesions suspicious for SCC and BCC should be biopsied and the diagnosis confirmed by histopathologic analysis. These NMSCs are locally destructive, but rarely metastatic with a generally good prognosis. The standard treatment for both is surgical excision with consideration for other treatment modalities, such as topical therapies, chemotherapy, and radiation, depending on tumor characteristics as well as whether the patient is a good surgical candidate.1,3

 

Melanoma is rising in incidence each year, with nearly 100,000 new cases expected in the United States this year.4 It is the leading cause of skin cancer related mortality.5 The most common suspicious lesions are variably pigmented macules with irregular borders. Biopsy and subsequent histopathologic analysis will confirm the diagnosis.

When a lesion is clinically suspicious for melanoma, it is particularly important to consider an excisional biopsy to allow for proper staging.5 Examples of appropriate excisional biopsies include elliptical excisions, punch biopsies, and deep shave biopsies.5 Definitive treatment involves a wider and deeper excision with histologically confirmed clear margins.5

Continue to: This case required a multidisciplinary team

 

 

This case required a multidisciplinary team

The patient underwent magnetic resonance imaging and positron emission tomography/computed tomography; the scans revealed no metastatic disease. She was evaluated by a multidisciplinary head and neck cancer team, and various treatment options were explored. Resection typically is the definitive treatment for localized cutaneous melanoma; however, given the configuration of the lesions, it was deemed impractical to resect this patient’s melanoma and not the other lesions. Radiotherapy can be effective for BCC and SCC, but it is traditionally not as effective for melanoma.6 The options presented to the patient were radiotherapy or surgical resection to all 3 lesions, and she decided to pursue resection.

The patient was cleared for surgery; however, after the patient held her warfarin in preparation for the resection, she suffered a left frontal operculum infarction. At this point, she was re-evaluated by her head and neck physician, cardiologist, and anesthesiologist. Consensus was reached that the patient was at high perioperative risk for morbidity and mortality, and surgical intervention was no longer considered a viable option.

The patient then opted for palliative radiation therapy to all 3 lesions, with the understanding that the local control offered by radiotherapy would be inferior to what resection would provide for the melanoma lesion. Although not curative, radiotherapy was expected to provide local symptom relief for the melanoma, consistent with the patient’s palliative goals of care. In the past, melanoma was thought to be resistant to radiation, but recent evidence suggests that it may be at least partially susceptible to hypofractionated courses of radiation.6

Radiation oncology recommended a 6 to 15 fraction regimen and she had a good clinical response with > 50% decrease in the size of all 3 lesions along with cessation of bleeding.

The take-home lesson. The findings in this case serve as an important reminder to biopsy lesions with varying morphologies—even when they are in close proximity to one another. Foregoing any of the biopsies in this case would have led to a missed diagnosis, which has implications for optimal management and treatment.

CORRESPONDENCE
Jennifer L. Hsiao, MD, 2020 Santa Monica Boulevard, Suite 510, Santa Monica, CA 90404; jhsiao@mednet.ucla.edu

A 97-year-old woman with a history of atrial fibrillation and nonmelanoma skin cancer presented to our clinic from an assisted living facility with a several-month history of rapidly growing forehead lesions. She denied symptoms, other than some bleeding and crusting, but was concerned about their appearance. She reported a notable history of sun exposure.

The patient had 3 confluent, but distinct, lesions on her forehead: an erythematous crateriform nodule with overlying hyperkeratotic scale (FIGURE, Lesion A); a nodular hyperpigmented plaque with irregular color and borders (Lesion B); and a pearly well-vascularized erythematous nodule with surrounding hemorrhagic crust (Lesion C).

JFP06806293_f1.JPG

She also had scattered, thin, gritty pink papules and plaques on the face that were thought to be actinic keratosis and nonmelanoma skin cancers based on clinical morphology; however, the patient deferred workup and treatment of these lesions to focus on the forehead lesions. The decision was made to biopsy all 3 clinical morphologies seen. The risks and benefits of biopsy were reviewed with the patient and her daughter, and they opted to proceed. The areas were anesthetized with an injection of 1% lidocaine and epinephrine 1:100,000; 3 shave biopsies were performed. Hemostasis was obtained with electrodesiccation.

WHAT IS YOUR DIAGNOSIS?
HOW WOULD YOU TREAT THIS PATIENT?

 

 

Diagnosis: Skin cancer

A histopathology report revealed that Lesion A was squamous cell carcinoma (SCC), Lesion B was a melanoma with a Breslow depth of at least 1.2 mm, and Lesion C was basal cell carcinoma (BCC). It is unusual to have a patient present with BCC, SCC, and melanoma concurrently in the same anatomic region.

Two of the lesions were nonmelanoma skin cancers (NMSC). BCC is the most common NMSC in the United States, affecting more than 3.3 million people per year.1 Although there are several subtypes of BCC with varying clinical presentations, the most classic appearance is a pearly papule with or without surface telangiectasias.2

The findings in this case serve as an important reminder to biopsy lesions with varying morphologies even when they are in close proximity to one another.

SCC has an incidence of 200,000 to 400,000 cases per year in the United States and the lifetime risk is 9% to 14% in men and 4% to 9% in women.3 SCC most commonly presents as a hyperkeratotic papule or plaque.2 Lesions suspicious for SCC and BCC should be biopsied and the diagnosis confirmed by histopathologic analysis. These NMSCs are locally destructive, but rarely metastatic with a generally good prognosis. The standard treatment for both is surgical excision with consideration for other treatment modalities, such as topical therapies, chemotherapy, and radiation, depending on tumor characteristics as well as whether the patient is a good surgical candidate.1,3

 

Melanoma is rising in incidence each year, with nearly 100,000 new cases expected in the United States this year.4 It is the leading cause of skin cancer related mortality.5 The most common suspicious lesions are variably pigmented macules with irregular borders. Biopsy and subsequent histopathologic analysis will confirm the diagnosis.

When a lesion is clinically suspicious for melanoma, it is particularly important to consider an excisional biopsy to allow for proper staging.5 Examples of appropriate excisional biopsies include elliptical excisions, punch biopsies, and deep shave biopsies.5 Definitive treatment involves a wider and deeper excision with histologically confirmed clear margins.5

Continue to: This case required a multidisciplinary team

 

 

This case required a multidisciplinary team

The patient underwent magnetic resonance imaging and positron emission tomography/computed tomography; the scans revealed no metastatic disease. She was evaluated by a multidisciplinary head and neck cancer team, and various treatment options were explored. Resection typically is the definitive treatment for localized cutaneous melanoma; however, given the configuration of the lesions, it was deemed impractical to resect this patient’s melanoma and not the other lesions. Radiotherapy can be effective for BCC and SCC, but it is traditionally not as effective for melanoma.6 The options presented to the patient were radiotherapy or surgical resection to all 3 lesions, and she decided to pursue resection.

The patient was cleared for surgery; however, after the patient held her warfarin in preparation for the resection, she suffered a left frontal operculum infarction. At this point, she was re-evaluated by her head and neck physician, cardiologist, and anesthesiologist. Consensus was reached that the patient was at high perioperative risk for morbidity and mortality, and surgical intervention was no longer considered a viable option.

The patient then opted for palliative radiation therapy to all 3 lesions, with the understanding that the local control offered by radiotherapy would be inferior to what resection would provide for the melanoma lesion. Although not curative, radiotherapy was expected to provide local symptom relief for the melanoma, consistent with the patient’s palliative goals of care. In the past, melanoma was thought to be resistant to radiation, but recent evidence suggests that it may be at least partially susceptible to hypofractionated courses of radiation.6

Radiation oncology recommended a 6 to 15 fraction regimen and she had a good clinical response with > 50% decrease in the size of all 3 lesions along with cessation of bleeding.

The take-home lesson. The findings in this case serve as an important reminder to biopsy lesions with varying morphologies—even when they are in close proximity to one another. Foregoing any of the biopsies in this case would have led to a missed diagnosis, which has implications for optimal management and treatment.

CORRESPONDENCE
Jennifer L. Hsiao, MD, 2020 Santa Monica Boulevard, Suite 510, Santa Monica, CA 90404; jhsiao@mednet.ucla.edu

References

1. Kim JYS, Kozlow JH, Mittal B, et al. Guidelines of care for the management of basal cell carcinoma. J Am Acad Dermatol. 2018;78:540-559.

2. Firnhaber JM. Diagnosis and treatment of basal cell and squamous cell carcinoma. Am Fam Physician. 2012;86:161-168.

3. Kim JYS, Kozlow JH, Mittal B, et al. Guidelines of care for the management of cutaneous squamous cell carcinoma. J Am Acad Dermatol. 2018;78:560-578.

4. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69:7-34.

5. Swetter SM, Tsao H, Bichakjian CK, et al. Guidelines of care for the management of primary cutaneous melanoma. J Am Acad Dermatol. 2019;80:208-250.

6. Vuong W, Lin J, Wei RL. Palliative radiotherapy for skin malignancies. Ann Palliat Med. 2017;6:165-172.

References

1. Kim JYS, Kozlow JH, Mittal B, et al. Guidelines of care for the management of basal cell carcinoma. J Am Acad Dermatol. 2018;78:540-559.

2. Firnhaber JM. Diagnosis and treatment of basal cell and squamous cell carcinoma. Am Fam Physician. 2012;86:161-168.

3. Kim JYS, Kozlow JH, Mittal B, et al. Guidelines of care for the management of cutaneous squamous cell carcinoma. J Am Acad Dermatol. 2018;78:560-578.

4. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69:7-34.

5. Swetter SM, Tsao H, Bichakjian CK, et al. Guidelines of care for the management of primary cutaneous melanoma. J Am Acad Dermatol. 2019;80:208-250.

6. Vuong W, Lin J, Wei RL. Palliative radiotherapy for skin malignancies. Ann Palliat Med. 2017;6:165-172.

Issue
The Journal of Family Practice - 68(5)
Issue
The Journal of Family Practice - 68(5)
Page Number
293-295
Page Number
293-295
Publications
Publications
Topics
Article Type
Display Headline
Rapidly growing lesions on the forehead
Display Headline
Rapidly growing lesions on the forehead
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
PubMed ID
31287446
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media
Image
Teambase ID
180013AC.SIG
Disable zoom
Off

Levofloxacin-Induced Purpura Annularis Telangiectodes of Majocchi

Article Type
Changed
Thu, 01/10/2019 - 13:46
Display Headline
Levofloxacin-Induced Purpura Annularis Telangiectodes of Majocchi

To the Editor:

Purpura annularis telangiectodes of Majocchi (PATM) is a type of pigmented purpuric dermatosis (PPD). Patients present with nonblanchable, annular, symmetric, purpuric, and telangiectatic patches, often on the legs, with histology revealing a perivascular lymphocytic infiltrate and extravasated erythrocytes.1,2 A variety of medications have been linked to the development of PPD. We describe a case of levofloxacin-induced PATM.

RELATED ARTICLE: Granulomatous Changes Associated With Pigmented Purpuric Dermatosis

A 42-year-old man presented with a rash on the arms, trunk, abdomen, and legs of 1 month’s duration. He reported no associated itching, bleeding, or pain, and no history of a similar rash. He had a history of hypothyroidism and had been taking levothyroxine for years. He had no known allergies and no history of childhood eczema, asthma, or allergic rhinitis. Notably, the rash started shortly after the patient finished a 2-week course of levofloxacin, an antibiotic he had not taken in the past. The patient resided with his wife, 3 children, and a pet dog, and no family members had the rash. Prior to presentation, the patient had tried econazole cream and then triamcinolone acetonide cream 0.5% without any clinical improvement.

A complete review of systems was unremarkable. Physical examination revealed scattered, reddish brown, annular, nonscaly patches on the back, abdomen (Figure 1), arms, and legs with nonblanching petechiae within the patches.

ct100004010_e_fig1.png
Figure 1. Purpura annularis telangiectodes of Majocchi with scattered, reddish brown, annular, nonscaly patches on the trunk and nonblanching petechiae within the patches.

A punch biopsy of the left inner thigh demonstrated patchy interface dermatitis, superficial perivascular inflammation, and numerous extravasated red blood cells in the papillary dermis (Figure 2). The histologic features were compatible with the clinical impression of PATM. The patient presented for a follow-up visit 2 weeks later with no new lesions and the old lesions were rapidly fading (Figure 3).

ct100004010_e_fig2.png
Figure 2. Purpura annularis telangiectodes of Majocchi histology demonstrated patchy interface dermatitis, superficial perivascular inflammation, and numerous extravasated red blood cells in the papillary dermis (A and B)(both H&E, original magnifications ×10 and ×20).

ct100004010_e_fig3.png
Figure 3. Clearance of purpura annularis telangiectodes of Majocchi lesions on the abdomen after discontinuation of levofloxacin.

Pigmented purpuric dermatoses are a group of conditions that have different clinical morphologies but similar histopathologic examinations.2 All PPDs are characterized by nonblanching, nonpalpable, purpuric lesions that often are bilaterally symmetrical and present on the legs.2,3 Although the precise etiology of these conditions is not known, most cases include a perivascular lymphocytic infiltrate along with the presence of extravasated erythrocytes and hemosiderin deposition in the dermis.2 Of note, PATM often is idiopathic and patients usually present with no associated comorbidities.3 The currently established PPDs include progressive pigmentary dermatosis (Schamberg disease), PATM, pigmented purpuric lichenoid dermatosis of Gougerot and Blum, lichen aureus, and eczematidlike purpura of Doucas and Kapetanakis.2,4

RELATED ARTICLE: Granulomatous Pigmented Purpuric Dermatosis

 

 

The lesions of PATM are symmetrically distributed on the bilateral legs and may be symptomatic in most cases, with severe pruritus being reported in several drug-induced PATM cases.3,5 Although the exact etiology of PPDs currently is unknown, some contributing factors that are thought to play a role include exercise, venous stasis, gravitational dependence, capillary fragility, hypertension, drugs, chemical exposure or ingestions, and contact allergy to dyes.3 Some of the drugs known to cause drug-induced PPDs fall into the class of sedatives, stimulants, antibiotics, cardiovascular drugs, vitamins, and nutritional supplements.3,6 Some medications that have been reported to cause PPDs include acetaminophen, aspirin, carbamazepine, diltiazem, furosemide, glipizide, hydralazine, infliximab, isotretinoin, lorazepam, minocycline, nitroglycerine, and sildenafil.3,7-15

Although the mechanism of drug-induced PPD is not completely understood, it is thought that the ingested substance leads to an immunologic response in the capillary endothelium, which results in a cell-mediated immune response causing vascular damage.3 The ingested substance may act as a hapten, stimulating antibody formation and immune-mediated injury, leading to the clinical presentation of nonblanching, symmetric, purpuric, telangiectatic, and atrophic patches at the site of injury.1,3

Levofloxacin is a broad-spectrum antibiotic that has activity against both gram-positive and gram-negative bacteria. It inhibits the enzymes DNA gyrase and topoisomerase IV, preventing bacteria from undergoing proper DNA synthesis.16 Our patient’s rash began shortly after a 2-week course of levofloxacin and faded within a few weeks of discontinuing the drug; the clinical presentation, time course, and histologic appearance of the lesions were consistent with the diagnosis of drug-induced PPD. Of note, solar capillaritis has been reported following a phototoxic reaction induced by levofloxacin.17 Our case differs in that our patient had annular lesions on both photoprotected and photoexposed skin.

The first-line interventions for the treatment of PPDs are nonpharmacologic, such as discontinuation of an offending drug or allergen or wearing supportive stockings if there are signs of venous stasis. Other interventions include the use of a medium- or high-potency topical corticosteroid once to twice daily to affected areas for 4 to 6 weeks.18 Some case series also have shown improvement with narrowband UVB treatment after 24 to 28 treatment sessions or with psoralen plus UVA phototherapy within 7 to 20 treatments.19,20 If the above measures are unsuccessful in resolving symptoms, other treatment alternatives may include pentoxifylline, griseofulvin, colchicine, cyclosporine, and methotrexate. The potential benefit of treatment must be weighed against the side-effect profile of these medications.2,21-24 Of note, oral rutoside (50 mg twice daily) and ascorbic acid (500 mg twice daily) were administered to 3 patients with chronic progressive pigmented purpura. At the end of the 4-week treatment period, complete clearance of skin lesions was seen in all patients with no adverse reactions noted.25

Despite these treatment options, PATM does not necessitate treatment given its benign course and often self-resolving nature.26 In cases of drug-induced PPD such as in our patient, discontinuation of the offending drug often may lead to resolution.

In summary, PATM is a PPD that has been associated with different etiologic factors. If PATM is suspected to be caused by a drug, discontinuation of the offending agent usually results in resolution of symptoms, as it did in our case with fading of lesions within a few weeks after the patient was no longer taking levofloxacin.

References
  1. Hale EK. Purpura annularis telangiectodes of Majocchi. Dermatol Online J. 2003;9:17.
  2. Hoesly FJ, Huerter CJ, Shehan JM. Purpura annularis telangiectodes of Majocchi: case report and review of the literature. Int J Dermatol. 2009;48:1129-1133.
  3. Kaplan R, Meehan SA, Leger M. A case of isotretinoin-induced purpura annularis telangiectodes of Majocchi and review of substance-induced pigmented purpuric dermatosis. JAMA Dermatol. 2014;150:182-184.
  4. Newton RC, Raimer SS. Pigmented purpuric eruptions. Dermatol Clin. 1985;3:165-169.
  5. Ratnam KV, Su WP, Peters MS. Purpura simplex (inflammatory purpura without vasculitis): a clinicopathologic study of 174 cases. J Am Acad Dermatol. 1991;25:642-647.
  6. Pang BK, Su D, Ratnam KV. Drug-induced purpura simplex: clinical and histological characteristics. Ann Acad Med Singapore. 1993;22:870-872.
  7. Abeck D, Gross GE, Kuwert C, et al. Acetaminophen-induced progressive pigmentary purpura (Schamberg’s disease). J Am Acad Dermatol. 1992;27:123-124.
  8. Lipsker D, Cribier B, Heid E, et al. Cutaneous lymphoma manifesting as pigmented, purpuric capillaries [in French]. Ann Dermatol Venereol. 1999;126:321-326.
  9. Peterson WC Jr, Manick KP. Purpuric eruptions associated with use of carbromal and meprobamate. Arch Dermatol. 1967;95:40-42.
  10. Nishioka K, Katayama I, Masuzawa M, et al. Drug-induced chronic pigmented purpura. J Dermatol. 1989;16:220-222.
  11. Voelter WW. Pigmented purpuric dermatosis-like reaction to topical fluorouracil. Arch Dermatol. 1983;119:875-876.
  12. Adams BB, Gadenne AS. Glipizide-induced pigmented purpuric dermatosis. J Am Acad Dermatol. 1999;41(5, pt 2):827-829.
  13. Tsao H, Lerner LH. Pigmented purpuric eruption associated with injection medroxyprogesterone acetate. J Am Acad Dermatol. 2000;43(2, pt 1):308-310.
  14. Koçak AY, Akay BN, Heper AO. Sildenafil-induced pigmented purpuric dermatosis. Cutan Ocul Toxicol. 2013;32:91-92.
  15. Nishioka K, Sarashi C, Katayama I. Chronic pigmented purpura induced by chemical substances. Clin Exp Dermatol. 1980;5:213-218.
  16. Drlica K, Zhao X. DNA gyrase, topoisomerase IV, and the 4-quinolones. Microbiol Mol Biol Rev. 1997;61:377-392.
  17. Rubegni P, Feci L, Pellegrino M, et al. Photolocalized purpura during levofloxacin therapy. Photodermatol Photoimmunol Photomed. 2012;28:105-107.
  18. Sardana K, Sarkar R, Sehgal VN. Pigmented purpuric dermatoses: an overview. Int J Dermatol. 2004;43:482-488.
  19. Fathy H, Abdelgaber S. Treatment of pigmented purpuric dermatoses with narrow-band UVB: a report of six cases. J Eur Acad Dermatol Venereol. 2011;25:603-606.
  20. Krizsa J, Hunyadi J, Dobozy A. PUVA treatment of pigmented purpuric lichenoid dermatitis (Gougerot-Blum). J Am Acad Dermatol. 1992;27(5, pt 1):778-780.
  21. Panda S, Malakar S, Lahiri K. Oral pentoxifylline vs topical betamethasone in Schamberg disease: a comparative randomized investigator-blinded parallel-group trial. Arch Dermatol. 2004;140:491-493.
  22. Tamaki K, Yasaka N, Osada A, et al. Successful treatment of pigmented purpuric dermatosis with griseofulvin. Br J Dermatol. 1995;132:159-160.
  23. Geller M. Benefit of colchicine in the treatment of Schamberg’s disease. Ann Allergy Asthma Immunol. 2000;85:246.
  24. Okada K, Ishikawa O, Miyachi Y. Purpura pigmentosa chronica successfully treated with oral cyclosporin A. Br J Dermatol. 1996;134:180-181.
  25. Reinhold U, Seiter S, Ugurel S, et al. Treatment of progressive pigmented purpura with oral bioflavonoids and ascorbic acid: an open pilot study in 3 patients. J Am Acad Dermatol. 1999;41(2, pt 1):207-208.
  26. Wang A, Shuja F, Chan A, et al. Unilateral purpura annularis telangiectodes of Majocchi in an elderly male: an atypical presentation. Dermatol Online J. 2013;19:19263.
Article PDF
Author and Disclosure Information

From the Division of Dermatology, David Geffen School of Medicine, University of California, Los Angeles, and Southern California Permanente Medical Group of South Bay, Gardena.

The authors report no conflict of interest.

Correspondence: Ki-Young Yoo, MD, Southern California Permanente Medical Group of South Bay, Department of Dermatology, 18600 S Figueroa St, Gardena, CA 90248 (ki-young.yoo@kp.org).

Issue
Cutis - 100(4)
Publications
Topics
Page Number
E10-E12
Sections
Author and Disclosure Information

From the Division of Dermatology, David Geffen School of Medicine, University of California, Los Angeles, and Southern California Permanente Medical Group of South Bay, Gardena.

The authors report no conflict of interest.

Correspondence: Ki-Young Yoo, MD, Southern California Permanente Medical Group of South Bay, Department of Dermatology, 18600 S Figueroa St, Gardena, CA 90248 (ki-young.yoo@kp.org).

Author and Disclosure Information

From the Division of Dermatology, David Geffen School of Medicine, University of California, Los Angeles, and Southern California Permanente Medical Group of South Bay, Gardena.

The authors report no conflict of interest.

Correspondence: Ki-Young Yoo, MD, Southern California Permanente Medical Group of South Bay, Department of Dermatology, 18600 S Figueroa St, Gardena, CA 90248 (ki-young.yoo@kp.org).

Article PDF
Article PDF

To the Editor:

Purpura annularis telangiectodes of Majocchi (PATM) is a type of pigmented purpuric dermatosis (PPD). Patients present with nonblanchable, annular, symmetric, purpuric, and telangiectatic patches, often on the legs, with histology revealing a perivascular lymphocytic infiltrate and extravasated erythrocytes.1,2 A variety of medications have been linked to the development of PPD. We describe a case of levofloxacin-induced PATM.

RELATED ARTICLE: Granulomatous Changes Associated With Pigmented Purpuric Dermatosis

A 42-year-old man presented with a rash on the arms, trunk, abdomen, and legs of 1 month’s duration. He reported no associated itching, bleeding, or pain, and no history of a similar rash. He had a history of hypothyroidism and had been taking levothyroxine for years. He had no known allergies and no history of childhood eczema, asthma, or allergic rhinitis. Notably, the rash started shortly after the patient finished a 2-week course of levofloxacin, an antibiotic he had not taken in the past. The patient resided with his wife, 3 children, and a pet dog, and no family members had the rash. Prior to presentation, the patient had tried econazole cream and then triamcinolone acetonide cream 0.5% without any clinical improvement.

A complete review of systems was unremarkable. Physical examination revealed scattered, reddish brown, annular, nonscaly patches on the back, abdomen (Figure 1), arms, and legs with nonblanching petechiae within the patches.

ct100004010_e_fig1.png
Figure 1. Purpura annularis telangiectodes of Majocchi with scattered, reddish brown, annular, nonscaly patches on the trunk and nonblanching petechiae within the patches.

A punch biopsy of the left inner thigh demonstrated patchy interface dermatitis, superficial perivascular inflammation, and numerous extravasated red blood cells in the papillary dermis (Figure 2). The histologic features were compatible with the clinical impression of PATM. The patient presented for a follow-up visit 2 weeks later with no new lesions and the old lesions were rapidly fading (Figure 3).

ct100004010_e_fig2.png
Figure 2. Purpura annularis telangiectodes of Majocchi histology demonstrated patchy interface dermatitis, superficial perivascular inflammation, and numerous extravasated red blood cells in the papillary dermis (A and B)(both H&E, original magnifications ×10 and ×20).

ct100004010_e_fig3.png
Figure 3. Clearance of purpura annularis telangiectodes of Majocchi lesions on the abdomen after discontinuation of levofloxacin.

Pigmented purpuric dermatoses are a group of conditions that have different clinical morphologies but similar histopathologic examinations.2 All PPDs are characterized by nonblanching, nonpalpable, purpuric lesions that often are bilaterally symmetrical and present on the legs.2,3 Although the precise etiology of these conditions is not known, most cases include a perivascular lymphocytic infiltrate along with the presence of extravasated erythrocytes and hemosiderin deposition in the dermis.2 Of note, PATM often is idiopathic and patients usually present with no associated comorbidities.3 The currently established PPDs include progressive pigmentary dermatosis (Schamberg disease), PATM, pigmented purpuric lichenoid dermatosis of Gougerot and Blum, lichen aureus, and eczematidlike purpura of Doucas and Kapetanakis.2,4

RELATED ARTICLE: Granulomatous Pigmented Purpuric Dermatosis

 

 

The lesions of PATM are symmetrically distributed on the bilateral legs and may be symptomatic in most cases, with severe pruritus being reported in several drug-induced PATM cases.3,5 Although the exact etiology of PPDs currently is unknown, some contributing factors that are thought to play a role include exercise, venous stasis, gravitational dependence, capillary fragility, hypertension, drugs, chemical exposure or ingestions, and contact allergy to dyes.3 Some of the drugs known to cause drug-induced PPDs fall into the class of sedatives, stimulants, antibiotics, cardiovascular drugs, vitamins, and nutritional supplements.3,6 Some medications that have been reported to cause PPDs include acetaminophen, aspirin, carbamazepine, diltiazem, furosemide, glipizide, hydralazine, infliximab, isotretinoin, lorazepam, minocycline, nitroglycerine, and sildenafil.3,7-15

Although the mechanism of drug-induced PPD is not completely understood, it is thought that the ingested substance leads to an immunologic response in the capillary endothelium, which results in a cell-mediated immune response causing vascular damage.3 The ingested substance may act as a hapten, stimulating antibody formation and immune-mediated injury, leading to the clinical presentation of nonblanching, symmetric, purpuric, telangiectatic, and atrophic patches at the site of injury.1,3

Levofloxacin is a broad-spectrum antibiotic that has activity against both gram-positive and gram-negative bacteria. It inhibits the enzymes DNA gyrase and topoisomerase IV, preventing bacteria from undergoing proper DNA synthesis.16 Our patient’s rash began shortly after a 2-week course of levofloxacin and faded within a few weeks of discontinuing the drug; the clinical presentation, time course, and histologic appearance of the lesions were consistent with the diagnosis of drug-induced PPD. Of note, solar capillaritis has been reported following a phototoxic reaction induced by levofloxacin.17 Our case differs in that our patient had annular lesions on both photoprotected and photoexposed skin.

The first-line interventions for the treatment of PPDs are nonpharmacologic, such as discontinuation of an offending drug or allergen or wearing supportive stockings if there are signs of venous stasis. Other interventions include the use of a medium- or high-potency topical corticosteroid once to twice daily to affected areas for 4 to 6 weeks.18 Some case series also have shown improvement with narrowband UVB treatment after 24 to 28 treatment sessions or with psoralen plus UVA phototherapy within 7 to 20 treatments.19,20 If the above measures are unsuccessful in resolving symptoms, other treatment alternatives may include pentoxifylline, griseofulvin, colchicine, cyclosporine, and methotrexate. The potential benefit of treatment must be weighed against the side-effect profile of these medications.2,21-24 Of note, oral rutoside (50 mg twice daily) and ascorbic acid (500 mg twice daily) were administered to 3 patients with chronic progressive pigmented purpura. At the end of the 4-week treatment period, complete clearance of skin lesions was seen in all patients with no adverse reactions noted.25

Despite these treatment options, PATM does not necessitate treatment given its benign course and often self-resolving nature.26 In cases of drug-induced PPD such as in our patient, discontinuation of the offending drug often may lead to resolution.

In summary, PATM is a PPD that has been associated with different etiologic factors. If PATM is suspected to be caused by a drug, discontinuation of the offending agent usually results in resolution of symptoms, as it did in our case with fading of lesions within a few weeks after the patient was no longer taking levofloxacin.

To the Editor:

Purpura annularis telangiectodes of Majocchi (PATM) is a type of pigmented purpuric dermatosis (PPD). Patients present with nonblanchable, annular, symmetric, purpuric, and telangiectatic patches, often on the legs, with histology revealing a perivascular lymphocytic infiltrate and extravasated erythrocytes.1,2 A variety of medications have been linked to the development of PPD. We describe a case of levofloxacin-induced PATM.

RELATED ARTICLE: Granulomatous Changes Associated With Pigmented Purpuric Dermatosis

A 42-year-old man presented with a rash on the arms, trunk, abdomen, and legs of 1 month’s duration. He reported no associated itching, bleeding, or pain, and no history of a similar rash. He had a history of hypothyroidism and had been taking levothyroxine for years. He had no known allergies and no history of childhood eczema, asthma, or allergic rhinitis. Notably, the rash started shortly after the patient finished a 2-week course of levofloxacin, an antibiotic he had not taken in the past. The patient resided with his wife, 3 children, and a pet dog, and no family members had the rash. Prior to presentation, the patient had tried econazole cream and then triamcinolone acetonide cream 0.5% without any clinical improvement.

A complete review of systems was unremarkable. Physical examination revealed scattered, reddish brown, annular, nonscaly patches on the back, abdomen (Figure 1), arms, and legs with nonblanching petechiae within the patches.

ct100004010_e_fig1.png
Figure 1. Purpura annularis telangiectodes of Majocchi with scattered, reddish brown, annular, nonscaly patches on the trunk and nonblanching petechiae within the patches.

A punch biopsy of the left inner thigh demonstrated patchy interface dermatitis, superficial perivascular inflammation, and numerous extravasated red blood cells in the papillary dermis (Figure 2). The histologic features were compatible with the clinical impression of PATM. The patient presented for a follow-up visit 2 weeks later with no new lesions and the old lesions were rapidly fading (Figure 3).

ct100004010_e_fig2.png
Figure 2. Purpura annularis telangiectodes of Majocchi histology demonstrated patchy interface dermatitis, superficial perivascular inflammation, and numerous extravasated red blood cells in the papillary dermis (A and B)(both H&E, original magnifications ×10 and ×20).

ct100004010_e_fig3.png
Figure 3. Clearance of purpura annularis telangiectodes of Majocchi lesions on the abdomen after discontinuation of levofloxacin.

Pigmented purpuric dermatoses are a group of conditions that have different clinical morphologies but similar histopathologic examinations.2 All PPDs are characterized by nonblanching, nonpalpable, purpuric lesions that often are bilaterally symmetrical and present on the legs.2,3 Although the precise etiology of these conditions is not known, most cases include a perivascular lymphocytic infiltrate along with the presence of extravasated erythrocytes and hemosiderin deposition in the dermis.2 Of note, PATM often is idiopathic and patients usually present with no associated comorbidities.3 The currently established PPDs include progressive pigmentary dermatosis (Schamberg disease), PATM, pigmented purpuric lichenoid dermatosis of Gougerot and Blum, lichen aureus, and eczematidlike purpura of Doucas and Kapetanakis.2,4

RELATED ARTICLE: Granulomatous Pigmented Purpuric Dermatosis

 

 

The lesions of PATM are symmetrically distributed on the bilateral legs and may be symptomatic in most cases, with severe pruritus being reported in several drug-induced PATM cases.3,5 Although the exact etiology of PPDs currently is unknown, some contributing factors that are thought to play a role include exercise, venous stasis, gravitational dependence, capillary fragility, hypertension, drugs, chemical exposure or ingestions, and contact allergy to dyes.3 Some of the drugs known to cause drug-induced PPDs fall into the class of sedatives, stimulants, antibiotics, cardiovascular drugs, vitamins, and nutritional supplements.3,6 Some medications that have been reported to cause PPDs include acetaminophen, aspirin, carbamazepine, diltiazem, furosemide, glipizide, hydralazine, infliximab, isotretinoin, lorazepam, minocycline, nitroglycerine, and sildenafil.3,7-15

Although the mechanism of drug-induced PPD is not completely understood, it is thought that the ingested substance leads to an immunologic response in the capillary endothelium, which results in a cell-mediated immune response causing vascular damage.3 The ingested substance may act as a hapten, stimulating antibody formation and immune-mediated injury, leading to the clinical presentation of nonblanching, symmetric, purpuric, telangiectatic, and atrophic patches at the site of injury.1,3

Levofloxacin is a broad-spectrum antibiotic that has activity against both gram-positive and gram-negative bacteria. It inhibits the enzymes DNA gyrase and topoisomerase IV, preventing bacteria from undergoing proper DNA synthesis.16 Our patient’s rash began shortly after a 2-week course of levofloxacin and faded within a few weeks of discontinuing the drug; the clinical presentation, time course, and histologic appearance of the lesions were consistent with the diagnosis of drug-induced PPD. Of note, solar capillaritis has been reported following a phototoxic reaction induced by levofloxacin.17 Our case differs in that our patient had annular lesions on both photoprotected and photoexposed skin.

The first-line interventions for the treatment of PPDs are nonpharmacologic, such as discontinuation of an offending drug or allergen or wearing supportive stockings if there are signs of venous stasis. Other interventions include the use of a medium- or high-potency topical corticosteroid once to twice daily to affected areas for 4 to 6 weeks.18 Some case series also have shown improvement with narrowband UVB treatment after 24 to 28 treatment sessions or with psoralen plus UVA phototherapy within 7 to 20 treatments.19,20 If the above measures are unsuccessful in resolving symptoms, other treatment alternatives may include pentoxifylline, griseofulvin, colchicine, cyclosporine, and methotrexate. The potential benefit of treatment must be weighed against the side-effect profile of these medications.2,21-24 Of note, oral rutoside (50 mg twice daily) and ascorbic acid (500 mg twice daily) were administered to 3 patients with chronic progressive pigmented purpura. At the end of the 4-week treatment period, complete clearance of skin lesions was seen in all patients with no adverse reactions noted.25

Despite these treatment options, PATM does not necessitate treatment given its benign course and often self-resolving nature.26 In cases of drug-induced PPD such as in our patient, discontinuation of the offending drug often may lead to resolution.

In summary, PATM is a PPD that has been associated with different etiologic factors. If PATM is suspected to be caused by a drug, discontinuation of the offending agent usually results in resolution of symptoms, as it did in our case with fading of lesions within a few weeks after the patient was no longer taking levofloxacin.

References
  1. Hale EK. Purpura annularis telangiectodes of Majocchi. Dermatol Online J. 2003;9:17.
  2. Hoesly FJ, Huerter CJ, Shehan JM. Purpura annularis telangiectodes of Majocchi: case report and review of the literature. Int J Dermatol. 2009;48:1129-1133.
  3. Kaplan R, Meehan SA, Leger M. A case of isotretinoin-induced purpura annularis telangiectodes of Majocchi and review of substance-induced pigmented purpuric dermatosis. JAMA Dermatol. 2014;150:182-184.
  4. Newton RC, Raimer SS. Pigmented purpuric eruptions. Dermatol Clin. 1985;3:165-169.
  5. Ratnam KV, Su WP, Peters MS. Purpura simplex (inflammatory purpura without vasculitis): a clinicopathologic study of 174 cases. J Am Acad Dermatol. 1991;25:642-647.
  6. Pang BK, Su D, Ratnam KV. Drug-induced purpura simplex: clinical and histological characteristics. Ann Acad Med Singapore. 1993;22:870-872.
  7. Abeck D, Gross GE, Kuwert C, et al. Acetaminophen-induced progressive pigmentary purpura (Schamberg’s disease). J Am Acad Dermatol. 1992;27:123-124.
  8. Lipsker D, Cribier B, Heid E, et al. Cutaneous lymphoma manifesting as pigmented, purpuric capillaries [in French]. Ann Dermatol Venereol. 1999;126:321-326.
  9. Peterson WC Jr, Manick KP. Purpuric eruptions associated with use of carbromal and meprobamate. Arch Dermatol. 1967;95:40-42.
  10. Nishioka K, Katayama I, Masuzawa M, et al. Drug-induced chronic pigmented purpura. J Dermatol. 1989;16:220-222.
  11. Voelter WW. Pigmented purpuric dermatosis-like reaction to topical fluorouracil. Arch Dermatol. 1983;119:875-876.
  12. Adams BB, Gadenne AS. Glipizide-induced pigmented purpuric dermatosis. J Am Acad Dermatol. 1999;41(5, pt 2):827-829.
  13. Tsao H, Lerner LH. Pigmented purpuric eruption associated with injection medroxyprogesterone acetate. J Am Acad Dermatol. 2000;43(2, pt 1):308-310.
  14. Koçak AY, Akay BN, Heper AO. Sildenafil-induced pigmented purpuric dermatosis. Cutan Ocul Toxicol. 2013;32:91-92.
  15. Nishioka K, Sarashi C, Katayama I. Chronic pigmented purpura induced by chemical substances. Clin Exp Dermatol. 1980;5:213-218.
  16. Drlica K, Zhao X. DNA gyrase, topoisomerase IV, and the 4-quinolones. Microbiol Mol Biol Rev. 1997;61:377-392.
  17. Rubegni P, Feci L, Pellegrino M, et al. Photolocalized purpura during levofloxacin therapy. Photodermatol Photoimmunol Photomed. 2012;28:105-107.
  18. Sardana K, Sarkar R, Sehgal VN. Pigmented purpuric dermatoses: an overview. Int J Dermatol. 2004;43:482-488.
  19. Fathy H, Abdelgaber S. Treatment of pigmented purpuric dermatoses with narrow-band UVB: a report of six cases. J Eur Acad Dermatol Venereol. 2011;25:603-606.
  20. Krizsa J, Hunyadi J, Dobozy A. PUVA treatment of pigmented purpuric lichenoid dermatitis (Gougerot-Blum). J Am Acad Dermatol. 1992;27(5, pt 1):778-780.
  21. Panda S, Malakar S, Lahiri K. Oral pentoxifylline vs topical betamethasone in Schamberg disease: a comparative randomized investigator-blinded parallel-group trial. Arch Dermatol. 2004;140:491-493.
  22. Tamaki K, Yasaka N, Osada A, et al. Successful treatment of pigmented purpuric dermatosis with griseofulvin. Br J Dermatol. 1995;132:159-160.
  23. Geller M. Benefit of colchicine in the treatment of Schamberg’s disease. Ann Allergy Asthma Immunol. 2000;85:246.
  24. Okada K, Ishikawa O, Miyachi Y. Purpura pigmentosa chronica successfully treated with oral cyclosporin A. Br J Dermatol. 1996;134:180-181.
  25. Reinhold U, Seiter S, Ugurel S, et al. Treatment of progressive pigmented purpura with oral bioflavonoids and ascorbic acid: an open pilot study in 3 patients. J Am Acad Dermatol. 1999;41(2, pt 1):207-208.
  26. Wang A, Shuja F, Chan A, et al. Unilateral purpura annularis telangiectodes of Majocchi in an elderly male: an atypical presentation. Dermatol Online J. 2013;19:19263.
References
  1. Hale EK. Purpura annularis telangiectodes of Majocchi. Dermatol Online J. 2003;9:17.
  2. Hoesly FJ, Huerter CJ, Shehan JM. Purpura annularis telangiectodes of Majocchi: case report and review of the literature. Int J Dermatol. 2009;48:1129-1133.
  3. Kaplan R, Meehan SA, Leger M. A case of isotretinoin-induced purpura annularis telangiectodes of Majocchi and review of substance-induced pigmented purpuric dermatosis. JAMA Dermatol. 2014;150:182-184.
  4. Newton RC, Raimer SS. Pigmented purpuric eruptions. Dermatol Clin. 1985;3:165-169.
  5. Ratnam KV, Su WP, Peters MS. Purpura simplex (inflammatory purpura without vasculitis): a clinicopathologic study of 174 cases. J Am Acad Dermatol. 1991;25:642-647.
  6. Pang BK, Su D, Ratnam KV. Drug-induced purpura simplex: clinical and histological characteristics. Ann Acad Med Singapore. 1993;22:870-872.
  7. Abeck D, Gross GE, Kuwert C, et al. Acetaminophen-induced progressive pigmentary purpura (Schamberg’s disease). J Am Acad Dermatol. 1992;27:123-124.
  8. Lipsker D, Cribier B, Heid E, et al. Cutaneous lymphoma manifesting as pigmented, purpuric capillaries [in French]. Ann Dermatol Venereol. 1999;126:321-326.
  9. Peterson WC Jr, Manick KP. Purpuric eruptions associated with use of carbromal and meprobamate. Arch Dermatol. 1967;95:40-42.
  10. Nishioka K, Katayama I, Masuzawa M, et al. Drug-induced chronic pigmented purpura. J Dermatol. 1989;16:220-222.
  11. Voelter WW. Pigmented purpuric dermatosis-like reaction to topical fluorouracil. Arch Dermatol. 1983;119:875-876.
  12. Adams BB, Gadenne AS. Glipizide-induced pigmented purpuric dermatosis. J Am Acad Dermatol. 1999;41(5, pt 2):827-829.
  13. Tsao H, Lerner LH. Pigmented purpuric eruption associated with injection medroxyprogesterone acetate. J Am Acad Dermatol. 2000;43(2, pt 1):308-310.
  14. Koçak AY, Akay BN, Heper AO. Sildenafil-induced pigmented purpuric dermatosis. Cutan Ocul Toxicol. 2013;32:91-92.
  15. Nishioka K, Sarashi C, Katayama I. Chronic pigmented purpura induced by chemical substances. Clin Exp Dermatol. 1980;5:213-218.
  16. Drlica K, Zhao X. DNA gyrase, topoisomerase IV, and the 4-quinolones. Microbiol Mol Biol Rev. 1997;61:377-392.
  17. Rubegni P, Feci L, Pellegrino M, et al. Photolocalized purpura during levofloxacin therapy. Photodermatol Photoimmunol Photomed. 2012;28:105-107.
  18. Sardana K, Sarkar R, Sehgal VN. Pigmented purpuric dermatoses: an overview. Int J Dermatol. 2004;43:482-488.
  19. Fathy H, Abdelgaber S. Treatment of pigmented purpuric dermatoses with narrow-band UVB: a report of six cases. J Eur Acad Dermatol Venereol. 2011;25:603-606.
  20. Krizsa J, Hunyadi J, Dobozy A. PUVA treatment of pigmented purpuric lichenoid dermatitis (Gougerot-Blum). J Am Acad Dermatol. 1992;27(5, pt 1):778-780.
  21. Panda S, Malakar S, Lahiri K. Oral pentoxifylline vs topical betamethasone in Schamberg disease: a comparative randomized investigator-blinded parallel-group trial. Arch Dermatol. 2004;140:491-493.
  22. Tamaki K, Yasaka N, Osada A, et al. Successful treatment of pigmented purpuric dermatosis with griseofulvin. Br J Dermatol. 1995;132:159-160.
  23. Geller M. Benefit of colchicine in the treatment of Schamberg’s disease. Ann Allergy Asthma Immunol. 2000;85:246.
  24. Okada K, Ishikawa O, Miyachi Y. Purpura pigmentosa chronica successfully treated with oral cyclosporin A. Br J Dermatol. 1996;134:180-181.
  25. Reinhold U, Seiter S, Ugurel S, et al. Treatment of progressive pigmented purpura with oral bioflavonoids and ascorbic acid: an open pilot study in 3 patients. J Am Acad Dermatol. 1999;41(2, pt 1):207-208.
  26. Wang A, Shuja F, Chan A, et al. Unilateral purpura annularis telangiectodes of Majocchi in an elderly male: an atypical presentation. Dermatol Online J. 2013;19:19263.
Issue
Cutis - 100(4)
Issue
Cutis - 100(4)
Page Number
E10-E12
Page Number
E10-E12
Publications
Publications
Topics
Article Type
Display Headline
Levofloxacin-Induced Purpura Annularis Telangiectodes of Majocchi
Display Headline
Levofloxacin-Induced Purpura Annularis Telangiectodes of Majocchi
Sections
Inside the Article

Practice Point

  • Purpura annularis telangiectodes of Majocchi, a type of pigmented purpuric dermatosis, may on occasion be triggered by a medication; therefore, a careful medication history may prove to be an important part of the workup for this eruption.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article PDF Media
Image
Teambase ID
18000BCB.SIG
Disable zoom
Off

Antiphospholipid Syndrome in a Patient With Rheumatoid Arthritis

Article Type
Changed
Thu, 01/10/2019 - 13:39
Display Headline
Antiphospholipid Syndrome in a Patient With Rheumatoid Arthritis

Case Report

A 39-year-old woman with a 20-year history of rheumatoid arthritis (RA) presented to a university-affiliated tertiary care hospital with painful ulcerations on the bilateral dorsal feet that started as bullae 16 weeks prior to presentation. Initial skin biopsy performed by an outside dermatologist 8 weeks prior to presentation showed vasculitis and culture was positive for methicillin-sensitive Staphylococcus aureus. She was started on a prednisone taper and cephalexin, which did not improve the lower extremity ulcerations and the pain became progressively worse. At the time of presentation to our dermatology department, the patient was taking prednisone, hydroxychloroquine, hydrocodone-acetaminophen, and gabapentin. Prior therapy with sulfasalazine failed; etanercept and methotrexate were discontinued years prior due to side effects. The patient had no history of deep vein thrombosis, pulmonary embolism, or miscarriage.

At presentation, the patient was afebrile and her vital signs were stable. Physical examination showed multiple ulcers and erosions on the bilateral dorsal feet with a few scattered retiform red-purple patches (Figure). One bulla was present on the right dorsal foot. All lesions were tender to the touch and edema was present on the bilateral feet. No oral ulcerations were present and no focal neuropathies or palpable cords were appreciated in the lower extremities. There were no other cutaneous abnormalities.

ct099003021_e_fig.png
Multiple ulcers and erosions on the bilateral dorsal feet (A) and a bulla on the right dorsal foot (B).

Laboratory studies showed a white blood cell count of 9.54×103/µL (reference range, 4.16-9.95×103/µL), hemoglobin count of 12.4 g/dL (reference range, 11.6-15.2 g/dL), and a platelet count of 175×103/µL (reference range, 143-398×103/µL). A basic metabolic panel was normal except for an elevated glucose level of 185 mg/dL (reference range, 65-100 mg/dL). Urinalysis was normal. Erythrocyte sedimentation rate and C-reactive protein level were not elevated. Antinuclear antibodies and double-stranded DNA antibodies were normal. Prothrombin time was 10.4 seconds (reference range, 9.2-11.5 seconds) and dilute viper's venom time was negative. Rheumatoid factor level was elevated at 76 IU/mL (reference range, <25 IU/mL) and anti-citrullinated peptide antibody was moderately elevated at 42 U/mL (negative, <20 U/mL; weak positive, 20-39 U/mL; moderate positive, 40-59 U/mL; strong positive, >59 U/mL). The cardiolipin antibodies IgG, IgM, and IgA were within reference range. Results of β2-glycoprotein I IgG and IgM antibody tests were normal, but IgA was elevated at 34 µg/mL (reference range, <20 µg/mL). Wound cultures grew moderate Enterobacter cloacae and Staphylococcus lugdunensis.

Slides from 2 prior punch biopsies obtained by an outside hospital approximately 8 weeks prior from the right and left dorsal foot lesions were reviewed. Both biopsies were histologically similar. Postcapillary venules showed extensive vasculitis with numerous fibrin thrombi in the lumens in both biopsy specimens. The biopsy from the right foot showed prominent ulceration of the epidermis, with a few of the affected vessels showing minimal accompanying nuclear dust; however, the predominant pattern was not that of leukocytoclastic vasculitis. Biopsy from the left foot showed prominent epidermal necrosis with focal reepithelialization and scattered eosinophils. The pathologist felt that a vasculitis secondary to coagulopathy was most likely but that a drug reaction and rheumatoid vasculitis would be other entities to consider in the differential. A review of the laboratory findings from the outside hospital from approximately 12 weeks prior to presentation showed IgM was normal but IgG was elevated at 28 U/mL (reference range, 0-15 U/mL) and IgA was elevated at 8 U/mL (reference range, 0-7 U/mL); β2-glycoprotein I IgG antibodies were elevated at 37 mg/dL (reference range, 0-25.0 mg/dL) and β2-glycoprotein I IgA antibodies were elevated at 5 mg/dL (reference range, 0-4.0 mg/dL).

 

 

The clinical suspicion of a thrombotic event on the dorsal feet, which was confirmed histologically, and the persistently positive antiphospholipid (aPL) antibody titers helped to establish the diagnosis of antiphospholipid syndrome (APS) in the setting of RA. The dose of prednisone was increased from 10 mg daily on admission to 40 mg daily. The patient was started on enoxaparin 60 mg subcutaneously twice daily at initial presentation and was bridged to oral warfarin 2 mg daily after the diagnosis of APS was established. Oral doxycycline 100 mg twice daily was started for wound infection. The ulcerations gradually improved over the course of her 7-day hospitalization. She was continued on prednisone, hydroxychloroquine, and warfarin as an outpatient and has had no recurrence of lesions after 3 years of follow-up on this regimen.

Comment

Antiphospholipid syndrome is an autoimmune condition defined by a venous and/or arterial thrombotic event and/or pregnancy morbidity in the presence of persistently elevated aPL antibody titers. The most frequently detected subgroups of aPL are anticardiolipin (aCL) antibodies, anti-β2-glycoprotein I antibodies, and lupus anticoagulants.1 Primary APS occurs as an isolated entity, whereas secondary APS occurs in the setting of a preexisting autoimmune disease, infection, malignancy, or medication.2 The diagnostic criteria for APS requires positive aPL titers at least 12 weeks apart and a clinically confirmed thrombotic event or pregnancy morbidity.3

About one-third to half of patients with APS exhibit cutaneous manifestations.4,5 Livedo reticularis is most commonly observed and represents the first clinical sign of APS in 17.5% of cases.6 Cutaneous findings of APS also include anetoderma, cutaneous ulceration and necrosis, necrotizing vasculitis, livedoid vasculitis, thrombophlebitis, purpura, ecchymoses, painful skin nodules, and subungual hemorrhages.7 The various cutaneous manifestations of APS are associated with a range of histopathologic findings, but noninflammatory thrombosis in small arteries and/or veins in the dermis and subcutaneous fat tissue is the most common histologic feature.4 Our patient exhibited cutaneous ulceration and necrosis, and biopsy clearly showed the presence of vasculitis and fibrin thrombi within postcapillary venules. These findings along with the persistently elevated β2-glycoprotein I IgA solidified the diagnosis of APS.

The most common cutaneous manifestations of RA are nodules (32%), Raynaud phenomenon (10%), and vasculitis (3%).8 The mean prevalence of aPL antibodies in patients with RA is 28%, though reports range from 5% to 75%.1 The presence of aPL or aCL does not predict the development of thrombosis and/or thrombocytopenia in RA patients9,10; however, aCL antibodies in RA patients are associated with a higher risk for developing rheumatoid nodules. It is hypothesized that the majority of aCL antibodies identified in RA patients have different specificities than those identified in other diseases that are associated with thrombotic events.1

Anticoagulation has been proven to decrease the risk for recurrent thrombotic events in patients with APS.11 Patients should discontinue the use of estrogen-containing oral contraceptives; avoid smoking cigarettes; and treat hypertension, hyperlipidemia, and diabetes mellitus, if present. The type and duration of anticoagulation therapy, especially for the treatment of the cutaneous manifestations of APS, is less well defined. Antiplatelet therapies such as low-dose aspirin or dipyridamole often are used for less severe cutaneous manifestations such as livedoid vasculopathy. Warfarin with a target international normalized ratio of 2.0 to 3.0 is most commonly used following major thrombotic events, including cutaneous necrosis and digital gangrene. The role of corticosteroids and immunosuppressants is unclear; one study showed that these therapies did not prevent further thrombotic events in patients with systemic lupus erythematosus.4

Conclusion

Although aPL antibodies are most prevalent in patients with systemic lupus erythematosus, an estimated 28% of patients with RA have elevated aPL titers. The aPL antibodies recognized in RA patients are thought to have a different specificity than those recognized in other APS-associated diseases because elevated aPL antibody titers are not associated with an increased incidence of thrombotic events in RA patients; however, larger studies are needed to clarify this phenomenon. It remains to be determined if this case of APS and RA represents a coincidence or a true disease association, but the recognition of the cutaneous and histological features of APS is crucial for establishing a diagnosis and initiating anticoagulation therapy to prevent further morbidity and mortality.

References
  1. Olech E, Merrill JT. The prevalence and clinical significance of antiphospholipid antibodies in rheumatoid arthritis. Curr Rheumatol Rep. 2006;8:100-108.
  2. Thornsberry LA, LoSicco KI, English JC. The skin and hypercoagulable states. J Am Acad Dermatol. 2013;69:450-462.
  3. Miyakis S, Lockshin MD, Atsumi T, et al. International consensus statement on an update of the classification criteria for definite antiphospholipid syndrome (APS). J Thromb Haemost. 2006;4:295-306.
  4. Asherson A, Francès C, Iaccarino FL, et al. Theantiphospholipid antibody syndrome: diagnosis, skin manifestations and current therapy. Clin Exp Rheumatol. 2006;24(1 suppl 40):S46-S51.
  5. Cervera R, Piette JC, Font J, et al; Euro-Phospholipid Project Group. Antiphospholipid syndrome: clinical and immunologic manifestations and patterns of disease expression in a cohort of 1,000 patients. Arthritis Rheum. 2002;46:1019-1027.
  6. Francès C, Niang S, Laffitte E, et al. Dermatologic manifestations of antiphospholipid syndrome. two hundred consecutive cases. Arthritis Rheum. 2005;52:1785-1793.
  7. Gibson GE, Su WP, Pittelkow MR. Antiphospholipid syndrome and the skin. J Am Acad Dermatol. 1997;36(6, pt 1):970-982.
  8. Young A. Extra-articular manifestations and complications of rheumatoid arthritis. Best Pract Res Clin Rheumatol. 2007;21:907-927.
  9. Palomo I, Pinochet C, Alarcón M, et al. Prevalence of antiphospholipid antibodies in Chilean patients with rheumatoid arthritis. J Clin Lab Anal. 2006;20:190-194.
  10. Wolf P, Gretler J, Aglas F, et al. Anticardiolipin antibodies in rheumatoid arthritis: their relation to rheumatoid nodules and cutaneous vascular manifestations. Br J Dermatol. 1994;131:48-51.
  11. Lim W, Crowther MA, Eikelboom JW. Management of antiphospholipid antibody syndrome: a systematic review. JAMA. 2006;295:1050-1057.
Article PDF
Author and Disclosure Information

From the Department of Internal Medicine, Division of Dermatology, David Geffen School of Medicine, University of California, Los Angeles.

The authors report no conflict of interest.

Correspondence: Scott D. Worswick, MD, Division of Dermatology, David Geffen School of Medicine at UCLA, 200 Medical Plaza, Ste 450, Los Angeles, CA 90095 (sworswick@mednet.ucla.edu).

Issue
Cutis - 99(3)
Publications
Topics
Page Number
E21-E24
Sections
Author and Disclosure Information

From the Department of Internal Medicine, Division of Dermatology, David Geffen School of Medicine, University of California, Los Angeles.

The authors report no conflict of interest.

Correspondence: Scott D. Worswick, MD, Division of Dermatology, David Geffen School of Medicine at UCLA, 200 Medical Plaza, Ste 450, Los Angeles, CA 90095 (sworswick@mednet.ucla.edu).

Author and Disclosure Information

From the Department of Internal Medicine, Division of Dermatology, David Geffen School of Medicine, University of California, Los Angeles.

The authors report no conflict of interest.

Correspondence: Scott D. Worswick, MD, Division of Dermatology, David Geffen School of Medicine at UCLA, 200 Medical Plaza, Ste 450, Los Angeles, CA 90095 (sworswick@mednet.ucla.edu).

Article PDF
Article PDF
Related Articles

Case Report

A 39-year-old woman with a 20-year history of rheumatoid arthritis (RA) presented to a university-affiliated tertiary care hospital with painful ulcerations on the bilateral dorsal feet that started as bullae 16 weeks prior to presentation. Initial skin biopsy performed by an outside dermatologist 8 weeks prior to presentation showed vasculitis and culture was positive for methicillin-sensitive Staphylococcus aureus. She was started on a prednisone taper and cephalexin, which did not improve the lower extremity ulcerations and the pain became progressively worse. At the time of presentation to our dermatology department, the patient was taking prednisone, hydroxychloroquine, hydrocodone-acetaminophen, and gabapentin. Prior therapy with sulfasalazine failed; etanercept and methotrexate were discontinued years prior due to side effects. The patient had no history of deep vein thrombosis, pulmonary embolism, or miscarriage.

At presentation, the patient was afebrile and her vital signs were stable. Physical examination showed multiple ulcers and erosions on the bilateral dorsal feet with a few scattered retiform red-purple patches (Figure). One bulla was present on the right dorsal foot. All lesions were tender to the touch and edema was present on the bilateral feet. No oral ulcerations were present and no focal neuropathies or palpable cords were appreciated in the lower extremities. There were no other cutaneous abnormalities.

ct099003021_e_fig.png
Multiple ulcers and erosions on the bilateral dorsal feet (A) and a bulla on the right dorsal foot (B).

Laboratory studies showed a white blood cell count of 9.54×103/µL (reference range, 4.16-9.95×103/µL), hemoglobin count of 12.4 g/dL (reference range, 11.6-15.2 g/dL), and a platelet count of 175×103/µL (reference range, 143-398×103/µL). A basic metabolic panel was normal except for an elevated glucose level of 185 mg/dL (reference range, 65-100 mg/dL). Urinalysis was normal. Erythrocyte sedimentation rate and C-reactive protein level were not elevated. Antinuclear antibodies and double-stranded DNA antibodies were normal. Prothrombin time was 10.4 seconds (reference range, 9.2-11.5 seconds) and dilute viper's venom time was negative. Rheumatoid factor level was elevated at 76 IU/mL (reference range, <25 IU/mL) and anti-citrullinated peptide antibody was moderately elevated at 42 U/mL (negative, <20 U/mL; weak positive, 20-39 U/mL; moderate positive, 40-59 U/mL; strong positive, >59 U/mL). The cardiolipin antibodies IgG, IgM, and IgA were within reference range. Results of β2-glycoprotein I IgG and IgM antibody tests were normal, but IgA was elevated at 34 µg/mL (reference range, <20 µg/mL). Wound cultures grew moderate Enterobacter cloacae and Staphylococcus lugdunensis.

Slides from 2 prior punch biopsies obtained by an outside hospital approximately 8 weeks prior from the right and left dorsal foot lesions were reviewed. Both biopsies were histologically similar. Postcapillary venules showed extensive vasculitis with numerous fibrin thrombi in the lumens in both biopsy specimens. The biopsy from the right foot showed prominent ulceration of the epidermis, with a few of the affected vessels showing minimal accompanying nuclear dust; however, the predominant pattern was not that of leukocytoclastic vasculitis. Biopsy from the left foot showed prominent epidermal necrosis with focal reepithelialization and scattered eosinophils. The pathologist felt that a vasculitis secondary to coagulopathy was most likely but that a drug reaction and rheumatoid vasculitis would be other entities to consider in the differential. A review of the laboratory findings from the outside hospital from approximately 12 weeks prior to presentation showed IgM was normal but IgG was elevated at 28 U/mL (reference range, 0-15 U/mL) and IgA was elevated at 8 U/mL (reference range, 0-7 U/mL); β2-glycoprotein I IgG antibodies were elevated at 37 mg/dL (reference range, 0-25.0 mg/dL) and β2-glycoprotein I IgA antibodies were elevated at 5 mg/dL (reference range, 0-4.0 mg/dL).

 

 

The clinical suspicion of a thrombotic event on the dorsal feet, which was confirmed histologically, and the persistently positive antiphospholipid (aPL) antibody titers helped to establish the diagnosis of antiphospholipid syndrome (APS) in the setting of RA. The dose of prednisone was increased from 10 mg daily on admission to 40 mg daily. The patient was started on enoxaparin 60 mg subcutaneously twice daily at initial presentation and was bridged to oral warfarin 2 mg daily after the diagnosis of APS was established. Oral doxycycline 100 mg twice daily was started for wound infection. The ulcerations gradually improved over the course of her 7-day hospitalization. She was continued on prednisone, hydroxychloroquine, and warfarin as an outpatient and has had no recurrence of lesions after 3 years of follow-up on this regimen.

Comment

Antiphospholipid syndrome is an autoimmune condition defined by a venous and/or arterial thrombotic event and/or pregnancy morbidity in the presence of persistently elevated aPL antibody titers. The most frequently detected subgroups of aPL are anticardiolipin (aCL) antibodies, anti-β2-glycoprotein I antibodies, and lupus anticoagulants.1 Primary APS occurs as an isolated entity, whereas secondary APS occurs in the setting of a preexisting autoimmune disease, infection, malignancy, or medication.2 The diagnostic criteria for APS requires positive aPL titers at least 12 weeks apart and a clinically confirmed thrombotic event or pregnancy morbidity.3

About one-third to half of patients with APS exhibit cutaneous manifestations.4,5 Livedo reticularis is most commonly observed and represents the first clinical sign of APS in 17.5% of cases.6 Cutaneous findings of APS also include anetoderma, cutaneous ulceration and necrosis, necrotizing vasculitis, livedoid vasculitis, thrombophlebitis, purpura, ecchymoses, painful skin nodules, and subungual hemorrhages.7 The various cutaneous manifestations of APS are associated with a range of histopathologic findings, but noninflammatory thrombosis in small arteries and/or veins in the dermis and subcutaneous fat tissue is the most common histologic feature.4 Our patient exhibited cutaneous ulceration and necrosis, and biopsy clearly showed the presence of vasculitis and fibrin thrombi within postcapillary venules. These findings along with the persistently elevated β2-glycoprotein I IgA solidified the diagnosis of APS.

The most common cutaneous manifestations of RA are nodules (32%), Raynaud phenomenon (10%), and vasculitis (3%).8 The mean prevalence of aPL antibodies in patients with RA is 28%, though reports range from 5% to 75%.1 The presence of aPL or aCL does not predict the development of thrombosis and/or thrombocytopenia in RA patients9,10; however, aCL antibodies in RA patients are associated with a higher risk for developing rheumatoid nodules. It is hypothesized that the majority of aCL antibodies identified in RA patients have different specificities than those identified in other diseases that are associated with thrombotic events.1

Anticoagulation has been proven to decrease the risk for recurrent thrombotic events in patients with APS.11 Patients should discontinue the use of estrogen-containing oral contraceptives; avoid smoking cigarettes; and treat hypertension, hyperlipidemia, and diabetes mellitus, if present. The type and duration of anticoagulation therapy, especially for the treatment of the cutaneous manifestations of APS, is less well defined. Antiplatelet therapies such as low-dose aspirin or dipyridamole often are used for less severe cutaneous manifestations such as livedoid vasculopathy. Warfarin with a target international normalized ratio of 2.0 to 3.0 is most commonly used following major thrombotic events, including cutaneous necrosis and digital gangrene. The role of corticosteroids and immunosuppressants is unclear; one study showed that these therapies did not prevent further thrombotic events in patients with systemic lupus erythematosus.4

Conclusion

Although aPL antibodies are most prevalent in patients with systemic lupus erythematosus, an estimated 28% of patients with RA have elevated aPL titers. The aPL antibodies recognized in RA patients are thought to have a different specificity than those recognized in other APS-associated diseases because elevated aPL antibody titers are not associated with an increased incidence of thrombotic events in RA patients; however, larger studies are needed to clarify this phenomenon. It remains to be determined if this case of APS and RA represents a coincidence or a true disease association, but the recognition of the cutaneous and histological features of APS is crucial for establishing a diagnosis and initiating anticoagulation therapy to prevent further morbidity and mortality.

Case Report

A 39-year-old woman with a 20-year history of rheumatoid arthritis (RA) presented to a university-affiliated tertiary care hospital with painful ulcerations on the bilateral dorsal feet that started as bullae 16 weeks prior to presentation. Initial skin biopsy performed by an outside dermatologist 8 weeks prior to presentation showed vasculitis and culture was positive for methicillin-sensitive Staphylococcus aureus. She was started on a prednisone taper and cephalexin, which did not improve the lower extremity ulcerations and the pain became progressively worse. At the time of presentation to our dermatology department, the patient was taking prednisone, hydroxychloroquine, hydrocodone-acetaminophen, and gabapentin. Prior therapy with sulfasalazine failed; etanercept and methotrexate were discontinued years prior due to side effects. The patient had no history of deep vein thrombosis, pulmonary embolism, or miscarriage.

At presentation, the patient was afebrile and her vital signs were stable. Physical examination showed multiple ulcers and erosions on the bilateral dorsal feet with a few scattered retiform red-purple patches (Figure). One bulla was present on the right dorsal foot. All lesions were tender to the touch and edema was present on the bilateral feet. No oral ulcerations were present and no focal neuropathies or palpable cords were appreciated in the lower extremities. There were no other cutaneous abnormalities.

ct099003021_e_fig.png
Multiple ulcers and erosions on the bilateral dorsal feet (A) and a bulla on the right dorsal foot (B).

Laboratory studies showed a white blood cell count of 9.54×103/µL (reference range, 4.16-9.95×103/µL), hemoglobin count of 12.4 g/dL (reference range, 11.6-15.2 g/dL), and a platelet count of 175×103/µL (reference range, 143-398×103/µL). A basic metabolic panel was normal except for an elevated glucose level of 185 mg/dL (reference range, 65-100 mg/dL). Urinalysis was normal. Erythrocyte sedimentation rate and C-reactive protein level were not elevated. Antinuclear antibodies and double-stranded DNA antibodies were normal. Prothrombin time was 10.4 seconds (reference range, 9.2-11.5 seconds) and dilute viper's venom time was negative. Rheumatoid factor level was elevated at 76 IU/mL (reference range, <25 IU/mL) and anti-citrullinated peptide antibody was moderately elevated at 42 U/mL (negative, <20 U/mL; weak positive, 20-39 U/mL; moderate positive, 40-59 U/mL; strong positive, >59 U/mL). The cardiolipin antibodies IgG, IgM, and IgA were within reference range. Results of β2-glycoprotein I IgG and IgM antibody tests were normal, but IgA was elevated at 34 µg/mL (reference range, <20 µg/mL). Wound cultures grew moderate Enterobacter cloacae and Staphylococcus lugdunensis.

Slides from 2 prior punch biopsies obtained by an outside hospital approximately 8 weeks prior from the right and left dorsal foot lesions were reviewed. Both biopsies were histologically similar. Postcapillary venules showed extensive vasculitis with numerous fibrin thrombi in the lumens in both biopsy specimens. The biopsy from the right foot showed prominent ulceration of the epidermis, with a few of the affected vessels showing minimal accompanying nuclear dust; however, the predominant pattern was not that of leukocytoclastic vasculitis. Biopsy from the left foot showed prominent epidermal necrosis with focal reepithelialization and scattered eosinophils. The pathologist felt that a vasculitis secondary to coagulopathy was most likely but that a drug reaction and rheumatoid vasculitis would be other entities to consider in the differential. A review of the laboratory findings from the outside hospital from approximately 12 weeks prior to presentation showed IgM was normal but IgG was elevated at 28 U/mL (reference range, 0-15 U/mL) and IgA was elevated at 8 U/mL (reference range, 0-7 U/mL); β2-glycoprotein I IgG antibodies were elevated at 37 mg/dL (reference range, 0-25.0 mg/dL) and β2-glycoprotein I IgA antibodies were elevated at 5 mg/dL (reference range, 0-4.0 mg/dL).

 

 

The clinical suspicion of a thrombotic event on the dorsal feet, which was confirmed histologically, and the persistently positive antiphospholipid (aPL) antibody titers helped to establish the diagnosis of antiphospholipid syndrome (APS) in the setting of RA. The dose of prednisone was increased from 10 mg daily on admission to 40 mg daily. The patient was started on enoxaparin 60 mg subcutaneously twice daily at initial presentation and was bridged to oral warfarin 2 mg daily after the diagnosis of APS was established. Oral doxycycline 100 mg twice daily was started for wound infection. The ulcerations gradually improved over the course of her 7-day hospitalization. She was continued on prednisone, hydroxychloroquine, and warfarin as an outpatient and has had no recurrence of lesions after 3 years of follow-up on this regimen.

Comment

Antiphospholipid syndrome is an autoimmune condition defined by a venous and/or arterial thrombotic event and/or pregnancy morbidity in the presence of persistently elevated aPL antibody titers. The most frequently detected subgroups of aPL are anticardiolipin (aCL) antibodies, anti-β2-glycoprotein I antibodies, and lupus anticoagulants.1 Primary APS occurs as an isolated entity, whereas secondary APS occurs in the setting of a preexisting autoimmune disease, infection, malignancy, or medication.2 The diagnostic criteria for APS requires positive aPL titers at least 12 weeks apart and a clinically confirmed thrombotic event or pregnancy morbidity.3

About one-third to half of patients with APS exhibit cutaneous manifestations.4,5 Livedo reticularis is most commonly observed and represents the first clinical sign of APS in 17.5% of cases.6 Cutaneous findings of APS also include anetoderma, cutaneous ulceration and necrosis, necrotizing vasculitis, livedoid vasculitis, thrombophlebitis, purpura, ecchymoses, painful skin nodules, and subungual hemorrhages.7 The various cutaneous manifestations of APS are associated with a range of histopathologic findings, but noninflammatory thrombosis in small arteries and/or veins in the dermis and subcutaneous fat tissue is the most common histologic feature.4 Our patient exhibited cutaneous ulceration and necrosis, and biopsy clearly showed the presence of vasculitis and fibrin thrombi within postcapillary venules. These findings along with the persistently elevated β2-glycoprotein I IgA solidified the diagnosis of APS.

The most common cutaneous manifestations of RA are nodules (32%), Raynaud phenomenon (10%), and vasculitis (3%).8 The mean prevalence of aPL antibodies in patients with RA is 28%, though reports range from 5% to 75%.1 The presence of aPL or aCL does not predict the development of thrombosis and/or thrombocytopenia in RA patients9,10; however, aCL antibodies in RA patients are associated with a higher risk for developing rheumatoid nodules. It is hypothesized that the majority of aCL antibodies identified in RA patients have different specificities than those identified in other diseases that are associated with thrombotic events.1

Anticoagulation has been proven to decrease the risk for recurrent thrombotic events in patients with APS.11 Patients should discontinue the use of estrogen-containing oral contraceptives; avoid smoking cigarettes; and treat hypertension, hyperlipidemia, and diabetes mellitus, if present. The type and duration of anticoagulation therapy, especially for the treatment of the cutaneous manifestations of APS, is less well defined. Antiplatelet therapies such as low-dose aspirin or dipyridamole often are used for less severe cutaneous manifestations such as livedoid vasculopathy. Warfarin with a target international normalized ratio of 2.0 to 3.0 is most commonly used following major thrombotic events, including cutaneous necrosis and digital gangrene. The role of corticosteroids and immunosuppressants is unclear; one study showed that these therapies did not prevent further thrombotic events in patients with systemic lupus erythematosus.4

Conclusion

Although aPL antibodies are most prevalent in patients with systemic lupus erythematosus, an estimated 28% of patients with RA have elevated aPL titers. The aPL antibodies recognized in RA patients are thought to have a different specificity than those recognized in other APS-associated diseases because elevated aPL antibody titers are not associated with an increased incidence of thrombotic events in RA patients; however, larger studies are needed to clarify this phenomenon. It remains to be determined if this case of APS and RA represents a coincidence or a true disease association, but the recognition of the cutaneous and histological features of APS is crucial for establishing a diagnosis and initiating anticoagulation therapy to prevent further morbidity and mortality.

References
  1. Olech E, Merrill JT. The prevalence and clinical significance of antiphospholipid antibodies in rheumatoid arthritis. Curr Rheumatol Rep. 2006;8:100-108.
  2. Thornsberry LA, LoSicco KI, English JC. The skin and hypercoagulable states. J Am Acad Dermatol. 2013;69:450-462.
  3. Miyakis S, Lockshin MD, Atsumi T, et al. International consensus statement on an update of the classification criteria for definite antiphospholipid syndrome (APS). J Thromb Haemost. 2006;4:295-306.
  4. Asherson A, Francès C, Iaccarino FL, et al. Theantiphospholipid antibody syndrome: diagnosis, skin manifestations and current therapy. Clin Exp Rheumatol. 2006;24(1 suppl 40):S46-S51.
  5. Cervera R, Piette JC, Font J, et al; Euro-Phospholipid Project Group. Antiphospholipid syndrome: clinical and immunologic manifestations and patterns of disease expression in a cohort of 1,000 patients. Arthritis Rheum. 2002;46:1019-1027.
  6. Francès C, Niang S, Laffitte E, et al. Dermatologic manifestations of antiphospholipid syndrome. two hundred consecutive cases. Arthritis Rheum. 2005;52:1785-1793.
  7. Gibson GE, Su WP, Pittelkow MR. Antiphospholipid syndrome and the skin. J Am Acad Dermatol. 1997;36(6, pt 1):970-982.
  8. Young A. Extra-articular manifestations and complications of rheumatoid arthritis. Best Pract Res Clin Rheumatol. 2007;21:907-927.
  9. Palomo I, Pinochet C, Alarcón M, et al. Prevalence of antiphospholipid antibodies in Chilean patients with rheumatoid arthritis. J Clin Lab Anal. 2006;20:190-194.
  10. Wolf P, Gretler J, Aglas F, et al. Anticardiolipin antibodies in rheumatoid arthritis: their relation to rheumatoid nodules and cutaneous vascular manifestations. Br J Dermatol. 1994;131:48-51.
  11. Lim W, Crowther MA, Eikelboom JW. Management of antiphospholipid antibody syndrome: a systematic review. JAMA. 2006;295:1050-1057.
References
  1. Olech E, Merrill JT. The prevalence and clinical significance of antiphospholipid antibodies in rheumatoid arthritis. Curr Rheumatol Rep. 2006;8:100-108.
  2. Thornsberry LA, LoSicco KI, English JC. The skin and hypercoagulable states. J Am Acad Dermatol. 2013;69:450-462.
  3. Miyakis S, Lockshin MD, Atsumi T, et al. International consensus statement on an update of the classification criteria for definite antiphospholipid syndrome (APS). J Thromb Haemost. 2006;4:295-306.
  4. Asherson A, Francès C, Iaccarino FL, et al. Theantiphospholipid antibody syndrome: diagnosis, skin manifestations and current therapy. Clin Exp Rheumatol. 2006;24(1 suppl 40):S46-S51.
  5. Cervera R, Piette JC, Font J, et al; Euro-Phospholipid Project Group. Antiphospholipid syndrome: clinical and immunologic manifestations and patterns of disease expression in a cohort of 1,000 patients. Arthritis Rheum. 2002;46:1019-1027.
  6. Francès C, Niang S, Laffitte E, et al. Dermatologic manifestations of antiphospholipid syndrome. two hundred consecutive cases. Arthritis Rheum. 2005;52:1785-1793.
  7. Gibson GE, Su WP, Pittelkow MR. Antiphospholipid syndrome and the skin. J Am Acad Dermatol. 1997;36(6, pt 1):970-982.
  8. Young A. Extra-articular manifestations and complications of rheumatoid arthritis. Best Pract Res Clin Rheumatol. 2007;21:907-927.
  9. Palomo I, Pinochet C, Alarcón M, et al. Prevalence of antiphospholipid antibodies in Chilean patients with rheumatoid arthritis. J Clin Lab Anal. 2006;20:190-194.
  10. Wolf P, Gretler J, Aglas F, et al. Anticardiolipin antibodies in rheumatoid arthritis: their relation to rheumatoid nodules and cutaneous vascular manifestations. Br J Dermatol. 1994;131:48-51.
  11. Lim W, Crowther MA, Eikelboom JW. Management of antiphospholipid antibody syndrome: a systematic review. JAMA. 2006;295:1050-1057.
Issue
Cutis - 99(3)
Issue
Cutis - 99(3)
Page Number
E21-E24
Page Number
E21-E24
Publications
Publications
Topics
Article Type
Display Headline
Antiphospholipid Syndrome in a Patient With Rheumatoid Arthritis
Display Headline
Antiphospholipid Syndrome in a Patient With Rheumatoid Arthritis
Sections
Inside the Article

Practice Points

  • Antiphospholipid syndrome (APS) is an autoimmune condition defined by a venous and/or arterial thrombotic event and/or pregnancy morbidity in the presence of persistently elevated antiphospholipid antibody titers.
  • Cutaneous findings of APS include livedo reticularis most commonly but also anetoderma, cutaneous ulceration and necrosis, necrotizing vasculitis, livedoid vasculitis, thrombophlebitis, purpura, ecchymoses, painful skin nodules, and subungual hemorrhages.
  • The various cutaneous manifestations of APS are associated with a range of histopathologic findings, but noninflammatory thrombosis in small arteries and/or veins in the dermis and subcutaneous fat tissue is the most common histologic feature.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Image
Teambase ID
18000899.SIG
Disable zoom
Off