Central Centrifugal Cicatricial Alopecia in Males: Analysis of Time to Diagnosis and Disease Severity

Article Type
Changed
Fri, 06/14/2024 - 12:41
Display Headline
Central Centrifugal Cicatricial Alopecia in Males: Analysis of Time to Diagnosis and Disease Severity

To the Editor:

Central centrifugal cicatricial alopecia (CCCA) is a chronic progressive type of scarring alopecia that primarily affects women of African descent.1 The disorder rarely is reported in men, which may be due to misdiagnosis or delayed diagnosis. Early diagnosis and treatment are the cornerstones to slow or halt disease progression and prevent permanent damage to hair follicles. This study aimed to investigate the time to diagnosis and disease severity among males with CCCA.

We conducted a retrospective chart review of male patients older than 18 years seen in outpatient clinics at an academic dermatology department (Philadelphia, Pennsylvania) between January 2012 and December 2022. An electronic query using the International Classification of Diseases, Ninth and Tenth Revisions, code L66.9 (cicatricial alopecia, unspecified) was performed. Patients were included if they had a clinical diagnosis of CCCA, histologic evidence of CCCA, and scalp photographs from the initial dermatology visit. Patients with folliculitis decalvans, scalp biopsy features that limited characterization, or no scalp biopsy were excluded from the study. Onset of CCCA was defined as the patient-reported start time of hair loss and/or scalp symptoms. To determine alopecia severity, the degree of central scalp hair loss was independently assessed by 2 dermatologists (S.C.T., T.O.) using the central scalp alopecia photographic scale in African American women.2,3 This 6-point photographic scale displays images with grades ranging from 0 (normal) to 5 (bald scalp); higher grades indicate probable and more severe CCCA. The scale also divides the central hair loss in a frontal-accentuation or vertex-predominant pattern, which corresponds to the A or B designations, respectively; thus, a score of 5A indicates probable severe CCCA with a frontal accentuation pattern, while 5B indicates probable severe CCCA with hair loss focused on the vertex scalp. This study was approved by the University of Pennsylvania institutional review board (approval #850730).

Of 108 male patients, 12 met the eligibility criteria. Nearly all patients (91.7% [11/12]) had a CCCA severity grade of 3 or higher at the initial dermatology visit, indicating extensive hair loss (Table). The clinical appearance of severity grades 2 through 5 is demonstrated in the Figure. Among patients with a known disease duration prior to diagnosis, 72.7% (8/11) were diagnosed more than 1 year after onset of CCCA, and 45.4% (5/11) were diagnosed more than 5 years after onset. On average (SD), it took 6.4 (5.9) years for patients to receive a diagnosis of CCCA after the onset of scalp symptoms and/or hair loss.

Randomized controlled trials evaluating treatment of CCCA are lacking, and anecdotal evidence posits a better treatment response in early CCCA; however, our results suggest that most male patients present with advanced CCCA and receive a diagnosis years after disease onset. Similar research in alopecia areata has shown that 72.4% (105/145) of patients received their diagnosis within a year after onset of symptoms, and the mean time from onset of symptoms to diagnosis was 1 year.4 In contrast, male patients with CCCA experience considerable diagnostic delays. This disparity indicates the need for clinicians to increase recognition of CCCA in men and quickly refer them to a dermatologist for prompt treatment.

lafrucrehoshawovihathegotreclabusephabruclemavowrudecidijeshouogetrastipewracropemulepregusholetedafrunushudedechuuijocemaretopewrisiwujastaprustaclimuswaspouunekawoshisledicliclavicawastespome

hekochepruvagacewriducaswugagomopuchafranuhuvuuucrabedruclatostatoshistoswajapebresedocakicluhes
%3Cp%3EA%E2%80%93D%2C%20Clinical%20appearance%20of%20central%20centrifugal%20cicatricial%20alopecia%20grades%202A%2C%203A%2FB%2C%204B%2C%20and%205B%2C%20respectively%2C%20based%20on%20comparison%20of%20the%20patients%E2%80%99%20hair%20loss%20to%20the%20images%20in%20the%20scale.%3C%2Fp%3E

Androgenetic alopecia (AGA) commonly is at the top of the differential diagnosis for hair loss on the vertex of the scalp in males, but clinicians should maintain a high index of suspicion for CCCA, especially when scalp symptoms or atypical features of AGA are present.5 Androgenetic alopecia typically is asymptomatic, whereas the symptoms of CCCA may include itching, tenderness, and/or burning.6,7 Trichoscopy is useful to evaluate for scarring, and a scalp biopsy may reveal other features to lower AGA on the differential. Educating patients, barbers, and hairstylists about the importance of early intervention also may encourage earlier visits before the scarring process is advanced. Further exploration into factors impacting diagnosis and CCCA severity may uncover implications for prognosis and treatment.

This study was limited by a small sample size, retrospective design, and single-center analysis. Some patients had comorbid hair loss conditions, which could affect disease severity. Moreover, the central scalp alopecia photographic scale2 was not validated in men or designed for assessment of the nonclassical hair loss distributions noted in some of our patients. Nonetheless, we hope these data will support clinicians in efforts to advocate for early diagnosis and treatment in patients with CCCA to ultimately help improve outcomes.

References
  1. Ogunleye TA, McMichael A, Olsen EA. Central centrifugal cicatricial alopecia: what has been achieved, current clues for future research. Dermatol Clin. 2014;32:173-181. doi:10.1016/j.det.2013.12.005
  2. Olsen EA, Callender V, McMichael A, et al. Central hair loss in African American women: incidence and potential risk factors. J Am Acad Dermatol. 2011;64:245-252. doi:10.1016/j.jaad.2009.11.693
  3. Olsen EA, Callendar V, Sperling L, et al. Central scalp alopecia photographic scale in African American women. Dermatol Ther. 2008;21:264-267. doi:10.1111/j.1529-8019.2008.00208.x
  4. Andersen YMF, Nymand L, DeLozier AM, et al. Patient characteristics and disease burden of alopecia areata in the Danish Skin Cohort. BMJ Open. 2022;12:E053137. doi:10.1136/bmjopen-2021-053137
  5. Davis EC, Reid SD, Callender VD, et al. Differentiating central centrifugal cicatricial alopecia and androgenetic alopecia in African American men. J Clin Aesthetic Dermatol. 2012;5:37-40.
  6. Jackson TK, Sow Y, Ayoade KO, et al. Central centrifugal cicatricial alopecia in males. J Am Acad Dermatol. 2023;89:1136-1140. doi:10.1016/j.jaad.2023.07.1011
  7. Lawson CN, Bakayoko A, Callender VD. Central centrifugal cicatricial alopecia: challenges and treatments. Dermatol Clin. 2021;39:389-405. doi:10.1016/j.det.2021.03.004
Article PDF
Author and Disclosure Information

 

Dr. Jackson is from the University of Illinois College of Medicine, Peoria. Dr. Sow is from the Morehouse School of Medicine, Atlanta, Georgia. Drs. Taylor and Ogunleye are from the Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia.

Drs. Jackson, Sow, and Ogunleye report no conflicts of interest. Dr. Taylor is an advisory board member, consultant, employee, investigator, and/or speaker for AbbVie; Allergan Aesthetics; Arcutis Biotherapeutics, Inc; Armis Biopharma; Avita Medical; Beiersdorf, Inc; Biorez, Inc; Bristol-Myers Squibb; Cara Therapeutics; Catalyst Medical Education LLC; Concert Pharmaceuticals/Sun Pharma; Croma-Pharma GmbH; Dior; Eli Lilly and Company; EPI Health; Evolus, Inc; Galderma Laboratories; GloGetter; Hugel America, Inc; Incyte; Johnson & Johnson Consumer Products Company; L’Oreal USA; Mercer Strategies; Pfizer; Piction Health; Sanofi; Scientis US; UCB; and Vichy Laboratoires.

Correspondence: Temitayo Ogunleye, MD, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, 7th Floor PCAM South, Room 773, Philadelphia, PA 19104-5162 (temitayo.ogunleye@pennmedicine.upenn.edu).

Cutis. 2024 June;113(6):246-248. doi:10.12788/cutis.1031

Issue
Cutis - 113(6)
Publications
Topics
Page Number
246-248
Sections
Author and Disclosure Information

 

Dr. Jackson is from the University of Illinois College of Medicine, Peoria. Dr. Sow is from the Morehouse School of Medicine, Atlanta, Georgia. Drs. Taylor and Ogunleye are from the Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia.

Drs. Jackson, Sow, and Ogunleye report no conflicts of interest. Dr. Taylor is an advisory board member, consultant, employee, investigator, and/or speaker for AbbVie; Allergan Aesthetics; Arcutis Biotherapeutics, Inc; Armis Biopharma; Avita Medical; Beiersdorf, Inc; Biorez, Inc; Bristol-Myers Squibb; Cara Therapeutics; Catalyst Medical Education LLC; Concert Pharmaceuticals/Sun Pharma; Croma-Pharma GmbH; Dior; Eli Lilly and Company; EPI Health; Evolus, Inc; Galderma Laboratories; GloGetter; Hugel America, Inc; Incyte; Johnson & Johnson Consumer Products Company; L’Oreal USA; Mercer Strategies; Pfizer; Piction Health; Sanofi; Scientis US; UCB; and Vichy Laboratoires.

Correspondence: Temitayo Ogunleye, MD, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, 7th Floor PCAM South, Room 773, Philadelphia, PA 19104-5162 (temitayo.ogunleye@pennmedicine.upenn.edu).

Cutis. 2024 June;113(6):246-248. doi:10.12788/cutis.1031

Author and Disclosure Information

 

Dr. Jackson is from the University of Illinois College of Medicine, Peoria. Dr. Sow is from the Morehouse School of Medicine, Atlanta, Georgia. Drs. Taylor and Ogunleye are from the Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia.

Drs. Jackson, Sow, and Ogunleye report no conflicts of interest. Dr. Taylor is an advisory board member, consultant, employee, investigator, and/or speaker for AbbVie; Allergan Aesthetics; Arcutis Biotherapeutics, Inc; Armis Biopharma; Avita Medical; Beiersdorf, Inc; Biorez, Inc; Bristol-Myers Squibb; Cara Therapeutics; Catalyst Medical Education LLC; Concert Pharmaceuticals/Sun Pharma; Croma-Pharma GmbH; Dior; Eli Lilly and Company; EPI Health; Evolus, Inc; Galderma Laboratories; GloGetter; Hugel America, Inc; Incyte; Johnson & Johnson Consumer Products Company; L’Oreal USA; Mercer Strategies; Pfizer; Piction Health; Sanofi; Scientis US; UCB; and Vichy Laboratoires.

Correspondence: Temitayo Ogunleye, MD, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, 7th Floor PCAM South, Room 773, Philadelphia, PA 19104-5162 (temitayo.ogunleye@pennmedicine.upenn.edu).

Cutis. 2024 June;113(6):246-248. doi:10.12788/cutis.1031

Article PDF
Article PDF

To the Editor:

Central centrifugal cicatricial alopecia (CCCA) is a chronic progressive type of scarring alopecia that primarily affects women of African descent.1 The disorder rarely is reported in men, which may be due to misdiagnosis or delayed diagnosis. Early diagnosis and treatment are the cornerstones to slow or halt disease progression and prevent permanent damage to hair follicles. This study aimed to investigate the time to diagnosis and disease severity among males with CCCA.

We conducted a retrospective chart review of male patients older than 18 years seen in outpatient clinics at an academic dermatology department (Philadelphia, Pennsylvania) between January 2012 and December 2022. An electronic query using the International Classification of Diseases, Ninth and Tenth Revisions, code L66.9 (cicatricial alopecia, unspecified) was performed. Patients were included if they had a clinical diagnosis of CCCA, histologic evidence of CCCA, and scalp photographs from the initial dermatology visit. Patients with folliculitis decalvans, scalp biopsy features that limited characterization, or no scalp biopsy were excluded from the study. Onset of CCCA was defined as the patient-reported start time of hair loss and/or scalp symptoms. To determine alopecia severity, the degree of central scalp hair loss was independently assessed by 2 dermatologists (S.C.T., T.O.) using the central scalp alopecia photographic scale in African American women.2,3 This 6-point photographic scale displays images with grades ranging from 0 (normal) to 5 (bald scalp); higher grades indicate probable and more severe CCCA. The scale also divides the central hair loss in a frontal-accentuation or vertex-predominant pattern, which corresponds to the A or B designations, respectively; thus, a score of 5A indicates probable severe CCCA with a frontal accentuation pattern, while 5B indicates probable severe CCCA with hair loss focused on the vertex scalp. This study was approved by the University of Pennsylvania institutional review board (approval #850730).

Of 108 male patients, 12 met the eligibility criteria. Nearly all patients (91.7% [11/12]) had a CCCA severity grade of 3 or higher at the initial dermatology visit, indicating extensive hair loss (Table). The clinical appearance of severity grades 2 through 5 is demonstrated in the Figure. Among patients with a known disease duration prior to diagnosis, 72.7% (8/11) were diagnosed more than 1 year after onset of CCCA, and 45.4% (5/11) were diagnosed more than 5 years after onset. On average (SD), it took 6.4 (5.9) years for patients to receive a diagnosis of CCCA after the onset of scalp symptoms and/or hair loss.

Randomized controlled trials evaluating treatment of CCCA are lacking, and anecdotal evidence posits a better treatment response in early CCCA; however, our results suggest that most male patients present with advanced CCCA and receive a diagnosis years after disease onset. Similar research in alopecia areata has shown that 72.4% (105/145) of patients received their diagnosis within a year after onset of symptoms, and the mean time from onset of symptoms to diagnosis was 1 year.4 In contrast, male patients with CCCA experience considerable diagnostic delays. This disparity indicates the need for clinicians to increase recognition of CCCA in men and quickly refer them to a dermatologist for prompt treatment.

lafrucrehoshawovihathegotreclabusephabruclemavowrudecidijeshouogetrastipewracropemulepregusholetedafrunushudedechuuijocemaretopewrisiwujastaprustaclimuswaspouunekawoshisledicliclavicawastespome

hekochepruvagacewriducaswugagomopuchafranuhuvuuucrabedruclatostatoshistoswajapebresedocakicluhes
%3Cp%3EA%E2%80%93D%2C%20Clinical%20appearance%20of%20central%20centrifugal%20cicatricial%20alopecia%20grades%202A%2C%203A%2FB%2C%204B%2C%20and%205B%2C%20respectively%2C%20based%20on%20comparison%20of%20the%20patients%E2%80%99%20hair%20loss%20to%20the%20images%20in%20the%20scale.%3C%2Fp%3E

Androgenetic alopecia (AGA) commonly is at the top of the differential diagnosis for hair loss on the vertex of the scalp in males, but clinicians should maintain a high index of suspicion for CCCA, especially when scalp symptoms or atypical features of AGA are present.5 Androgenetic alopecia typically is asymptomatic, whereas the symptoms of CCCA may include itching, tenderness, and/or burning.6,7 Trichoscopy is useful to evaluate for scarring, and a scalp biopsy may reveal other features to lower AGA on the differential. Educating patients, barbers, and hairstylists about the importance of early intervention also may encourage earlier visits before the scarring process is advanced. Further exploration into factors impacting diagnosis and CCCA severity may uncover implications for prognosis and treatment.

This study was limited by a small sample size, retrospective design, and single-center analysis. Some patients had comorbid hair loss conditions, which could affect disease severity. Moreover, the central scalp alopecia photographic scale2 was not validated in men or designed for assessment of the nonclassical hair loss distributions noted in some of our patients. Nonetheless, we hope these data will support clinicians in efforts to advocate for early diagnosis and treatment in patients with CCCA to ultimately help improve outcomes.

To the Editor:

Central centrifugal cicatricial alopecia (CCCA) is a chronic progressive type of scarring alopecia that primarily affects women of African descent.1 The disorder rarely is reported in men, which may be due to misdiagnosis or delayed diagnosis. Early diagnosis and treatment are the cornerstones to slow or halt disease progression and prevent permanent damage to hair follicles. This study aimed to investigate the time to diagnosis and disease severity among males with CCCA.

We conducted a retrospective chart review of male patients older than 18 years seen in outpatient clinics at an academic dermatology department (Philadelphia, Pennsylvania) between January 2012 and December 2022. An electronic query using the International Classification of Diseases, Ninth and Tenth Revisions, code L66.9 (cicatricial alopecia, unspecified) was performed. Patients were included if they had a clinical diagnosis of CCCA, histologic evidence of CCCA, and scalp photographs from the initial dermatology visit. Patients with folliculitis decalvans, scalp biopsy features that limited characterization, or no scalp biopsy were excluded from the study. Onset of CCCA was defined as the patient-reported start time of hair loss and/or scalp symptoms. To determine alopecia severity, the degree of central scalp hair loss was independently assessed by 2 dermatologists (S.C.T., T.O.) using the central scalp alopecia photographic scale in African American women.2,3 This 6-point photographic scale displays images with grades ranging from 0 (normal) to 5 (bald scalp); higher grades indicate probable and more severe CCCA. The scale also divides the central hair loss in a frontal-accentuation or vertex-predominant pattern, which corresponds to the A or B designations, respectively; thus, a score of 5A indicates probable severe CCCA with a frontal accentuation pattern, while 5B indicates probable severe CCCA with hair loss focused on the vertex scalp. This study was approved by the University of Pennsylvania institutional review board (approval #850730).

Of 108 male patients, 12 met the eligibility criteria. Nearly all patients (91.7% [11/12]) had a CCCA severity grade of 3 or higher at the initial dermatology visit, indicating extensive hair loss (Table). The clinical appearance of severity grades 2 through 5 is demonstrated in the Figure. Among patients with a known disease duration prior to diagnosis, 72.7% (8/11) were diagnosed more than 1 year after onset of CCCA, and 45.4% (5/11) were diagnosed more than 5 years after onset. On average (SD), it took 6.4 (5.9) years for patients to receive a diagnosis of CCCA after the onset of scalp symptoms and/or hair loss.

Randomized controlled trials evaluating treatment of CCCA are lacking, and anecdotal evidence posits a better treatment response in early CCCA; however, our results suggest that most male patients present with advanced CCCA and receive a diagnosis years after disease onset. Similar research in alopecia areata has shown that 72.4% (105/145) of patients received their diagnosis within a year after onset of symptoms, and the mean time from onset of symptoms to diagnosis was 1 year.4 In contrast, male patients with CCCA experience considerable diagnostic delays. This disparity indicates the need for clinicians to increase recognition of CCCA in men and quickly refer them to a dermatologist for prompt treatment.

lafrucrehoshawovihathegotreclabusephabruclemavowrudecidijeshouogetrastipewracropemulepregusholetedafrunushudedechuuijocemaretopewrisiwujastaprustaclimuswaspouunekawoshisledicliclavicawastespome

hekochepruvagacewriducaswugagomopuchafranuhuvuuucrabedruclatostatoshistoswajapebresedocakicluhes
%3Cp%3EA%E2%80%93D%2C%20Clinical%20appearance%20of%20central%20centrifugal%20cicatricial%20alopecia%20grades%202A%2C%203A%2FB%2C%204B%2C%20and%205B%2C%20respectively%2C%20based%20on%20comparison%20of%20the%20patients%E2%80%99%20hair%20loss%20to%20the%20images%20in%20the%20scale.%3C%2Fp%3E

Androgenetic alopecia (AGA) commonly is at the top of the differential diagnosis for hair loss on the vertex of the scalp in males, but clinicians should maintain a high index of suspicion for CCCA, especially when scalp symptoms or atypical features of AGA are present.5 Androgenetic alopecia typically is asymptomatic, whereas the symptoms of CCCA may include itching, tenderness, and/or burning.6,7 Trichoscopy is useful to evaluate for scarring, and a scalp biopsy may reveal other features to lower AGA on the differential. Educating patients, barbers, and hairstylists about the importance of early intervention also may encourage earlier visits before the scarring process is advanced. Further exploration into factors impacting diagnosis and CCCA severity may uncover implications for prognosis and treatment.

This study was limited by a small sample size, retrospective design, and single-center analysis. Some patients had comorbid hair loss conditions, which could affect disease severity. Moreover, the central scalp alopecia photographic scale2 was not validated in men or designed for assessment of the nonclassical hair loss distributions noted in some of our patients. Nonetheless, we hope these data will support clinicians in efforts to advocate for early diagnosis and treatment in patients with CCCA to ultimately help improve outcomes.

References
  1. Ogunleye TA, McMichael A, Olsen EA. Central centrifugal cicatricial alopecia: what has been achieved, current clues for future research. Dermatol Clin. 2014;32:173-181. doi:10.1016/j.det.2013.12.005
  2. Olsen EA, Callender V, McMichael A, et al. Central hair loss in African American women: incidence and potential risk factors. J Am Acad Dermatol. 2011;64:245-252. doi:10.1016/j.jaad.2009.11.693
  3. Olsen EA, Callendar V, Sperling L, et al. Central scalp alopecia photographic scale in African American women. Dermatol Ther. 2008;21:264-267. doi:10.1111/j.1529-8019.2008.00208.x
  4. Andersen YMF, Nymand L, DeLozier AM, et al. Patient characteristics and disease burden of alopecia areata in the Danish Skin Cohort. BMJ Open. 2022;12:E053137. doi:10.1136/bmjopen-2021-053137
  5. Davis EC, Reid SD, Callender VD, et al. Differentiating central centrifugal cicatricial alopecia and androgenetic alopecia in African American men. J Clin Aesthetic Dermatol. 2012;5:37-40.
  6. Jackson TK, Sow Y, Ayoade KO, et al. Central centrifugal cicatricial alopecia in males. J Am Acad Dermatol. 2023;89:1136-1140. doi:10.1016/j.jaad.2023.07.1011
  7. Lawson CN, Bakayoko A, Callender VD. Central centrifugal cicatricial alopecia: challenges and treatments. Dermatol Clin. 2021;39:389-405. doi:10.1016/j.det.2021.03.004
References
  1. Ogunleye TA, McMichael A, Olsen EA. Central centrifugal cicatricial alopecia: what has been achieved, current clues for future research. Dermatol Clin. 2014;32:173-181. doi:10.1016/j.det.2013.12.005
  2. Olsen EA, Callender V, McMichael A, et al. Central hair loss in African American women: incidence and potential risk factors. J Am Acad Dermatol. 2011;64:245-252. doi:10.1016/j.jaad.2009.11.693
  3. Olsen EA, Callendar V, Sperling L, et al. Central scalp alopecia photographic scale in African American women. Dermatol Ther. 2008;21:264-267. doi:10.1111/j.1529-8019.2008.00208.x
  4. Andersen YMF, Nymand L, DeLozier AM, et al. Patient characteristics and disease burden of alopecia areata in the Danish Skin Cohort. BMJ Open. 2022;12:E053137. doi:10.1136/bmjopen-2021-053137
  5. Davis EC, Reid SD, Callender VD, et al. Differentiating central centrifugal cicatricial alopecia and androgenetic alopecia in African American men. J Clin Aesthetic Dermatol. 2012;5:37-40.
  6. Jackson TK, Sow Y, Ayoade KO, et al. Central centrifugal cicatricial alopecia in males. J Am Acad Dermatol. 2023;89:1136-1140. doi:10.1016/j.jaad.2023.07.1011
  7. Lawson CN, Bakayoko A, Callender VD. Central centrifugal cicatricial alopecia: challenges and treatments. Dermatol Clin. 2021;39:389-405. doi:10.1016/j.det.2021.03.004
Issue
Cutis - 113(6)
Issue
Cutis - 113(6)
Page Number
246-248
Page Number
246-248
Publications
Publications
Topics
Article Type
Display Headline
Central Centrifugal Cicatricial Alopecia in Males: Analysis of Time to Diagnosis and Disease Severity
Display Headline
Central Centrifugal Cicatricial Alopecia in Males: Analysis of Time to Diagnosis and Disease Severity
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>Jackson</fileName> <TBEID>0C02F7C0.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F7C0</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Jackson</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240614T091251</firstPublished> <LastPublished>20240614T091251</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240614T091250</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Tiaranesha Jackson, MD, MPH</byline> <bylineText>Tiaranesha Jackson, MD, MPH; Yacine Sow, MD; Susan C. Taylor, MD; Temitayo Ogunleye, MD</bylineText> <bylineFull>Tiaranesha Jackson, MD, MPH</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>246-248</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>To the Editor:Central centrifugal cicatricial alopecia (CCCA) is a chronic progressive type of scarring alopecia that primarily affects women of African descent</metaDescription> <articlePDF>301774</articlePDF> <teaserImage/> <title>Central Centrifugal Cicatricial Alopecia in Males: Analysis of Time to Diagnosis and Disease Severity</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>June</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>6</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2161</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>January 2019</pubIssueName> <pubArticleType>Original Articles | 2161</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">104</term> </sections> <topics> <term canonical="true">219</term> <term>66772</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002745.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Central Centrifugal Cicatricial Alopecia in Males: Analysis of Time to Diagnosis and Disease Severity</title> <deck/> </itemMeta> <itemContent> <p>To the Editor:<br/><br/>Central centrifugal cicatricial alopecia (CCCA) is a chronic progressive type of scarring alopecia that primarily affects women of African descent.<sup>1</sup> The disorder rarely is reported in men, which may be due to misdiagnosis or delayed diagnosis. Early diagnosis and treatment are the cornerstones to slow or halt disease progression and prevent permanent damage to hair follicles. This study aimed to investigate the time to diagnosis and disease severity among males with CCCA.</p> <p>We conducted a retrospective chart review of male patients older than 18 years seen in outpatient clinics at an academic dermatology department (Philadelphia, Pennsylvania) between January 2012 and December 2022. An electronic query using the <i>International Classification of Diseases, Ninth </i>and<i> Tenth Revisions</i>, code L66.9 (cicatricial alopecia, unspecified) was performed. Patients were included if they had a clinical diagnosis of CCCA, histologic evidence of CCCA, and scalp photographs from the initial dermatology visit. Patients with folliculitis decalvans, scalp biopsy features that limited characterization, or no scalp biopsy were excluded from the study. Onset of CCCA was defined as the patient-reported start time of hair loss and/or scalp symptoms. To determine alopecia severity, the degree of central scalp hair loss was independently assessed by 2 dermatologists (S.C.T., T.O.) using the central scalp alopecia photographic scale in African American women.<sup>2,3</sup> This 6-point photographic scale displays images with grades ranging from 0 (normal) to 5 (bald scalp); higher grades indicate probable and more severe CCCA. The scale also divides the central hair loss in a frontal-accentuation or vertex-predominant pattern, which corresponds to the A or B designations, respectively; thus, a score of 5A indicates probable severe CCCA with a frontal accentuation pattern, while 5B indicates probable severe CCCA with hair loss focused on the vertex scalp. This study was approved by the University of Pennsylvania institutional review board (approval #850730). </p> <p>Of 108 male patients, 12 met the eligibility criteria. Nearly all patients (91.7% [11/12]) had a CCCA severity grade of 3 or higher at the initial dermatology visit, indicating extensive hair loss (Table). The clinical appearance of severity grades 2 through 5 is demonstrated in the Figure. Among patients with a known disease duration prior to diagnosis, 72.7% (8/11) were diagnosed more than 1 year after onset of CCCA, and 45.4% (5/11) were diagnosed more than 5 years after onset. On average (SD), it took 6.4 (5.9) years for patients to receive a diagnosis of CCCA after the onset of scalp symptoms and/or hair loss. <br/><br/>Randomized controlled trials evaluating treatment of CCCA are lacking, and anecdotal evidence posits a better treatment response in early CCCA; however, our results suggest that most male patients present with advanced CCCA and receive a diagnosis years after disease onset. Similar research in alopecia areata has shown that 72.4% (105/145) of patients received their diagnosis within a year after onset of symptoms, and the mean time from onset of symptoms to diagnosis was 1 year.<sup>4</sup> In contrast, male patients with CCCA experience considerable diagnostic delays. This disparity indicates the need for clinicians to increase recognition of CCCA in men and quickly refer them to a dermatologist for prompt treatment. <br/><br/>Androgenetic alopecia (AGA) commonly is at the top of the differential diagnosis for hair loss on the vertex of the scalp in males, but clinicians should maintain a high index of suspicion for CCCA, especially when scalp symptoms or atypical features of AGA are present.<sup>5 </sup>Androgenetic alopecia typically is asymptomatic, whereas the symptoms of CCCA may include itching, tenderness, and/or burning.<sup>6,7</sup> Trichoscopy is useful to evaluate for scarring, and a scalp biopsy may reveal other features to lower AGA on the differential. Educating patients, barbers, and hairstylists about the importance of early intervention also may encourage earlier visits before the scarring process is advanced. Further exploration into factors impacting diagnosis and CCCA severity may uncover implications for prognosis and treatment. <br/><br/>This study was limited by a small sample size, retrospective design, and single-center analysis. Some patients had comorbid hair loss conditions, which could affect disease severity. Moreover, the central scalp alopecia photographic scale<sup>2</sup> was not validated in men or designed for assessment of the nonclassical hair loss distributions noted in some of our patients. Nonetheless, we hope these data will support clinicians in efforts to advocate for early diagnosis and treatment in patients with CCCA to ultimately help improve outcomes.</p> <h2>References</h2> <p class="reference"> 1. Ogunleye TA, McMichael A, Olsen EA. Central centrifugal cicatricial alopecia: what has been achieved, current clues for future research. <i>Dermatol Clin</i>. 2014;32:173-181. doi:10.1016/j.det.2013.12.005<br/><br/> 2. Olsen EA, Callender V, McMichael A, et al. Central hair loss in African American women: incidence and potential risk factors. <i>J Am Acad Dermatol</i>. 2011;64:245-252. doi:10.1016/j.jaad.2009.11.693<br/><br/> 3. Olsen EA, Callendar V, Sperling L, et al. Central scalp alopecia photographic scale in African American women. <i>Dermatol Ther</i>. 2008;21:264-267. doi:10.1111/j.1529-8019.2008.00208.x<br/><br/> 4. Andersen YMF, Nymand L, DeLozier AM, et al. Patient characteristics and disease burden of alopecia areata in the Danish Skin Cohort. <i>BMJ Open</i>. 2022;12:E053137. doi:10.1136/bmjopen-2021-053137<br/><br/> 5. Davis EC, Reid SD, Callender VD, et al. Differentiating central centrifugal cicatricial alopecia and androgenetic alopecia in African American men. <i>J Clin Aesthetic Dermatol</i>. 2012;5:37-40.<br/><br/> 6. Jackson TK, Sow Y, Ayoade KO, et al. Central centrifugal cicatricial alopecia in males. <i>J Am Acad Dermatol</i>. 2023;89:1136-1140. doi:10.1016/j.jaad.2023.07.1011<br/><br/> 7. Lawson CN, Bakayoko A, Callender VD. Central centrifugal cicatricial alopecia: challenges and treatments. <i>Dermatol Clin</i>. 2021;39:389-405. doi:10.1016/j.det.2021.03.004</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Dr. Jackson is from the University of Illinois College of Medicine, Peoria. Dr. Sow is from the Morehouse School of Medicine, Atlanta, Georgia. Drs. Taylor and Ogunleye are from the Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia.</p> <p class="disclosure">Drs. Jackson, Sow, and Ogunleye report no conflicts of interest. Dr. Taylor is an advisory board member, consultant, employee, investigator, and/or speaker for AbbVie; Allergan Aesthetics; Arcutis Biotherapeutics, Inc; Armis Biopharma; Avita Medical; Beiersdorf, Inc; Biorez, Inc; Bristol-Myers Squibb; Cara Therapeutics; Catalyst Medical Education LLC; Concert Pharmaceuticals/Sun Pharma; Croma-Pharma GmbH; Dior; Eli Lilly and Company; EPI Health; Evolus, Inc; Galderma Laboratories; GloGetter; Hugel America, Inc; Incyte; Johnson &amp; Johnson Consumer Products Company; L’Oreal USA; Mercer Strategies; Pfizer; Piction Health; Sanofi; Scientis US; UCB; and Vichy Laboratoires.<br/><br/>Correspondence: Temitayo Ogunleye, MD, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, 7th Floor PCAM South, Room 773, Philadelphia, PA 19104-5162 (temitayo.ogunleye@pennmedicine.upenn.edu).<br/><br/><em>Cutis. </em>2024 June;113(6):246-248. doi:10.12788/cutis.1031</p> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • Most males with central centrifugal cicatricial alopecia (CCCA) experience considerable diagnostic delays and typically present to dermatology with late-stage disease.
  • Dermatologists should consider CCCA in the differential diagnosis for adult Black males with alopecia.
  • More research is needed to explore advanced CCCA in males, including factors limiting timely diagnosis and the impact on quality of life in this population.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Disable zoom
Off

Overuse of Hematocrit Testing After Elective General Surgery at a Veterans Affairs Medical Center

Article Type
Changed
Mon, 06/10/2024 - 16:16

It is common practice to routinely measure postoperative hematocrit levels at US Department of Veterans Affairs (VA) hospitals for a wide range of elective general surgeries. While hematocrit measurement is a low-cost test, the high frequency with which these tests are performed may drastically increase overall costs.

Numerous studies have suggested that physicians overuse laboratory testing.1-10 Kohli and colleagues recommended that the routine practice of obtaining postoperative hematocrit tests following elective gynecologic surgery be abandoned.1 A similar recommendation was made by Olus and colleagues after studying uneventful, unplanned cesarean sections and by Wu and colleagues after investigating routine laboratory tests post total hip arthroplasty.2,3

To our knowledge, a study assessing routine postoperative hematocrit testing in elective general surgery has not yet been conducted. Many laboratory tests ordered in the perioperative period are not indicated, including complete blood count (CBC), electrolytes, and coagulation studies.4 Based on the results of these studies, we expected that the routine measurement of postoperative hematocrit levels after elective general surgeries at VA medical centers would not be cost effective. A PubMed search for articles published from 1990 to 2023 using the search terms “hematocrit,” “hemoglobin,” “general,” “surgery,” “routine,” and “cost” or “cost-effectiveness,” suggests that the clinical usefulness of postoperative hematocrit testing has not been well studied in the general surgery setting. The purpose of this study was to determine the clinical utility and associated cost of measuring routine postoperative hematocrit levels in order to generate a guide as to when the practice is warranted following common elective general surgery.

 

Although gynecologic textbooks may describe recommendations of routine hematocrit checking after elective gynecologic operations, one has difficulty finding the same recommendations in general surgery textbooks.1 However, it is common practice for surgical residents and attending surgeons to routinely order hematocrit on postoperative day-1 to ensure that the operation did not result in unsuspected anemia that then would need treatment (either with fluids or a blood transfusion). Many other surgeons rely on clinical factors such as tachycardia, oliguria, or hypotension to trigger a hematocrit (and other laboratory) tests. Our hypothesis is that the latter group has chosen the most cost-effective and prudent practice. One problem with checking the hematocrit routinely, as with any other screening test, is what to do with an abnormal result, assuming an asymptomatic patient? If the postoperative hematocrit is lower than expected given the estimated blood loss (EBL), what is one to do?

 

 

Methods

This retrospective case-control study conducted at the New Mexico VA Health Care System (NMVAHCS) in Albuquerque compared data for patients who received transfusion within 72 hours of elective surgeries vs patients who did not. Patients who underwent elective general surgery from January 2011 through December 2014 were included. An elective general surgery was defined as surgery performed following an outpatient preoperative anesthesia evaluation ≥ 30 days prior to operation. Patients who underwent emergency operations, and those with baseline anemia (preoperative hematocrit < 30%), and those transfused > 72 hours after their operation were excluded. The NMVAHCSInstitutional Review Board approved this study (No. 15-H184).

A detailed record review was conducted to collect data on demographics and other preoperative risk factors, including age, sex, body mass index (BMI), race and ethnicity, cardiac and pulmonary comorbidities, tobacco use, alcohol intake, diabetes, American Society of Anesthesiologists Physical Status Classification, metabolic equivalent of task, hematologic conditions, and renal disease.

For each procedure, we recorded the type of elective general surgery performed, the diagnosis/indication, pre- and postoperative hemoglobin/hematocrit, intraoperative EBL, length of operation, surgical wound class, length of hospital stay (LOS), intensive care unit (ICU) status, number of hematocrit tests, cardiovascular risk of operation (defined by anesthesia assessment), presence or absence of malignancy, preoperative platelet count, albumin level, preoperative prothrombin time/activated partial thromboplastin time (aPTT), international normalized ratio (INR), hemoglobin A1c, and incidence of transfusion. Signs and symptoms of anemia were recorded as present if the postoperative vital signs suggested low intravascular volume (pulse > 120 beats/minute, systolic blood pressure < 90 mm Hg, or vasoactive medication requirement [per anesthesia postoperative note]) or if the patient reported or exhibited symptoms of dizziness or fatigue or evidence of clinically apparent bleeding (ie, hematoma formation). Laboratory charges for hematocrit tests and CBC at the NMAVAHCS were used to assess cost.11

gashiuimorevaphithacraslokopagosabetritresedapoclustaslapukafratracreburicrolashefruuastitrumogouimasloswotegucrotheboshiprobrimarerophapruhosutremospevofrouuvatradespogeslugelavecretiswileclesethishevekererecrucrogiroki

To stratify the transfusion risk, patients were distributed among 3 groups based on the following criteria: discharged home the same day as surgery; admitted but did not have postoperative hematocrit testing; and admitted and had postoperative hematocrit testing. We also stratified operations into low or high risk based on the risk for postoperative transfusion (Figure). Recognizing that the American College of Chest Physicians guidelines for perioperative management of antithrombotic therapy places bowel resection in a high-risk category, we designated a surgery as high risk when ≥ 2 patients in the transfusion group had that type of surgery over the 4 years of the study.12 Otherwise, the operations were deemed low risk.

 

Statistical Analysis

Numeric analysis used t tests and Binary and categorical variables used Fisher exact tests. P value ≤ .05 was considered statistically significant. SAS software was used for all statistical analyses.

 

 

Results

From 2011 through 2014, 1531 patients had elective general surgery at NMVAHCS. Twenty-two patients with preoperative anemia (hematocrit < 30%) and 1 patient who received a transfusion > 72 hours after the operation were excluded. Most elective operations (70%, n = 1075) were performed on an outpatient basis; none involved transfusion. Inguinal hernia repair was most common with 479 operations; 17 patients were treated inpatient of which 2 patients had routine postoperative hematocrit checks; (neither received transfusion). One patient with inguinal hernia surgery received transfusion without routine postoperative hematocrit monitoring.

Of 112 partial colon resections, 1 patient had a postoperative transfusion; and all but 3 received postoperative hematocrit monitoring. Nineteen patients undergoing partial colon resection had a clinical indication for postoperative hematocrit monitoring. None of the 5 patients with partial gastrectomy received a postoperative transfusion. Of 121 elective cholecystectomies, no patients had postoperative transfusion, whereas 34 had postoperative hematocrit monitoring; only 2 patients had a clinical reason for the hematocrit monitoring.

Of 430 elective inpatient operations, 12 received transfusions and 288 patients had ≥ 1 postoperative hematocrit test (67%). All hematocrit tests were requested by the attending surgeon, resident surgeon, or the surgical ICU team. Of the group that had postoperative hematocrit monitoring, there was an average of 4.4 postoperative hematocrit tests per patient (range, 1-44).

sweshoniuinegitheuicraswevoslefrokostashubopijaruwiwigouilucupophacrashefruswalecrefrifrithogutewebetethaheladraswepauurowiuadrofredreguphoprospophocla

There were 12 transfusions for inpatients (2.8%), which is similar to the findings of a recent study of VA general surgery (2.3%).13 Five of the 12 patients received intraoperative transfusions while 7 were transfused within 72 hours postoperation. All but 1 patient receiving transfusion had EBL > 199 mL (range, 5-3000; mean, 950 mL; median, 500 mL) and/or signs or symptoms of anemia or other indications for measurement of the postoperative hematocrit. There were no statistically significant differences in patients’ age, sex, BMI, or race and ethnicity between groups receiving and not receiving transfusion (Table 1).

rospedrijuhespaspikojejithuproclaclumitrerobowrokecespishucroswalirocrojeshotraleropuuostabuphocicahewabrikophadojabewresipuvahunodruwirochuchudobuhuphonefroswuu

When comparing the transfusion vs the nontransfusion groups (after excluding those with clinical preoperative anemia) the risk factors for transfusion included: relatively low mean preoperative hematocrit (mean, 36.9% vs 42.7%, respectively; P = .003), low postoperative hematocrit (mean, 30.2% vs 37.1%, respectively; P < .001), high EBL (mean, 844 mL vs 109 mL, respectively; P = .005), large infusion of intraoperative fluids (mean, 4625 mL vs 2505 mL, respectively; P = .005), longer duration of operation (mean, 397 min vs 183 min, respectively; P < .001), and longer LOS (mean, 14.5 d vs 4.9 d, respectively; P < .001) (Table 2). Similarly, we found an increased risk for transfusion with high/intermediate cardiovascular risk (vs low), any wound not classified as clean, ICU stay, and postoperative symptoms of anemia.

 

We found no increased risk for transfusion with ethanol, tobacco, warfarin, or clopidogrel use; polycythemia; thrombocytopenia; preoperative INR; preoperative aPTT; preoperative albumin; Hemoglobin A1c; or diabetes mellitus; or for operations performed for malignancy. Ten patients in the ICU received transfusion (5.8%) compared with 2 patients (0.8%) not admitted to the ICU.

Operations were deemed high risk when ≥ 2 of patients having that operation received transfusions within 72 hours of their operation. There were 15 abdominoperineal resections; 3 of these received transfusions (20%). There were 7 total abdominal colectomies; 3 of these received transfusions (43%). We therefore had 22 high-risk operations, 6 of which were transfused (27%).

 

 

Discussion

Routine measurement of postoperative hematocrit levels after elective general surgery at NMVAHCS was not necessary. There were 12 transfusions for inpatients (2.8%), which is similar to the findings of a recent study of VA general surgery (2.3%).13 We found that routine postoperative hematocrit measurements to assess anemia had little or no effect on clinical decision-making or clinical outcomes.

According to our results, 88% of initial hematocrit tests after elective partial colectomies could have been eliminated; only 32 of 146 patients demonstrated a clinical reason for postoperative hematocrit testing. Similarly, 36 of 40 postcholecystectomy hematocrit tests (90%) could have been eliminated had the surgeons relied on clinical signs indicating possible postoperative anemia (none were transfused). Excluding patients with major intraoperative blood loss (> 300 mL), only 29 of 288 (10%) patients who had postoperative hematocrit tests had a clinical indication for a postoperative hematocrit test (ie, symptoms of anemia and/or active bleeding). One patient with inguinal hernia surgery who received transfusion was taking an anticoagulant and had a clinically indicated hematocrit test for a large hematoma that eventually required reoperation.

Our study found that routine hematocrit checks may actually increase the risk that a patient would receive an unnecessary transfusion. For instance, one elderly patient, after a right colectomy, had 6 hematocrit levels while on a heparin drip and received transfusion despite being asymptomatic. His lowest hematocrit level prior to transfusion was 23.7%. This patient had a total of 18 hematocrit tests. His EBL was 350 mL and his first postoperative HCT level was 33.1%. In another instance, a patient undergoing abdominoperineal resection had a transfusion on postoperative day 1, despite being hypertensive, with a hematocrit that ranged from 26% before transfusion to 31% after the transfusion. These 2 cases illustrate what has been shown in a recent study: A substantial number of patients with colorectal cancer receive unnecessary transfusions.14 On the other hand, one ileostomy closure patient had 33 hematocrit tests, yet his initial postoperative hematocrit was 37%, and he never received a transfusion. With low-risk surgeries, clinical judgment should dictate when a postoperative hematocrit level is needed. This strategy would have eliminated 206 unnecessary initial postoperative hematocrit tests (72%), could have decreased the number of unnecessary transfusions, and would have saved NMVAHCS about $1600 annually.

Abdominoperineal resections and total abdominal colectomies accounted for a high proportion of transfusions in our study. Inpatient elective operations can be risk stratified and have routine hematocrit tests ordered for patients at high risk. The probability of transfusion was greater in high-risk vs low-risk surgeries; 27% (6 of 22 patients) vs 2% (6 of 408 patients), respectively (P < .001). Since 14 of the 22 patients undergoing high-risk operation already had clinical reasons for a postoperative hematocrit test, we only need to add the remaining 8 patients with high-risk operations to the 74 who had a clinical reason for a hematocrit test and conclude that 82 of 430 patients (19%) had a clinical reason for a hematocrit test, either from signs or symptoms of blood loss or because they were in a high-risk group.

 


While our elective general surgery cases may not represent many general surgery programs in the US and VA health care systems, we can extrapolate cost savings using the same cost analyses outlined by Kohli and colleagues.1 Assuming 1.9 million elective inpatient general surgeries per year in the United States with an average cost of $21 per CBC, the annual cost of universal postoperative hematocrit testing would be $40 million.11,15 If postoperative hematocrit testing were 70% consistent with our findings, the annual cost for hematocrit tests on 51% of the inpatient general surgeries would be approximately $20.4 million. A reduction in routine hematocrit testing to 25% of all inpatient general surgeries (vs our finding that 19% were deemed necessary) results in an annual savings of $30 million. This conservative estimate could be even higher since there were 4.4 hematocrit tests per patient; therefore, we have about $132 million in savings.

Assuming 181,384 elective VA inpatient general surgeries each year, costing $7.14 per CBC (the NMVAHCS cost), the VA could save $1.3 million annually. If postoperative HCT testing were 70% consistent with our findings, the annual cost for hematocrit tests on 50.4% of inpatient general surgery operations would be about $653,000. A reduction in routine hematocrit testing to 25% of all inpatient general surgeries (vs our 19%) results in annual VA savings of $330,000. This conservative estimate could be even higher since there were on average 4.4 hematocrit levels per patient; therefore, we estimate that annual savings for the VA of about $1.45 million.

 

 

Limitations

The retrospective chart review nature of this study may have led to selection bias. Only a small number of patients received a transfusion, which may have skewed the data. This study population comes from a single VA medical center; this patient population may not be reflective of other VA medical centers or the US population as a whole. Given that NMVAHCS does not perform hepatic, esophageal, pancreas, or transplant operations, the potential savings to both the US and the VA may be overestimated, but this could be studied in the future by VA medical centers that perform more complex operations.

 

Conclusions

This study found that over a 4-year period routine postoperative hematocrit tests for patients undergoing elective general surgery at a VA medical center were not necessary. General surgeons routinely order various pre- and postoperative laboratory tests despite their limited utility. Reduction in unneeded routine tests could result in notable savings to the VA without compromising quality of care.

Only general surgery patients undergoing operations that carry a high risk for needing a blood transfusion should have a routine postoperative hematocrit testing. In our study population, the chance of an elective colectomy, cholecystectomy, or hernia patient needing a transfusion was rare. This strategy could eliminate a considerable number of unnecessary blood tests and would potentially yield significant savings.

References

1. Kohli N, Mallipeddi PK, Neff JM, Sze EH, Roat TW. Routine hematocrit after elective gynecologic surgery. Obstet Gynecol. 2000;95(6 Pt 1):847-850. doi:10.1016/s0029-7844(00)00796-1

2. Olus A, Orhan, U, Murat A, et al. Do asymptomatic patients require routine hemoglobin testing following uneventful, unplanned cesarean sections? Arch Gynecol Obstet. 2010;281(2):195-199. doi:10.1007/s00404-009-1093-1

3. Wu XD, Zhu ZL, Xiao P, Liu JC, Wang JW, Huang W. Are routine postoperative laboratory tests necessary after primary total hip arthroplasty? J Arthroplasty. 2020;35(10):2892-2898. doi:10.1016/j.arth.2020.04.097

4. Kumar A, Srivastava U. Role of routine laboratory investigations in preoperative evaluation. J Anesthesiol Clin Pharmacol. 2011;27(2):174-179. doi:10.4103/0970-9185.81824

5. Aghajanian A, Grimes DA. Routine prothrombin time determination before elective gynecologic operations. Obstet Gynecol. 1991;78(5 Pt 1):837-839.

6. Ransom SB, McNeeley SG, Malone JM Jr. A cost-effectiveness evaluation of preoperative type-and-screen testing for vaginal hysterectomy. Am J Obstet Gynecol. 1996;175(5):1201-1203. doi:10.1016/s0002-9378(96)70028-5

7. Ransom SB, McNeeley SG, Hosseini RB. Cost-effectiveness of routine blood type and screen testing before elective laparoscopy. Obstet Gynecol. 1995;86(3):346-348. doi:10.1016/0029-7844(95)00187-V

<--pagebreak-->

8. Committee on Standards and Practice Parameters, Apfelbaum JL, Connis RT, et al. Practice advisory for preanesthesia evaluation: an updated report by the American Society of Anesthesiologists Task Force on Preanesthesia Evaluation. Anesthesiology. 2012;116(3):522-538. doi:10.1097/ALN.0b013e31823c1067

9. Weil IA, Seicean S, Neuhauser D, Schiltz NK, Seicean A. Use and utility of hemostatic screening in adults undergoing elective, non-cardiac surgery. PLoS One. 2015;10(12):e0139139. doi:10.1371/journal.pone.0139139

10. Wu WC, Schifftner TL, Henderson WG, et al. Preoperative hematocrit levels and postoperative outcomes in older patients undergoing non-cardiac surgery. JAMA. 2007;297(22):2481-2488. doi:10.1001/jama.297.22.2481

11. Healthcare Bluebook. Complete blood count (CBC) with differential. Accessed March 28, 2024. https://www.healthcarebluebook.com/page_ProcedureDetails.aspx?id=214&dataset=lab

12. Douketis JD, Spyropoulos AC, Murad MH, et al. Perioperative management of antithrombotic therapy: an American College of Chest Physicians Clinical Practice Guideline. Chest. 2022;162(5):e207-e243. doi:10.1016/j.chest.2022.07.025

13. Randall JA, Wagner KT, Brody F. Perioperative transfusions in veterans following noncardiac procedures. J Laparoendosc Adv Surg Tech A. 2023;33(10):923-931. doi:10.1089/lap. 2023.0307

14. Tartter PI, Barron DM. Unnecessary blood transfusions in elective colorectal cancer surgery. Transfusion. 1985;25(2):113-115. doi:10.1046/j.1537-2995.1985.25285169199.x

15. Steiner CA, Karaca Z, Moore BJ, Imshaug MC, Pickens G. Surgeries in hospital-based ambulatory surgery and hospital inpatient settings, 2014. Healthcare Cost and Utilization Project statistical brief #223. May 2017. Revised July 2020. Agency for Healthcare Research and Quality. Accessed February 26, 2024. https://hcup-us.ahrq.gov/reports/statbriefs/sb223-Ambulatory-Inpatient-Surgeries-2014.pdf

16. US Department of Veterans Affairs, National Surgery Office. Quarterly report: Q3 of fiscal year 2017. VISN operative complexity summary [Source not verified].

Article PDF
Author and Disclosure Information

Anthony Vigil, MDa,b; Taylor Parnall, MDc; Clifford Qualls, PhDa,b; Robert Glew, PhDb; Robin Osofsky, MDd; Micah Guess, RNa;  Lauren Mercer, MDb

Correspondence:  Anthony Vigil  (anthony.vigil@va.gov) 

aNew Mexico Veterans Affairs Health Care System, Albuquerque

bUniversity of New Mexico School of Medicine, Albuquerque

cHarbor-UCLA Medical Center, Torrance, California

dOregon Health and Science University, Portland

Author contributions

Study conception and design: Vigil, Taylor; acquisition of data: Vigil, Taylor, Geuss, Mercer; analysis and interpretation of data: Vigil, Taylor, Osofsky, Qualls; drafting of manuscript: Vigil, Taylor; critical revision: Glew.

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Ethics and consent

The New Mexico Veterans Affairs Health Care System Institutional Review Board approved this study (No. 15-H184).

Publications
Topics
Page Number
S2-S8
Sections
Author and Disclosure Information

Anthony Vigil, MDa,b; Taylor Parnall, MDc; Clifford Qualls, PhDa,b; Robert Glew, PhDb; Robin Osofsky, MDd; Micah Guess, RNa;  Lauren Mercer, MDb

Correspondence:  Anthony Vigil  (anthony.vigil@va.gov) 

aNew Mexico Veterans Affairs Health Care System, Albuquerque

bUniversity of New Mexico School of Medicine, Albuquerque

cHarbor-UCLA Medical Center, Torrance, California

dOregon Health and Science University, Portland

Author contributions

Study conception and design: Vigil, Taylor; acquisition of data: Vigil, Taylor, Geuss, Mercer; analysis and interpretation of data: Vigil, Taylor, Osofsky, Qualls; drafting of manuscript: Vigil, Taylor; critical revision: Glew.

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Ethics and consent

The New Mexico Veterans Affairs Health Care System Institutional Review Board approved this study (No. 15-H184).

Author and Disclosure Information

Anthony Vigil, MDa,b; Taylor Parnall, MDc; Clifford Qualls, PhDa,b; Robert Glew, PhDb; Robin Osofsky, MDd; Micah Guess, RNa;  Lauren Mercer, MDb

Correspondence:  Anthony Vigil  (anthony.vigil@va.gov) 

aNew Mexico Veterans Affairs Health Care System, Albuquerque

bUniversity of New Mexico School of Medicine, Albuquerque

cHarbor-UCLA Medical Center, Torrance, California

dOregon Health and Science University, Portland

Author contributions

Study conception and design: Vigil, Taylor; acquisition of data: Vigil, Taylor, Geuss, Mercer; analysis and interpretation of data: Vigil, Taylor, Osofsky, Qualls; drafting of manuscript: Vigil, Taylor; critical revision: Glew.

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Ethics and consent

The New Mexico Veterans Affairs Health Care System Institutional Review Board approved this study (No. 15-H184).

Article PDF
Article PDF

It is common practice to routinely measure postoperative hematocrit levels at US Department of Veterans Affairs (VA) hospitals for a wide range of elective general surgeries. While hematocrit measurement is a low-cost test, the high frequency with which these tests are performed may drastically increase overall costs.

Numerous studies have suggested that physicians overuse laboratory testing.1-10 Kohli and colleagues recommended that the routine practice of obtaining postoperative hematocrit tests following elective gynecologic surgery be abandoned.1 A similar recommendation was made by Olus and colleagues after studying uneventful, unplanned cesarean sections and by Wu and colleagues after investigating routine laboratory tests post total hip arthroplasty.2,3

To our knowledge, a study assessing routine postoperative hematocrit testing in elective general surgery has not yet been conducted. Many laboratory tests ordered in the perioperative period are not indicated, including complete blood count (CBC), electrolytes, and coagulation studies.4 Based on the results of these studies, we expected that the routine measurement of postoperative hematocrit levels after elective general surgeries at VA medical centers would not be cost effective. A PubMed search for articles published from 1990 to 2023 using the search terms “hematocrit,” “hemoglobin,” “general,” “surgery,” “routine,” and “cost” or “cost-effectiveness,” suggests that the clinical usefulness of postoperative hematocrit testing has not been well studied in the general surgery setting. The purpose of this study was to determine the clinical utility and associated cost of measuring routine postoperative hematocrit levels in order to generate a guide as to when the practice is warranted following common elective general surgery.

 

Although gynecologic textbooks may describe recommendations of routine hematocrit checking after elective gynecologic operations, one has difficulty finding the same recommendations in general surgery textbooks.1 However, it is common practice for surgical residents and attending surgeons to routinely order hematocrit on postoperative day-1 to ensure that the operation did not result in unsuspected anemia that then would need treatment (either with fluids or a blood transfusion). Many other surgeons rely on clinical factors such as tachycardia, oliguria, or hypotension to trigger a hematocrit (and other laboratory) tests. Our hypothesis is that the latter group has chosen the most cost-effective and prudent practice. One problem with checking the hematocrit routinely, as with any other screening test, is what to do with an abnormal result, assuming an asymptomatic patient? If the postoperative hematocrit is lower than expected given the estimated blood loss (EBL), what is one to do?

 

 

Methods

This retrospective case-control study conducted at the New Mexico VA Health Care System (NMVAHCS) in Albuquerque compared data for patients who received transfusion within 72 hours of elective surgeries vs patients who did not. Patients who underwent elective general surgery from January 2011 through December 2014 were included. An elective general surgery was defined as surgery performed following an outpatient preoperative anesthesia evaluation ≥ 30 days prior to operation. Patients who underwent emergency operations, and those with baseline anemia (preoperative hematocrit < 30%), and those transfused > 72 hours after their operation were excluded. The NMVAHCSInstitutional Review Board approved this study (No. 15-H184).

A detailed record review was conducted to collect data on demographics and other preoperative risk factors, including age, sex, body mass index (BMI), race and ethnicity, cardiac and pulmonary comorbidities, tobacco use, alcohol intake, diabetes, American Society of Anesthesiologists Physical Status Classification, metabolic equivalent of task, hematologic conditions, and renal disease.

For each procedure, we recorded the type of elective general surgery performed, the diagnosis/indication, pre- and postoperative hemoglobin/hematocrit, intraoperative EBL, length of operation, surgical wound class, length of hospital stay (LOS), intensive care unit (ICU) status, number of hematocrit tests, cardiovascular risk of operation (defined by anesthesia assessment), presence or absence of malignancy, preoperative platelet count, albumin level, preoperative prothrombin time/activated partial thromboplastin time (aPTT), international normalized ratio (INR), hemoglobin A1c, and incidence of transfusion. Signs and symptoms of anemia were recorded as present if the postoperative vital signs suggested low intravascular volume (pulse > 120 beats/minute, systolic blood pressure < 90 mm Hg, or vasoactive medication requirement [per anesthesia postoperative note]) or if the patient reported or exhibited symptoms of dizziness or fatigue or evidence of clinically apparent bleeding (ie, hematoma formation). Laboratory charges for hematocrit tests and CBC at the NMAVAHCS were used to assess cost.11

gashiuimorevaphithacraslokopagosabetritresedapoclustaslapukafratracreburicrolashefruuastitrumogouimasloswotegucrotheboshiprobrimarerophapruhosutremospevofrouuvatradespogeslugelavecretiswileclesethishevekererecrucrogiroki

To stratify the transfusion risk, patients were distributed among 3 groups based on the following criteria: discharged home the same day as surgery; admitted but did not have postoperative hematocrit testing; and admitted and had postoperative hematocrit testing. We also stratified operations into low or high risk based on the risk for postoperative transfusion (Figure). Recognizing that the American College of Chest Physicians guidelines for perioperative management of antithrombotic therapy places bowel resection in a high-risk category, we designated a surgery as high risk when ≥ 2 patients in the transfusion group had that type of surgery over the 4 years of the study.12 Otherwise, the operations were deemed low risk.

 

Statistical Analysis

Numeric analysis used t tests and Binary and categorical variables used Fisher exact tests. P value ≤ .05 was considered statistically significant. SAS software was used for all statistical analyses.

 

 

Results

From 2011 through 2014, 1531 patients had elective general surgery at NMVAHCS. Twenty-two patients with preoperative anemia (hematocrit < 30%) and 1 patient who received a transfusion > 72 hours after the operation were excluded. Most elective operations (70%, n = 1075) were performed on an outpatient basis; none involved transfusion. Inguinal hernia repair was most common with 479 operations; 17 patients were treated inpatient of which 2 patients had routine postoperative hematocrit checks; (neither received transfusion). One patient with inguinal hernia surgery received transfusion without routine postoperative hematocrit monitoring.

Of 112 partial colon resections, 1 patient had a postoperative transfusion; and all but 3 received postoperative hematocrit monitoring. Nineteen patients undergoing partial colon resection had a clinical indication for postoperative hematocrit monitoring. None of the 5 patients with partial gastrectomy received a postoperative transfusion. Of 121 elective cholecystectomies, no patients had postoperative transfusion, whereas 34 had postoperative hematocrit monitoring; only 2 patients had a clinical reason for the hematocrit monitoring.

Of 430 elective inpatient operations, 12 received transfusions and 288 patients had ≥ 1 postoperative hematocrit test (67%). All hematocrit tests were requested by the attending surgeon, resident surgeon, or the surgical ICU team. Of the group that had postoperative hematocrit monitoring, there was an average of 4.4 postoperative hematocrit tests per patient (range, 1-44).

sweshoniuinegitheuicraswevoslefrokostashubopijaruwiwigouilucupophacrashefruswalecrefrifrithogutewebetethaheladraswepauurowiuadrofredreguphoprospophocla

There were 12 transfusions for inpatients (2.8%), which is similar to the findings of a recent study of VA general surgery (2.3%).13 Five of the 12 patients received intraoperative transfusions while 7 were transfused within 72 hours postoperation. All but 1 patient receiving transfusion had EBL > 199 mL (range, 5-3000; mean, 950 mL; median, 500 mL) and/or signs or symptoms of anemia or other indications for measurement of the postoperative hematocrit. There were no statistically significant differences in patients’ age, sex, BMI, or race and ethnicity between groups receiving and not receiving transfusion (Table 1).

rospedrijuhespaspikojejithuproclaclumitrerobowrokecespishucroswalirocrojeshotraleropuuostabuphocicahewabrikophadojabewresipuvahunodruwirochuchudobuhuphonefroswuu

When comparing the transfusion vs the nontransfusion groups (after excluding those with clinical preoperative anemia) the risk factors for transfusion included: relatively low mean preoperative hematocrit (mean, 36.9% vs 42.7%, respectively; P = .003), low postoperative hematocrit (mean, 30.2% vs 37.1%, respectively; P < .001), high EBL (mean, 844 mL vs 109 mL, respectively; P = .005), large infusion of intraoperative fluids (mean, 4625 mL vs 2505 mL, respectively; P = .005), longer duration of operation (mean, 397 min vs 183 min, respectively; P < .001), and longer LOS (mean, 14.5 d vs 4.9 d, respectively; P < .001) (Table 2). Similarly, we found an increased risk for transfusion with high/intermediate cardiovascular risk (vs low), any wound not classified as clean, ICU stay, and postoperative symptoms of anemia.

 

We found no increased risk for transfusion with ethanol, tobacco, warfarin, or clopidogrel use; polycythemia; thrombocytopenia; preoperative INR; preoperative aPTT; preoperative albumin; Hemoglobin A1c; or diabetes mellitus; or for operations performed for malignancy. Ten patients in the ICU received transfusion (5.8%) compared with 2 patients (0.8%) not admitted to the ICU.

Operations were deemed high risk when ≥ 2 of patients having that operation received transfusions within 72 hours of their operation. There were 15 abdominoperineal resections; 3 of these received transfusions (20%). There were 7 total abdominal colectomies; 3 of these received transfusions (43%). We therefore had 22 high-risk operations, 6 of which were transfused (27%).

 

 

Discussion

Routine measurement of postoperative hematocrit levels after elective general surgery at NMVAHCS was not necessary. There were 12 transfusions for inpatients (2.8%), which is similar to the findings of a recent study of VA general surgery (2.3%).13 We found that routine postoperative hematocrit measurements to assess anemia had little or no effect on clinical decision-making or clinical outcomes.

According to our results, 88% of initial hematocrit tests after elective partial colectomies could have been eliminated; only 32 of 146 patients demonstrated a clinical reason for postoperative hematocrit testing. Similarly, 36 of 40 postcholecystectomy hematocrit tests (90%) could have been eliminated had the surgeons relied on clinical signs indicating possible postoperative anemia (none were transfused). Excluding patients with major intraoperative blood loss (> 300 mL), only 29 of 288 (10%) patients who had postoperative hematocrit tests had a clinical indication for a postoperative hematocrit test (ie, symptoms of anemia and/or active bleeding). One patient with inguinal hernia surgery who received transfusion was taking an anticoagulant and had a clinically indicated hematocrit test for a large hematoma that eventually required reoperation.

Our study found that routine hematocrit checks may actually increase the risk that a patient would receive an unnecessary transfusion. For instance, one elderly patient, after a right colectomy, had 6 hematocrit levels while on a heparin drip and received transfusion despite being asymptomatic. His lowest hematocrit level prior to transfusion was 23.7%. This patient had a total of 18 hematocrit tests. His EBL was 350 mL and his first postoperative HCT level was 33.1%. In another instance, a patient undergoing abdominoperineal resection had a transfusion on postoperative day 1, despite being hypertensive, with a hematocrit that ranged from 26% before transfusion to 31% after the transfusion. These 2 cases illustrate what has been shown in a recent study: A substantial number of patients with colorectal cancer receive unnecessary transfusions.14 On the other hand, one ileostomy closure patient had 33 hematocrit tests, yet his initial postoperative hematocrit was 37%, and he never received a transfusion. With low-risk surgeries, clinical judgment should dictate when a postoperative hematocrit level is needed. This strategy would have eliminated 206 unnecessary initial postoperative hematocrit tests (72%), could have decreased the number of unnecessary transfusions, and would have saved NMVAHCS about $1600 annually.

Abdominoperineal resections and total abdominal colectomies accounted for a high proportion of transfusions in our study. Inpatient elective operations can be risk stratified and have routine hematocrit tests ordered for patients at high risk. The probability of transfusion was greater in high-risk vs low-risk surgeries; 27% (6 of 22 patients) vs 2% (6 of 408 patients), respectively (P < .001). Since 14 of the 22 patients undergoing high-risk operation already had clinical reasons for a postoperative hematocrit test, we only need to add the remaining 8 patients with high-risk operations to the 74 who had a clinical reason for a hematocrit test and conclude that 82 of 430 patients (19%) had a clinical reason for a hematocrit test, either from signs or symptoms of blood loss or because they were in a high-risk group.

 


While our elective general surgery cases may not represent many general surgery programs in the US and VA health care systems, we can extrapolate cost savings using the same cost analyses outlined by Kohli and colleagues.1 Assuming 1.9 million elective inpatient general surgeries per year in the United States with an average cost of $21 per CBC, the annual cost of universal postoperative hematocrit testing would be $40 million.11,15 If postoperative hematocrit testing were 70% consistent with our findings, the annual cost for hematocrit tests on 51% of the inpatient general surgeries would be approximately $20.4 million. A reduction in routine hematocrit testing to 25% of all inpatient general surgeries (vs our finding that 19% were deemed necessary) results in an annual savings of $30 million. This conservative estimate could be even higher since there were 4.4 hematocrit tests per patient; therefore, we have about $132 million in savings.

Assuming 181,384 elective VA inpatient general surgeries each year, costing $7.14 per CBC (the NMVAHCS cost), the VA could save $1.3 million annually. If postoperative HCT testing were 70% consistent with our findings, the annual cost for hematocrit tests on 50.4% of inpatient general surgery operations would be about $653,000. A reduction in routine hematocrit testing to 25% of all inpatient general surgeries (vs our 19%) results in annual VA savings of $330,000. This conservative estimate could be even higher since there were on average 4.4 hematocrit levels per patient; therefore, we estimate that annual savings for the VA of about $1.45 million.

 

 

Limitations

The retrospective chart review nature of this study may have led to selection bias. Only a small number of patients received a transfusion, which may have skewed the data. This study population comes from a single VA medical center; this patient population may not be reflective of other VA medical centers or the US population as a whole. Given that NMVAHCS does not perform hepatic, esophageal, pancreas, or transplant operations, the potential savings to both the US and the VA may be overestimated, but this could be studied in the future by VA medical centers that perform more complex operations.

 

Conclusions

This study found that over a 4-year period routine postoperative hematocrit tests for patients undergoing elective general surgery at a VA medical center were not necessary. General surgeons routinely order various pre- and postoperative laboratory tests despite their limited utility. Reduction in unneeded routine tests could result in notable savings to the VA without compromising quality of care.

Only general surgery patients undergoing operations that carry a high risk for needing a blood transfusion should have a routine postoperative hematocrit testing. In our study population, the chance of an elective colectomy, cholecystectomy, or hernia patient needing a transfusion was rare. This strategy could eliminate a considerable number of unnecessary blood tests and would potentially yield significant savings.

It is common practice to routinely measure postoperative hematocrit levels at US Department of Veterans Affairs (VA) hospitals for a wide range of elective general surgeries. While hematocrit measurement is a low-cost test, the high frequency with which these tests are performed may drastically increase overall costs.

Numerous studies have suggested that physicians overuse laboratory testing.1-10 Kohli and colleagues recommended that the routine practice of obtaining postoperative hematocrit tests following elective gynecologic surgery be abandoned.1 A similar recommendation was made by Olus and colleagues after studying uneventful, unplanned cesarean sections and by Wu and colleagues after investigating routine laboratory tests post total hip arthroplasty.2,3

To our knowledge, a study assessing routine postoperative hematocrit testing in elective general surgery has not yet been conducted. Many laboratory tests ordered in the perioperative period are not indicated, including complete blood count (CBC), electrolytes, and coagulation studies.4 Based on the results of these studies, we expected that the routine measurement of postoperative hematocrit levels after elective general surgeries at VA medical centers would not be cost effective. A PubMed search for articles published from 1990 to 2023 using the search terms “hematocrit,” “hemoglobin,” “general,” “surgery,” “routine,” and “cost” or “cost-effectiveness,” suggests that the clinical usefulness of postoperative hematocrit testing has not been well studied in the general surgery setting. The purpose of this study was to determine the clinical utility and associated cost of measuring routine postoperative hematocrit levels in order to generate a guide as to when the practice is warranted following common elective general surgery.

 

Although gynecologic textbooks may describe recommendations of routine hematocrit checking after elective gynecologic operations, one has difficulty finding the same recommendations in general surgery textbooks.1 However, it is common practice for surgical residents and attending surgeons to routinely order hematocrit on postoperative day-1 to ensure that the operation did not result in unsuspected anemia that then would need treatment (either with fluids or a blood transfusion). Many other surgeons rely on clinical factors such as tachycardia, oliguria, or hypotension to trigger a hematocrit (and other laboratory) tests. Our hypothesis is that the latter group has chosen the most cost-effective and prudent practice. One problem with checking the hematocrit routinely, as with any other screening test, is what to do with an abnormal result, assuming an asymptomatic patient? If the postoperative hematocrit is lower than expected given the estimated blood loss (EBL), what is one to do?

 

 

Methods

This retrospective case-control study conducted at the New Mexico VA Health Care System (NMVAHCS) in Albuquerque compared data for patients who received transfusion within 72 hours of elective surgeries vs patients who did not. Patients who underwent elective general surgery from January 2011 through December 2014 were included. An elective general surgery was defined as surgery performed following an outpatient preoperative anesthesia evaluation ≥ 30 days prior to operation. Patients who underwent emergency operations, and those with baseline anemia (preoperative hematocrit < 30%), and those transfused > 72 hours after their operation were excluded. The NMVAHCSInstitutional Review Board approved this study (No. 15-H184).

A detailed record review was conducted to collect data on demographics and other preoperative risk factors, including age, sex, body mass index (BMI), race and ethnicity, cardiac and pulmonary comorbidities, tobacco use, alcohol intake, diabetes, American Society of Anesthesiologists Physical Status Classification, metabolic equivalent of task, hematologic conditions, and renal disease.

For each procedure, we recorded the type of elective general surgery performed, the diagnosis/indication, pre- and postoperative hemoglobin/hematocrit, intraoperative EBL, length of operation, surgical wound class, length of hospital stay (LOS), intensive care unit (ICU) status, number of hematocrit tests, cardiovascular risk of operation (defined by anesthesia assessment), presence or absence of malignancy, preoperative platelet count, albumin level, preoperative prothrombin time/activated partial thromboplastin time (aPTT), international normalized ratio (INR), hemoglobin A1c, and incidence of transfusion. Signs and symptoms of anemia were recorded as present if the postoperative vital signs suggested low intravascular volume (pulse > 120 beats/minute, systolic blood pressure < 90 mm Hg, or vasoactive medication requirement [per anesthesia postoperative note]) or if the patient reported or exhibited symptoms of dizziness or fatigue or evidence of clinically apparent bleeding (ie, hematoma formation). Laboratory charges for hematocrit tests and CBC at the NMAVAHCS were used to assess cost.11

gashiuimorevaphithacraslokopagosabetritresedapoclustaslapukafratracreburicrolashefruuastitrumogouimasloswotegucrotheboshiprobrimarerophapruhosutremospevofrouuvatradespogeslugelavecretiswileclesethishevekererecrucrogiroki

To stratify the transfusion risk, patients were distributed among 3 groups based on the following criteria: discharged home the same day as surgery; admitted but did not have postoperative hematocrit testing; and admitted and had postoperative hematocrit testing. We also stratified operations into low or high risk based on the risk for postoperative transfusion (Figure). Recognizing that the American College of Chest Physicians guidelines for perioperative management of antithrombotic therapy places bowel resection in a high-risk category, we designated a surgery as high risk when ≥ 2 patients in the transfusion group had that type of surgery over the 4 years of the study.12 Otherwise, the operations were deemed low risk.

 

Statistical Analysis

Numeric analysis used t tests and Binary and categorical variables used Fisher exact tests. P value ≤ .05 was considered statistically significant. SAS software was used for all statistical analyses.

 

 

Results

From 2011 through 2014, 1531 patients had elective general surgery at NMVAHCS. Twenty-two patients with preoperative anemia (hematocrit < 30%) and 1 patient who received a transfusion > 72 hours after the operation were excluded. Most elective operations (70%, n = 1075) were performed on an outpatient basis; none involved transfusion. Inguinal hernia repair was most common with 479 operations; 17 patients were treated inpatient of which 2 patients had routine postoperative hematocrit checks; (neither received transfusion). One patient with inguinal hernia surgery received transfusion without routine postoperative hematocrit monitoring.

Of 112 partial colon resections, 1 patient had a postoperative transfusion; and all but 3 received postoperative hematocrit monitoring. Nineteen patients undergoing partial colon resection had a clinical indication for postoperative hematocrit monitoring. None of the 5 patients with partial gastrectomy received a postoperative transfusion. Of 121 elective cholecystectomies, no patients had postoperative transfusion, whereas 34 had postoperative hematocrit monitoring; only 2 patients had a clinical reason for the hematocrit monitoring.

Of 430 elective inpatient operations, 12 received transfusions and 288 patients had ≥ 1 postoperative hematocrit test (67%). All hematocrit tests were requested by the attending surgeon, resident surgeon, or the surgical ICU team. Of the group that had postoperative hematocrit monitoring, there was an average of 4.4 postoperative hematocrit tests per patient (range, 1-44).

sweshoniuinegitheuicraswevoslefrokostashubopijaruwiwigouilucupophacrashefruswalecrefrifrithogutewebetethaheladraswepauurowiuadrofredreguphoprospophocla

There were 12 transfusions for inpatients (2.8%), which is similar to the findings of a recent study of VA general surgery (2.3%).13 Five of the 12 patients received intraoperative transfusions while 7 were transfused within 72 hours postoperation. All but 1 patient receiving transfusion had EBL > 199 mL (range, 5-3000; mean, 950 mL; median, 500 mL) and/or signs or symptoms of anemia or other indications for measurement of the postoperative hematocrit. There were no statistically significant differences in patients’ age, sex, BMI, or race and ethnicity between groups receiving and not receiving transfusion (Table 1).

rospedrijuhespaspikojejithuproclaclumitrerobowrokecespishucroswalirocrojeshotraleropuuostabuphocicahewabrikophadojabewresipuvahunodruwirochuchudobuhuphonefroswuu

When comparing the transfusion vs the nontransfusion groups (after excluding those with clinical preoperative anemia) the risk factors for transfusion included: relatively low mean preoperative hematocrit (mean, 36.9% vs 42.7%, respectively; P = .003), low postoperative hematocrit (mean, 30.2% vs 37.1%, respectively; P < .001), high EBL (mean, 844 mL vs 109 mL, respectively; P = .005), large infusion of intraoperative fluids (mean, 4625 mL vs 2505 mL, respectively; P = .005), longer duration of operation (mean, 397 min vs 183 min, respectively; P < .001), and longer LOS (mean, 14.5 d vs 4.9 d, respectively; P < .001) (Table 2). Similarly, we found an increased risk for transfusion with high/intermediate cardiovascular risk (vs low), any wound not classified as clean, ICU stay, and postoperative symptoms of anemia.

 

We found no increased risk for transfusion with ethanol, tobacco, warfarin, or clopidogrel use; polycythemia; thrombocytopenia; preoperative INR; preoperative aPTT; preoperative albumin; Hemoglobin A1c; or diabetes mellitus; or for operations performed for malignancy. Ten patients in the ICU received transfusion (5.8%) compared with 2 patients (0.8%) not admitted to the ICU.

Operations were deemed high risk when ≥ 2 of patients having that operation received transfusions within 72 hours of their operation. There were 15 abdominoperineal resections; 3 of these received transfusions (20%). There were 7 total abdominal colectomies; 3 of these received transfusions (43%). We therefore had 22 high-risk operations, 6 of which were transfused (27%).

 

 

Discussion

Routine measurement of postoperative hematocrit levels after elective general surgery at NMVAHCS was not necessary. There were 12 transfusions for inpatients (2.8%), which is similar to the findings of a recent study of VA general surgery (2.3%).13 We found that routine postoperative hematocrit measurements to assess anemia had little or no effect on clinical decision-making or clinical outcomes.

According to our results, 88% of initial hematocrit tests after elective partial colectomies could have been eliminated; only 32 of 146 patients demonstrated a clinical reason for postoperative hematocrit testing. Similarly, 36 of 40 postcholecystectomy hematocrit tests (90%) could have been eliminated had the surgeons relied on clinical signs indicating possible postoperative anemia (none were transfused). Excluding patients with major intraoperative blood loss (> 300 mL), only 29 of 288 (10%) patients who had postoperative hematocrit tests had a clinical indication for a postoperative hematocrit test (ie, symptoms of anemia and/or active bleeding). One patient with inguinal hernia surgery who received transfusion was taking an anticoagulant and had a clinically indicated hematocrit test for a large hematoma that eventually required reoperation.

Our study found that routine hematocrit checks may actually increase the risk that a patient would receive an unnecessary transfusion. For instance, one elderly patient, after a right colectomy, had 6 hematocrit levels while on a heparin drip and received transfusion despite being asymptomatic. His lowest hematocrit level prior to transfusion was 23.7%. This patient had a total of 18 hematocrit tests. His EBL was 350 mL and his first postoperative HCT level was 33.1%. In another instance, a patient undergoing abdominoperineal resection had a transfusion on postoperative day 1, despite being hypertensive, with a hematocrit that ranged from 26% before transfusion to 31% after the transfusion. These 2 cases illustrate what has been shown in a recent study: A substantial number of patients with colorectal cancer receive unnecessary transfusions.14 On the other hand, one ileostomy closure patient had 33 hematocrit tests, yet his initial postoperative hematocrit was 37%, and he never received a transfusion. With low-risk surgeries, clinical judgment should dictate when a postoperative hematocrit level is needed. This strategy would have eliminated 206 unnecessary initial postoperative hematocrit tests (72%), could have decreased the number of unnecessary transfusions, and would have saved NMVAHCS about $1600 annually.

Abdominoperineal resections and total abdominal colectomies accounted for a high proportion of transfusions in our study. Inpatient elective operations can be risk stratified and have routine hematocrit tests ordered for patients at high risk. The probability of transfusion was greater in high-risk vs low-risk surgeries; 27% (6 of 22 patients) vs 2% (6 of 408 patients), respectively (P < .001). Since 14 of the 22 patients undergoing high-risk operation already had clinical reasons for a postoperative hematocrit test, we only need to add the remaining 8 patients with high-risk operations to the 74 who had a clinical reason for a hematocrit test and conclude that 82 of 430 patients (19%) had a clinical reason for a hematocrit test, either from signs or symptoms of blood loss or because they were in a high-risk group.

 


While our elective general surgery cases may not represent many general surgery programs in the US and VA health care systems, we can extrapolate cost savings using the same cost analyses outlined by Kohli and colleagues.1 Assuming 1.9 million elective inpatient general surgeries per year in the United States with an average cost of $21 per CBC, the annual cost of universal postoperative hematocrit testing would be $40 million.11,15 If postoperative hematocrit testing were 70% consistent with our findings, the annual cost for hematocrit tests on 51% of the inpatient general surgeries would be approximately $20.4 million. A reduction in routine hematocrit testing to 25% of all inpatient general surgeries (vs our finding that 19% were deemed necessary) results in an annual savings of $30 million. This conservative estimate could be even higher since there were 4.4 hematocrit tests per patient; therefore, we have about $132 million in savings.

Assuming 181,384 elective VA inpatient general surgeries each year, costing $7.14 per CBC (the NMVAHCS cost), the VA could save $1.3 million annually. If postoperative HCT testing were 70% consistent with our findings, the annual cost for hematocrit tests on 50.4% of inpatient general surgery operations would be about $653,000. A reduction in routine hematocrit testing to 25% of all inpatient general surgeries (vs our 19%) results in annual VA savings of $330,000. This conservative estimate could be even higher since there were on average 4.4 hematocrit levels per patient; therefore, we estimate that annual savings for the VA of about $1.45 million.

 

 

Limitations

The retrospective chart review nature of this study may have led to selection bias. Only a small number of patients received a transfusion, which may have skewed the data. This study population comes from a single VA medical center; this patient population may not be reflective of other VA medical centers or the US population as a whole. Given that NMVAHCS does not perform hepatic, esophageal, pancreas, or transplant operations, the potential savings to both the US and the VA may be overestimated, but this could be studied in the future by VA medical centers that perform more complex operations.

 

Conclusions

This study found that over a 4-year period routine postoperative hematocrit tests for patients undergoing elective general surgery at a VA medical center were not necessary. General surgeons routinely order various pre- and postoperative laboratory tests despite their limited utility. Reduction in unneeded routine tests could result in notable savings to the VA without compromising quality of care.

Only general surgery patients undergoing operations that carry a high risk for needing a blood transfusion should have a routine postoperative hematocrit testing. In our study population, the chance of an elective colectomy, cholecystectomy, or hernia patient needing a transfusion was rare. This strategy could eliminate a considerable number of unnecessary blood tests and would potentially yield significant savings.

References

1. Kohli N, Mallipeddi PK, Neff JM, Sze EH, Roat TW. Routine hematocrit after elective gynecologic surgery. Obstet Gynecol. 2000;95(6 Pt 1):847-850. doi:10.1016/s0029-7844(00)00796-1

2. Olus A, Orhan, U, Murat A, et al. Do asymptomatic patients require routine hemoglobin testing following uneventful, unplanned cesarean sections? Arch Gynecol Obstet. 2010;281(2):195-199. doi:10.1007/s00404-009-1093-1

3. Wu XD, Zhu ZL, Xiao P, Liu JC, Wang JW, Huang W. Are routine postoperative laboratory tests necessary after primary total hip arthroplasty? J Arthroplasty. 2020;35(10):2892-2898. doi:10.1016/j.arth.2020.04.097

4. Kumar A, Srivastava U. Role of routine laboratory investigations in preoperative evaluation. J Anesthesiol Clin Pharmacol. 2011;27(2):174-179. doi:10.4103/0970-9185.81824

5. Aghajanian A, Grimes DA. Routine prothrombin time determination before elective gynecologic operations. Obstet Gynecol. 1991;78(5 Pt 1):837-839.

6. Ransom SB, McNeeley SG, Malone JM Jr. A cost-effectiveness evaluation of preoperative type-and-screen testing for vaginal hysterectomy. Am J Obstet Gynecol. 1996;175(5):1201-1203. doi:10.1016/s0002-9378(96)70028-5

7. Ransom SB, McNeeley SG, Hosseini RB. Cost-effectiveness of routine blood type and screen testing before elective laparoscopy. Obstet Gynecol. 1995;86(3):346-348. doi:10.1016/0029-7844(95)00187-V

<--pagebreak-->

8. Committee on Standards and Practice Parameters, Apfelbaum JL, Connis RT, et al. Practice advisory for preanesthesia evaluation: an updated report by the American Society of Anesthesiologists Task Force on Preanesthesia Evaluation. Anesthesiology. 2012;116(3):522-538. doi:10.1097/ALN.0b013e31823c1067

9. Weil IA, Seicean S, Neuhauser D, Schiltz NK, Seicean A. Use and utility of hemostatic screening in adults undergoing elective, non-cardiac surgery. PLoS One. 2015;10(12):e0139139. doi:10.1371/journal.pone.0139139

10. Wu WC, Schifftner TL, Henderson WG, et al. Preoperative hematocrit levels and postoperative outcomes in older patients undergoing non-cardiac surgery. JAMA. 2007;297(22):2481-2488. doi:10.1001/jama.297.22.2481

11. Healthcare Bluebook. Complete blood count (CBC) with differential. Accessed March 28, 2024. https://www.healthcarebluebook.com/page_ProcedureDetails.aspx?id=214&dataset=lab

12. Douketis JD, Spyropoulos AC, Murad MH, et al. Perioperative management of antithrombotic therapy: an American College of Chest Physicians Clinical Practice Guideline. Chest. 2022;162(5):e207-e243. doi:10.1016/j.chest.2022.07.025

13. Randall JA, Wagner KT, Brody F. Perioperative transfusions in veterans following noncardiac procedures. J Laparoendosc Adv Surg Tech A. 2023;33(10):923-931. doi:10.1089/lap. 2023.0307

14. Tartter PI, Barron DM. Unnecessary blood transfusions in elective colorectal cancer surgery. Transfusion. 1985;25(2):113-115. doi:10.1046/j.1537-2995.1985.25285169199.x

15. Steiner CA, Karaca Z, Moore BJ, Imshaug MC, Pickens G. Surgeries in hospital-based ambulatory surgery and hospital inpatient settings, 2014. Healthcare Cost and Utilization Project statistical brief #223. May 2017. Revised July 2020. Agency for Healthcare Research and Quality. Accessed February 26, 2024. https://hcup-us.ahrq.gov/reports/statbriefs/sb223-Ambulatory-Inpatient-Surgeries-2014.pdf

16. US Department of Veterans Affairs, National Surgery Office. Quarterly report: Q3 of fiscal year 2017. VISN operative complexity summary [Source not verified].

References

1. Kohli N, Mallipeddi PK, Neff JM, Sze EH, Roat TW. Routine hematocrit after elective gynecologic surgery. Obstet Gynecol. 2000;95(6 Pt 1):847-850. doi:10.1016/s0029-7844(00)00796-1

2. Olus A, Orhan, U, Murat A, et al. Do asymptomatic patients require routine hemoglobin testing following uneventful, unplanned cesarean sections? Arch Gynecol Obstet. 2010;281(2):195-199. doi:10.1007/s00404-009-1093-1

3. Wu XD, Zhu ZL, Xiao P, Liu JC, Wang JW, Huang W. Are routine postoperative laboratory tests necessary after primary total hip arthroplasty? J Arthroplasty. 2020;35(10):2892-2898. doi:10.1016/j.arth.2020.04.097

4. Kumar A, Srivastava U. Role of routine laboratory investigations in preoperative evaluation. J Anesthesiol Clin Pharmacol. 2011;27(2):174-179. doi:10.4103/0970-9185.81824

5. Aghajanian A, Grimes DA. Routine prothrombin time determination before elective gynecologic operations. Obstet Gynecol. 1991;78(5 Pt 1):837-839.

6. Ransom SB, McNeeley SG, Malone JM Jr. A cost-effectiveness evaluation of preoperative type-and-screen testing for vaginal hysterectomy. Am J Obstet Gynecol. 1996;175(5):1201-1203. doi:10.1016/s0002-9378(96)70028-5

7. Ransom SB, McNeeley SG, Hosseini RB. Cost-effectiveness of routine blood type and screen testing before elective laparoscopy. Obstet Gynecol. 1995;86(3):346-348. doi:10.1016/0029-7844(95)00187-V

<--pagebreak-->

8. Committee on Standards and Practice Parameters, Apfelbaum JL, Connis RT, et al. Practice advisory for preanesthesia evaluation: an updated report by the American Society of Anesthesiologists Task Force on Preanesthesia Evaluation. Anesthesiology. 2012;116(3):522-538. doi:10.1097/ALN.0b013e31823c1067

9. Weil IA, Seicean S, Neuhauser D, Schiltz NK, Seicean A. Use and utility of hemostatic screening in adults undergoing elective, non-cardiac surgery. PLoS One. 2015;10(12):e0139139. doi:10.1371/journal.pone.0139139

10. Wu WC, Schifftner TL, Henderson WG, et al. Preoperative hematocrit levels and postoperative outcomes in older patients undergoing non-cardiac surgery. JAMA. 2007;297(22):2481-2488. doi:10.1001/jama.297.22.2481

11. Healthcare Bluebook. Complete blood count (CBC) with differential. Accessed March 28, 2024. https://www.healthcarebluebook.com/page_ProcedureDetails.aspx?id=214&dataset=lab

12. Douketis JD, Spyropoulos AC, Murad MH, et al. Perioperative management of antithrombotic therapy: an American College of Chest Physicians Clinical Practice Guideline. Chest. 2022;162(5):e207-e243. doi:10.1016/j.chest.2022.07.025

13. Randall JA, Wagner KT, Brody F. Perioperative transfusions in veterans following noncardiac procedures. J Laparoendosc Adv Surg Tech A. 2023;33(10):923-931. doi:10.1089/lap. 2023.0307

14. Tartter PI, Barron DM. Unnecessary blood transfusions in elective colorectal cancer surgery. Transfusion. 1985;25(2):113-115. doi:10.1046/j.1537-2995.1985.25285169199.x

15. Steiner CA, Karaca Z, Moore BJ, Imshaug MC, Pickens G. Surgeries in hospital-based ambulatory surgery and hospital inpatient settings, 2014. Healthcare Cost and Utilization Project statistical brief #223. May 2017. Revised July 2020. Agency for Healthcare Research and Quality. Accessed February 26, 2024. https://hcup-us.ahrq.gov/reports/statbriefs/sb223-Ambulatory-Inpatient-Surgeries-2014.pdf

16. US Department of Veterans Affairs, National Surgery Office. Quarterly report: Q3 of fiscal year 2017. VISN operative complexity summary [Source not verified].

Page Number
S2-S8
Page Number
S2-S8
Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>0524 FED Hematocrit</fileName> <TBEID>0C02F5A6.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F5A6</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname/> <articleType>1</articleType> <TBLocation>Copyfitting-FED</TBLocation> <QCDate/> <firstPublished>20240610T154406</firstPublished> <LastPublished>20240610T154406</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240610T154406</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline/> <bylineText>Anthony Vigil, MDa,b; Taylor Parnall, MDc; Clifford Qualls, PhDa,b; Robert Glew, PhDb; Robin Osofsky, MDd; Micah Guess, RNa; Lauren Mercer, MDb</bylineText> <bylineFull/> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>It is common practice to routinely measure postoperative hematocrit levels at US Department of Veterans Affairs (VA) hospitals for a wide range of elective gene</metaDescription> <articlePDF/> <teaserImage/> <title>Overuse of Hematocrit Testing After Elective General Surgery at a Veterans Affairs Medical Center</title> <deck/> <eyebrow>Original Research</eyebrow> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>Aug</pubPubdateMonth> <pubPubdateDay/> <pubVolume>41</pubVolume> <pubNumber>8S</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2951</CMSID> <CMSID>3639</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>FED</publicationCode> <pubIssueName>August 2024</pubIssueName> <pubArticleType>Feature Articles | 3639</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Feature | 2951<pubSubsection/></pubSection> </pubSections> <journalTitle>Fed Pract</journalTitle> <journalFullTitle>Federal Practitioner</journalFullTitle> <copyrightStatement>Copyright 2017 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">16</term> </publications> <sections> <term canonical="true">104</term> </sections> <topics> <term>327</term> <term canonical="true">27442</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Overuse of Hematocrit Testing After Elective General Surgery at a Veterans Affairs Medical Center</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><b>Objective:</b> To evaluate the clinical usefulness and costs of routine postoperative hematocrit testing after elective general surgery. <br/><br/><b>Methods:</b> We reviewed charts of all patients who had elective general surgery at New Mexico Veterans Affairs Health Care System, Albuquerque hospital from 2011 through 2014. Demographic data and patient characteristics (eg, comorbidities, smoking/drinking history), estimated blood loss (EBL), pre- and postoperative hematocrit levels, and signs and symptoms of anemia were compared in patients who did or did not receive a blood transfusion within 72 hours of the operation. <br/><br/><b>Results:</b> Of 1531 patients who had an elective general surgery between 2011 and 2014, ≥ 1 postoperative hematocrit levels were measured in 288 individuals. There were 1312 postoperative hematocrit measurements before discharge (mean, 8.7; range, 1-44). There were 12 transfusions (0.8%) for patients without moderate to severe pre-existing anemia (hematocrit &lt; 30%). Five of 12 transfused patients received intraoperative transfusions and 7 patients were transfused within 72 hours postoperation. No patients were transfused preoperatively. Of 12 patients receiving transfusion, 11 had EBL &gt; 199 mL and/or signs of anemia. Risk factors for postoperative transfusion included lower preoperative hematocrit, increased EBL, and having either abdominoperineal resection or a total proctocolectomy.<br/><br/><b>Conclusions:</b> Routine postoperative hematocrit measurements after elective general surgery at US Department of Veterans Affairs medical centers are of negligible clinical value and should be reconsidered. Clinical judgment, laboratory-documented pre-existing anemia, a high-risk operation, or symptoms of anemia should prompt monitoring of patient postoperative hematocrit testing. This strategy could have eliminated 206 initial hematocrit checks over the 4 years of the study. </p> <p><span class="Drop">I</span>t is common practice to routinely measure postoperative hematocrit levels at US Department of Veterans Affairs (VA) hospitals for a wide range of elective general surgeries. While hematocrit measurement is a low-cost test, the high frequency with which these tests are performed may drastically increase overall costs. </p> <p>Numerous studies have suggested that physicians overuse laboratory testing.<sup>1-10</sup> Kohli and colleagues recommended that the routine practice of obtaining postoperative hematocrit tests following elective gynecologic surgery be abandoned.<sup>1</sup> A similar recommendation was made by Olus and colleagues after studying uneventful, unplanned cesarean sections and by Wu and colleagues after investigating routine laboratory tests post total hip arthroplasty.<sup>2,3</sup> <br/><br/>To our knowledge, a study assessing routine postoperative hematocrit testing in elective general surgery has not yet been conducted. Many laboratory tests ordered in the perioperative period are not indicated, including complete blood count (CBC), electrolytes, and coagulation studies.<sup>4</sup> Based on the results of these studies, we expected that the routine measurement of postoperative hematocrit levels after elective general surgeries at VA medical centers would not be cost effective. A PubMed search for articles published from 1990 to 2023 using the search terms “hematocrit,” “hemoglobin,” “general,” “surgery,” “routine,” and “cost” or “cost-effectiveness,” suggests that the clinical usefulness of postoperative hematocrit testing has not been well studied in the general surgery setting. The purpose of this study was to determine the clinical utility and associated cost of measuring routine postoperative hematocrit levels in order to generate a guide as to when the practice is warranted following common elective general surgery. Although gynecologic textbooks may describe recommendations of routine hematocrit checking after elective gynecologic operations, one has difficulty finding the same recommendations in general surgery textbooks.<sup>1</sup> However, it is common practice for surgical residents and attending surgeons to routinely order hematocrit on postoperative day-1 to ensure that the operation did not result in unsuspected anemia that then would need treatment (either with fluids or a blood transfusion). Many other surgeons rely on clinical factors such as tachycardia, oliguria, or hypotension to trigger a hematocrit (and other laboratory) tests. Our hypothesis is that the latter group has chosen the most cost-effective and prudent practice. One problem with checking the hematocrit routinely, as with any other screening test, is what to do with an abnormal result, assuming an asymptomatic patient? If the postoperative hematocrit is lower than expected given the estimated blood loss (EBL), what is one to do? </p> <h2>Methods</h2> <p>This retrospective case-control study conducted at the New Mexico VA Health Care System (NMVAHCS) in Albuquerque compared data for patients who received transfusion within 72 hours of elective surgeries vs patients who did not. Patients who underwent elective general surgery from January 2011 through December 2014 were included. An elective general surgery was defined as surgery performed following an outpatient preoperative anesthesia evaluation ≥ 30 days prior to operation. Patients who underwent emergency operations, and those with baseline anemia (preoperative hematocrit &lt; 30%), and those transfused &gt; 72 hours after their operation were excluded. The NMVAHCSInstitutional Review Board approved this study (No. 15-H184).</p> <p>A detailed record review was conducted to collect data on demographics and other preoperative risk factors, including age, sex, body mass index (BMI), race and ethnicity, cardiac and pulmonary comorbidities, tobacco use, alcohol intake, diabetes, American Society of Anesthesiologists Physical Status Classification, metabolic equivalent of task, hematologic conditions, and renal disease.<br/><br/>For each procedure, we recorded the type of elective general surgery performed, the diagnosis/indication, pre- and postoperative hemoglobin/hematocrit, intraoperative EBL, length of operation, surgical wound class, length of hospital stay (LOS), intensive care unit (ICU) status, number of hematocrit tests, cardiovascular risk of operation (defined by anesthesia assessment), presence or absence of malignancy, preoperative platelet count, albumin level, preoperative prothrombin time/activated partial thromboplastin time (aPTT), international normalized ratio (INR), hemoglobin A<sub>1c</sub>, and incidence of transfusion. Signs and symptoms of anemia were recorded as present if the postoperative vital signs suggested low intravascular volume (pulse &gt; 120 beats/minute, systolic blood pressure &lt; 90 mm Hg, or vasoactive medication requirement [per anesthesia postoperative note]) or if the patient reported or exhibited symptoms of dizziness or fatigue or evidence of clinically apparent bleeding (ie, hematoma formation). Laboratory charges for hematocrit tests and CBC at the NMAVAHCS were used to assess cost.<sup>11</sup>To stratify the transfusion risk, patients were distributed among 3 groups based on the following criteria: discharged home the same day as surgery; admitted but did not have postoperative hematocrit testing; and admitted and had postoperative hematocrit testing. We also stratified operations into low or high risk based on the risk for postoperative transfusion (Figure). Recognizing that the American College of Chest Physicians guidelines for perioperative management of antithrombotic therapy places bowel resection in a high-risk category, we designated a surgery as high risk when ≥ 2 patients in the transfusion group had that type of surgery over the 4 years of the study.<sup>12</sup> Otherwise, the operations were deemed low risk.</p> <h3>Statistical Analysis</h3> <p>Numeric analysis used <i>t</i> tests and Binary and categorical variables used Fisher exact tests. <i>P</i> value ≤ .05 was considered statistically significant. SAS software was used for all statistical analyses.</p> <h2>Results</h2> <p>From 2011 through 2014, 1531 patients had elective general surgery at NMVAHCS. Twenty-two patients with preoperative anemia (hematocrit &lt; 30%) and 1 patient who received a transfusion &gt; 72 hours after the operation were excluded. Most elective operations (70%, n = 1075) were performed on an outpatient basis; none involved transfusion. Inguinal hernia repair was most common with 479 operations; 17 patients were treated inpatient of which 2 patients had routine postoperative hematocrit checks; (neither received transfusion). One patient with inguinal hernia surgery received transfusion without routine postoperative hematocrit monitoring.</p> <p>Of 112 partial colon resections, 1 patient had a postoperative transfusion; and all but 3 received postoperative hematocrit monitoring. Nineteen patients undergoing partial colon resection had a clinical indication for postoperative hematocrit monitoring. None of the 5 patients with partial gastrectomy received a postoperative transfusion. Of 121 elective cholecystectomies, no patients had postoperative transfusion, whereas 34 had postoperative hematocrit monitoring; only 2 patients had a clinical reason for the hematocrit monitoring. <br/><br/>Of 430 elective inpatient operations, 12 received transfusions and 288 patients had ≥ 1 postoperative hematocrit test (67%). All hematocrit tests were requested by the attending surgeon, resident surgeon, or the surgical ICU team. Of the group that had postoperative hematocrit monitoring, there was an average of 4.4 postoperative hematocrit tests per patient (range, 1-44). <br/><br/>There were 12 transfusions for inpatients (2.8%), which is similar to the findings of a recent study of VA general surgery (2.3%).<sup>13</sup> Five of the 12 patients received intraoperative transfusions while 7 were transfused within 72 hours postoperation. All but 1 patient receiving transfusion had EBL &gt; 199 mL (range, 5-3000; mean, 950 mL; median, 500 mL) and/or signs or symptoms of anemia or other indications for measurement of the postoperative hematocrit. There were no statistically significant differences in patients’ age, sex, BMI, or race and ethnicity between groups receiving and not receiving transfusion (Table 1).<br/><br/>When comparing the transfusion vs the nontransfusion groups (after excluding those with clinical preoperative anemia) the risk factors for transfusion included: relatively low mean preoperative hematocrit (mean, 36.9% vs 42.7%, respectively; <i>P</i><i> </i>= .003), low postoperative hematocrit (mean, 30.2% vs 37.1%, respectively; <i>P</i><i> </i>&lt; .001), high EBL (mean, 844 mL vs 109 mL, respectively; <i>P </i>= .005), large infusion of intraoperative fluids (mean, 4625 mL vs 2505 mL, respectively; <i>P </i>= .005), longer duration of operation (mean, 397 min vs 183 min, respectively; <i>P </i>&lt; .001), and longer LOS (mean, 14.5 d vs 4.9 d, respectively; <i>P </i>&lt; .001) (Table 2). Similarly, we found an increased risk for transfusion with high/intermediate cardiovascular risk (vs low), any wound not classified as clean, ICU stay, and postoperative symptoms of anemia.<br/><br/>We found no increased risk for transfusion with ethanol, tobacco, warfarin, or clopidogrel use; polycythemia; thrombocytopenia; preoperative INR; preoperative aPTT; preoperative albumin; Hemoglobin A<sub>1c</sub>; or diabetes mellitus; or for operations performed for malignancy. Ten patients in the ICU received transfusion (5.8%) compared with 2 patients (0.8%) not admitted to the ICU. <br/><br/>Operations were deemed high risk when ≥ 2 of patients having that operation received transfusions within 72 hours of their operation. There were 15 abdominoperineal resections; 3 of these received transfusions (20%). There were 7 total abdominal colectomies; 3 of these received transfusions (43%). We therefore had 22 high-risk operations, 6 of which were transfused (27%).</p> <h2>Discussion </h2> <p>Routine measurement of postoperative hematocrit levels after elective general surgery at NMVAHCS was not necessary. There were 12 transfusions for inpatients (2.8%), which is similar to the findings of a recent study of VA general surgery (2.3%).<sup>13</sup> We found that routine postoperative hematocrit measurements to assess anemia had little or no effect on clinical decision-making or clinical outcomes. </p> <p>According to our results, 88% of initial hematocrit tests after elective partial colectomies could have been eliminated; only 32 of 146 patients demonstrated a clinical reason for postoperative hematocrit testing. Similarly, 36 of 40 postcholecystectomy hematocrit tests (90%) could have been eliminated had the surgeons relied on clinical signs indicating possible postoperative anemia (none were transfused). Excluding patients with major intraoperative blood loss (&gt; 300 mL), only 29 of 288 (10%) patients who had postoperative hematocrit tests had a clinical indication for a postoperative hematocrit test (ie, symptoms of anemia and/or active bleeding). One patient with inguinal hernia surgery who received transfusion was taking an anticoagulant and had a clinically indicated hematocrit test for a large hematoma that eventually required reoperation.<br/><br/>Our study found that routine hematocrit checks may actually increase the risk that a patient would receive an unnecessary transfusion. For instance, one elderly patient, after a right colectomy, had 6 hematocrit levels while on a heparin drip and received transfusion despite being asymptomatic. His lowest hematocrit level prior to transfusion was 23.7%. This patient had a total of 18 hematocrit tests. His EBL was 350 mL and his first postoperative HCT level was 33.1%. In another instance, a patient undergoing abdominoperineal resection had a transfusion on postoperative day 1, despite being hypertensive, with a hematocrit that ranged from 26% before transfusion to 31% after the transfusion. These 2 cases illustrate what has been shown in a recent study: A substantial number of patients with colorectal cancer receive unnecessary transfusions.<sup>14</sup> On the other hand, one ileostomy closure patient had 33 hematocrit tests, yet his initial postoperative hematocrit was 37%, and he never received a transfusion. With low-risk surgeries, clinical judgment should dictate when a postoperative hematocrit level is needed. This strategy would have eliminated 206 unnecessary initial postoperative hematocrit tests (72%), could have decreased the number of unnecessary transfusions, and would have saved NMVAHCS about $1600 annually. <br/><br/>Abdominoperineal resections and total abdominal colectomies accounted for a high proportion of transfusions in our study. Inpatient elective operations can be risk stratified and have routine hematocrit tests ordered for patients at high risk. The probability of transfusion was greater in high-risk vs low-risk surgeries; 27% (6 of 22 patients) vs 2% (6 of 408 patients), respectively (<i>P</i> &lt; .001). Since 14 of the 22 patients undergoing high-risk operation already had clinical reasons for a postoperative hematocrit test, we only need to add the remaining 8 patients with high-risk operations to the 74 who had a clinical reason for a hematocrit test and conclude that 82 of 430 patients (19%) had a clinical reason for a hematocrit test, either from signs or symptoms of blood loss or because they were in a high-risk group. <br/><br/>While our elective general surgery cases may not represent many general surgery programs in the US and VA health care systems, we can extrapolate cost savings using the same cost analyses outlined by Kohli and colleagues.<sup>1</sup> Assuming 1.9 million elective inpatient general surgeries per year in the United States with an average cost of $21 per CBC, the annual cost of universal postoperative hematocrit testing would be $40 million.<sup>11,15</sup> If postoperative hematocrit testing were 70% consistent with our findings, the annual cost for hematocrit tests on 51% of the inpatient general surgeries would be approximately $20.4 million. A reduction in routine hematocrit testing to 25% of all inpatient general surgeries (vs our finding that 19% were deemed necessary) results in an annual savings of $30 million. This conservative estimate could be even higher since there were 4.4 hematocrit tests per patient; therefore, we have about $132 million in savings.<br/><br/>Assuming 181,384 elective VA inpatient general surgeries each year, costing $7.14 per CBC (the NMVAHCS cost), the VA could save $1.3 million annually. If postoperative HCT testing were 70% consistent with our findings, the annual cost for hematocrit tests on 50.4% of inpatient general surgery operations would be about $653,000. A reduction in routine hematocrit testing to 25% of all inpatient general surgeries (vs our 19%) results in annual VA savings of $330,000. This conservative estimate could be even higher since there were on average 4.4 hematocrit levels per patient; therefore, we estimate that annual savings for the VA of about $1.45 million.</p> <h3>Limitations </h3> <p>The retrospective chart review nature of this study may have led to selection bias. Only a small number of patients received a transfusion, which may have skewed the data. This study population comes from a single VA medical center; this patient population may not be reflective of other VA medical centers or the US population as a whole. Given that NMVAHCS does not perform hepatic, esophageal, pancreas, or transplant operations, the potential savings to both the US and the VA may be overestimated, but this could be studied in the future by VA medical centers that perform more complex operations. </p> <h2>Conclusions</h2> <p>This study found that over a 4-year period routine postoperative hematocrit tests for patients undergoing elective general surgery at a VA medical center were not necessary. General surgeons routinely order various pre- and postoperative laboratory tests despite their limited utility. Reduction in unneeded routine tests could result in notable savings to the VA without compromising quality of care.</p> <p>Only general surgery patients undergoing operations that carry a high risk for needing a blood transfusion should have a routine postoperative hematocrit testing. In our study population, the chance of an elective colectomy, cholecystectomy, or hernia patient needing a transfusion was rare. This strategy could eliminate a considerable number of unnecessary blood tests and would potentially yield significant savings.</p> <p class="isub">Author affiliations</p> <p> <em><sup>a</sup>New Mexico Veterans Affairs Health Care System, Albuquerque<br/><br/><sup>b</sup>University of New Mexico School of Medicine, Albuquerque<br/><br/><sup>c</sup>Harbor-UCLA Medical Center, Torrance, California<br/><br/><sup>d</sup>Oregon Health and Science University, Portland</em> </p> <p class="isub">Author contributions</p> <p> <em><i>Study conception and design</i>: Vigil, Taylor; <i>acquisition of data</i>: Vigil, Taylor, Geuss, Mercer; <i>analysis and interpretation of data</i>: Vigil, Taylor, Osofsky, Qualls; <i>drafting of manuscript</i>: Vigil, Taylor; <i>critical revision</i>: Glew.</em> </p> <p class="isub">Author disclosures </p> <p> <em>The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.</em> </p> <p class="isub">Disclaimer</p> <p> <em>The opinions expressed herein are those of the authors and do not necessarily reflect those of <i>Federal Practitioner,</i> Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.</em> </p> <p class="isub">Ethics and consent</p> <p> <em>The New Mexico Veterans Affairs Health Care System Institutional Review Board approved this study (No. 15-H184).</em> </p> <h2>References</h2> <p class="reference"> 1. Kohli N, Mallipeddi PK, Neff JM, Sze EH, Roat TW. Routine hematocrit after elective gynecologic surgery. Obstet Gynecol. 2000;95(6 Pt 1):847-850. doi:10.1016/s0029-7844(00)00796-1<br/><br/> 2. Olus A, Orhan, U, Murat A, et al. Do asymptomatic patients require routine hemoglobin testing following uneventful, unplanned cesarean sections? <i>Arch Gynecol Obstet.</i> 2010;281(2):195-199. doi:10.1007/s00404-009-1093-1<br/><br/> 3. Wu XD, Zhu ZL, Xiao P, Liu JC, Wang JW, Huang W. Are routine postoperative laboratory tests necessary after primary total hip arthroplasty? <i>J Arthroplasty.</i> 2020;35(10):2892-2898. doi:10.1016/j.arth.2020.04.097<br/><br/> 4. Kumar A, Srivastava U. Role of routine laboratory investigations in preoperative evaluation. <i>J Anesthesiol Clin Pharmacol.</i> 2011;27(2):174-179. doi:10.4103/0970-9185.81824<br/><br/> 5. Aghajanian A, Grimes DA. Routine prothrombin time determination before elective gynecologic operations. <i>Obstet Gynecol. </i>1991;78(5 Pt 1):837-839.<br/><br/> 6. Ransom SB, McNeeley SG, Malone JM Jr. A cost-effectiveness evaluation of preoperative type-and-screen testing for vaginal hysterectomy. <i>Am J Obstet Gynecol</i>. 1996;175(5):1201-1203. doi:10.1016/s0002-9378(96)70028-5<br/><br/> 7. Ransom SB, McNeeley SG, Hosseini RB. Cost-effectiveness of routine blood type and screen testing before elective laparoscopy. <i>Obstet Gynecol.</i> 1995;86(3):346-348. doi:10.1016/0029-7844(95)00187-V</p> <p class="reference"> 8. Committee on Standards and Practice Parameters, Apfelbaum JL, Connis RT, et al. Practice advisory for preanesthesia evaluation: an updated report by the American Society of Anesthesiologists Task Force on Preanesthesia Evaluation. <i>Anesthesiology.</i> 2012;116(3):522-538. doi:10.1097/ALN.0b013e31823c1067 <br/><br/> 9. Weil IA, Seicean S, Neuhauser D, Schiltz NK, Seicean A. Use and utility of hemostatic screening in adults undergoing elective, non-cardiac surgery. <i>PLoS One.</i> 2015;10(12):e0139139. doi:10.1371/journal.pone.0139139<br/><br/>10. Wu WC, Schifftner TL, Henderson WG, et al. Preoperative hematocrit levels and postoperative outcomes in older patients undergoing non-cardiac surgery. <i>JAMA</i>. 2007;297(22):2481-2488. doi:10.1001/jama.297.22.2481<br/><br/>11. Healthcare Bluebook. Complete blood count (CBC) with differential. Accessed March 28, 2024. https://www.healthcarebluebook.com/page_ProcedureDetails.aspx?id=214&amp;dataset=lab<br/><br/>12. Douketis JD, Spyropoulos AC, Murad MH, et al. Perioperative management of antithrombotic therapy: an American College of Chest Physicians Clinical Practice Guideline. <i>Chest. </i>2022;162(5):e207-e243. doi:10.1016/j.chest.2022.07.025<br/><br/>13. Randall JA, Wagner KT, Brody F. Perioperative transfusions in veterans following noncardiac procedures. <i>J Laparoendosc Adv Surg Tech A</i>. 2023;33(10):923-931. doi:10.1089/lap. 2023.0307<br/><br/>14. Tartter PI, Barron DM. Unnecessary blood transfusions in elective colorectal cancer surgery. <i>Transfusion.</i> 1985;25(2):113-115. doi:10.1046/j.1537-2995.1985.25285169199.x<br/><br/>15. Steiner CA, Karaca Z, Moore BJ, Imshaug MC, Pickens G. Surgeries in hospital-based ambulatory surgery and hospital inpatient settings, 2014. Healthcare Cost and Utilization Project statistical brief #223. May 2017. Revised July 2020. Agency for Healthcare Research and Quality. Accessed February 26, 2024. https://hcup-us.ahrq.gov/reports/statbriefs/sb223-Ambulatory-Inpatient-Surgeries-2014.pdf<br/><br/>16. US Department of Veterans Affairs, National Surgery Office. Quarterly report: Q3 of fiscal year 2017. VISN operative complexity summary [Source not verified].</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
Disable zoom
Off

Oxidative Stress in Patients With Melasma: An Evaluation of the Correlation of the Thiol/Disulfide Homeostasis Parameters and Modified MASI Score

Article Type
Changed
Fri, 06/14/2024 - 12:34
Display Headline
Oxidative Stress in Patients With Melasma: An Evaluation of the Correlation of the Thiol/Disulfide Homeostasis Parameters and Modified MASI Score

Melasma is an acquired hyperpigmentation disorder characterized by irregular brown macules and patches that usually appear on sun-exposed areas of the skin. The term melasma originates from the Greek word melas meaning black.1 Facial melasma is divided into 2 groups according to its clinical distribution: centrofacial lesions are located in the center of the face (eg, the glabellar, frontal, nasal, zygomatic, upper lip, chin areas), and peripheral lesions manifest on the frontotemporal, preauricular, and mandibular regions.1,2 There is debate on the categorization of zygomatic (or malar) melasma; some researchers argue it should be categorized independent of other areas, while others include malar melasma in the centrofacial group because of its frequent association with the centrofacial type, especially with glabellar lesions.2 Mandibular melasma is rare and occurs mostly in postmenopausal women after intense sun exposure.1,2 Although the etiopathogenesis of the disease is not clearly known, increased melanogenesis, extracellular matrix alterations, inflammation, and angiogenesis are assumed to play a role.3 Various risk factors such as genetic predisposition, UV radiation (UVR) exposure, pregnancy, thyroid dysfunction, and exogenous hormones (eg, oral contraceptives, hormone replacement therapy) have been identified; phototoxic drugs, anticonvulsants, and some cosmetics also have been implicated.4,5 Exposure to UVR is thought to be the main triggering environmental factor by inducing both melanin production and oxidative stress.5 However, it also has been shown that visible light can induce hyperpigmentation in darker skin types.6

The presence of oxidative stress in melasma recently has become an intriguing topic of interest. First, the presence of oxidative stress in the etiopathogenesis of melasma was thought to be based on the effectiveness of antioxidants in treatment. A few studies also have confirmed the presence of oxidative stress in melasma.7-10 Classically, oxidative stress can be described as a disturbance in the balance between oxidants and antioxidants. Reactive oxygen species (ROS) are highly reactive molecules due to the unpaired electrons in their structure. Although ROS are present at low levels in physiologic conditions and are involved in critical physiologic events, they damage cellular components such as fat, protein, and nucleic acid at high concentrations.5

Dynamic thiol/disulfide homeostasis is one of the most important markers of oxidative stress in biological systems. Thiols are organic compounds containing a sulfhydryl (-SH) group. Thiols are considered highly potent antioxidants because they reduce unstable free radicals by donating electrons. They are the first antioxidants to be depleted in an oxidative environment.11,12 In case of oxidative stress, they transform into reversible forms called disulfide bridges between 2 thiol groups. Disulfide bridges can be reduced back to thiol groups, which is how dynamic thiol/disulfide homeostasis is maintained. Dynamic thiol/disulfide homeostasis is responsible for cellular events such as antioxidant defense, signal transduction, regulation of enzyme function, and apoptosis.11,12

The aim of this study was to evaluate the presence of oxidative stress in melasma by comparing dynamic thiol/disulfide homeostasis in patients with melasma compared with age- and sex-matched healthy controls.

Materials and Methods

Participants and Eligibility Criteria—We conducted a prospective study in a tertiary-care hospital (Ankara Bilkent City Hospital [Ankara, Turkey]) of patients with melasma who were followed from October 2021 to October 2022 compared with age- and sex-matched healthy volunteers. Ethics committee approval was obtained from Ankara Bilkent City Hospital before the study (E2-21-881)(13.10.2021). Written informed consent was obtained from all participants, and all were older than 18 years. Patients were excluded if there was the presence of any systemic disease or dermatologic disease other than melasma; smoking or alcohol use; any use of vitamins, food supplements, or any medication in the last 3 months; or pregnancy.

Melasma Severity—The modified melasma area and severity index (mMASI) score was used to determine the severity of melasma. The score is calculated from assessments of the darkness of the pigmentation and the percentage of affected area on the face. The mMASI score is the sum of the darkness score (D); area score (A); and separate fixed coefficients for the forehead, as well as the right malar, left malar, and chin regions.13 The mMASI score, with a range of 0 to 24, is a reliable and objective marker in the calculation of melasma severity.4

Biochemical Analysis of Samples—The 6-cc peripheral fasting venous blood samples obtained from the study participants were centrifuged at 1500 g for 10 minutes, and the separated sera were stored in a freezer at 80 °C until the time of analysis. When the study was completed, the disulfide and thiol values were analyzed. Serum native and total thiol concentrations indicating thiol/disulfide homeostasis were calculated by a new fully automatic colorimetric method developed by Erel and Neselioglu.14 Using this method, short disulfide bonds are first reduced with sodium borohydride solution to form free-functional thiol groups, and then the unused sodium borohydride is removed using formaldehyde. Finally, all thiol groups are reacted with 5,5’-dithiobis-(2-nitrobenzoic) acid (Ellman reagent), and all thiol groups are detected after reaction with 5,5’-dithiobis-(2-nitrobenzoic) acid. When a disulfide bond (SS) is reduced, 2 thiol groups are formed. For this reason, half of the difference between total thiol (-SH + the amount of thiol formed by the reduction of disulfides) and native thiol (-SH) corresponds to the dynamic disulfide amount (total thiol − native thiol/2).14

Statistical Analysis—Statistical analysis was performed using SPSS software (version 24.0). Descriptive statistics were presented as numbers and percentages for categorical variables, and numerical variables were presented as mean, SD, median, minimum, maximum, 25th quartile, and 75th quartile. The conformity of the variables to normal distribution was examined using visual (histograms and probability plots) and analytical methods (Kolmogorov-Smirnov/Shapiro-Wilk tests). In pairwise group comparisons for numerical variables, a Mann-Whitney U test was used when normal distribution was not met, and a t test was used when normal distribution was met. The statistical significance level was accepted as P<.05.

Results

Our study included 67 patients with melasma and 41 healthy age- and sex-matched controls. Of the participants with melasma, 60 (89.5%) were female and 7 (10.5%) were male. The control group was similar to the melasma group in terms of sex (87.8% female vs 12.2% male [P=.59]). The mean age (SD) was 33.1 (6.7) years in the melasma group and 31.9 (6.7) years in the control group. Age was similar across both groups (P=.41). All participants were of Asian race, and Fitzpatrick skin types (types II–IV) were similar across both groups.

Fifty-four (80.6%) participants had centrofacial melasma and 13 (19.4%) had mixed-type melasma. The mMASI score ranged from 3 to 20; the mean (SD) mMASI score was 11.28 (3.2). Disease duration ranged from 2 to 72 months; the mean (SD) disease duration was 12.26 (6.3) months. The demographics and clinical characteristics of the study group are shown in eTable 1.

eTable 2 provides a summary of disulfide, native thiol, and total thiol levels, as well as disulfide/native thiol, disulfide/total thiol, and native thiol/total thiol ratios in the study population. Disulfide/native thiol and disulfide/total thiol ratios were higher in melasma patients (Figure 1), whereas the native thiol/total thiol ratio was higher in the control group (P=.025, P=.025, and P=.026, respectively).

All correlations between age, disease duration, and mMASI scores and disulfide, native thiol, and total thiol levels, as well as disulfide/native thiol, disulfide/total thiol, and native thiol/total thiol ratios, are summarized in eTable 3. No significant correlation was observed between age and disease duration and disulfide, native thiol, and total thiol levels or disulfide/native thiol, disulfide/total thiol, and native thiol/total thiol ratios.

We independently assessed whether Fitzpatrick skin types II, III, and IV exhibited distinct levels of oxidative stress in clinical melasma. There were no significant correlations with Fitzpatrick skin type (disulfide/native thiol, P=.25; disulfide/total thiol, P=.19). We further evaluated if the thiol/disulfide parameters were correlated with duration of melasma by dividing the melasma patients into 3 groups (<6 months [n=12], 6–18 months [n=32], >18 months [n=23]), but there was not any significant correlation (disulfide/native thiol, P=.15; disulfide/total thiol, P=.15). We also divided our patients into 3 groups according to age (<27 years [n=14], 27–36 years [n=33], >36 years [n=20]). There was no correlation of the parameters with age (disulfide/native thiol, P=.15; disulfide/total thiol, P=.14).

There was a positive correlation between mMASI score and disulfide, native thiol, and total thiol levels and disulfide/native thiol and disulfide/total thiol ratios, as well as a negative correlation between mMASI score and native thiol/total thiol ratio. The correlations between mMASI scores and disulfide/native thiol and disulfide/total thiol ratios are shown in Figure 2 and eTable 3.

Comment

Melasma is a common condition that may cause psychosocial problems in affected patients and negatively affect quality of life.1 It occurs in all races but is more common in individuals with darker skin types (eg, Fitzpatrick skin types III and IV). Although melasma is more common in women during reproductive years (50%–70%), it also has been observed in 10% to 30% of men.5

Treatment options include topical bleaching agents, chemical peels, and laser therapy, as well as discontinuation of medications that may potentially trigger melasma; use of broad-spectrum sunscreens also is recommended.4 Vitamins A, C, and E, as well as niacinamide, are used in the treatment of melasma, especially for their antioxidant properties. The key role of antioxidants in the treatment of melasma supports the importance of oxidative stress in the pathogenesis.7 Melasma often is challenging to treat, particularly the mixed or dermal types, due to their stubborn nature. This condition poses a considerable therapeutic challenge for dermatologists.4

kocradrupathedriphuhatriwebripudrudrimechametehosothahapedaslihebulalakathapuretumod
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%2C%20Disulfide%2Fnative%20thiol%20homeostasis%20parameters%20in%20participants%20with%20melasma%20and%20controls.%20B%2C%20Disulfide%2Ftotal%20thiol%20homeostasis%20parameters%20in%20participants%20with%20melasma%20and%20controls.%20Higher%20scores%20indicate%20that%20in%20patients%20with%20melasma%2C%20oxidative%20stress%20shifts%20the%20thiol%2F%20disulfide%20balance%20to%20disulfide%20formation%2C%20causing%20thiols%20to%20oxidize%20into%20disulfide%20bonds.%20The%20horizontal%20bar%20inside%20the%20boxes%20indicates%20the%20mean%2C%20and%20the%20lower%20and%20upper%20ends%20of%20the%20boxes%20are%20the%2025th%20and%2075th%20quartiles.%20The%20whiskers%20indicate%20the%20range%20of%20the%20parameters%20of%20thiol%2Fdisulfide%20homeostasis.%20Asterisk%20indicates%20P%3D.025.%3C%2Fp%3E

pimifrumafricrijeuamostoshimubarocrobiwegospogotrikesheclouoswivuvuhonucohasuthujestaspimocradifrutrekuthubrebrocrospichukalifrahalaceputhalacruswospahapheswi
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20A%2C%20Correlations%20between%20modified%20melasma%20area%20and%20severity%20index%20(mMASI)%20scores%20and%20disulfide%2Fnative%20thiol%20ratios%20(P%26lt%3B.001%3B%20r%3D0.42).%20B%2C%20Correlations%20between%20mMASI%20scores%20and%20disulfide%2Ftotal%20thiol%20ratios%20(P%26lt%3B.001%3B%20r%3D0.42).%20The%20correlation%20of%20mMASI%20scores%20with%20disulfide%2Fnative%20thiol%20and%20disulfide%2Ftotal%20thiol%20values%20in%20the%20melasma%20group%20indicates%20that%20oxidative%20stress%20is%20linked%20to%20melasma%20severity.%20The%20red%20diagonal%20lines%20indicate%20correlation%2C%20showing%20that%20as%20one%20value%20increases%2C%20the%20other%20also%20increases.%3C%2Fp%3E

Oxidative stress and oxidant-antioxidant imbalance previously have been studied in various diseases, but research investigating the presence of oxidative stress in melasma are limited.7-10 Exposure of the skin to polluted air and intense UVR, as well as some food by-products, cosmetics, and drugs (eg, oral contraceptives), can directly or indirectly cause ROS production in the skin. Reactive oxygen species are thought to be involved in the pathophysiology of melasma by affecting apoptotic pathways and causing cell proliferation. The intermediate heme pathway has pro-oxidant effects and produces ROS and metabolites such as redox-active quinines. Exposure to UVR leads to the generation of ROS, highlighting the role of oxidative stress in the onset of melasma. 5

In any cutaneous disease in which oxidative stress plays a role, oxidant and antioxidant levels may be expected to vary both locally and systemically; however, measurement of oxidative stress markers in serum instead of skin is technically and economically more advantageous.8 Firstly, serum collection is less invasive and technically simpler than skin biopsies. Drawing blood is a routine procedure that requires minimal specialized equipment and training compared to the extraction and processing of skin samples. Secondly, analyzing serum samples generally is less expensive than processing skin tissue.8

In our study, we evaluated dynamic thiol/disulfide homeostasis in serum to investigate the presence of oxidative stress in the setting of melasma. Functional sulfhydryl (-SH) groups in thiols act as substrates for antioxidant enzymes and as free-radical scavengers. They constitute one of the most powerful defense systems against the unwanted effects of ROS. Thiols, which become the main target of ROS under oxidative stress, oxidize with oxidant molecules and form disulfide bridges.15

Thiol/disulfide homeostasis has been studied many times in dermatologic diseases,16-19 and the results obtained from these studies are heterogenous depending on the extent of oxidative damage. It has been shown that thiol/disulfide homeostasis plays a role in oxidative stress in conditions such as psoriasis,17 seborrheic dermatitis,11 atopic dermatitits,18 and rosacea.19 In our study, disulfide/native thiol and disulfide/total thiol levels were significantly higher (both P=.025) in the melasma group compared with the control group, which indicates that the thiol/disulfide balance in patients with melasma is shifted to disulfide formation and thiols are oxidized to disulfide bonds in the presence of oxidative stress.

Seçkin et al7 evaluated the role of oxidative stress in the pathogenesis of melasma and found that the serum levels of the antioxidants superoxide dismutase and glutathione peroxidase were significantly higher in the patient group compared with the control group (both P<.001). They also found that the levels of nitric oxide (another antioxidant) were increased in the patient group and the levels of protein carbonyl (an oxidative metabolite) were significantly lower (both P<.001). These findings indicated that free-radical damage may be involved in the pathogenesis of melasma.7

In a study of 75 patients with melasma, serum levels of the antioxidants melatonin and catalase were significantly (P<.001 and P=.001, respectively) lower in the melasma group compared with the control group, while serum levels of the oxidants protein carbonyl and nitric oxide were significantly higher (P=.002 and P=.001, respectively). No significant correlation was found between oxidative stress parameters and melasma severity.8

Choubey et al9 found that serum malondialdehyde (an end product of lipid peroxidation), superoxide dismutase, and glutathione peroxidase levels were significantly higher in the melasma group (n=50) compared with the control group (n=50)(all P<.001). In addition, a significant positive correlation (correlation coefficient, +0.307; P<.05) was found between serum malondialdehyde levels and melasma severity. The mean age (SD) of the patients was 32.22 (6.377) years, and the female (n=41) to male (n=9) ratio was 4.55:1. The most common melasma pattern was centrofacial, followed by malar.9

In a study with 50 melasma patients and 50 controls, Rahimi et al10 examined bilirubin and uric acid levels, which are major extracellular antioxidants. The mean age (SD) at disease onset was 32.6 (6.7) years, and the mean MASI score (SD) was 18.1 (9). Serum bilirubin levels were found to be higher in the melasma group than in the control group and were correlated with disease severity. No significant difference in uric acid levels was found between the groups, and no correlation was found between MASI score and bilirubin and uric acid levels.10

In our study, the melasma group was similar to those in other reportsin the literature regarding gender distribution, mean age, and melasma pattern.7-10 Additionally, the correlation of mMASI score with disulfide/native thiol and disulfide/total thiol values in the melasma group suggested that oxidative stress also is correlated with melasma severity.

Thiol-based treatments such as n-acetyl cysteine, which contains a thiol compound, may be helpful in melasma.20 In a double-blind, placebo-controlled study, topical n-acetyl cysteine combined with hydroquinone 2% was used in 10 female patients with melasma. Mild to strong bleaching of the skin was observed in 90% (9/10) of the patients.21 Systemic use of n-acetyl cysteine in melasma also may be a potential research topic.

Major limitations of our study were the small sample size and lack of measurement of oxidative stress parameters in the skin concurrently with serum.

Conclusion

In our study, the presence of oxidative stress in melasma was demonstrated by evaluating thiol/disulfide homeostasis—one of the strongest markers of oxidative stress. Oxidative stress also correlated with melasma disease severity in our analysis. The data obtained in this study may contribute to understanding the etiopathogenesis of melasma and may open new horizons in treatment; however, more comprehensive studies should be conducted to support our findings.

 

gowribriphosaslaswarucusparucrocubiseruclajowrothageclumaslabotenochukanethepronechaphijiuuclakiwruslaclusleshistospasophojephu

modrawespospuvuravachacrojogistewowospuwuviphohastos

ph

References
  1. Handel AC, Miot LD, Miot HA. Melasma: a clinical and epidemiological review. An Bras Dermatol. 2014;89:771-782.
  2. Tamega Ade A, Miot LD, Bonfietti C, et al. Clinical patterns and epidemiological characteristics of facial melasma in Brazilian women. J Eur Acad Dermatol Venereol. 2013;27:151-156.
  3. Rajanala S, Maymone MBC, Vashi NA. Melasma pathogenesis: a review of the latest research, pathological findings, and investigational therapies. Dermatol Online J. 2019;25:13030/qt47b7r28c.
  4. Abou-Taleb DA, Ibrahim AK, Youssef EM, et al. Reliability, validity, and sensitivity to change overtime of the modified melasma area and severity index score. Dermatol Surg. 2017;43:210-217.
  5. Katiyar S, Yadav D. Correlation of oxidative stress with melasma: an overview. Curr Pharm Des. 2022;28:225-231.
  6. Mahmoud BH, Ruvolo E, Hexsel CL, et al. Impact of long-wavelength UVA and visible light on melanocompetent skin. J Invest Dermatol. 2010;130:2092-2097.
  7. Seçkin HY, Kalkan G, Bas¸ Y, et al. Oxidative stress status in patients with melasma. Cutan Ocul Toxicol. 2014;33:212-217.
  8. Sarkar R, Devadasan S, Choubey V, et al. Melatonin and oxidative stress in melasma—an unexplored territory; a prospective study. Int J Dermatol. 2020;59:572-575.
  9. Choubey V, Sarkar R, Garg V, et al. Role of oxidative stress in melasma: a prospective study on serum and blood markers of oxidative stress in melasma patients. Int J Dermatol. 2017;56:939-943.
  10. Rahimi H, Mirnezami M, Yazdabadi A. Bilirubin as a new antioxidant in melasma. J Cosmet Dermatol. 2022;21:5800-5803.
  11. Emre S, Kalkan G, Erdog˘an S, et al. Dynamic thiol/disulfide balance in patients with seborrheic dermatitis: a case-control study. Saudi J Med Med Sci. 2020;8:12-16.
  12. Erel Ö, Erdog˘an S. Thiol-disulfide homeostasis: an integrated approach with biochemical and clinical aspects. Turk J Med Sci. 2020;50:1728-1738.
  13. Pandya AG, Hynan LS, Bhore R, et al. Reliability assessment and validation of the Melasma Area and Severity Index (MASI) and a new modified MASI scoring method. J Am Acad Dermatol. 2011;64:78-83, 83.E1-E2.
  14. Erel O, Neselioglu S. A novel and automated assay for thiol/disulphide homeostasis. Clin Biochem. 2014;47:326-332.
  15. Guzelcicek A, Cakirca G, Erel O, et al. Assessment of thiol/disulfide balance as an oxidative stress marker in children with β-thalassemia major. Pak J Med Sci. 2019;35:161-165.
  16. Georgescu SR, Mitran CI, Mitran MI, et al. Thiol-Disulfide homeostasis in skin diseases. J Clin Med. 2022;11:1507.
  17. Üstüner P, Balevi A, Özdemir M, et al. The role of thiol/disulfide homeostasis in psoriasis: can it be a new marker for inflammation? Turk Arch Dermatol Venereol. 2018;52:120-125.
  18. Karacan G, Ercan N, Bostanci I, et al. A novel oxidative stress marker of atopic dermatitis in infants: Thiol–disulfide balance. Arch Dermatol Res. 2020;312:697-703.
  19. Demir Pektas S, Cinar N, Pektas G, et al. Thiol/disulfide homeostasis and its relationship with insulin resistance in patients with rosacea. J Cosmet Dermatol. 2021;11:14477.
  20. Adil M, Amin SS, Mohtashim M. N-acetylcysteine in dermatology. Indian J Dermatol Venereol Leprol. 2018;84:652-659.
  21. Njoo MD, Menke HE, Pavel W, et al. N-acetylcysteine as a bleaching agent in the treatment of melasma. J Eur Acad Dermatol Venereol. 1997;9:86-87.
Article PDF
Author and Disclosure Information

Drs. Erduran, Hayran, Eren, and  Iyidal are from Ankara Bilkent City Hospital, Turkey. Drs. Erduran, Hayran, and Iyidal are from the Department of Dermatology, and Dr. Eren is from the Department of Medical Biochemistry. Drs. Emre and Erel are from Ankara Yıldırım Beyazıt University Faculty of Medicine, Turkey. Dr. Emre is from the Department of Dermatology, and Dr. Erel is from the Department of Medical Biochemistry.

The authors report no conflict of interest.

The eTables are available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Funda Erduran, MD, Ankara Bilkent City Hospital, Department of Dermatology, Üniversiteler Mah, Çankaya, Ankara, 06800, Turkey (fnderdrn@mail.com).

Cutis. 2024 June;113(6):264-268, E6-E7. doi:10.12788/cutis.1036

Issue
Cutis - 113(6)
Publications
Topics
Page Number
264-268
Sections
Author and Disclosure Information

Drs. Erduran, Hayran, Eren, and  Iyidal are from Ankara Bilkent City Hospital, Turkey. Drs. Erduran, Hayran, and Iyidal are from the Department of Dermatology, and Dr. Eren is from the Department of Medical Biochemistry. Drs. Emre and Erel are from Ankara Yıldırım Beyazıt University Faculty of Medicine, Turkey. Dr. Emre is from the Department of Dermatology, and Dr. Erel is from the Department of Medical Biochemistry.

The authors report no conflict of interest.

The eTables are available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Funda Erduran, MD, Ankara Bilkent City Hospital, Department of Dermatology, Üniversiteler Mah, Çankaya, Ankara, 06800, Turkey (fnderdrn@mail.com).

Cutis. 2024 June;113(6):264-268, E6-E7. doi:10.12788/cutis.1036

Author and Disclosure Information

Drs. Erduran, Hayran, Eren, and  Iyidal are from Ankara Bilkent City Hospital, Turkey. Drs. Erduran, Hayran, and Iyidal are from the Department of Dermatology, and Dr. Eren is from the Department of Medical Biochemistry. Drs. Emre and Erel are from Ankara Yıldırım Beyazıt University Faculty of Medicine, Turkey. Dr. Emre is from the Department of Dermatology, and Dr. Erel is from the Department of Medical Biochemistry.

The authors report no conflict of interest.

The eTables are available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Funda Erduran, MD, Ankara Bilkent City Hospital, Department of Dermatology, Üniversiteler Mah, Çankaya, Ankara, 06800, Turkey (fnderdrn@mail.com).

Cutis. 2024 June;113(6):264-268, E6-E7. doi:10.12788/cutis.1036

Article PDF
Article PDF

Melasma is an acquired hyperpigmentation disorder characterized by irregular brown macules and patches that usually appear on sun-exposed areas of the skin. The term melasma originates from the Greek word melas meaning black.1 Facial melasma is divided into 2 groups according to its clinical distribution: centrofacial lesions are located in the center of the face (eg, the glabellar, frontal, nasal, zygomatic, upper lip, chin areas), and peripheral lesions manifest on the frontotemporal, preauricular, and mandibular regions.1,2 There is debate on the categorization of zygomatic (or malar) melasma; some researchers argue it should be categorized independent of other areas, while others include malar melasma in the centrofacial group because of its frequent association with the centrofacial type, especially with glabellar lesions.2 Mandibular melasma is rare and occurs mostly in postmenopausal women after intense sun exposure.1,2 Although the etiopathogenesis of the disease is not clearly known, increased melanogenesis, extracellular matrix alterations, inflammation, and angiogenesis are assumed to play a role.3 Various risk factors such as genetic predisposition, UV radiation (UVR) exposure, pregnancy, thyroid dysfunction, and exogenous hormones (eg, oral contraceptives, hormone replacement therapy) have been identified; phototoxic drugs, anticonvulsants, and some cosmetics also have been implicated.4,5 Exposure to UVR is thought to be the main triggering environmental factor by inducing both melanin production and oxidative stress.5 However, it also has been shown that visible light can induce hyperpigmentation in darker skin types.6

The presence of oxidative stress in melasma recently has become an intriguing topic of interest. First, the presence of oxidative stress in the etiopathogenesis of melasma was thought to be based on the effectiveness of antioxidants in treatment. A few studies also have confirmed the presence of oxidative stress in melasma.7-10 Classically, oxidative stress can be described as a disturbance in the balance between oxidants and antioxidants. Reactive oxygen species (ROS) are highly reactive molecules due to the unpaired electrons in their structure. Although ROS are present at low levels in physiologic conditions and are involved in critical physiologic events, they damage cellular components such as fat, protein, and nucleic acid at high concentrations.5

Dynamic thiol/disulfide homeostasis is one of the most important markers of oxidative stress in biological systems. Thiols are organic compounds containing a sulfhydryl (-SH) group. Thiols are considered highly potent antioxidants because they reduce unstable free radicals by donating electrons. They are the first antioxidants to be depleted in an oxidative environment.11,12 In case of oxidative stress, they transform into reversible forms called disulfide bridges between 2 thiol groups. Disulfide bridges can be reduced back to thiol groups, which is how dynamic thiol/disulfide homeostasis is maintained. Dynamic thiol/disulfide homeostasis is responsible for cellular events such as antioxidant defense, signal transduction, regulation of enzyme function, and apoptosis.11,12

The aim of this study was to evaluate the presence of oxidative stress in melasma by comparing dynamic thiol/disulfide homeostasis in patients with melasma compared with age- and sex-matched healthy controls.

Materials and Methods

Participants and Eligibility Criteria—We conducted a prospective study in a tertiary-care hospital (Ankara Bilkent City Hospital [Ankara, Turkey]) of patients with melasma who were followed from October 2021 to October 2022 compared with age- and sex-matched healthy volunteers. Ethics committee approval was obtained from Ankara Bilkent City Hospital before the study (E2-21-881)(13.10.2021). Written informed consent was obtained from all participants, and all were older than 18 years. Patients were excluded if there was the presence of any systemic disease or dermatologic disease other than melasma; smoking or alcohol use; any use of vitamins, food supplements, or any medication in the last 3 months; or pregnancy.

Melasma Severity—The modified melasma area and severity index (mMASI) score was used to determine the severity of melasma. The score is calculated from assessments of the darkness of the pigmentation and the percentage of affected area on the face. The mMASI score is the sum of the darkness score (D); area score (A); and separate fixed coefficients for the forehead, as well as the right malar, left malar, and chin regions.13 The mMASI score, with a range of 0 to 24, is a reliable and objective marker in the calculation of melasma severity.4

Biochemical Analysis of Samples—The 6-cc peripheral fasting venous blood samples obtained from the study participants were centrifuged at 1500 g for 10 minutes, and the separated sera were stored in a freezer at 80 °C until the time of analysis. When the study was completed, the disulfide and thiol values were analyzed. Serum native and total thiol concentrations indicating thiol/disulfide homeostasis were calculated by a new fully automatic colorimetric method developed by Erel and Neselioglu.14 Using this method, short disulfide bonds are first reduced with sodium borohydride solution to form free-functional thiol groups, and then the unused sodium borohydride is removed using formaldehyde. Finally, all thiol groups are reacted with 5,5’-dithiobis-(2-nitrobenzoic) acid (Ellman reagent), and all thiol groups are detected after reaction with 5,5’-dithiobis-(2-nitrobenzoic) acid. When a disulfide bond (SS) is reduced, 2 thiol groups are formed. For this reason, half of the difference between total thiol (-SH + the amount of thiol formed by the reduction of disulfides) and native thiol (-SH) corresponds to the dynamic disulfide amount (total thiol − native thiol/2).14

Statistical Analysis—Statistical analysis was performed using SPSS software (version 24.0). Descriptive statistics were presented as numbers and percentages for categorical variables, and numerical variables were presented as mean, SD, median, minimum, maximum, 25th quartile, and 75th quartile. The conformity of the variables to normal distribution was examined using visual (histograms and probability plots) and analytical methods (Kolmogorov-Smirnov/Shapiro-Wilk tests). In pairwise group comparisons for numerical variables, a Mann-Whitney U test was used when normal distribution was not met, and a t test was used when normal distribution was met. The statistical significance level was accepted as P<.05.

Results

Our study included 67 patients with melasma and 41 healthy age- and sex-matched controls. Of the participants with melasma, 60 (89.5%) were female and 7 (10.5%) were male. The control group was similar to the melasma group in terms of sex (87.8% female vs 12.2% male [P=.59]). The mean age (SD) was 33.1 (6.7) years in the melasma group and 31.9 (6.7) years in the control group. Age was similar across both groups (P=.41). All participants were of Asian race, and Fitzpatrick skin types (types II–IV) were similar across both groups.

Fifty-four (80.6%) participants had centrofacial melasma and 13 (19.4%) had mixed-type melasma. The mMASI score ranged from 3 to 20; the mean (SD) mMASI score was 11.28 (3.2). Disease duration ranged from 2 to 72 months; the mean (SD) disease duration was 12.26 (6.3) months. The demographics and clinical characteristics of the study group are shown in eTable 1.

eTable 2 provides a summary of disulfide, native thiol, and total thiol levels, as well as disulfide/native thiol, disulfide/total thiol, and native thiol/total thiol ratios in the study population. Disulfide/native thiol and disulfide/total thiol ratios were higher in melasma patients (Figure 1), whereas the native thiol/total thiol ratio was higher in the control group (P=.025, P=.025, and P=.026, respectively).

All correlations between age, disease duration, and mMASI scores and disulfide, native thiol, and total thiol levels, as well as disulfide/native thiol, disulfide/total thiol, and native thiol/total thiol ratios, are summarized in eTable 3. No significant correlation was observed between age and disease duration and disulfide, native thiol, and total thiol levels or disulfide/native thiol, disulfide/total thiol, and native thiol/total thiol ratios.

We independently assessed whether Fitzpatrick skin types II, III, and IV exhibited distinct levels of oxidative stress in clinical melasma. There were no significant correlations with Fitzpatrick skin type (disulfide/native thiol, P=.25; disulfide/total thiol, P=.19). We further evaluated if the thiol/disulfide parameters were correlated with duration of melasma by dividing the melasma patients into 3 groups (<6 months [n=12], 6–18 months [n=32], >18 months [n=23]), but there was not any significant correlation (disulfide/native thiol, P=.15; disulfide/total thiol, P=.15). We also divided our patients into 3 groups according to age (<27 years [n=14], 27–36 years [n=33], >36 years [n=20]). There was no correlation of the parameters with age (disulfide/native thiol, P=.15; disulfide/total thiol, P=.14).

There was a positive correlation between mMASI score and disulfide, native thiol, and total thiol levels and disulfide/native thiol and disulfide/total thiol ratios, as well as a negative correlation between mMASI score and native thiol/total thiol ratio. The correlations between mMASI scores and disulfide/native thiol and disulfide/total thiol ratios are shown in Figure 2 and eTable 3.

Comment

Melasma is a common condition that may cause psychosocial problems in affected patients and negatively affect quality of life.1 It occurs in all races but is more common in individuals with darker skin types (eg, Fitzpatrick skin types III and IV). Although melasma is more common in women during reproductive years (50%–70%), it also has been observed in 10% to 30% of men.5

Treatment options include topical bleaching agents, chemical peels, and laser therapy, as well as discontinuation of medications that may potentially trigger melasma; use of broad-spectrum sunscreens also is recommended.4 Vitamins A, C, and E, as well as niacinamide, are used in the treatment of melasma, especially for their antioxidant properties. The key role of antioxidants in the treatment of melasma supports the importance of oxidative stress in the pathogenesis.7 Melasma often is challenging to treat, particularly the mixed or dermal types, due to their stubborn nature. This condition poses a considerable therapeutic challenge for dermatologists.4

kocradrupathedriphuhatriwebripudrudrimechametehosothahapedaslihebulalakathapuretumod
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%2C%20Disulfide%2Fnative%20thiol%20homeostasis%20parameters%20in%20participants%20with%20melasma%20and%20controls.%20B%2C%20Disulfide%2Ftotal%20thiol%20homeostasis%20parameters%20in%20participants%20with%20melasma%20and%20controls.%20Higher%20scores%20indicate%20that%20in%20patients%20with%20melasma%2C%20oxidative%20stress%20shifts%20the%20thiol%2F%20disulfide%20balance%20to%20disulfide%20formation%2C%20causing%20thiols%20to%20oxidize%20into%20disulfide%20bonds.%20The%20horizontal%20bar%20inside%20the%20boxes%20indicates%20the%20mean%2C%20and%20the%20lower%20and%20upper%20ends%20of%20the%20boxes%20are%20the%2025th%20and%2075th%20quartiles.%20The%20whiskers%20indicate%20the%20range%20of%20the%20parameters%20of%20thiol%2Fdisulfide%20homeostasis.%20Asterisk%20indicates%20P%3D.025.%3C%2Fp%3E

pimifrumafricrijeuamostoshimubarocrobiwegospogotrikesheclouoswivuvuhonucohasuthujestaspimocradifrutrekuthubrebrocrospichukalifrahalaceputhalacruswospahapheswi
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20A%2C%20Correlations%20between%20modified%20melasma%20area%20and%20severity%20index%20(mMASI)%20scores%20and%20disulfide%2Fnative%20thiol%20ratios%20(P%26lt%3B.001%3B%20r%3D0.42).%20B%2C%20Correlations%20between%20mMASI%20scores%20and%20disulfide%2Ftotal%20thiol%20ratios%20(P%26lt%3B.001%3B%20r%3D0.42).%20The%20correlation%20of%20mMASI%20scores%20with%20disulfide%2Fnative%20thiol%20and%20disulfide%2Ftotal%20thiol%20values%20in%20the%20melasma%20group%20indicates%20that%20oxidative%20stress%20is%20linked%20to%20melasma%20severity.%20The%20red%20diagonal%20lines%20indicate%20correlation%2C%20showing%20that%20as%20one%20value%20increases%2C%20the%20other%20also%20increases.%3C%2Fp%3E

Oxidative stress and oxidant-antioxidant imbalance previously have been studied in various diseases, but research investigating the presence of oxidative stress in melasma are limited.7-10 Exposure of the skin to polluted air and intense UVR, as well as some food by-products, cosmetics, and drugs (eg, oral contraceptives), can directly or indirectly cause ROS production in the skin. Reactive oxygen species are thought to be involved in the pathophysiology of melasma by affecting apoptotic pathways and causing cell proliferation. The intermediate heme pathway has pro-oxidant effects and produces ROS and metabolites such as redox-active quinines. Exposure to UVR leads to the generation of ROS, highlighting the role of oxidative stress in the onset of melasma. 5

In any cutaneous disease in which oxidative stress plays a role, oxidant and antioxidant levels may be expected to vary both locally and systemically; however, measurement of oxidative stress markers in serum instead of skin is technically and economically more advantageous.8 Firstly, serum collection is less invasive and technically simpler than skin biopsies. Drawing blood is a routine procedure that requires minimal specialized equipment and training compared to the extraction and processing of skin samples. Secondly, analyzing serum samples generally is less expensive than processing skin tissue.8

In our study, we evaluated dynamic thiol/disulfide homeostasis in serum to investigate the presence of oxidative stress in the setting of melasma. Functional sulfhydryl (-SH) groups in thiols act as substrates for antioxidant enzymes and as free-radical scavengers. They constitute one of the most powerful defense systems against the unwanted effects of ROS. Thiols, which become the main target of ROS under oxidative stress, oxidize with oxidant molecules and form disulfide bridges.15

Thiol/disulfide homeostasis has been studied many times in dermatologic diseases,16-19 and the results obtained from these studies are heterogenous depending on the extent of oxidative damage. It has been shown that thiol/disulfide homeostasis plays a role in oxidative stress in conditions such as psoriasis,17 seborrheic dermatitis,11 atopic dermatitits,18 and rosacea.19 In our study, disulfide/native thiol and disulfide/total thiol levels were significantly higher (both P=.025) in the melasma group compared with the control group, which indicates that the thiol/disulfide balance in patients with melasma is shifted to disulfide formation and thiols are oxidized to disulfide bonds in the presence of oxidative stress.

Seçkin et al7 evaluated the role of oxidative stress in the pathogenesis of melasma and found that the serum levels of the antioxidants superoxide dismutase and glutathione peroxidase were significantly higher in the patient group compared with the control group (both P<.001). They also found that the levels of nitric oxide (another antioxidant) were increased in the patient group and the levels of protein carbonyl (an oxidative metabolite) were significantly lower (both P<.001). These findings indicated that free-radical damage may be involved in the pathogenesis of melasma.7

In a study of 75 patients with melasma, serum levels of the antioxidants melatonin and catalase were significantly (P<.001 and P=.001, respectively) lower in the melasma group compared with the control group, while serum levels of the oxidants protein carbonyl and nitric oxide were significantly higher (P=.002 and P=.001, respectively). No significant correlation was found between oxidative stress parameters and melasma severity.8

Choubey et al9 found that serum malondialdehyde (an end product of lipid peroxidation), superoxide dismutase, and glutathione peroxidase levels were significantly higher in the melasma group (n=50) compared with the control group (n=50)(all P<.001). In addition, a significant positive correlation (correlation coefficient, +0.307; P<.05) was found between serum malondialdehyde levels and melasma severity. The mean age (SD) of the patients was 32.22 (6.377) years, and the female (n=41) to male (n=9) ratio was 4.55:1. The most common melasma pattern was centrofacial, followed by malar.9

In a study with 50 melasma patients and 50 controls, Rahimi et al10 examined bilirubin and uric acid levels, which are major extracellular antioxidants. The mean age (SD) at disease onset was 32.6 (6.7) years, and the mean MASI score (SD) was 18.1 (9). Serum bilirubin levels were found to be higher in the melasma group than in the control group and were correlated with disease severity. No significant difference in uric acid levels was found between the groups, and no correlation was found between MASI score and bilirubin and uric acid levels.10

In our study, the melasma group was similar to those in other reportsin the literature regarding gender distribution, mean age, and melasma pattern.7-10 Additionally, the correlation of mMASI score with disulfide/native thiol and disulfide/total thiol values in the melasma group suggested that oxidative stress also is correlated with melasma severity.

Thiol-based treatments such as n-acetyl cysteine, which contains a thiol compound, may be helpful in melasma.20 In a double-blind, placebo-controlled study, topical n-acetyl cysteine combined with hydroquinone 2% was used in 10 female patients with melasma. Mild to strong bleaching of the skin was observed in 90% (9/10) of the patients.21 Systemic use of n-acetyl cysteine in melasma also may be a potential research topic.

Major limitations of our study were the small sample size and lack of measurement of oxidative stress parameters in the skin concurrently with serum.

Conclusion

In our study, the presence of oxidative stress in melasma was demonstrated by evaluating thiol/disulfide homeostasis—one of the strongest markers of oxidative stress. Oxidative stress also correlated with melasma disease severity in our analysis. The data obtained in this study may contribute to understanding the etiopathogenesis of melasma and may open new horizons in treatment; however, more comprehensive studies should be conducted to support our findings.

 

gowribriphosaslaswarucusparucrocubiseruclajowrothageclumaslabotenochukanethepronechaphijiuuclakiwruslaclusleshistospasophojephu

modrawespospuvuravachacrojogistewowospuwuviphohastos

ph

Melasma is an acquired hyperpigmentation disorder characterized by irregular brown macules and patches that usually appear on sun-exposed areas of the skin. The term melasma originates from the Greek word melas meaning black.1 Facial melasma is divided into 2 groups according to its clinical distribution: centrofacial lesions are located in the center of the face (eg, the glabellar, frontal, nasal, zygomatic, upper lip, chin areas), and peripheral lesions manifest on the frontotemporal, preauricular, and mandibular regions.1,2 There is debate on the categorization of zygomatic (or malar) melasma; some researchers argue it should be categorized independent of other areas, while others include malar melasma in the centrofacial group because of its frequent association with the centrofacial type, especially with glabellar lesions.2 Mandibular melasma is rare and occurs mostly in postmenopausal women after intense sun exposure.1,2 Although the etiopathogenesis of the disease is not clearly known, increased melanogenesis, extracellular matrix alterations, inflammation, and angiogenesis are assumed to play a role.3 Various risk factors such as genetic predisposition, UV radiation (UVR) exposure, pregnancy, thyroid dysfunction, and exogenous hormones (eg, oral contraceptives, hormone replacement therapy) have been identified; phototoxic drugs, anticonvulsants, and some cosmetics also have been implicated.4,5 Exposure to UVR is thought to be the main triggering environmental factor by inducing both melanin production and oxidative stress.5 However, it also has been shown that visible light can induce hyperpigmentation in darker skin types.6

The presence of oxidative stress in melasma recently has become an intriguing topic of interest. First, the presence of oxidative stress in the etiopathogenesis of melasma was thought to be based on the effectiveness of antioxidants in treatment. A few studies also have confirmed the presence of oxidative stress in melasma.7-10 Classically, oxidative stress can be described as a disturbance in the balance between oxidants and antioxidants. Reactive oxygen species (ROS) are highly reactive molecules due to the unpaired electrons in their structure. Although ROS are present at low levels in physiologic conditions and are involved in critical physiologic events, they damage cellular components such as fat, protein, and nucleic acid at high concentrations.5

Dynamic thiol/disulfide homeostasis is one of the most important markers of oxidative stress in biological systems. Thiols are organic compounds containing a sulfhydryl (-SH) group. Thiols are considered highly potent antioxidants because they reduce unstable free radicals by donating electrons. They are the first antioxidants to be depleted in an oxidative environment.11,12 In case of oxidative stress, they transform into reversible forms called disulfide bridges between 2 thiol groups. Disulfide bridges can be reduced back to thiol groups, which is how dynamic thiol/disulfide homeostasis is maintained. Dynamic thiol/disulfide homeostasis is responsible for cellular events such as antioxidant defense, signal transduction, regulation of enzyme function, and apoptosis.11,12

The aim of this study was to evaluate the presence of oxidative stress in melasma by comparing dynamic thiol/disulfide homeostasis in patients with melasma compared with age- and sex-matched healthy controls.

Materials and Methods

Participants and Eligibility Criteria—We conducted a prospective study in a tertiary-care hospital (Ankara Bilkent City Hospital [Ankara, Turkey]) of patients with melasma who were followed from October 2021 to October 2022 compared with age- and sex-matched healthy volunteers. Ethics committee approval was obtained from Ankara Bilkent City Hospital before the study (E2-21-881)(13.10.2021). Written informed consent was obtained from all participants, and all were older than 18 years. Patients were excluded if there was the presence of any systemic disease or dermatologic disease other than melasma; smoking or alcohol use; any use of vitamins, food supplements, or any medication in the last 3 months; or pregnancy.

Melasma Severity—The modified melasma area and severity index (mMASI) score was used to determine the severity of melasma. The score is calculated from assessments of the darkness of the pigmentation and the percentage of affected area on the face. The mMASI score is the sum of the darkness score (D); area score (A); and separate fixed coefficients for the forehead, as well as the right malar, left malar, and chin regions.13 The mMASI score, with a range of 0 to 24, is a reliable and objective marker in the calculation of melasma severity.4

Biochemical Analysis of Samples—The 6-cc peripheral fasting venous blood samples obtained from the study participants were centrifuged at 1500 g for 10 minutes, and the separated sera were stored in a freezer at 80 °C until the time of analysis. When the study was completed, the disulfide and thiol values were analyzed. Serum native and total thiol concentrations indicating thiol/disulfide homeostasis were calculated by a new fully automatic colorimetric method developed by Erel and Neselioglu.14 Using this method, short disulfide bonds are first reduced with sodium borohydride solution to form free-functional thiol groups, and then the unused sodium borohydride is removed using formaldehyde. Finally, all thiol groups are reacted with 5,5’-dithiobis-(2-nitrobenzoic) acid (Ellman reagent), and all thiol groups are detected after reaction with 5,5’-dithiobis-(2-nitrobenzoic) acid. When a disulfide bond (SS) is reduced, 2 thiol groups are formed. For this reason, half of the difference between total thiol (-SH + the amount of thiol formed by the reduction of disulfides) and native thiol (-SH) corresponds to the dynamic disulfide amount (total thiol − native thiol/2).14

Statistical Analysis—Statistical analysis was performed using SPSS software (version 24.0). Descriptive statistics were presented as numbers and percentages for categorical variables, and numerical variables were presented as mean, SD, median, minimum, maximum, 25th quartile, and 75th quartile. The conformity of the variables to normal distribution was examined using visual (histograms and probability plots) and analytical methods (Kolmogorov-Smirnov/Shapiro-Wilk tests). In pairwise group comparisons for numerical variables, a Mann-Whitney U test was used when normal distribution was not met, and a t test was used when normal distribution was met. The statistical significance level was accepted as P<.05.

Results

Our study included 67 patients with melasma and 41 healthy age- and sex-matched controls. Of the participants with melasma, 60 (89.5%) were female and 7 (10.5%) were male. The control group was similar to the melasma group in terms of sex (87.8% female vs 12.2% male [P=.59]). The mean age (SD) was 33.1 (6.7) years in the melasma group and 31.9 (6.7) years in the control group. Age was similar across both groups (P=.41). All participants were of Asian race, and Fitzpatrick skin types (types II–IV) were similar across both groups.

Fifty-four (80.6%) participants had centrofacial melasma and 13 (19.4%) had mixed-type melasma. The mMASI score ranged from 3 to 20; the mean (SD) mMASI score was 11.28 (3.2). Disease duration ranged from 2 to 72 months; the mean (SD) disease duration was 12.26 (6.3) months. The demographics and clinical characteristics of the study group are shown in eTable 1.

eTable 2 provides a summary of disulfide, native thiol, and total thiol levels, as well as disulfide/native thiol, disulfide/total thiol, and native thiol/total thiol ratios in the study population. Disulfide/native thiol and disulfide/total thiol ratios were higher in melasma patients (Figure 1), whereas the native thiol/total thiol ratio was higher in the control group (P=.025, P=.025, and P=.026, respectively).

All correlations between age, disease duration, and mMASI scores and disulfide, native thiol, and total thiol levels, as well as disulfide/native thiol, disulfide/total thiol, and native thiol/total thiol ratios, are summarized in eTable 3. No significant correlation was observed between age and disease duration and disulfide, native thiol, and total thiol levels or disulfide/native thiol, disulfide/total thiol, and native thiol/total thiol ratios.

We independently assessed whether Fitzpatrick skin types II, III, and IV exhibited distinct levels of oxidative stress in clinical melasma. There were no significant correlations with Fitzpatrick skin type (disulfide/native thiol, P=.25; disulfide/total thiol, P=.19). We further evaluated if the thiol/disulfide parameters were correlated with duration of melasma by dividing the melasma patients into 3 groups (<6 months [n=12], 6–18 months [n=32], >18 months [n=23]), but there was not any significant correlation (disulfide/native thiol, P=.15; disulfide/total thiol, P=.15). We also divided our patients into 3 groups according to age (<27 years [n=14], 27–36 years [n=33], >36 years [n=20]). There was no correlation of the parameters with age (disulfide/native thiol, P=.15; disulfide/total thiol, P=.14).

There was a positive correlation between mMASI score and disulfide, native thiol, and total thiol levels and disulfide/native thiol and disulfide/total thiol ratios, as well as a negative correlation between mMASI score and native thiol/total thiol ratio. The correlations between mMASI scores and disulfide/native thiol and disulfide/total thiol ratios are shown in Figure 2 and eTable 3.

Comment

Melasma is a common condition that may cause psychosocial problems in affected patients and negatively affect quality of life.1 It occurs in all races but is more common in individuals with darker skin types (eg, Fitzpatrick skin types III and IV). Although melasma is more common in women during reproductive years (50%–70%), it also has been observed in 10% to 30% of men.5

Treatment options include topical bleaching agents, chemical peels, and laser therapy, as well as discontinuation of medications that may potentially trigger melasma; use of broad-spectrum sunscreens also is recommended.4 Vitamins A, C, and E, as well as niacinamide, are used in the treatment of melasma, especially for their antioxidant properties. The key role of antioxidants in the treatment of melasma supports the importance of oxidative stress in the pathogenesis.7 Melasma often is challenging to treat, particularly the mixed or dermal types, due to their stubborn nature. This condition poses a considerable therapeutic challenge for dermatologists.4

kocradrupathedriphuhatriwebripudrudrimechametehosothahapedaslihebulalakathapuretumod
%3Cp%3E%3Cstrong%3EFIGURE%201.%3C%2Fstrong%3E%20A%2C%20Disulfide%2Fnative%20thiol%20homeostasis%20parameters%20in%20participants%20with%20melasma%20and%20controls.%20B%2C%20Disulfide%2Ftotal%20thiol%20homeostasis%20parameters%20in%20participants%20with%20melasma%20and%20controls.%20Higher%20scores%20indicate%20that%20in%20patients%20with%20melasma%2C%20oxidative%20stress%20shifts%20the%20thiol%2F%20disulfide%20balance%20to%20disulfide%20formation%2C%20causing%20thiols%20to%20oxidize%20into%20disulfide%20bonds.%20The%20horizontal%20bar%20inside%20the%20boxes%20indicates%20the%20mean%2C%20and%20the%20lower%20and%20upper%20ends%20of%20the%20boxes%20are%20the%2025th%20and%2075th%20quartiles.%20The%20whiskers%20indicate%20the%20range%20of%20the%20parameters%20of%20thiol%2Fdisulfide%20homeostasis.%20Asterisk%20indicates%20P%3D.025.%3C%2Fp%3E

pimifrumafricrijeuamostoshimubarocrobiwegospogotrikesheclouoswivuvuhonucohasuthujestaspimocradifrutrekuthubrebrocrospichukalifrahalaceputhalacruswospahapheswi
%3Cp%3E%3Cstrong%3EFIGURE%202.%3C%2Fstrong%3E%20A%2C%20Correlations%20between%20modified%20melasma%20area%20and%20severity%20index%20(mMASI)%20scores%20and%20disulfide%2Fnative%20thiol%20ratios%20(P%26lt%3B.001%3B%20r%3D0.42).%20B%2C%20Correlations%20between%20mMASI%20scores%20and%20disulfide%2Ftotal%20thiol%20ratios%20(P%26lt%3B.001%3B%20r%3D0.42).%20The%20correlation%20of%20mMASI%20scores%20with%20disulfide%2Fnative%20thiol%20and%20disulfide%2Ftotal%20thiol%20values%20in%20the%20melasma%20group%20indicates%20that%20oxidative%20stress%20is%20linked%20to%20melasma%20severity.%20The%20red%20diagonal%20lines%20indicate%20correlation%2C%20showing%20that%20as%20one%20value%20increases%2C%20the%20other%20also%20increases.%3C%2Fp%3E

Oxidative stress and oxidant-antioxidant imbalance previously have been studied in various diseases, but research investigating the presence of oxidative stress in melasma are limited.7-10 Exposure of the skin to polluted air and intense UVR, as well as some food by-products, cosmetics, and drugs (eg, oral contraceptives), can directly or indirectly cause ROS production in the skin. Reactive oxygen species are thought to be involved in the pathophysiology of melasma by affecting apoptotic pathways and causing cell proliferation. The intermediate heme pathway has pro-oxidant effects and produces ROS and metabolites such as redox-active quinines. Exposure to UVR leads to the generation of ROS, highlighting the role of oxidative stress in the onset of melasma. 5

In any cutaneous disease in which oxidative stress plays a role, oxidant and antioxidant levels may be expected to vary both locally and systemically; however, measurement of oxidative stress markers in serum instead of skin is technically and economically more advantageous.8 Firstly, serum collection is less invasive and technically simpler than skin biopsies. Drawing blood is a routine procedure that requires minimal specialized equipment and training compared to the extraction and processing of skin samples. Secondly, analyzing serum samples generally is less expensive than processing skin tissue.8

In our study, we evaluated dynamic thiol/disulfide homeostasis in serum to investigate the presence of oxidative stress in the setting of melasma. Functional sulfhydryl (-SH) groups in thiols act as substrates for antioxidant enzymes and as free-radical scavengers. They constitute one of the most powerful defense systems against the unwanted effects of ROS. Thiols, which become the main target of ROS under oxidative stress, oxidize with oxidant molecules and form disulfide bridges.15

Thiol/disulfide homeostasis has been studied many times in dermatologic diseases,16-19 and the results obtained from these studies are heterogenous depending on the extent of oxidative damage. It has been shown that thiol/disulfide homeostasis plays a role in oxidative stress in conditions such as psoriasis,17 seborrheic dermatitis,11 atopic dermatitits,18 and rosacea.19 In our study, disulfide/native thiol and disulfide/total thiol levels were significantly higher (both P=.025) in the melasma group compared with the control group, which indicates that the thiol/disulfide balance in patients with melasma is shifted to disulfide formation and thiols are oxidized to disulfide bonds in the presence of oxidative stress.

Seçkin et al7 evaluated the role of oxidative stress in the pathogenesis of melasma and found that the serum levels of the antioxidants superoxide dismutase and glutathione peroxidase were significantly higher in the patient group compared with the control group (both P<.001). They also found that the levels of nitric oxide (another antioxidant) were increased in the patient group and the levels of protein carbonyl (an oxidative metabolite) were significantly lower (both P<.001). These findings indicated that free-radical damage may be involved in the pathogenesis of melasma.7

In a study of 75 patients with melasma, serum levels of the antioxidants melatonin and catalase were significantly (P<.001 and P=.001, respectively) lower in the melasma group compared with the control group, while serum levels of the oxidants protein carbonyl and nitric oxide were significantly higher (P=.002 and P=.001, respectively). No significant correlation was found between oxidative stress parameters and melasma severity.8

Choubey et al9 found that serum malondialdehyde (an end product of lipid peroxidation), superoxide dismutase, and glutathione peroxidase levels were significantly higher in the melasma group (n=50) compared with the control group (n=50)(all P<.001). In addition, a significant positive correlation (correlation coefficient, +0.307; P<.05) was found between serum malondialdehyde levels and melasma severity. The mean age (SD) of the patients was 32.22 (6.377) years, and the female (n=41) to male (n=9) ratio was 4.55:1. The most common melasma pattern was centrofacial, followed by malar.9

In a study with 50 melasma patients and 50 controls, Rahimi et al10 examined bilirubin and uric acid levels, which are major extracellular antioxidants. The mean age (SD) at disease onset was 32.6 (6.7) years, and the mean MASI score (SD) was 18.1 (9). Serum bilirubin levels were found to be higher in the melasma group than in the control group and were correlated with disease severity. No significant difference in uric acid levels was found between the groups, and no correlation was found between MASI score and bilirubin and uric acid levels.10

In our study, the melasma group was similar to those in other reportsin the literature regarding gender distribution, mean age, and melasma pattern.7-10 Additionally, the correlation of mMASI score with disulfide/native thiol and disulfide/total thiol values in the melasma group suggested that oxidative stress also is correlated with melasma severity.

Thiol-based treatments such as n-acetyl cysteine, which contains a thiol compound, may be helpful in melasma.20 In a double-blind, placebo-controlled study, topical n-acetyl cysteine combined with hydroquinone 2% was used in 10 female patients with melasma. Mild to strong bleaching of the skin was observed in 90% (9/10) of the patients.21 Systemic use of n-acetyl cysteine in melasma also may be a potential research topic.

Major limitations of our study were the small sample size and lack of measurement of oxidative stress parameters in the skin concurrently with serum.

Conclusion

In our study, the presence of oxidative stress in melasma was demonstrated by evaluating thiol/disulfide homeostasis—one of the strongest markers of oxidative stress. Oxidative stress also correlated with melasma disease severity in our analysis. The data obtained in this study may contribute to understanding the etiopathogenesis of melasma and may open new horizons in treatment; however, more comprehensive studies should be conducted to support our findings.

 

gowribriphosaslaswarucusparucrocubiseruclajowrothageclumaslabotenochukanethepronechaphijiuuclakiwruslaclusleshistospasophojephu

modrawespospuvuravachacrojogistewowospuwuviphohastos

ph

References
  1. Handel AC, Miot LD, Miot HA. Melasma: a clinical and epidemiological review. An Bras Dermatol. 2014;89:771-782.
  2. Tamega Ade A, Miot LD, Bonfietti C, et al. Clinical patterns and epidemiological characteristics of facial melasma in Brazilian women. J Eur Acad Dermatol Venereol. 2013;27:151-156.
  3. Rajanala S, Maymone MBC, Vashi NA. Melasma pathogenesis: a review of the latest research, pathological findings, and investigational therapies. Dermatol Online J. 2019;25:13030/qt47b7r28c.
  4. Abou-Taleb DA, Ibrahim AK, Youssef EM, et al. Reliability, validity, and sensitivity to change overtime of the modified melasma area and severity index score. Dermatol Surg. 2017;43:210-217.
  5. Katiyar S, Yadav D. Correlation of oxidative stress with melasma: an overview. Curr Pharm Des. 2022;28:225-231.
  6. Mahmoud BH, Ruvolo E, Hexsel CL, et al. Impact of long-wavelength UVA and visible light on melanocompetent skin. J Invest Dermatol. 2010;130:2092-2097.
  7. Seçkin HY, Kalkan G, Bas¸ Y, et al. Oxidative stress status in patients with melasma. Cutan Ocul Toxicol. 2014;33:212-217.
  8. Sarkar R, Devadasan S, Choubey V, et al. Melatonin and oxidative stress in melasma—an unexplored territory; a prospective study. Int J Dermatol. 2020;59:572-575.
  9. Choubey V, Sarkar R, Garg V, et al. Role of oxidative stress in melasma: a prospective study on serum and blood markers of oxidative stress in melasma patients. Int J Dermatol. 2017;56:939-943.
  10. Rahimi H, Mirnezami M, Yazdabadi A. Bilirubin as a new antioxidant in melasma. J Cosmet Dermatol. 2022;21:5800-5803.
  11. Emre S, Kalkan G, Erdog˘an S, et al. Dynamic thiol/disulfide balance in patients with seborrheic dermatitis: a case-control study. Saudi J Med Med Sci. 2020;8:12-16.
  12. Erel Ö, Erdog˘an S. Thiol-disulfide homeostasis: an integrated approach with biochemical and clinical aspects. Turk J Med Sci. 2020;50:1728-1738.
  13. Pandya AG, Hynan LS, Bhore R, et al. Reliability assessment and validation of the Melasma Area and Severity Index (MASI) and a new modified MASI scoring method. J Am Acad Dermatol. 2011;64:78-83, 83.E1-E2.
  14. Erel O, Neselioglu S. A novel and automated assay for thiol/disulphide homeostasis. Clin Biochem. 2014;47:326-332.
  15. Guzelcicek A, Cakirca G, Erel O, et al. Assessment of thiol/disulfide balance as an oxidative stress marker in children with β-thalassemia major. Pak J Med Sci. 2019;35:161-165.
  16. Georgescu SR, Mitran CI, Mitran MI, et al. Thiol-Disulfide homeostasis in skin diseases. J Clin Med. 2022;11:1507.
  17. Üstüner P, Balevi A, Özdemir M, et al. The role of thiol/disulfide homeostasis in psoriasis: can it be a new marker for inflammation? Turk Arch Dermatol Venereol. 2018;52:120-125.
  18. Karacan G, Ercan N, Bostanci I, et al. A novel oxidative stress marker of atopic dermatitis in infants: Thiol–disulfide balance. Arch Dermatol Res. 2020;312:697-703.
  19. Demir Pektas S, Cinar N, Pektas G, et al. Thiol/disulfide homeostasis and its relationship with insulin resistance in patients with rosacea. J Cosmet Dermatol. 2021;11:14477.
  20. Adil M, Amin SS, Mohtashim M. N-acetylcysteine in dermatology. Indian J Dermatol Venereol Leprol. 2018;84:652-659.
  21. Njoo MD, Menke HE, Pavel W, et al. N-acetylcysteine as a bleaching agent in the treatment of melasma. J Eur Acad Dermatol Venereol. 1997;9:86-87.
References
  1. Handel AC, Miot LD, Miot HA. Melasma: a clinical and epidemiological review. An Bras Dermatol. 2014;89:771-782.
  2. Tamega Ade A, Miot LD, Bonfietti C, et al. Clinical patterns and epidemiological characteristics of facial melasma in Brazilian women. J Eur Acad Dermatol Venereol. 2013;27:151-156.
  3. Rajanala S, Maymone MBC, Vashi NA. Melasma pathogenesis: a review of the latest research, pathological findings, and investigational therapies. Dermatol Online J. 2019;25:13030/qt47b7r28c.
  4. Abou-Taleb DA, Ibrahim AK, Youssef EM, et al. Reliability, validity, and sensitivity to change overtime of the modified melasma area and severity index score. Dermatol Surg. 2017;43:210-217.
  5. Katiyar S, Yadav D. Correlation of oxidative stress with melasma: an overview. Curr Pharm Des. 2022;28:225-231.
  6. Mahmoud BH, Ruvolo E, Hexsel CL, et al. Impact of long-wavelength UVA and visible light on melanocompetent skin. J Invest Dermatol. 2010;130:2092-2097.
  7. Seçkin HY, Kalkan G, Bas¸ Y, et al. Oxidative stress status in patients with melasma. Cutan Ocul Toxicol. 2014;33:212-217.
  8. Sarkar R, Devadasan S, Choubey V, et al. Melatonin and oxidative stress in melasma—an unexplored territory; a prospective study. Int J Dermatol. 2020;59:572-575.
  9. Choubey V, Sarkar R, Garg V, et al. Role of oxidative stress in melasma: a prospective study on serum and blood markers of oxidative stress in melasma patients. Int J Dermatol. 2017;56:939-943.
  10. Rahimi H, Mirnezami M, Yazdabadi A. Bilirubin as a new antioxidant in melasma. J Cosmet Dermatol. 2022;21:5800-5803.
  11. Emre S, Kalkan G, Erdog˘an S, et al. Dynamic thiol/disulfide balance in patients with seborrheic dermatitis: a case-control study. Saudi J Med Med Sci. 2020;8:12-16.
  12. Erel Ö, Erdog˘an S. Thiol-disulfide homeostasis: an integrated approach with biochemical and clinical aspects. Turk J Med Sci. 2020;50:1728-1738.
  13. Pandya AG, Hynan LS, Bhore R, et al. Reliability assessment and validation of the Melasma Area and Severity Index (MASI) and a new modified MASI scoring method. J Am Acad Dermatol. 2011;64:78-83, 83.E1-E2.
  14. Erel O, Neselioglu S. A novel and automated assay for thiol/disulphide homeostasis. Clin Biochem. 2014;47:326-332.
  15. Guzelcicek A, Cakirca G, Erel O, et al. Assessment of thiol/disulfide balance as an oxidative stress marker in children with β-thalassemia major. Pak J Med Sci. 2019;35:161-165.
  16. Georgescu SR, Mitran CI, Mitran MI, et al. Thiol-Disulfide homeostasis in skin diseases. J Clin Med. 2022;11:1507.
  17. Üstüner P, Balevi A, Özdemir M, et al. The role of thiol/disulfide homeostasis in psoriasis: can it be a new marker for inflammation? Turk Arch Dermatol Venereol. 2018;52:120-125.
  18. Karacan G, Ercan N, Bostanci I, et al. A novel oxidative stress marker of atopic dermatitis in infants: Thiol–disulfide balance. Arch Dermatol Res. 2020;312:697-703.
  19. Demir Pektas S, Cinar N, Pektas G, et al. Thiol/disulfide homeostasis and its relationship with insulin resistance in patients with rosacea. J Cosmet Dermatol. 2021;11:14477.
  20. Adil M, Amin SS, Mohtashim M. N-acetylcysteine in dermatology. Indian J Dermatol Venereol Leprol. 2018;84:652-659.
  21. Njoo MD, Menke HE, Pavel W, et al. N-acetylcysteine as a bleaching agent in the treatment of melasma. J Eur Acad Dermatol Venereol. 1997;9:86-87.
Issue
Cutis - 113(6)
Issue
Cutis - 113(6)
Page Number
264-268
Page Number
264-268
Publications
Publications
Topics
Article Type
Display Headline
Oxidative Stress in Patients With Melasma: An Evaluation of the Correlation of the Thiol/Disulfide Homeostasis Parameters and Modified MASI Score
Display Headline
Oxidative Stress in Patients With Melasma: An Evaluation of the Correlation of the Thiol/Disulfide Homeostasis Parameters and Modified MASI Score
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>Erduran</fileName> <TBEID>0C02F829.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F829</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Erduran</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240610T110127</firstPublished> <LastPublished>20240610T110127</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240610T110126</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Funda Erduran, MD</byline> <bylineText>Funda Erduran, MD; Yıldız Hayran, MD; Selma Emre, MD; Funda Eren, MD; Ays¸egül Yalçınkaya I˙yidal, MD; Özcan Erel, MD</bylineText> <bylineFull>Funda Erduran, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>264-268</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Melasma is an acquired hyperpigmentation disorder characterized by irregular brown macules and patches that usually appear on sun-exposed areas of the skin. The</metaDescription> <articlePDF>301782</articlePDF> <teaserImage/> <title>Oxidative Stress in Patients With Melasma: An Evaluation of the Correlation of the Thiol/Disulfide Homeostasis Parameters and Modified MASI Score</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>June</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>6</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2161</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>June 2024</pubIssueName> <pubArticleType>Original Articles | 2161</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">104</term> </sections> <topics> <term canonical="true">276</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/1800274d.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Oxidative Stress in Patients With Melasma: An Evaluation of the Correlation of the Thiol/Disulfide Homeostasis Parameters and Modified MASI Score</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Melasma is a common acquired hyperpigmentation disorder that affects mostly women and individuals with darker skin types. Oxidative stress may play a role in the pathogenesis of melasma. Dynamic thiol/disulfide homeostasis is one of the most important indicators of oxidative stress. This study aimed to investigate the presence of oxidative stress in patients with melasma by evaluating thiol/disulfide homeostasis. Sixty-seven patients with melasma and 41 healthy age- and sex-matched controls were included in the study. Disease severity was evaluated using the modified melasma area and severity index (mMASI). Thiol/disulfide homeostasis parameters of the melasma and control groups were measured using a novel, fully automated spectrophotometric method. Our data indicated the presence of oxidative stress in melasma, which may be correlated with disease severity. Because research on the presence of oxidative stress in melasma is limited, further studies are needed to support these conclusions.</p> <p>Melasma is an acquired hyperpigmentation disorder characterized by irregular brown macules and patches that usually appear on sun-exposed areas of the skin. The term <em>melasma</em> originates from the Greek word <em>melas</em> meaning black.<sup>1</sup> Facial melasma is divided into 2 groups according to its clinical distribution: centrofacial lesions are located in the center of the face (eg, the glabellar, frontal, nasal, zygomatic, upper lip, chin areas), and peripheral lesions manifest on the frontotemporal, preauricular, and mandibular regions.<sup>1,2</sup> There is debate on the categorization of zygomatic (or malar) melasma; some researchers argue it should be categorized independent of other areas, while others include malar melasma in the centrofacial group because of its frequent association with the centrofacial type, especially with glabellar lesions.<sup>2</sup> Mandibular melasma is rare and occurs mostly in postmenopausal women after intense sun exposure.<sup>1,2</sup> Although the etiopathogenesis of the disease is not clearly known, increased melanogenesis, extracellular matrix alterations, inflammation, and angiogenesis are assumed to play a role.<sup>3</sup> Various risk factors such as genetic predisposition, UV radiation (UVR) exposure, pregnancy, thyroid dysfunction, and exogenous hormones (eg, oral contraceptives, hormone replacement therapy) have been identified; phototoxic drugs, anticonvulsants, and some cosmetics also have been implicated.<sup>4,5</sup> Exposure to UVR is thought to be the main triggering environmental factor by inducing both melanin production and oxidative stress.<sup>5</sup> However, it also has been shown that visible light can induce hyperpigmentation in darker skin types.<sup>6</sup> </p> <p>The presence of oxidative stress in melasma recently has become an intriguing topic of interest. First, the presence of oxidative stress in the etiopathogenesis of melasma was thought to be based on the effectiveness of antioxidants in treatment. A few studies also have confirmed the presence of oxidative stress in melasma.<sup>7-10</sup> Classically, oxidative stress can be described as a disturbance in the balance between oxidants and antioxidants. Reactive oxygen species (ROS) are highly reactive molecules due to the unpaired electrons in their structure. Although ROS are present at low levels in physiologic conditions and are involved in critical physiologic events, they damage cellular components such as fat, protein, and nucleic acid at high concentrations.<sup>5 <br/><br/></sup>Dynamic thiol/disulfide homeostasis is one of the most important markers of oxidative stress in biological systems. Thiols are organic compounds containing a sulfhydryl (-SH) group. Thiols are considered highly potent antioxidants because they reduce unstable free radicals by donating electrons. They are the first antioxidants to be depleted in an oxidative environment.<sup>11,12</sup> In case of oxidative stress, they transform into reversible forms called disulfide bridges between 2 thiol groups. Disulfide bridges can be reduced back to thiol groups, which is how dynamic thiol/disulfide homeostasis is maintained. Dynamic thiol/disulfide homeostasis is responsible for cellular events such as antioxidant defense, signal transduction, regulation of enzyme function, and apoptosis.<sup>11,12 <br/><br/></sup>The aim of this study was to evaluate the presence of oxidative stress in melasma by comparing dynamic thiol/disulfide homeostasis in patients with melasma compared with age- and sex-matched healthy controls. </p> <h3>Materials and Methods</h3> <p><i>Participants and Eligibility Criteria—</i>We conducted a prospective study in a tertiary-care hospital (Ankara Bilkent City Hospital [Ankara, Turkey]) of patients with melasma who were followed from October 2021 to October 2022 compared with age- and sex-matched healthy volunteers. Ethics committee approval was obtained from Ankara Bilkent City Hospital before the study (E2-21-881)(13.10.2021). Written informed consent was obtained from all participants, and all were older than 18 years. Patients were excluded if there was the presence of any systemic disease or dermatologic disease other than melasma; smoking or alcohol use; any use of vitamins, food supplements, or any medication in the last 3 months; or pregnancy. </p> <p><em>Melasma Severity</em>—The modified melasma area and severity index (mMASI) score was used to determine the severity of melasma. The score is calculated from assessments of the darkness of the pigmentation and the percentage of affected area on the face. The mMASI score is the sum of the darkness score (D); area score (A); and separate fixed coefficients for the forehead, as well as the right malar, left malar, and chin regions.<sup>13</sup> The mMASI score, with a range of 0 to 24, is a reliable and objective marker in the calculation of melasma severity.<sup>4</sup> <br/><br/><em>Biochemical Analysis of Samples</em>—The 6-cc peripheral fasting venous blood samples obtained from the study participants were centrifuged at 1500 g for 10 minutes, and the separated sera were stored in a freezer at <span class="body">−</span>80 <span class="body">°</span>C until the time of analysis. When the study was completed, the disulfide and thiol values were analyzed. Serum native and total thiol concentrations indicating thiol/disulfide homeostasis were calculated by a new fully automatic colorimetric method developed by Erel and Neselioglu.<sup>14</sup> Using this method, short disulfide bonds are first reduced with sodium borohydride solution to form free-functional thiol groups, and then the unused sodium borohydride is removed using formaldehyde. Finally, all thiol groups are reacted with 5,5’-dithiobis-(2-nitrobenzoic) acid (Ellman reagent), and all thiol groups are detected after reaction with 5,5’-dithiobis-(2-nitrobenzoic) acid. When a disulfide bond (<span class="body">−</span>S<span class="body">−</span>S<span class="body">−</span>) is reduced, 2 thiol groups are formed. For this reason, half of the difference between total thiol (-SH <span class="body">+</span> the amount of thiol formed by the reduction of disulfides) and native thiol (-SH) corresponds to the dynamic disulfide amount (total thiol − native thiol/2).<sup>14</sup> <br/><br/><em>Statistical Analysis</em>—Statistical analysis was performed using SPSS software (version 24.0). Descriptive statistics were presented as numbers and percentages for categorical variables, and numerical variables were presented as mean, SD, median, minimum, maximum, 25th quartile, and 75th quartile. The conformity of the variables to normal distribution was examined using visual (histograms and probability plots) and analytical methods (Kolmogorov-Smirnov/Shapiro-Wilk tests). In pairwise group comparisons for numerical variables, a Mann-Whitney <em>U</em> test was used when normal distribution was not met, and a <i>t</i> test was used when normal distribution was met. The statistical significance level was accepted as <i>P</i><span class="body">&lt;</span>.05.</p> <h3>Results</h3> <p>Our study included 67 patients with melasma and 41 healthy age- and sex-matched controls. Of the participants with melasma, 60 (89.5%) were female and 7 (10.5%) were male. The control group was similar to the melasma group in terms of sex (87.8% female vs 12.2% male [<i>P</i><span class="body">=</span>.59]). The mean age (SD) was 33.1 (6.7) years in the melasma group and 31.9 (6.7) years in the control group. Age was similar across both groups (<i>P</i><span class="body">=</span>.41). All participants were of Asian race, and Fitzpatrick skin types (types II–IV) were similar across both groups. </p> <p>Fifty-four (80.6%) participants had centrofacial melasma and 13 (19.4%) had mixed-type melasma. The mMASI score ranged from 3 to 20; the mean (SD) mMASI score was 11.28 (3.2). Disease duration ranged from 2 to 72 months; the mean (SD) disease duration was 12.26 (6.3) months. The demographics and clinical characteristics of the study group are shown in eTable 1. <br/><br/>eTable 2 provides a summary of disulfide, native thiol, and total thiol levels, as well as disulfide/native thiol, disulfide/total thiol, and native thiol/total thiol ratios in the study population. Disulfide/native thiol and disulfide/total thiol ratios were higher in melasma patients (Figure 1), whereas the native thiol/total thiol ratio was higher in the control group (<i>P</i><span class="body">=</span>.025, <i>P</i><span class="body">=</span>.025, and <i>P</i><span class="body">=</span>.026, respectively).<br/><br/>All correlations between age, disease duration, and mMASI scores and disulfide, native thiol, and total thiol levels, as well as disulfide/native thiol, disulfide/total thiol, and native thiol/total thiol ratios, are summarized in eTable 3. No significant correlation was observed between age and disease duration and disulfide, native thiol, and total thiol levels or disulfide/native thiol, disulfide/total thiol, and native thiol/total thiol ratios.<br/><br/>We independently assessed whether Fitzpatrick skin types II, III, and IV exhibited distinct levels of oxidative stress in clinical melasma. There were no significant correlations with Fitzpatrick skin type (disulfide/native thiol,<i> P</i><span class="body">=</span>.25; disulfide/total thiol,<i> P</i><span class="body">=</span>.19). We further evaluated if the thiol/disulfide parameters were correlated with duration of melasma by dividing the melasma patients into 3 groups (<span class="body">&lt;</span>6 months [n<span class="body">=</span>12], 6–18 months [n<span class="body">=</span>32], <span class="body">&gt;</span>18 months [n<span class="body">=</span>23]), but there was not any significant correlation (disulfide/native thiol,<i> P</i><span class="body">=</span>.15; disulfide/total thiol,<i> P</i><span class="body">=</span>.15). We also divided our patients into 3 groups according to age (<span class="body">&lt;</span>27 years [n<span class="body">=</span>14], 27–36 years [n<span class="body">=</span>33], <span class="body">&gt;</span>36 years [n<span class="body">=</span>20]). There was no correlation of the parameters with age (disulfide/native thiol,<i> P</i><span class="body">=</span>.15; disulfide/total thiol,<i> P</i><span class="body">=</span>.14). <br/><br/>There was a positive correlation between mMASI score and disulfide, native thiol, and total thiol levels and disulfide/native thiol and disulfide/total thiol ratios, as well as a negative correlation between mMASI score and native thiol/total thiol ratio. The correlations between mMASI scores and disulfide/native thiol and disulfide/total thiol ratios are shown in Figure 2 and eTable 3.</p> <h3>Comment</h3> <p>Melasma is a common condition that may cause psychosocial problems in affected patients and negatively affect quality of life.<sup>1</sup> It occurs in all races but is more common in individuals with darker skin types (eg, Fitzpatrick skin types III and IV). Although melasma is more common in women during reproductive years (50%–70%), it also has been observed in 10% to 30% of men.<sup>5 </sup></p> <p>Treatment options include topical bleaching agents, chemical peels, and laser therapy, as well as discontinuation of medications that may potentially trigger melasma; use of broad-spectrum sunscreens also is recommended.<sup>4</sup> Vitamins A, C, and E, as well as niacinamide, are used in the treatment of melasma, especially for their antioxidant properties. The key role of antioxidants in the treatment of melasma supports the importance of oxidative stress in the pathogenesis.<sup>7</sup> Melasma often is challenging to treat, particularly the mixed or dermal types, due to their stubborn nature. This condition poses a considerable therapeutic challenge for dermatologists.<sup>4<br/><br/></sup>Oxidative stress and oxidant-antioxidant imbalance previously have been studied in various diseases, but research investigating the presence of oxidative stress in melasma are limited.<sup>7-10</sup> Exposure of the skin to polluted air and intense UVR, as well as some food by-products, cosmetics, and drugs (eg, oral contraceptives), can directly or indirectly cause ROS production in the skin. Reactive oxygen species are thought to be involved in the pathophysiology of melasma by affecting apoptotic pathways and causing cell proliferation. The intermediate heme pathway has pro-oxidant effects and produces ROS and metabolites such as redox-active quinines. Exposure to UVR leads to the generation of ROS, highlighting the role of oxidative stress in the onset of melasma.<sup> 5</sup> <br/><br/>In any cutaneous disease in which oxidative stress plays a role, oxidant and antioxidant levels may be expected to vary both locally and systemically; however, measurement of oxidative stress markers in serum instead of skin is technically and economically more advantageous.<sup>8</sup> Firstly, serum collection is less invasive and technically simpler than skin biopsies. Drawing blood is a routine procedure that requires minimal specialized equipment and training compared to the extraction and processing of skin samples. Secondly, analyzing serum samples generally is less expensive than processing skin tissue.<sup>8</sup> <br/><br/>In our study, we evaluated dynamic thiol/disulfide homeostasis in serum to investigate the presence of oxidative stress in the setting of melasma. Functional sulfhydryl (-SH) groups in thiols act as substrates for antioxidant enzymes and as free-radical scavengers. They constitute one of the most powerful defense systems against the unwanted effects of ROS. Thiols, which become the main target of ROS under oxidative stress, oxidize with oxidant molecules and form disulfide bridges.<sup>15<br/><br/></sup>Thiol/disulfide homeostasis has been studied many times in dermatologic diseases,<sup>16-19</sup> and the results obtained from these studies are heterogenous depending on the extent of oxidative damage. It has been shown that thiol/disulfide homeostasis plays a role in oxidative stress in conditions such as psoriasis,<sup>17</sup> seborrheic dermatitis,<sup>11</sup> atopic dermatitits,<sup>18</sup> and rosacea.<sup>19</sup> In our study, disulfide/native thiol and disulfide/total thiol levels were significantly higher (both <i>P</i><span class="body">=</span>.025) in the melasma group compared with the control group, which indicates that the thiol/disulfide balance in patients with melasma is shifted to disulfide formation and thiols are oxidized to disulfide bonds in the presence of oxidative stress. <br/><br/>Seçkin et al<sup>7</sup> evaluated the role of oxidative stress in the pathogenesis of melasma and found that the serum levels of the antioxidants superoxide dismutase and glutathione peroxidase were significantly higher in the patient group compared with the control group (both <i>P</i><span class="body">&lt;</span>.001). They also found that the levels of nitric oxide (another antioxidant) were increased in the patient group and the levels of protein carbonyl (an oxidative metabolite) were significantly lower (both <i>P</i><span class="body">&lt;</span>.001). These findings indicated that free-radical damage may be involved in the pathogenesis of melasma<hl name="3"/>.<sup>7<br/><br/></sup>In a study of 75 patients with melasma, serum levels of the antioxidants melatonin and catalase were significantly (<i>P</i><span class="body">&lt;</span>.001 and <i>P</i><span class="body">=</span>.001, respectively) lower in the melasma group compared with the control group, while serum levels of the oxidants protein carbonyl and nitric oxide were significantly higher (<i>P</i><span class="body">=</span>.002 and <i>P</i><span class="body">=</span>.001, respectively). No significant correlation was found between oxidative stress parameters and melasma severity.<sup>8<br/><br/></sup>Choubey et al<sup>9</sup> found that serum malondialdehyde (an end product of lipid peroxidation), superoxide dismutase, and glutathione peroxidase levels were significantly higher in the melasma group (n<span class="body">=</span>50) compared with the control group (n<span class="body">=</span>50)(all <i>P</i><span class="body">&lt;</span>.001). In addition, a significant positive correlation (correlation coefficient, +0.307; <i>P</i><span class="body">&lt;</span>.05) was found between serum malondialdehyde levels and melasma severity. The mean age (SD) of the patients was 32.22 (6.377) years, and the female (n<span class="body">=</span>41) to male (n<span class="body">=</span>9) ratio was 4.55:1. The most common melasma pattern was centrofacial, followed by malar.<sup>9<br/><br/></sup>In a study with 50 melasma patients and 50 controls, Rahimi et al<sup>10</sup> examined bilirubin and uric acid levels, which are major extracellular antioxidants. The mean age (SD) at disease onset was 32.6 (6.7) years, and the mean MASI score (SD) was 18.1 (9). Serum bilirubin levels were found to be higher in the melasma group than in the control group and were correlated with disease severity. No significant difference in uric acid levels was found between the groups, and no correlation was found between MASI score and bilirubin and uric acid levels.<sup>10<br/><br/></sup>In our study, the melasma group was similar to those in other reports<sup> </sup>in the literature regarding gender distribution, mean age, and melasma pattern.<sup>7-10</sup> Additionally, the correlation of mMASI score with disulfide/native thiol and disulfide/total thiol values in the melasma group suggested that oxidative stress also is correlated with melasma severity. <br/><br/>Thiol-based treatments such as n-acetyl cysteine, which contains a thiol compound, may be helpful in melasma.<sup>20</sup> In a double-blind, placebo-controlled study, topical n-acetyl cysteine combined with hydroquinone 2% was used in 10 female patients with melasma. Mild to strong bleaching of the skin was observed in 90% (9/10) of the patients.<sup>21</sup> Systemic use of n-acetyl cysteine in melasma also may be a potential research topic.<br/><br/>Major limitations of our study were the small sample size and lack of measurement of oxidative stress parameters in the skin concurrently with serum. </p> <h3>Conclusion</h3> <p>In our study, the presence of oxidative stress in melasma was demonstrated by evaluating thiol/disulfide homeostasis—one of the strongest markers of oxidative stress. Oxidative stress also correlated with melasma disease severity in our analysis. The data obtained in this study may contribute to understanding the etiopathogenesis of melasma and may open new horizons in treatment; however, more comprehensive studies should be conducted to support our findings.</p> <h2>References</h2> <p class="reference"> 1. Handel AC, Miot LD, Miot HA. Melasma: a clinical and epidemiological review. <i>An Bras Dermatol. </i>2014;89:771-782.</p> <p class="reference"> 2. Tamega Ade A, Miot LD, Bonfietti C, et al. Clinical patterns and epidemiological characteristics of facial melasma in Brazilian women. <i>J Eur Acad Dermatol Venereol. </i>2013;27:151-156.<br/><br/> 3. Rajanala S, Maymone MBC, Vashi NA. Melasma pathogenesis: a review of the latest research, pathological findings, and investigational therapies. <i>Dermatol Online J.</i> 2019;25:13030/qt47b7r28c.<br/><br/> 4. Abou-Taleb DA, Ibrahim AK, Youssef EM, et al. Reliability, validity, and sensitivity to change overtime of the modified melasma area and severity index score. <i>Dermatol Surg.</i> 2017;43:210-217. <br/><br/> 5. Katiyar S, Yadav D. Correlation of oxidative stress with melasma: an overview. <i>Curr Pharm Des. </i>2022;28:225-231. <br/><br/> 6. Mahmoud BH, Ruvolo E, Hexsel CL, et al. Impact of long-wavelength UVA and visible light on melanocompetent skin. <i>J Invest Dermatol.</i> 2010;130:2092-2097.<br/><br/> 7. Seçkin HY, Kalkan G, Bas¸ Y, et al. Oxidative stress status in patients with melasma. <i>Cutan Ocul Toxicol.</i> 2014;33:212-217.<br/><br/> 8. Sarkar R, Devadasan S, Choubey V, et al. Melatonin and oxidative stress in melasma—an unexplored territory; a prospective study. <i>Int J Dermatol.</i> 2020;59:572-575. <br/><br/> 9. Choubey V, Sarkar R, Garg V, et al. Role of oxidative stress in melasma: a prospective study on serum and blood markers of oxidative stress in melasma patients. <i>Int J Dermatol. </i>2017;56:939-943. <br/><br/>10. Rahimi H, Mirnezami M, Yazdabadi A. Bilirubin as a new antioxidant in melasma. <i>J Cosmet Dermatol.</i> 2022;21:5800-5803. <br/><br/>11. Emre S, Kalkan G, Erdog˘an S, et al. Dynamic thiol/disulfide balance in patients with seborrheic dermatitis: a case-control study. <i>Saudi J Med Med Sci.</i> 2020;8:12-16. <br/><br/>12. Erel Ö, Erdog˘an S. Thiol-disulfide homeostasis: an integrated approach with biochemical and clinical aspects. <i>Turk J Med Sci.</i> 2020;50:1728-1738. <br/><br/>13. Pandya AG, Hynan LS, Bhore R, et al. Reliability assessment and validation of the Melasma Area and Severity Index (MASI) and a new modified MASI scoring method. <i>J Am Acad Dermatol.</i> 2011;64:78-83, 83.E1-E2.<br/><br/>14. Erel O, Neselioglu S. A novel and automated assay for thiol/disulphide homeostasis. <i>Clin Biochem.</i> 2014;47:326-332. <br/><br/>15. Guzelcicek A, Cakirca G, Erel O, et al. Assessment of thiol/disulfide balance as an oxidative stress marker in children with <span class="body">β</span>-thalassemia major. <i>Pak J Med Sci</i>. 2019;35:161-165.<br/><br/>16. Georgescu SR, Mitran CI, Mitran MI, et al. Thiol-Disulfide homeostasis in skin diseases. <i>J Clin Med. </i>2022;11:1507. <br/><br/>17. Üstüner P, Balevi A, Özdemir M, et al. The role of thiol/disulfide homeostasis in psoriasis: can it be a new marker for inflammation? <span class="ref-journal"><i>Turk Arch Dermatol Venereol. </i></span>2018;<span class="ref-vol">52</span>:120-125. <br/><br/><span class="element-citation">18. Karacan G, Ercan N, Bostanci I, et al. A novel oxidative stress marker of atopic dermatitis in infants: Thiol–disulfide balance. </span><span class="ref-journal"><i>Arch Dermatol Res. </i></span><span class="element-citation">2020;</span><span class="ref-vol">312</span><span class="element-citation">:697-703.<br/><br/></span>19. Demir Pektas S, Cinar N, Pektas G, et al. Thiol/disulfide homeostasis and its relationship with insulin resistance in patients with rosacea. <span class="ref-journal"><i>J Cosmet Dermatol. </i></span>2021;<span class="ref-vol">11</span>:14477. <br/><br/>20. Adil M, Amin SS, Mohtashim M. N-acetylcysteine in dermatology. <i>Indian J Dermatol Venereol Leprol. </i>2018;84:652-659. <br/><br/>21. Njoo MD, Menke HE, Pavel W, et al. N-acetylcysteine as a bleaching agent in the treatment of melasma. <i>J Eur Acad Dermatol Venereol.</i> 1997;9:86-87.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Drs. Erduran, Hayran, Eren, and I˙yidal are from Ankara Bilkent City Hospital, Turkey. Drs. Erduran, Hayran, and I˙yidal are from the Department of Dermatology, and Dr. Eren is from the Department of Medical Biochemistry. Drs. Emre and Erel are from Ankara Yıldırım Beyazıt University Faculty of Medicine, Turkey. Dr. Emre is from the Department of Dermatology, and Dr. Erel is from the Department of Medical Biochemistry.</p> <p class="disclosure">The authors report no conflict of interest. <br/><br/>The eTables are available in the Appendix online at www.mdedge.com/dermatology.<br/><br/>Correspondence: Funda Erduran, MD, Ankara Bilkent City Hospital, Department of Dermatology, Üniversiteler Mah, Çankaya, Ankara, 06800, Turkey (fnderdrn@mail.com).<br/><br/><em>Cutis.</em> 2024 June;113(6):264-268, E6-E7. doi:10.12788/cutis.1036</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>Melasma is a common pigmentation disorder that causes brown or grayish patches on the skin.</li> <li>Disulfide/native thiol and disulfide/total thiol ratios were higher in patients with melasma compared with controls, which indicated the presence of oxidative stress in melasma.</li> <li>The evaluation of modified melasma area and severity index score with disulfide/native thiol and disulfide/total thiol values suggests that oxidative stress is correlated with melasma disease severity.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • Melasma is a common pigmentation disorder that causes brown or grayish patches on the skin.
  • Disulfide/native thiol and disulfide/total thiol ratios were higher in patients with melasma compared with controls, which indicated the presence of oxidative stress in melasma.
  • The evaluation of modified melasma area and severity index score with disulfide/native thiol and disulfide/total thiol values suggests that oxidative stress is correlated with melasma disease severity.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Disable zoom
Off

Small Fiber Neuropathy in Veterans With Gulf War Illness

Article Type
Changed
Mon, 05/13/2024 - 14:10

Following deployment to operations Desert Shield and Desert Storm (Gulf War) in 1990 and 1991, many Gulf War veterans (GWVs) developed chronic, complex symptoms, including pain, dyscognition, and fatigue, with gastrointestinal, skin, and respiratory manifestations. This Gulf War Illness (GWI) is reported to affect about 30% of those deployed. More than 30 years later, there is no consensus as to the etiology of GWI, although some deployment-related exposures have been implicated.1

Accepted research definitions for GWI include the Centers for Disease Control and Prevention and Kansas definitions.2 The US Department of Veterans Affairs (VA) uses the terminology chronic multisymptom illness (CMI), which is an overarching diagnosis under which GWI falls. Although there is no consensus case definition for CMI, there is overlap with conditions such as fibromyalgia, myalgic encephalomyelitis/chronic fatigue syndrome, and irritable bowel syndrome; the VA considers these as qualifying clinical diagnoses.3 The pathophysiology of GWI is also unknown, though a frequently reported unifying feature is that of autonomic nervous system (ANS) dysfunction. Studies have demonstrated differences between veterans with GWI and those without GWI in both the reporting of symptoms attributable to ANS dysfunction and in physiologic evaluations of the ANS.4-10

Small fiber neuropathy (SFN), a condition with damage to the A-δ and C small nerve fibers, has been proposed as a potential mechanism for the pain and ANS dysfunction experienced in GWI.11-13 Symptoms of SFN are similar to those of GWI, with pain and ANS symptoms commonly reported.14,15 There are multiple diagnostic criteria for SFN, the most commonly used requiring the presence of appropriate symptoms in the absence of large fiber neuropathy and a skin biopsy demonstrating reduced intraepidermal nerve fiber density.16-19 Several conditions reportedly cause SFN, most notably diabetes/prediabetes. Autoimmune disease, vitamin B12 deficiency, monoclonal gammopathies, celiac disease, paraneoplastic syndromes, and sodium channel gene mutations may also contribute to SFN.20 Hyperlipidemia has been identified as a contributor, although it has been variably reported.21,22

Idiopathic neuropathies, SFN included, may be secondary to neurotoxicant exposures. Agents whose exposure or consumption have been associated with SFN include alcohol most prominently, but also the organic solvent n-hexane, heavy metals, and excess vitamin B6.20,23-25 Agents associated with large fiber neuropathy may also have relevance for SFN, as small fibers have been likened to the “canary in the coal mine” in that they may be more susceptible to neurotoxicants and are affected earlier in the disease process.26 In this way, SFN may be the harbinger of large fiber neuropathy in some cases. Of specific relevance for GWVs, organophosphates and carbamates are known to produce a delayed onset large fiber neuropathy.27-30 Exposure to petrochemical solvents has also been associated with large fiber neuropathies.31,32

The War Related Illness and Injury Study Center (WRIISC) is a clinical, research, and education center established by Congress in 2001. Its primary focus is on military exposures and postdeployment health of veterans. It is located at 3 sites: East Orange, New Jersey; Washington, DC; and Palo Alto, California. The New Jersey WRIISC began a program to evaluate GWVs with characteristic symptoms for possible SFN with use of a skin biopsy.

We hypothesize that SFN may underly much of GWI symptomatology and may not be accounted for by the putative etiologies detailed in review of the medical literature. This retrospective review of clinical evaluations for SFN in GWVs who sought care at the New Jersey WRIISC explored and addressed the following questions: (1) how common is biopsy-confirmed SFN in veterans with GWI; (2) do veterans with GWI and SFN report more symptoms attributable to ANS dysfunction when compared with veterans with GWI and no SFN; and (3) can SFN in veterans with GWI and SFN be explained by conditions and substances commonly associated with SFN? Institutional review board approval and waiver of consent was obtained from the Veterans Affairs New Jersey Health Care Center for the study.

 

 

Methods

A retrospective chart review was conducted on veterans evaluated at the WRIISC from March 1, 2015, to January 31, 2019. Inclusion criteria were: deployment to operations Desert Shield and Desert Storm between August 2, 1990, and February 28, 1991, and skin biopsy conducted at the WRIISC. Skin biopsies were obtained at the discretion of an examining clinician based on clinical indications, including neuropathic pain, ANS symptoms, and/or a fibromyalgia/chronic pain–type presentation.

Electronic health record review explicitly abstracted GWI status, results of the skin biopsy, and ANS symptom burden as determined by the Composite Autonomic Symptom Scale 31 (COMPASS 31) completed at the time of the WRIISC evaluation. Determination of GWI was established as per the clinical opinion of the WRIISC lead clinician or environmental exposure clinician as evidenced by a diagnosis of fibromyalgia or chronic fatigue syndrome, or explicit statement of CMI/GWI in the patient assessment. A diagnosis of SFN was established if clinical signs were present and an intraepidermal nerve fiber density below the lower limits, as compared to normative data from the clinical diagnostic laboratory (Therapath Neuropathology), was documented.

COMPASS 31 assesses symptoms across 6 domains (orthostatic, vasomotor, secretomotor, gastrointestinal, bladder, andpupillomotor). Patients are asked about symptom frequency (rarely to almost always), severity (mild to severe), and improvement (much worse to completely gone). Individual domain scores and a total weighted score (0-100) have demonstrated good validity, reliability, and consistency in SFN.33,34

thuswichawrojomashis

In veterans with GWI and documented SFN, a health record review was performed to identify potential etiologies for SFN (Appendix).

 

Statistical Analysis

Microsoft Excel and IBM SPSS 12.0.1 for Windows were used for data collection and statistical analysis. Fisher exact test was used for comparing the prevalence of SFN in veterans with GWI vs without GWI. The independent samples t test was used for comparing COMPASS 31 scores for veterans with GWI by SFN status. α < .05 was used for determining statistical significance. For those GWVs documented with SFN and GWI, potential explanations were documented in total and by condition.

Results

tradibrawrustiphisosudrospenudivowobeswochitrodafrubusekurekurubrilistitiphosliwisespugaspidrastorochuuosepreselatriuarawruwresephisephuc

juprutestuuotrucliheshicradojepruwrotrijotihatrethuswumospejoriphagashugutejistemugistastushiuispakejathagobrithechecujej

From March 1, 2015, to January 31, 2019, 141 GWVs received a comprehensive in person clinical evaluation at the WRIISC and 51 veterans (36%) received a skin biopsy and were included in this retrospective observational study (Figure). The mean age was 48.6 years, and the majority were male and served in the US Army. Skin biopsies met clinical criteria for GWI for 42 (82%) and 24 of 42 (57%) were determined to have SFN. Four of 9 (44%) veterans without GWI had positive SFN biopsies, though this difference was not statistically significant (Table 1). Veterans with SFN but no GWI were not included in the further analysis.

tahulabrusanuthapalabradimouathukeswagoswuclepaspucethapofruprufrumatraswastustepocrib

Thirty-five veterans with GWI—18 with SFN and 17 without SFN—completed the COMPASS 31 (Table 2). COMPASS 31 data were not analyzed for veterans without GWI. Individual domain scores and the difference in COMPASS 31 scores for veterans with GWI and SFN vs GWI and no SFN (38.3 vs 37.8, respectively) were not statistically significant.

jugucraspethouutrufribrokusletatredugitripruneswuchahiwridrihecribrucrawrashetrouotruhiphiduclopodratushapabuwijespuchospupijuchavebru

Sixteen of 24 veterans with GWI and SFN (67%) had ≥ 1 conditions that could potentially be responsible for SFN (Table 3), including 11 veterans (46%) with prediabetes/diabetes. Hyperlipidemia is only variably reported as a cause of SFN; when included, 19 of 24 (79%) SFN cases were accounted for. We could not identify a medical explanation for SFN in 5 of 24 veterans (21%) with GWI, which were deemed to be idiopathic.

 

 

Discussion

Biopsy-confirmed SFN was present in more than half of our sample of veterans with GWI, which is broadly consistent with what has been reported in the literature.13,35-38 In this clinical observation study, SFN was similarly prevalent in veterans with and without GWI; although it should be noted that biopsies only were obtained when there was a strong clinical suspicion for SFN. Almost half of patients with GWI did not have SFN, so our study does not support SFN as the underlying explanation for all GWI. Although our data cannot provide clinical guidance as to when skin biopsy may be indicated in GWI, work done in fibromyalgia found symptoms of dysautonomia and paresthesias are more specific for SFN and may be useful to help guide medical decision making.39

Veterans with GWI in our clinical sample reported a high burden of clinical symptoms conceivably attributable to ANS dysfunction. This symptom reporting is consistent with that seen in other GWI studies, as well as in other studies of SFN.4,5,7-9,14,15,34,38,40 Our clinical sample of veterans with GWI found no differences in the ANS symptom reporting between those with and without SFN. Therefore, our study cannot support SFN alone as accounting for ANS symptom burden in patients with GWI.

Two-thirds of biopsy-confirmed SFN in our clinical sample of veterans with GWI could potentially be explained by established medical conditions. As in other studies of SFN, prediabetes and diabetes represented a plurality (46%). Even after considering hyperlipidemia as a potential explanation, about 21% of SFN cases in veterans with GWI still were deemed idiopathic.

Evidence supports certain environmental agents as causal factors for GWI. Neurotoxicants reportedly related to GWI include pesticides (particularly organophosphates and carbamates), pyridostigmine bromide (used during the Gulf War as a prophylactic agent against the use of chemical weapons), and low levels of the nerve agent sarin from environmental contamination due to chemical weapons detonations.1 Some of these agents have been implicated in neuropathy as well.1,28-30 It is biologically plausible that deployment-related exposures could trigger SFN, though the traditional consensus has been that remote exposure to neurotoxic substances is unlikely to produce neuropathy that presents many years after the exposure.41 In the WRIISC clinical experience, however, veterans often report that their neuropathic symptoms predate the diagnosis of the associated medical conditions, sometimes by decades. It is conceivable that remote exposures may trigger the condition that is then potentiated by ongoing exposures, metabolic factors, and/or other medical conditions. These may perpetuate neuropathic symptoms and the illness experience of affected veterans. Our clinical observation study cannot clarify the extent to which this may be the case. Despite these findings and arguments, an environmental contribution to SFN cannot be discounted, and further research is needed to explore a potential relationship.

Limitations

This study’s conclusions are limited by its observational/retrospective design in a relatively small clinical sample of veterans evaluated at a tertiary referral center for postdeployment exposure-related health concerns. The WRIISC clinical sample is not representative of all GWVs or even of all veterans with GWI, as there is inherent selection bias as to who gets referred to and evaluated at the WRIISC. As with studies based on retrospective chart review, data are reliant on clinical documentation andaccuracy/consistency of the reviewer. Evaluation for SFN with skin biopsy is an invasive procedure and was performed when a high index of clinical suspicion for this condition existed, possibly representing confirmation bias. Therefore, the relatively high prevalence ofbiopsy-confirmed SFN seen in our clinical sample cannot be generalized to GWVs as a whole or even to veterans with GWI.

 

 

Assessment of autonomic dysfunction was based on COMPASS 31 symptom reporting by an small subset of the clinical cohort. Symptom reporting may not be reflective of true abnormality in ANS function. Physiologic tests of the ANS were not performed; such studies could more objectively establish whether ANS dysfunction is more prevalent in GWI veterans with SFN.

Evaluation for all potential etiologic/contributory conditions to SFN was not exhaustive. For example, sodium channel gene mutations have been documented to account for up to one-third of all cases of idiopathic SFN.42 For those cases in which no compelling etiology was identified, it is plausible that medical explanations for SFN may be found on further investigation.

Clinical assessments at the WRIISC were performed on GWVs ≥ 26 years after their deployment-related exposures. Other conditions/exposures may have occurred in the interim. What is not clear is whether the SFN predated the onset of any of these medical conditions or other putative contributors. This observational study is not able to tease out a temporal association to make a cause-and-effect assessment.

 

Conclusions

Retrospective analysis of clinical data of veterans evaluated at a specialized center for postdeployment health demonstrated that skin biopsy–confirmed SFN was prevalent, but not ubiquitous, in veterans with GWI. Symptom that may be attributed to ANS dysfunction in this clinical sample was consistent with literature on SFN and with GWI, but we could not definitively attribute ANS symptoms to SFN. Our study does not support the hypothesis that GWI symptoms are solely due to SFN, though it may still be relevant in a subset of veterans with GWI with strongly suggestive clinical features. We were able to identify a potential etiology for SFN in most veterans with GWI. Further investigations are recommended to explore any potential relationship between Gulf War exposures and SFN.

References

1. White RF, Steele L, O’Callaghan JP, et al. Recent research on Gulf War illness and other health problems in veterans of the 1991 Gulf War: effects of toxicant exposures during deployment. Cortex. 2016;74:449-475. doi:10.1016/j.cortex.2015.08.022

2. Committee on the Development of a Consensus Case Definition for Chronic Multisymptom Illness in 1990-1991 Gulf War Veterans, Board on the Health of Select Populations, Institute of Medicine. Chronic Multisymptom Illness in Gulf War Veterans: Case Definitions Reexamined. National Academies Press; 2014.

3. Robbins R, Helmer D, Monahan P, et al. Management of chronic multisymptom illness: synopsis of the 2021 US Department of Veterans Affairs and US Department of Defense Clinical Practice Guideline. Mayo Clin Proc. 2022;97(5):991-1002. doi:10.1016/j.mayocp.2022.01.031

4. Fox A, Helmer D, Tseng CL, Patrick-DeLuca L, Osinubi O. Report of autonomic symptoms in a clinical sample of veterans with Gulf War Illness. Mil Med. 2018;183(3-4):e179-e185. doi:10.1093/milmed/usx052

5. Fox A, Helmer D, Tseng CL, McCarron K, Satcher S, Osinubi O. Autonomic symptoms in Gulf War veterans evaluated at the War Related Illness and Injury Study Center. Mil Med. 2019;184(3-4):e191-e196. doi:10.1093/milmed/usy227

6. Reyes L, Falvo M, Blatt M, Ghobreal B, Acosta A, Serrador J. Autonomic dysfunction in veterans with Gulf War illness [abstract]. FASEB J. 2014;28(S1):1068.19. doi:10.1096/fasebj.28.1_supplement.1068.19

7. Haley RW, Charuvastra E, Shell WE, et al. Cholinergic autonomic dysfunction in veterans with Gulf War illness: confirmation in a population-based sample. JAMA Neurol. 2013;70(2):191-200. doi:10.1001/jamaneurol.2013.596

8. Haley RW, Vongpatanasin W, Wolfe GI, et al. Blunted circadian variation in autonomic regulation of sinus node function in veterans with Gulf War syndrome. Am J Med. 2004;117(7):469-478. doi:10.1016/j.amjmed.2004.03.041

9. Avery TJ, Mathersul DC, Schulz-Heik RJ, Mahoney L, Bayley PJ. Self-reported autonomic dysregulation in Gulf War Illness. Mil Med. Published online December 30, 2021. doi:10.1093/milmed/usab546

10. Verne ZT, Fields JZ, Zhang BB, Zhou Q. Autonomic dysfunction and gastroparesis in Gulf War veterans. J Investig Med. 2023;71(1):7-10. doi:10.1136/jim-2021-002291

11. Levine TD. Small fiber neuropathy: disease classification beyond pain and burning. J Cent Nerv Syst Dis. 2018;10:1179573518771703. doi:10.1177/1179573518771703

12. Novak P. Autonomic disorders. Am J Med. 2019;132(4):420-436. doi:10.1016/j.amjmed.2018.09.027

13. Oaklander AL, Klein MM. Undiagnosed small-fiber polyneuropathy: is it a component of Gulf War Illness? Defense Technical Information Center. Accessed February 21, 2024. https://apps.dtic.mil/sti/citations/ADA613891

<--pagebreak-->14. Sène D. Small fiber neuropathy: diagnosis, causes, and treatment. Joint Bone Spine. 2018;85(5):553-559. doi:10.1016/j.jbspin.2017.11.002

15. Novak V, Freimer ML, Kissel JT, et al. Autonomic impairment in painful neuropathy. Neurology. 2001;56(7):861-868. doi:10.1212/wnl.56.7.861

16. Myers MI, Peltier AC. Uses of skin biopsy for sensory and autonomic nerve assessment. Curr Neurol Neurosci Rep. 2013;13(1):323. doi:10.1007/s11910-012-0323-2

17. Haroutounian S, Todorovic MS, Leinders M, et al. Diagnostic criteria for idiopathic small fiber neuropathy: a systematic review. Muscle Nerve. 2021;63(2):170-177. doi:10.1002/mus.27070

18. Levine TD, Saperstein DS. Routine use of punch biopsy to diagnose small fiber neuropathy in fibromyalgia patients. Clin Rheumatol. 2015;34(3):413-417. doi:10.1007/s10067-014-2850-5

19. England JD, Gronseth G S, Franklin G, et al. Practice parameter: the evaluation of distal symmetric polyneuropathy: the role of autonomic testing, nerve biopsy, and skin biopsy (an evidence-based review). Report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. PM R. 2009;1(1):14-22. doi:10.1016/j.pmrj.2008.11.011

20. de Greef BTA, Hoeijmakers JGJ, Gorissen-Brouwers CML, Geerts M, Faber CG, Merkies ISJ. Associated conditions in small fiber neuropathy - a large cohort study and review of the literature. Eur J Neurol. 2018;25(2):348-355. doi:10.1111/ene.13508

21. Morkavuk G, Leventoglu A. Small fiber neuropathy associated with hyperlipidemia: utility of cutaneous silent periods and autonomic tests. ISRN Neurol. 2014;2014:579242. doi:10.1155/2014/579242

22. Bednarik J, Vlckova-Moravcova E, Bursova S, Belobradkova J, Dusek L, Sommer C. Etiology of small-fiber neuropathy. J Peripher Nerv Syst. 2009;14(3):177-183. doi:10.1111/j.1529-8027.2009.00229.x

23. Kokotis P, Papantoniou M, Schmelz M, Buntziouka C, Tzavellas E, Paparrigopoulos T. Pure small fiber neuropathy in alcohol dependency detected by skin biopsy. Alcohol Fayettev N. 2023;111:67-73. doi:10.1016/j.alcohol.2023.05.006

24. Guimarães-Costa R, Schoindre Y, Metlaine A, et al. N-hexane exposure: a cause of small fiber neuropathy. J Peripher Nerv Syst. 2018;23(2):143-146. doi:10.1111/jns.12261

25. Koszewicz M, Markowska K, Waliszewska-Prosol M, et al. The impact of chronic co-exposure to different heavy metals on small fibers of peripheral nerves. A study of metal industry workers. J Occup Med Toxicol. 2021;16(1):12. doi:10.1186/s12995-021-00302-6

26. Johns Hopkins Medicine. Small nerve fibers defy neuropathy conventions. April 11, 2016. Accessed February 21, 2024. https://www.hopkinsmedicine.org/news/media/releases/small_nerve_fibers_defy_neuropathy_conventions

27. Jett DA. Neurotoxic pesticides and neurologic effects. Neurol Clin. 2011;29(3):667-677. doi:10.1016/j.ncl.2011.06.002

28. Berger AR, Schaumburg HH. Human toxic neuropathy caused by industrial agents. In: Dyck PJ, Thomas PK, eds. Peripheral Neuropathy. 4th ed. Saunders; 2005:2505-2525. doi:10.1016/B978-0-7216-9491-7.50115-0

29. Herskovitz S, Schaumburg HH. Neuropathy caused by drugs. In: Dyck PJ, Thomas PK, eds. Peripheral Neuropathy. 4th ed. Saunders; 2005:2553-2583.

30. Katona I, Weis J. Chapter 31 - Diseases of the peripheral nerves. Handb Clin Neurol. 2017;145:453-474. doi:10.1016/B978-0-12-802395-2.00031-6

31. Matikainen E, Juntunen J. Autonomic nervous system dysfunction in workers exposed to organic solvents. J Neurol Neurosurg Psychiatry. 1985;48(10):1021-1024. doi:10.1136/jnnp.48.10.1021

32. Murata K, Araki S, Yokoyama K, Maeda K. Autonomic and peripheral nervous system dysfunction in workers exposed to mixed organic solvents. Int Arch Occup Environ Health. 1991;63(5):335-340. doi:10.1007/BF00381584

33. Sletten DM, Suarez GA, Low PA, Mandrekar J, Singer W. COMPASS 31: a refined and abbreviated Composite Autonomic Symptom Score. Mayo Clin Proc. 2012;87(12):1196-1201. doi:10.1016/j.mayocp.2012.10.013

34. Treister R, O’Neil K, Downs HM, Oaklander AL. Validation of the Composite Autonomic Symptom Scale-31 (COMPASS-31) in patients with and without small-fiber polyneuropathy. Eur J Neurol. 2015;22(7):1124-1130. doi:10.1111/ene.12717

35. Joseph P, Arevalo C, Oliveira RKF, et al. Insights from invasive cardiopulmonary exercise testing of patients with myalgic encephalomyelitis/chronic fatigue syndrome. Chest. 2021;160(2):642-651. doi:10.1016/j.chest.2021.01.082

36. Giannoccaro MP, Donadio V, Incensi A, Avoni P, Liguori R. Small nerve fiber involvement in patients referred for fibromyalgia. Muscle Nerve. 2014;49(5):757-759. doi:10.1002/mus.24156

37. Oaklander AL, Herzog ZD, Downs HM, Klein MM. Objective evidence that small-fiber polyneuropathy underlies some illnesses currently labeled as fibromyalgia. Pain. 2013;154(11):2310-2316. doi:10.1016/j.pain.2013.06.001

38. Serrador JM. Diagnosis of late-stage, early-onset, small-fiber polyneuropathy. Defense Technical Information Center. December 1, 2019. Accessed February 21, 2024. https://apps.dtic.mil/sti/citations/AD1094831

39. Lodahl M, Treister R, Oaklander AL. Specific symptoms may discriminate between fibromyalgia patients with vs without objective test evidence of small-fiber polyneuropathy. Pain Rep. 2018;3(1):e633. doi:10.1097/PR9.0000000000000633

40. Sastre A, Cook MR. Autonomic dysfunction in Gulf War veterans. Defense Technical Information Center. April 1, 2004. Accessed February 21, 2024. https://apps.dtic.mil/sti/citations/ADA429525

41. Little AA, Albers JW. Clinical description of toxic neuropathies. Handb Clin Neurol. 2015;131:253-296. doi:10.1016/B978-0-444-62627-1.00015-9

42. Faber CG, Hoeijmakers JGJ, Ahn HS, et al. Gain of function NaV1.7 mutations in idiopathic small fiber neuropathy. Ann Neurol. 2012;71(1):26-39.

Article PDF
Author and Disclosure Information

Edward C. Shadiack III, DO, MPHa,b; Omowunmi Osinubi, MD, MPHa,b; Apollonia Gruber-Fox, PhDb; Chinmoy Bhate, MDa;  Lydia Patrick-DeLuca, MSN, RNa,b; Philip Cohen, MDa; Drew A. Helmer, MD, MSb,c

Correspondence:  Edward Shadiack  (edward.shadiack@va.gov)

aVeterans Affairs New Jersey Health Care Systems, East Orange

bWar Related Illness and Injury Study Center, East Orange, New Jersey

cMichael E. DeBakey Veterans Affairs Medical Center, Houston, Texas

Author contributions

Concept: Shadiack.

Data collection: Shadiack, Osinubi, Bhate, Patrick-DeLuca, Cohen.

Data analysis: Shadiack, Gruber-Fox, Helmer.

Drafting of manuscript: Shadiack, Osinubi.

Critical review of manuscript: Gruber-Fox, Patrick-DeLuca, Cohen, Helmer.

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Funding and ethics

This study was reviewed and approved by the Veterans Affairs New Jersey Health Care System Institutional Review Board (IRB# 01497). Funding provided by Veterans Affairs War Related Illness and Injury Study Center-New Jersey clinical resources; US Department of Veterans Affairs Health Services Research & Development (CIN 13-413). Poster presented virtually at the American Autonomic Society’s annual meeting, November 2020.

Issue
Federal Practitioner - 41(5)a
Publications
Topics
Page Number
136-141
Sections
Author and Disclosure Information

Edward C. Shadiack III, DO, MPHa,b; Omowunmi Osinubi, MD, MPHa,b; Apollonia Gruber-Fox, PhDb; Chinmoy Bhate, MDa;  Lydia Patrick-DeLuca, MSN, RNa,b; Philip Cohen, MDa; Drew A. Helmer, MD, MSb,c

Correspondence:  Edward Shadiack  (edward.shadiack@va.gov)

aVeterans Affairs New Jersey Health Care Systems, East Orange

bWar Related Illness and Injury Study Center, East Orange, New Jersey

cMichael E. DeBakey Veterans Affairs Medical Center, Houston, Texas

Author contributions

Concept: Shadiack.

Data collection: Shadiack, Osinubi, Bhate, Patrick-DeLuca, Cohen.

Data analysis: Shadiack, Gruber-Fox, Helmer.

Drafting of manuscript: Shadiack, Osinubi.

Critical review of manuscript: Gruber-Fox, Patrick-DeLuca, Cohen, Helmer.

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Funding and ethics

This study was reviewed and approved by the Veterans Affairs New Jersey Health Care System Institutional Review Board (IRB# 01497). Funding provided by Veterans Affairs War Related Illness and Injury Study Center-New Jersey clinical resources; US Department of Veterans Affairs Health Services Research & Development (CIN 13-413). Poster presented virtually at the American Autonomic Society’s annual meeting, November 2020.

Author and Disclosure Information

Edward C. Shadiack III, DO, MPHa,b; Omowunmi Osinubi, MD, MPHa,b; Apollonia Gruber-Fox, PhDb; Chinmoy Bhate, MDa;  Lydia Patrick-DeLuca, MSN, RNa,b; Philip Cohen, MDa; Drew A. Helmer, MD, MSb,c

Correspondence:  Edward Shadiack  (edward.shadiack@va.gov)

aVeterans Affairs New Jersey Health Care Systems, East Orange

bWar Related Illness and Injury Study Center, East Orange, New Jersey

cMichael E. DeBakey Veterans Affairs Medical Center, Houston, Texas

Author contributions

Concept: Shadiack.

Data collection: Shadiack, Osinubi, Bhate, Patrick-DeLuca, Cohen.

Data analysis: Shadiack, Gruber-Fox, Helmer.

Drafting of manuscript: Shadiack, Osinubi.

Critical review of manuscript: Gruber-Fox, Patrick-DeLuca, Cohen, Helmer.

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Funding and ethics

This study was reviewed and approved by the Veterans Affairs New Jersey Health Care System Institutional Review Board (IRB# 01497). Funding provided by Veterans Affairs War Related Illness and Injury Study Center-New Jersey clinical resources; US Department of Veterans Affairs Health Services Research & Development (CIN 13-413). Poster presented virtually at the American Autonomic Society’s annual meeting, November 2020.

Article PDF
Article PDF
Related Articles

Following deployment to operations Desert Shield and Desert Storm (Gulf War) in 1990 and 1991, many Gulf War veterans (GWVs) developed chronic, complex symptoms, including pain, dyscognition, and fatigue, with gastrointestinal, skin, and respiratory manifestations. This Gulf War Illness (GWI) is reported to affect about 30% of those deployed. More than 30 years later, there is no consensus as to the etiology of GWI, although some deployment-related exposures have been implicated.1

Accepted research definitions for GWI include the Centers for Disease Control and Prevention and Kansas definitions.2 The US Department of Veterans Affairs (VA) uses the terminology chronic multisymptom illness (CMI), which is an overarching diagnosis under which GWI falls. Although there is no consensus case definition for CMI, there is overlap with conditions such as fibromyalgia, myalgic encephalomyelitis/chronic fatigue syndrome, and irritable bowel syndrome; the VA considers these as qualifying clinical diagnoses.3 The pathophysiology of GWI is also unknown, though a frequently reported unifying feature is that of autonomic nervous system (ANS) dysfunction. Studies have demonstrated differences between veterans with GWI and those without GWI in both the reporting of symptoms attributable to ANS dysfunction and in physiologic evaluations of the ANS.4-10

Small fiber neuropathy (SFN), a condition with damage to the A-δ and C small nerve fibers, has been proposed as a potential mechanism for the pain and ANS dysfunction experienced in GWI.11-13 Symptoms of SFN are similar to those of GWI, with pain and ANS symptoms commonly reported.14,15 There are multiple diagnostic criteria for SFN, the most commonly used requiring the presence of appropriate symptoms in the absence of large fiber neuropathy and a skin biopsy demonstrating reduced intraepidermal nerve fiber density.16-19 Several conditions reportedly cause SFN, most notably diabetes/prediabetes. Autoimmune disease, vitamin B12 deficiency, monoclonal gammopathies, celiac disease, paraneoplastic syndromes, and sodium channel gene mutations may also contribute to SFN.20 Hyperlipidemia has been identified as a contributor, although it has been variably reported.21,22

Idiopathic neuropathies, SFN included, may be secondary to neurotoxicant exposures. Agents whose exposure or consumption have been associated with SFN include alcohol most prominently, but also the organic solvent n-hexane, heavy metals, and excess vitamin B6.20,23-25 Agents associated with large fiber neuropathy may also have relevance for SFN, as small fibers have been likened to the “canary in the coal mine” in that they may be more susceptible to neurotoxicants and are affected earlier in the disease process.26 In this way, SFN may be the harbinger of large fiber neuropathy in some cases. Of specific relevance for GWVs, organophosphates and carbamates are known to produce a delayed onset large fiber neuropathy.27-30 Exposure to petrochemical solvents has also been associated with large fiber neuropathies.31,32

The War Related Illness and Injury Study Center (WRIISC) is a clinical, research, and education center established by Congress in 2001. Its primary focus is on military exposures and postdeployment health of veterans. It is located at 3 sites: East Orange, New Jersey; Washington, DC; and Palo Alto, California. The New Jersey WRIISC began a program to evaluate GWVs with characteristic symptoms for possible SFN with use of a skin biopsy.

We hypothesize that SFN may underly much of GWI symptomatology and may not be accounted for by the putative etiologies detailed in review of the medical literature. This retrospective review of clinical evaluations for SFN in GWVs who sought care at the New Jersey WRIISC explored and addressed the following questions: (1) how common is biopsy-confirmed SFN in veterans with GWI; (2) do veterans with GWI and SFN report more symptoms attributable to ANS dysfunction when compared with veterans with GWI and no SFN; and (3) can SFN in veterans with GWI and SFN be explained by conditions and substances commonly associated with SFN? Institutional review board approval and waiver of consent was obtained from the Veterans Affairs New Jersey Health Care Center for the study.

 

 

Methods

A retrospective chart review was conducted on veterans evaluated at the WRIISC from March 1, 2015, to January 31, 2019. Inclusion criteria were: deployment to operations Desert Shield and Desert Storm between August 2, 1990, and February 28, 1991, and skin biopsy conducted at the WRIISC. Skin biopsies were obtained at the discretion of an examining clinician based on clinical indications, including neuropathic pain, ANS symptoms, and/or a fibromyalgia/chronic pain–type presentation.

Electronic health record review explicitly abstracted GWI status, results of the skin biopsy, and ANS symptom burden as determined by the Composite Autonomic Symptom Scale 31 (COMPASS 31) completed at the time of the WRIISC evaluation. Determination of GWI was established as per the clinical opinion of the WRIISC lead clinician or environmental exposure clinician as evidenced by a diagnosis of fibromyalgia or chronic fatigue syndrome, or explicit statement of CMI/GWI in the patient assessment. A diagnosis of SFN was established if clinical signs were present and an intraepidermal nerve fiber density below the lower limits, as compared to normative data from the clinical diagnostic laboratory (Therapath Neuropathology), was documented.

COMPASS 31 assesses symptoms across 6 domains (orthostatic, vasomotor, secretomotor, gastrointestinal, bladder, andpupillomotor). Patients are asked about symptom frequency (rarely to almost always), severity (mild to severe), and improvement (much worse to completely gone). Individual domain scores and a total weighted score (0-100) have demonstrated good validity, reliability, and consistency in SFN.33,34

thuswichawrojomashis

In veterans with GWI and documented SFN, a health record review was performed to identify potential etiologies for SFN (Appendix).

 

Statistical Analysis

Microsoft Excel and IBM SPSS 12.0.1 for Windows were used for data collection and statistical analysis. Fisher exact test was used for comparing the prevalence of SFN in veterans with GWI vs without GWI. The independent samples t test was used for comparing COMPASS 31 scores for veterans with GWI by SFN status. α < .05 was used for determining statistical significance. For those GWVs documented with SFN and GWI, potential explanations were documented in total and by condition.

Results

tradibrawrustiphisosudrospenudivowobeswochitrodafrubusekurekurubrilistitiphosliwisespugaspidrastorochuuosepreselatriuarawruwresephisephuc

juprutestuuotrucliheshicradojepruwrotrijotihatrethuswumospejoriphagashugutejistemugistastushiuispakejathagobrithechecujej

From March 1, 2015, to January 31, 2019, 141 GWVs received a comprehensive in person clinical evaluation at the WRIISC and 51 veterans (36%) received a skin biopsy and were included in this retrospective observational study (Figure). The mean age was 48.6 years, and the majority were male and served in the US Army. Skin biopsies met clinical criteria for GWI for 42 (82%) and 24 of 42 (57%) were determined to have SFN. Four of 9 (44%) veterans without GWI had positive SFN biopsies, though this difference was not statistically significant (Table 1). Veterans with SFN but no GWI were not included in the further analysis.

tahulabrusanuthapalabradimouathukeswagoswuclepaspucethapofruprufrumatraswastustepocrib

Thirty-five veterans with GWI—18 with SFN and 17 without SFN—completed the COMPASS 31 (Table 2). COMPASS 31 data were not analyzed for veterans without GWI. Individual domain scores and the difference in COMPASS 31 scores for veterans with GWI and SFN vs GWI and no SFN (38.3 vs 37.8, respectively) were not statistically significant.

jugucraspethouutrufribrokusletatredugitripruneswuchahiwridrihecribrucrawrashetrouotruhiphiduclopodratushapabuwijespuchospupijuchavebru

Sixteen of 24 veterans with GWI and SFN (67%) had ≥ 1 conditions that could potentially be responsible for SFN (Table 3), including 11 veterans (46%) with prediabetes/diabetes. Hyperlipidemia is only variably reported as a cause of SFN; when included, 19 of 24 (79%) SFN cases were accounted for. We could not identify a medical explanation for SFN in 5 of 24 veterans (21%) with GWI, which were deemed to be idiopathic.

 

 

Discussion

Biopsy-confirmed SFN was present in more than half of our sample of veterans with GWI, which is broadly consistent with what has been reported in the literature.13,35-38 In this clinical observation study, SFN was similarly prevalent in veterans with and without GWI; although it should be noted that biopsies only were obtained when there was a strong clinical suspicion for SFN. Almost half of patients with GWI did not have SFN, so our study does not support SFN as the underlying explanation for all GWI. Although our data cannot provide clinical guidance as to when skin biopsy may be indicated in GWI, work done in fibromyalgia found symptoms of dysautonomia and paresthesias are more specific for SFN and may be useful to help guide medical decision making.39

Veterans with GWI in our clinical sample reported a high burden of clinical symptoms conceivably attributable to ANS dysfunction. This symptom reporting is consistent with that seen in other GWI studies, as well as in other studies of SFN.4,5,7-9,14,15,34,38,40 Our clinical sample of veterans with GWI found no differences in the ANS symptom reporting between those with and without SFN. Therefore, our study cannot support SFN alone as accounting for ANS symptom burden in patients with GWI.

Two-thirds of biopsy-confirmed SFN in our clinical sample of veterans with GWI could potentially be explained by established medical conditions. As in other studies of SFN, prediabetes and diabetes represented a plurality (46%). Even after considering hyperlipidemia as a potential explanation, about 21% of SFN cases in veterans with GWI still were deemed idiopathic.

Evidence supports certain environmental agents as causal factors for GWI. Neurotoxicants reportedly related to GWI include pesticides (particularly organophosphates and carbamates), pyridostigmine bromide (used during the Gulf War as a prophylactic agent against the use of chemical weapons), and low levels of the nerve agent sarin from environmental contamination due to chemical weapons detonations.1 Some of these agents have been implicated in neuropathy as well.1,28-30 It is biologically plausible that deployment-related exposures could trigger SFN, though the traditional consensus has been that remote exposure to neurotoxic substances is unlikely to produce neuropathy that presents many years after the exposure.41 In the WRIISC clinical experience, however, veterans often report that their neuropathic symptoms predate the diagnosis of the associated medical conditions, sometimes by decades. It is conceivable that remote exposures may trigger the condition that is then potentiated by ongoing exposures, metabolic factors, and/or other medical conditions. These may perpetuate neuropathic symptoms and the illness experience of affected veterans. Our clinical observation study cannot clarify the extent to which this may be the case. Despite these findings and arguments, an environmental contribution to SFN cannot be discounted, and further research is needed to explore a potential relationship.

Limitations

This study’s conclusions are limited by its observational/retrospective design in a relatively small clinical sample of veterans evaluated at a tertiary referral center for postdeployment exposure-related health concerns. The WRIISC clinical sample is not representative of all GWVs or even of all veterans with GWI, as there is inherent selection bias as to who gets referred to and evaluated at the WRIISC. As with studies based on retrospective chart review, data are reliant on clinical documentation andaccuracy/consistency of the reviewer. Evaluation for SFN with skin biopsy is an invasive procedure and was performed when a high index of clinical suspicion for this condition existed, possibly representing confirmation bias. Therefore, the relatively high prevalence ofbiopsy-confirmed SFN seen in our clinical sample cannot be generalized to GWVs as a whole or even to veterans with GWI.

 

 

Assessment of autonomic dysfunction was based on COMPASS 31 symptom reporting by an small subset of the clinical cohort. Symptom reporting may not be reflective of true abnormality in ANS function. Physiologic tests of the ANS were not performed; such studies could more objectively establish whether ANS dysfunction is more prevalent in GWI veterans with SFN.

Evaluation for all potential etiologic/contributory conditions to SFN was not exhaustive. For example, sodium channel gene mutations have been documented to account for up to one-third of all cases of idiopathic SFN.42 For those cases in which no compelling etiology was identified, it is plausible that medical explanations for SFN may be found on further investigation.

Clinical assessments at the WRIISC were performed on GWVs ≥ 26 years after their deployment-related exposures. Other conditions/exposures may have occurred in the interim. What is not clear is whether the SFN predated the onset of any of these medical conditions or other putative contributors. This observational study is not able to tease out a temporal association to make a cause-and-effect assessment.

 

Conclusions

Retrospective analysis of clinical data of veterans evaluated at a specialized center for postdeployment health demonstrated that skin biopsy–confirmed SFN was prevalent, but not ubiquitous, in veterans with GWI. Symptom that may be attributed to ANS dysfunction in this clinical sample was consistent with literature on SFN and with GWI, but we could not definitively attribute ANS symptoms to SFN. Our study does not support the hypothesis that GWI symptoms are solely due to SFN, though it may still be relevant in a subset of veterans with GWI with strongly suggestive clinical features. We were able to identify a potential etiology for SFN in most veterans with GWI. Further investigations are recommended to explore any potential relationship between Gulf War exposures and SFN.

Following deployment to operations Desert Shield and Desert Storm (Gulf War) in 1990 and 1991, many Gulf War veterans (GWVs) developed chronic, complex symptoms, including pain, dyscognition, and fatigue, with gastrointestinal, skin, and respiratory manifestations. This Gulf War Illness (GWI) is reported to affect about 30% of those deployed. More than 30 years later, there is no consensus as to the etiology of GWI, although some deployment-related exposures have been implicated.1

Accepted research definitions for GWI include the Centers for Disease Control and Prevention and Kansas definitions.2 The US Department of Veterans Affairs (VA) uses the terminology chronic multisymptom illness (CMI), which is an overarching diagnosis under which GWI falls. Although there is no consensus case definition for CMI, there is overlap with conditions such as fibromyalgia, myalgic encephalomyelitis/chronic fatigue syndrome, and irritable bowel syndrome; the VA considers these as qualifying clinical diagnoses.3 The pathophysiology of GWI is also unknown, though a frequently reported unifying feature is that of autonomic nervous system (ANS) dysfunction. Studies have demonstrated differences between veterans with GWI and those without GWI in both the reporting of symptoms attributable to ANS dysfunction and in physiologic evaluations of the ANS.4-10

Small fiber neuropathy (SFN), a condition with damage to the A-δ and C small nerve fibers, has been proposed as a potential mechanism for the pain and ANS dysfunction experienced in GWI.11-13 Symptoms of SFN are similar to those of GWI, with pain and ANS symptoms commonly reported.14,15 There are multiple diagnostic criteria for SFN, the most commonly used requiring the presence of appropriate symptoms in the absence of large fiber neuropathy and a skin biopsy demonstrating reduced intraepidermal nerve fiber density.16-19 Several conditions reportedly cause SFN, most notably diabetes/prediabetes. Autoimmune disease, vitamin B12 deficiency, monoclonal gammopathies, celiac disease, paraneoplastic syndromes, and sodium channel gene mutations may also contribute to SFN.20 Hyperlipidemia has been identified as a contributor, although it has been variably reported.21,22

Idiopathic neuropathies, SFN included, may be secondary to neurotoxicant exposures. Agents whose exposure or consumption have been associated with SFN include alcohol most prominently, but also the organic solvent n-hexane, heavy metals, and excess vitamin B6.20,23-25 Agents associated with large fiber neuropathy may also have relevance for SFN, as small fibers have been likened to the “canary in the coal mine” in that they may be more susceptible to neurotoxicants and are affected earlier in the disease process.26 In this way, SFN may be the harbinger of large fiber neuropathy in some cases. Of specific relevance for GWVs, organophosphates and carbamates are known to produce a delayed onset large fiber neuropathy.27-30 Exposure to petrochemical solvents has also been associated with large fiber neuropathies.31,32

The War Related Illness and Injury Study Center (WRIISC) is a clinical, research, and education center established by Congress in 2001. Its primary focus is on military exposures and postdeployment health of veterans. It is located at 3 sites: East Orange, New Jersey; Washington, DC; and Palo Alto, California. The New Jersey WRIISC began a program to evaluate GWVs with characteristic symptoms for possible SFN with use of a skin biopsy.

We hypothesize that SFN may underly much of GWI symptomatology and may not be accounted for by the putative etiologies detailed in review of the medical literature. This retrospective review of clinical evaluations for SFN in GWVs who sought care at the New Jersey WRIISC explored and addressed the following questions: (1) how common is biopsy-confirmed SFN in veterans with GWI; (2) do veterans with GWI and SFN report more symptoms attributable to ANS dysfunction when compared with veterans with GWI and no SFN; and (3) can SFN in veterans with GWI and SFN be explained by conditions and substances commonly associated with SFN? Institutional review board approval and waiver of consent was obtained from the Veterans Affairs New Jersey Health Care Center for the study.

 

 

Methods

A retrospective chart review was conducted on veterans evaluated at the WRIISC from March 1, 2015, to January 31, 2019. Inclusion criteria were: deployment to operations Desert Shield and Desert Storm between August 2, 1990, and February 28, 1991, and skin biopsy conducted at the WRIISC. Skin biopsies were obtained at the discretion of an examining clinician based on clinical indications, including neuropathic pain, ANS symptoms, and/or a fibromyalgia/chronic pain–type presentation.

Electronic health record review explicitly abstracted GWI status, results of the skin biopsy, and ANS symptom burden as determined by the Composite Autonomic Symptom Scale 31 (COMPASS 31) completed at the time of the WRIISC evaluation. Determination of GWI was established as per the clinical opinion of the WRIISC lead clinician or environmental exposure clinician as evidenced by a diagnosis of fibromyalgia or chronic fatigue syndrome, or explicit statement of CMI/GWI in the patient assessment. A diagnosis of SFN was established if clinical signs were present and an intraepidermal nerve fiber density below the lower limits, as compared to normative data from the clinical diagnostic laboratory (Therapath Neuropathology), was documented.

COMPASS 31 assesses symptoms across 6 domains (orthostatic, vasomotor, secretomotor, gastrointestinal, bladder, andpupillomotor). Patients are asked about symptom frequency (rarely to almost always), severity (mild to severe), and improvement (much worse to completely gone). Individual domain scores and a total weighted score (0-100) have demonstrated good validity, reliability, and consistency in SFN.33,34

thuswichawrojomashis

In veterans with GWI and documented SFN, a health record review was performed to identify potential etiologies for SFN (Appendix).

 

Statistical Analysis

Microsoft Excel and IBM SPSS 12.0.1 for Windows were used for data collection and statistical analysis. Fisher exact test was used for comparing the prevalence of SFN in veterans with GWI vs without GWI. The independent samples t test was used for comparing COMPASS 31 scores for veterans with GWI by SFN status. α < .05 was used for determining statistical significance. For those GWVs documented with SFN and GWI, potential explanations were documented in total and by condition.

Results

tradibrawrustiphisosudrospenudivowobeswochitrodafrubusekurekurubrilistitiphosliwisespugaspidrastorochuuosepreselatriuarawruwresephisephuc

juprutestuuotrucliheshicradojepruwrotrijotihatrethuswumospejoriphagashugutejistemugistastushiuispakejathagobrithechecujej

From March 1, 2015, to January 31, 2019, 141 GWVs received a comprehensive in person clinical evaluation at the WRIISC and 51 veterans (36%) received a skin biopsy and were included in this retrospective observational study (Figure). The mean age was 48.6 years, and the majority were male and served in the US Army. Skin biopsies met clinical criteria for GWI for 42 (82%) and 24 of 42 (57%) were determined to have SFN. Four of 9 (44%) veterans without GWI had positive SFN biopsies, though this difference was not statistically significant (Table 1). Veterans with SFN but no GWI were not included in the further analysis.

tahulabrusanuthapalabradimouathukeswagoswuclepaspucethapofruprufrumatraswastustepocrib

Thirty-five veterans with GWI—18 with SFN and 17 without SFN—completed the COMPASS 31 (Table 2). COMPASS 31 data were not analyzed for veterans without GWI. Individual domain scores and the difference in COMPASS 31 scores for veterans with GWI and SFN vs GWI and no SFN (38.3 vs 37.8, respectively) were not statistically significant.

jugucraspethouutrufribrokusletatredugitripruneswuchahiwridrihecribrucrawrashetrouotruhiphiduclopodratushapabuwijespuchospupijuchavebru

Sixteen of 24 veterans with GWI and SFN (67%) had ≥ 1 conditions that could potentially be responsible for SFN (Table 3), including 11 veterans (46%) with prediabetes/diabetes. Hyperlipidemia is only variably reported as a cause of SFN; when included, 19 of 24 (79%) SFN cases were accounted for. We could not identify a medical explanation for SFN in 5 of 24 veterans (21%) with GWI, which were deemed to be idiopathic.

 

 

Discussion

Biopsy-confirmed SFN was present in more than half of our sample of veterans with GWI, which is broadly consistent with what has been reported in the literature.13,35-38 In this clinical observation study, SFN was similarly prevalent in veterans with and without GWI; although it should be noted that biopsies only were obtained when there was a strong clinical suspicion for SFN. Almost half of patients with GWI did not have SFN, so our study does not support SFN as the underlying explanation for all GWI. Although our data cannot provide clinical guidance as to when skin biopsy may be indicated in GWI, work done in fibromyalgia found symptoms of dysautonomia and paresthesias are more specific for SFN and may be useful to help guide medical decision making.39

Veterans with GWI in our clinical sample reported a high burden of clinical symptoms conceivably attributable to ANS dysfunction. This symptom reporting is consistent with that seen in other GWI studies, as well as in other studies of SFN.4,5,7-9,14,15,34,38,40 Our clinical sample of veterans with GWI found no differences in the ANS symptom reporting between those with and without SFN. Therefore, our study cannot support SFN alone as accounting for ANS symptom burden in patients with GWI.

Two-thirds of biopsy-confirmed SFN in our clinical sample of veterans with GWI could potentially be explained by established medical conditions. As in other studies of SFN, prediabetes and diabetes represented a plurality (46%). Even after considering hyperlipidemia as a potential explanation, about 21% of SFN cases in veterans with GWI still were deemed idiopathic.

Evidence supports certain environmental agents as causal factors for GWI. Neurotoxicants reportedly related to GWI include pesticides (particularly organophosphates and carbamates), pyridostigmine bromide (used during the Gulf War as a prophylactic agent against the use of chemical weapons), and low levels of the nerve agent sarin from environmental contamination due to chemical weapons detonations.1 Some of these agents have been implicated in neuropathy as well.1,28-30 It is biologically plausible that deployment-related exposures could trigger SFN, though the traditional consensus has been that remote exposure to neurotoxic substances is unlikely to produce neuropathy that presents many years after the exposure.41 In the WRIISC clinical experience, however, veterans often report that their neuropathic symptoms predate the diagnosis of the associated medical conditions, sometimes by decades. It is conceivable that remote exposures may trigger the condition that is then potentiated by ongoing exposures, metabolic factors, and/or other medical conditions. These may perpetuate neuropathic symptoms and the illness experience of affected veterans. Our clinical observation study cannot clarify the extent to which this may be the case. Despite these findings and arguments, an environmental contribution to SFN cannot be discounted, and further research is needed to explore a potential relationship.

Limitations

This study’s conclusions are limited by its observational/retrospective design in a relatively small clinical sample of veterans evaluated at a tertiary referral center for postdeployment exposure-related health concerns. The WRIISC clinical sample is not representative of all GWVs or even of all veterans with GWI, as there is inherent selection bias as to who gets referred to and evaluated at the WRIISC. As with studies based on retrospective chart review, data are reliant on clinical documentation andaccuracy/consistency of the reviewer. Evaluation for SFN with skin biopsy is an invasive procedure and was performed when a high index of clinical suspicion for this condition existed, possibly representing confirmation bias. Therefore, the relatively high prevalence ofbiopsy-confirmed SFN seen in our clinical sample cannot be generalized to GWVs as a whole or even to veterans with GWI.

 

 

Assessment of autonomic dysfunction was based on COMPASS 31 symptom reporting by an small subset of the clinical cohort. Symptom reporting may not be reflective of true abnormality in ANS function. Physiologic tests of the ANS were not performed; such studies could more objectively establish whether ANS dysfunction is more prevalent in GWI veterans with SFN.

Evaluation for all potential etiologic/contributory conditions to SFN was not exhaustive. For example, sodium channel gene mutations have been documented to account for up to one-third of all cases of idiopathic SFN.42 For those cases in which no compelling etiology was identified, it is plausible that medical explanations for SFN may be found on further investigation.

Clinical assessments at the WRIISC were performed on GWVs ≥ 26 years after their deployment-related exposures. Other conditions/exposures may have occurred in the interim. What is not clear is whether the SFN predated the onset of any of these medical conditions or other putative contributors. This observational study is not able to tease out a temporal association to make a cause-and-effect assessment.

 

Conclusions

Retrospective analysis of clinical data of veterans evaluated at a specialized center for postdeployment health demonstrated that skin biopsy–confirmed SFN was prevalent, but not ubiquitous, in veterans with GWI. Symptom that may be attributed to ANS dysfunction in this clinical sample was consistent with literature on SFN and with GWI, but we could not definitively attribute ANS symptoms to SFN. Our study does not support the hypothesis that GWI symptoms are solely due to SFN, though it may still be relevant in a subset of veterans with GWI with strongly suggestive clinical features. We were able to identify a potential etiology for SFN in most veterans with GWI. Further investigations are recommended to explore any potential relationship between Gulf War exposures and SFN.

References

1. White RF, Steele L, O’Callaghan JP, et al. Recent research on Gulf War illness and other health problems in veterans of the 1991 Gulf War: effects of toxicant exposures during deployment. Cortex. 2016;74:449-475. doi:10.1016/j.cortex.2015.08.022

2. Committee on the Development of a Consensus Case Definition for Chronic Multisymptom Illness in 1990-1991 Gulf War Veterans, Board on the Health of Select Populations, Institute of Medicine. Chronic Multisymptom Illness in Gulf War Veterans: Case Definitions Reexamined. National Academies Press; 2014.

3. Robbins R, Helmer D, Monahan P, et al. Management of chronic multisymptom illness: synopsis of the 2021 US Department of Veterans Affairs and US Department of Defense Clinical Practice Guideline. Mayo Clin Proc. 2022;97(5):991-1002. doi:10.1016/j.mayocp.2022.01.031

4. Fox A, Helmer D, Tseng CL, Patrick-DeLuca L, Osinubi O. Report of autonomic symptoms in a clinical sample of veterans with Gulf War Illness. Mil Med. 2018;183(3-4):e179-e185. doi:10.1093/milmed/usx052

5. Fox A, Helmer D, Tseng CL, McCarron K, Satcher S, Osinubi O. Autonomic symptoms in Gulf War veterans evaluated at the War Related Illness and Injury Study Center. Mil Med. 2019;184(3-4):e191-e196. doi:10.1093/milmed/usy227

6. Reyes L, Falvo M, Blatt M, Ghobreal B, Acosta A, Serrador J. Autonomic dysfunction in veterans with Gulf War illness [abstract]. FASEB J. 2014;28(S1):1068.19. doi:10.1096/fasebj.28.1_supplement.1068.19

7. Haley RW, Charuvastra E, Shell WE, et al. Cholinergic autonomic dysfunction in veterans with Gulf War illness: confirmation in a population-based sample. JAMA Neurol. 2013;70(2):191-200. doi:10.1001/jamaneurol.2013.596

8. Haley RW, Vongpatanasin W, Wolfe GI, et al. Blunted circadian variation in autonomic regulation of sinus node function in veterans with Gulf War syndrome. Am J Med. 2004;117(7):469-478. doi:10.1016/j.amjmed.2004.03.041

9. Avery TJ, Mathersul DC, Schulz-Heik RJ, Mahoney L, Bayley PJ. Self-reported autonomic dysregulation in Gulf War Illness. Mil Med. Published online December 30, 2021. doi:10.1093/milmed/usab546

10. Verne ZT, Fields JZ, Zhang BB, Zhou Q. Autonomic dysfunction and gastroparesis in Gulf War veterans. J Investig Med. 2023;71(1):7-10. doi:10.1136/jim-2021-002291

11. Levine TD. Small fiber neuropathy: disease classification beyond pain and burning. J Cent Nerv Syst Dis. 2018;10:1179573518771703. doi:10.1177/1179573518771703

12. Novak P. Autonomic disorders. Am J Med. 2019;132(4):420-436. doi:10.1016/j.amjmed.2018.09.027

13. Oaklander AL, Klein MM. Undiagnosed small-fiber polyneuropathy: is it a component of Gulf War Illness? Defense Technical Information Center. Accessed February 21, 2024. https://apps.dtic.mil/sti/citations/ADA613891

<--pagebreak-->14. Sène D. Small fiber neuropathy: diagnosis, causes, and treatment. Joint Bone Spine. 2018;85(5):553-559. doi:10.1016/j.jbspin.2017.11.002

15. Novak V, Freimer ML, Kissel JT, et al. Autonomic impairment in painful neuropathy. Neurology. 2001;56(7):861-868. doi:10.1212/wnl.56.7.861

16. Myers MI, Peltier AC. Uses of skin biopsy for sensory and autonomic nerve assessment. Curr Neurol Neurosci Rep. 2013;13(1):323. doi:10.1007/s11910-012-0323-2

17. Haroutounian S, Todorovic MS, Leinders M, et al. Diagnostic criteria for idiopathic small fiber neuropathy: a systematic review. Muscle Nerve. 2021;63(2):170-177. doi:10.1002/mus.27070

18. Levine TD, Saperstein DS. Routine use of punch biopsy to diagnose small fiber neuropathy in fibromyalgia patients. Clin Rheumatol. 2015;34(3):413-417. doi:10.1007/s10067-014-2850-5

19. England JD, Gronseth G S, Franklin G, et al. Practice parameter: the evaluation of distal symmetric polyneuropathy: the role of autonomic testing, nerve biopsy, and skin biopsy (an evidence-based review). Report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. PM R. 2009;1(1):14-22. doi:10.1016/j.pmrj.2008.11.011

20. de Greef BTA, Hoeijmakers JGJ, Gorissen-Brouwers CML, Geerts M, Faber CG, Merkies ISJ. Associated conditions in small fiber neuropathy - a large cohort study and review of the literature. Eur J Neurol. 2018;25(2):348-355. doi:10.1111/ene.13508

21. Morkavuk G, Leventoglu A. Small fiber neuropathy associated with hyperlipidemia: utility of cutaneous silent periods and autonomic tests. ISRN Neurol. 2014;2014:579242. doi:10.1155/2014/579242

22. Bednarik J, Vlckova-Moravcova E, Bursova S, Belobradkova J, Dusek L, Sommer C. Etiology of small-fiber neuropathy. J Peripher Nerv Syst. 2009;14(3):177-183. doi:10.1111/j.1529-8027.2009.00229.x

23. Kokotis P, Papantoniou M, Schmelz M, Buntziouka C, Tzavellas E, Paparrigopoulos T. Pure small fiber neuropathy in alcohol dependency detected by skin biopsy. Alcohol Fayettev N. 2023;111:67-73. doi:10.1016/j.alcohol.2023.05.006

24. Guimarães-Costa R, Schoindre Y, Metlaine A, et al. N-hexane exposure: a cause of small fiber neuropathy. J Peripher Nerv Syst. 2018;23(2):143-146. doi:10.1111/jns.12261

25. Koszewicz M, Markowska K, Waliszewska-Prosol M, et al. The impact of chronic co-exposure to different heavy metals on small fibers of peripheral nerves. A study of metal industry workers. J Occup Med Toxicol. 2021;16(1):12. doi:10.1186/s12995-021-00302-6

26. Johns Hopkins Medicine. Small nerve fibers defy neuropathy conventions. April 11, 2016. Accessed February 21, 2024. https://www.hopkinsmedicine.org/news/media/releases/small_nerve_fibers_defy_neuropathy_conventions

27. Jett DA. Neurotoxic pesticides and neurologic effects. Neurol Clin. 2011;29(3):667-677. doi:10.1016/j.ncl.2011.06.002

28. Berger AR, Schaumburg HH. Human toxic neuropathy caused by industrial agents. In: Dyck PJ, Thomas PK, eds. Peripheral Neuropathy. 4th ed. Saunders; 2005:2505-2525. doi:10.1016/B978-0-7216-9491-7.50115-0

29. Herskovitz S, Schaumburg HH. Neuropathy caused by drugs. In: Dyck PJ, Thomas PK, eds. Peripheral Neuropathy. 4th ed. Saunders; 2005:2553-2583.

30. Katona I, Weis J. Chapter 31 - Diseases of the peripheral nerves. Handb Clin Neurol. 2017;145:453-474. doi:10.1016/B978-0-12-802395-2.00031-6

31. Matikainen E, Juntunen J. Autonomic nervous system dysfunction in workers exposed to organic solvents. J Neurol Neurosurg Psychiatry. 1985;48(10):1021-1024. doi:10.1136/jnnp.48.10.1021

32. Murata K, Araki S, Yokoyama K, Maeda K. Autonomic and peripheral nervous system dysfunction in workers exposed to mixed organic solvents. Int Arch Occup Environ Health. 1991;63(5):335-340. doi:10.1007/BF00381584

33. Sletten DM, Suarez GA, Low PA, Mandrekar J, Singer W. COMPASS 31: a refined and abbreviated Composite Autonomic Symptom Score. Mayo Clin Proc. 2012;87(12):1196-1201. doi:10.1016/j.mayocp.2012.10.013

34. Treister R, O’Neil K, Downs HM, Oaklander AL. Validation of the Composite Autonomic Symptom Scale-31 (COMPASS-31) in patients with and without small-fiber polyneuropathy. Eur J Neurol. 2015;22(7):1124-1130. doi:10.1111/ene.12717

35. Joseph P, Arevalo C, Oliveira RKF, et al. Insights from invasive cardiopulmonary exercise testing of patients with myalgic encephalomyelitis/chronic fatigue syndrome. Chest. 2021;160(2):642-651. doi:10.1016/j.chest.2021.01.082

36. Giannoccaro MP, Donadio V, Incensi A, Avoni P, Liguori R. Small nerve fiber involvement in patients referred for fibromyalgia. Muscle Nerve. 2014;49(5):757-759. doi:10.1002/mus.24156

37. Oaklander AL, Herzog ZD, Downs HM, Klein MM. Objective evidence that small-fiber polyneuropathy underlies some illnesses currently labeled as fibromyalgia. Pain. 2013;154(11):2310-2316. doi:10.1016/j.pain.2013.06.001

38. Serrador JM. Diagnosis of late-stage, early-onset, small-fiber polyneuropathy. Defense Technical Information Center. December 1, 2019. Accessed February 21, 2024. https://apps.dtic.mil/sti/citations/AD1094831

39. Lodahl M, Treister R, Oaklander AL. Specific symptoms may discriminate between fibromyalgia patients with vs without objective test evidence of small-fiber polyneuropathy. Pain Rep. 2018;3(1):e633. doi:10.1097/PR9.0000000000000633

40. Sastre A, Cook MR. Autonomic dysfunction in Gulf War veterans. Defense Technical Information Center. April 1, 2004. Accessed February 21, 2024. https://apps.dtic.mil/sti/citations/ADA429525

41. Little AA, Albers JW. Clinical description of toxic neuropathies. Handb Clin Neurol. 2015;131:253-296. doi:10.1016/B978-0-444-62627-1.00015-9

42. Faber CG, Hoeijmakers JGJ, Ahn HS, et al. Gain of function NaV1.7 mutations in idiopathic small fiber neuropathy. Ann Neurol. 2012;71(1):26-39.

References

1. White RF, Steele L, O’Callaghan JP, et al. Recent research on Gulf War illness and other health problems in veterans of the 1991 Gulf War: effects of toxicant exposures during deployment. Cortex. 2016;74:449-475. doi:10.1016/j.cortex.2015.08.022

2. Committee on the Development of a Consensus Case Definition for Chronic Multisymptom Illness in 1990-1991 Gulf War Veterans, Board on the Health of Select Populations, Institute of Medicine. Chronic Multisymptom Illness in Gulf War Veterans: Case Definitions Reexamined. National Academies Press; 2014.

3. Robbins R, Helmer D, Monahan P, et al. Management of chronic multisymptom illness: synopsis of the 2021 US Department of Veterans Affairs and US Department of Defense Clinical Practice Guideline. Mayo Clin Proc. 2022;97(5):991-1002. doi:10.1016/j.mayocp.2022.01.031

4. Fox A, Helmer D, Tseng CL, Patrick-DeLuca L, Osinubi O. Report of autonomic symptoms in a clinical sample of veterans with Gulf War Illness. Mil Med. 2018;183(3-4):e179-e185. doi:10.1093/milmed/usx052

5. Fox A, Helmer D, Tseng CL, McCarron K, Satcher S, Osinubi O. Autonomic symptoms in Gulf War veterans evaluated at the War Related Illness and Injury Study Center. Mil Med. 2019;184(3-4):e191-e196. doi:10.1093/milmed/usy227

6. Reyes L, Falvo M, Blatt M, Ghobreal B, Acosta A, Serrador J. Autonomic dysfunction in veterans with Gulf War illness [abstract]. FASEB J. 2014;28(S1):1068.19. doi:10.1096/fasebj.28.1_supplement.1068.19

7. Haley RW, Charuvastra E, Shell WE, et al. Cholinergic autonomic dysfunction in veterans with Gulf War illness: confirmation in a population-based sample. JAMA Neurol. 2013;70(2):191-200. doi:10.1001/jamaneurol.2013.596

8. Haley RW, Vongpatanasin W, Wolfe GI, et al. Blunted circadian variation in autonomic regulation of sinus node function in veterans with Gulf War syndrome. Am J Med. 2004;117(7):469-478. doi:10.1016/j.amjmed.2004.03.041

9. Avery TJ, Mathersul DC, Schulz-Heik RJ, Mahoney L, Bayley PJ. Self-reported autonomic dysregulation in Gulf War Illness. Mil Med. Published online December 30, 2021. doi:10.1093/milmed/usab546

10. Verne ZT, Fields JZ, Zhang BB, Zhou Q. Autonomic dysfunction and gastroparesis in Gulf War veterans. J Investig Med. 2023;71(1):7-10. doi:10.1136/jim-2021-002291

11. Levine TD. Small fiber neuropathy: disease classification beyond pain and burning. J Cent Nerv Syst Dis. 2018;10:1179573518771703. doi:10.1177/1179573518771703

12. Novak P. Autonomic disorders. Am J Med. 2019;132(4):420-436. doi:10.1016/j.amjmed.2018.09.027

13. Oaklander AL, Klein MM. Undiagnosed small-fiber polyneuropathy: is it a component of Gulf War Illness? Defense Technical Information Center. Accessed February 21, 2024. https://apps.dtic.mil/sti/citations/ADA613891

<--pagebreak-->14. Sène D. Small fiber neuropathy: diagnosis, causes, and treatment. Joint Bone Spine. 2018;85(5):553-559. doi:10.1016/j.jbspin.2017.11.002

15. Novak V, Freimer ML, Kissel JT, et al. Autonomic impairment in painful neuropathy. Neurology. 2001;56(7):861-868. doi:10.1212/wnl.56.7.861

16. Myers MI, Peltier AC. Uses of skin biopsy for sensory and autonomic nerve assessment. Curr Neurol Neurosci Rep. 2013;13(1):323. doi:10.1007/s11910-012-0323-2

17. Haroutounian S, Todorovic MS, Leinders M, et al. Diagnostic criteria for idiopathic small fiber neuropathy: a systematic review. Muscle Nerve. 2021;63(2):170-177. doi:10.1002/mus.27070

18. Levine TD, Saperstein DS. Routine use of punch biopsy to diagnose small fiber neuropathy in fibromyalgia patients. Clin Rheumatol. 2015;34(3):413-417. doi:10.1007/s10067-014-2850-5

19. England JD, Gronseth G S, Franklin G, et al. Practice parameter: the evaluation of distal symmetric polyneuropathy: the role of autonomic testing, nerve biopsy, and skin biopsy (an evidence-based review). Report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. PM R. 2009;1(1):14-22. doi:10.1016/j.pmrj.2008.11.011

20. de Greef BTA, Hoeijmakers JGJ, Gorissen-Brouwers CML, Geerts M, Faber CG, Merkies ISJ. Associated conditions in small fiber neuropathy - a large cohort study and review of the literature. Eur J Neurol. 2018;25(2):348-355. doi:10.1111/ene.13508

21. Morkavuk G, Leventoglu A. Small fiber neuropathy associated with hyperlipidemia: utility of cutaneous silent periods and autonomic tests. ISRN Neurol. 2014;2014:579242. doi:10.1155/2014/579242

22. Bednarik J, Vlckova-Moravcova E, Bursova S, Belobradkova J, Dusek L, Sommer C. Etiology of small-fiber neuropathy. J Peripher Nerv Syst. 2009;14(3):177-183. doi:10.1111/j.1529-8027.2009.00229.x

23. Kokotis P, Papantoniou M, Schmelz M, Buntziouka C, Tzavellas E, Paparrigopoulos T. Pure small fiber neuropathy in alcohol dependency detected by skin biopsy. Alcohol Fayettev N. 2023;111:67-73. doi:10.1016/j.alcohol.2023.05.006

24. Guimarães-Costa R, Schoindre Y, Metlaine A, et al. N-hexane exposure: a cause of small fiber neuropathy. J Peripher Nerv Syst. 2018;23(2):143-146. doi:10.1111/jns.12261

25. Koszewicz M, Markowska K, Waliszewska-Prosol M, et al. The impact of chronic co-exposure to different heavy metals on small fibers of peripheral nerves. A study of metal industry workers. J Occup Med Toxicol. 2021;16(1):12. doi:10.1186/s12995-021-00302-6

26. Johns Hopkins Medicine. Small nerve fibers defy neuropathy conventions. April 11, 2016. Accessed February 21, 2024. https://www.hopkinsmedicine.org/news/media/releases/small_nerve_fibers_defy_neuropathy_conventions

27. Jett DA. Neurotoxic pesticides and neurologic effects. Neurol Clin. 2011;29(3):667-677. doi:10.1016/j.ncl.2011.06.002

28. Berger AR, Schaumburg HH. Human toxic neuropathy caused by industrial agents. In: Dyck PJ, Thomas PK, eds. Peripheral Neuropathy. 4th ed. Saunders; 2005:2505-2525. doi:10.1016/B978-0-7216-9491-7.50115-0

29. Herskovitz S, Schaumburg HH. Neuropathy caused by drugs. In: Dyck PJ, Thomas PK, eds. Peripheral Neuropathy. 4th ed. Saunders; 2005:2553-2583.

30. Katona I, Weis J. Chapter 31 - Diseases of the peripheral nerves. Handb Clin Neurol. 2017;145:453-474. doi:10.1016/B978-0-12-802395-2.00031-6

31. Matikainen E, Juntunen J. Autonomic nervous system dysfunction in workers exposed to organic solvents. J Neurol Neurosurg Psychiatry. 1985;48(10):1021-1024. doi:10.1136/jnnp.48.10.1021

32. Murata K, Araki S, Yokoyama K, Maeda K. Autonomic and peripheral nervous system dysfunction in workers exposed to mixed organic solvents. Int Arch Occup Environ Health. 1991;63(5):335-340. doi:10.1007/BF00381584

33. Sletten DM, Suarez GA, Low PA, Mandrekar J, Singer W. COMPASS 31: a refined and abbreviated Composite Autonomic Symptom Score. Mayo Clin Proc. 2012;87(12):1196-1201. doi:10.1016/j.mayocp.2012.10.013

34. Treister R, O’Neil K, Downs HM, Oaklander AL. Validation of the Composite Autonomic Symptom Scale-31 (COMPASS-31) in patients with and without small-fiber polyneuropathy. Eur J Neurol. 2015;22(7):1124-1130. doi:10.1111/ene.12717

35. Joseph P, Arevalo C, Oliveira RKF, et al. Insights from invasive cardiopulmonary exercise testing of patients with myalgic encephalomyelitis/chronic fatigue syndrome. Chest. 2021;160(2):642-651. doi:10.1016/j.chest.2021.01.082

36. Giannoccaro MP, Donadio V, Incensi A, Avoni P, Liguori R. Small nerve fiber involvement in patients referred for fibromyalgia. Muscle Nerve. 2014;49(5):757-759. doi:10.1002/mus.24156

37. Oaklander AL, Herzog ZD, Downs HM, Klein MM. Objective evidence that small-fiber polyneuropathy underlies some illnesses currently labeled as fibromyalgia. Pain. 2013;154(11):2310-2316. doi:10.1016/j.pain.2013.06.001

38. Serrador JM. Diagnosis of late-stage, early-onset, small-fiber polyneuropathy. Defense Technical Information Center. December 1, 2019. Accessed February 21, 2024. https://apps.dtic.mil/sti/citations/AD1094831

39. Lodahl M, Treister R, Oaklander AL. Specific symptoms may discriminate between fibromyalgia patients with vs without objective test evidence of small-fiber polyneuropathy. Pain Rep. 2018;3(1):e633. doi:10.1097/PR9.0000000000000633

40. Sastre A, Cook MR. Autonomic dysfunction in Gulf War veterans. Defense Technical Information Center. April 1, 2004. Accessed February 21, 2024. https://apps.dtic.mil/sti/citations/ADA429525

41. Little AA, Albers JW. Clinical description of toxic neuropathies. Handb Clin Neurol. 2015;131:253-296. doi:10.1016/B978-0-444-62627-1.00015-9

42. Faber CG, Hoeijmakers JGJ, Ahn HS, et al. Gain of function NaV1.7 mutations in idiopathic small fiber neuropathy. Ann Neurol. 2012;71(1):26-39.

Issue
Federal Practitioner - 41(5)a
Issue
Federal Practitioner - 41(5)a
Page Number
136-141
Page Number
136-141
Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>0524 FED GWI</fileName> <TBEID>0C02F3C5.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F3C5</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname/> <articleType>1</articleType> <TBLocation>Copyfitting-FED</TBLocation> <QCDate/> <firstPublished>20240507T114059</firstPublished> <LastPublished>20240507T114059</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240507T114059</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline/> <bylineText>Edward C. Shadiack III, DO, MPHa,b; Omowunmi Osinubi, MD, MPHa,b; Apollonia Gruber-Fox, PhDb; Chinmoy Bhate, MDa; Lydia Patrick-DeLuca, MSN, RNa,b; Philip Cohen, MDa; Drew A. Helmer, MD, MSb,c</bylineText> <bylineFull/> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Following deployment to operations Desert Shield and Desert Storm (Gulf War) in 1990 and 1991, many Gulf War veterans (GWVs) developed chronic, complex symptoms</metaDescription> <articlePDF/> <teaserImage/> <title>Small Fiber Neuropathy in Veterans With Gulf War Illness</title> <deck/> <eyebrow>Original Research</eyebrow> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>May</pubPubdateMonth> <pubPubdateDay/> <pubVolume>41</pubVolume> <pubNumber>5</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2951</CMSID> <CMSID>3639</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>FED</publicationCode> <pubIssueName>May 2024</pubIssueName> <pubArticleType>Feature Articles | 3639</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Feature | 2951<pubSubsection/></pubSection> </pubSections> <journalTitle>Fed Pract</journalTitle> <journalFullTitle>Federal Practitioner</journalFullTitle> <copyrightStatement>Copyright 2017 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">16</term> </publications> <sections> <term canonical="true">104</term> </sections> <topics> <term canonical="true">258</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Small Fiber Neuropathy in Veterans With Gulf War Illness</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><b>Background:</b> Gulf War veterans deployed to operations Desert Shield and Desert Storm returned with chronic multisystemic symptoms. This Gulf War Illness (GWI) has defied attempts to identify an underlying etiology. Pain and other symptoms attributable to autonomic nervous system (ANS) dysfunction are common, which may suggest a pathophysiologic underpinning. <hl name="33706"/>Small fiber neuropathy (SFN) presents with similar symptoms. Toxic exposures have been implicated in both SFN and GWI. <br/><br/><b>Methods:</b> A retrospective chart review of clinical data at the New Jersey War Related Illness and Injury Study Center addressed the following questions: (1) how common was biopsy-confirmed SFN in veterans with GWI; (2) do veterans with GWI and SFN report more symptoms attributable to ANS dysfunction as compared to veterans with GWI and no SFN; and (3) can SFN in veterans with GWI and SFN be explained by conditions commonly associated with SFN? Chart review abstracted GWI status, skin biopsy results, and ANS symptom burden. For veterans with GWI and SFN, additional chart abstraction was explored for commonly reported contributing conditions.<br/><br/><b>Results:</b> From March 1, 2015, to January 31, 2019, 51 Gulf War veterans evaluated at the War Related Illness and Injury Study center had a skin biopsy. Of these, 42 (83%) were diagnosed with GWI and 24 of 42 (57%) also had SFN. No differences were observed in ANS symptoms when compared with veterans with GWI and no SFN. A potential etiology for SFN was identified in16 of 24 (67%) veterans with GWI and SFN, increasing to 19 (79%) when hyperlipidemia was included. Our analysis did not identify an explanation in 5 of 24 (21%) veterans with GWI and SFN.<b>Conclusions:</b> SFN was common in this clinical sample of veterans diagnosed with GWI. A well-established potential etiology was identified in most cases of SFN. About 20% of veterans with GWI in our clinical sample had idiopathic SFN, and it is plausible that deployment-related exposures could have contributed to this condition. Symptoms of ANS are prevalent in GWI, though SFN cannot solely account for this. Our study does not generally support SFN as etiologic for GWI, though this may still be relevant for some. Additional research is required to explore relationships between Gulf War exposures and SFN.</p> <p><span class="Drop">F</span>ollowing deployment to operations Desert Shield and Desert Storm (Gulf War) in 1990 and 1991, many Gulf War veterans (GWVs) developed chronic, complex symptoms, including pain, dyscognition, and fatigue, with gastrointestinal, skin, and respiratory manifestations. This Gulf War Illness (GWI) is reported to affect about 30% of those deployed. More than 30 years later, there is no consensus as to the etiology of GWI, although some deployment-related exposures have been implicated.<sup>1</sup></p> <p>Accepted research definitions for GWI include the Centers for Disease Control and Prevention and Kansas definitions.<sup>2</sup> The US Department of Veterans Affairs (VA) uses the terminology <i>chronic</i> <i>multisymptom</i> <i>illness</i> (CMI), which is an overarching diagnosis under which GWI falls. Although there is no consensus case definition for CMI, there is overlap with conditions such as fibromyalgia, myalgic encephalomyelitis/chronic fatigue syndrome, and irritable bowel syndrome; the VA considers these as qualifying clinical diagnoses.<sup>3</sup> The pathophysiology of GWI is also unknown, though a frequently reported unifying feature is that of autonomic nervous system (ANS) dysfunction. Studies have demonstrated differences between veterans with GWI and those without GWI in both the reporting of symptoms attributable to ANS dysfunction and in physiologic evaluations of the ANS.<sup>4-10</sup><br/><br/>Small fiber neuropathy (SFN), a condition with damage to the A-δ and C small nerve fibers, has been proposed as a potential mechanism for the pain and ANS dysfunction experienced in GWI.<sup>11-13</sup> Symptoms of SFN are similar to those of GWI, with pain and ANS symptoms commonly reported.<sup>14,15</sup> There are multiple diagnostic criteria for SFN, the most commonly used requiring the presence of appropriate symptoms in the absence of large fiber neuropathy and a skin biopsy demonstrating reduced intraepidermal nerve fiber density.<sup>16-19</sup> Several conditions reportedly cause SFN, most notably diabetes/prediabetes, autoimmune disease, vitamin B12 deficiency, monoclonal gammopathies, celiac disease, paraneoplastic syndromes, and sodium channel gene mutations.<sup>20</sup> Hyperlipidemia has been identified as a contributor, although it has been variably reported.<sup>21,22</sup><br/><br/>Idiopathic neuropathies, SFN included, may be secondary to neurotoxicant exposures.Agents whose exposure or consumption have been associated with SFN include alcohol most prominently, but also the organic solvent n-hexane, heavy metals, and excess vitamin B<sub>6</sub>.<sup>20,23-25</sup> Agents associated with large fiber neuropathy may also have relevance for SFN, as small fibers have been likened to the “canary in the coal mine” in that they may be more susceptible to neurotoxicants and are affected earlier in the disease process.<sup>26</sup> In this way, SFN may be the harbinger of large fiber neuropathy in some cases. Of specific relevance for GWVs, organophosphates and carbamates are known to produce a delayed onset large fiber neuropathy.<sup>27-30</sup> Exposure to petrochemical solvents has also been associated with large fiber neuropathies.<sup>31,32</sup> <br/><br/>The War Related Illness and Injury Study Center (WRIISC) is a clinical, research, and education center established by Congress in 2001. Its primary focus is on military exposures and postdeployment health of veterans. It is located at 3 sites: East Orange, New Jersey; Washington, DC; and Palo Alto, California. The New Jersey WRIISC began a program to evaluate GWVs with characteristic symptoms for possible SFN with use of a skin biopsy. <br/><br/>We hypothesized that SFN may underly much of GWI symptomatology and may not be accounted for by the putative etiologies detailed in review of the medical literature. This retrospective review of clinical evaluations for SFN in GWVs who sought care at the New Jersey WRIISC explored and addressed the following questions: (1) how common is biopsy-confirmed SFN in veterans with GWI; (2) do veterans with GWI and SFN report more symptoms attributable to ANS dysfunction when compared with veterans with GWI and no SFN; and (3) can SFN in veterans with GWI and SFN be explained by conditions and substances commonly associated with SFN? Institutional review board approval and waiver of consent was obtained from the Veterans Affairs New Jersey Health Care Center for the study.</p> <h2>Methods</h2> <p>A retrospective chart review was conducted on veterans evaluated at the WRIISC from March 1, 2015, to January 31, 2019. Inclusion criteria were: deployment to operations Desert Shield and Desert Storm between August 2, 1990, and February 28, 1991, and skin biopsy conducted at the WRIISC. Skin biopsies were obtained at the discretion of an examining clinician based on clinical indications, including neuropathic pain, ANS symptoms, and/or a fibromyalgia/chronic pain–type presentation. </p> <p>Electronic health record review explicitly abstracted GWI status, results of the skin biopsy, and ANS symptom burden as determined by the Composite Autonomic Symptom Scale 31 (COMPASS 31) completed at the time of the WRIISC evaluation.<hl name="33707"/> Determination of GWI was established as per the clinical opinion of the WRIISC lead clinician or environmental exposure clinician as evidenced by a diagnosis of fibromyalgia or chronic fatigue syndrome, or explicit statement of CMI/GWI in the patient assessment. A diagnosis of SFN was established if clinical signs were present and an intraepidermal nerve fiber density below the lower limits, as compared to normative data from the clinical diagnostic laboratory (Therapath Neuropathology), was documented. <br/><br/>COMPASS 31 assesses symptoms across 6 domains (orthostatic, vasomotor, secretomotor, gastrointestinal, bladder, andpupillomotor). Patients are asked about symptom frequency (rarely to almost always), severity (mild to severe), and improvement (much worse to completely gone). Individual domain scores and a total weighted score (0-100) have demonstrated good validity, reliability, and consistency in SFN.<sup>33,34</sup>In veterans with GWI and documented SFN, a health record review was performed to identify potential etiologies for SFN (Appendix). </p> <h3>Statistical Analysis</h3> <p>Microsoft Excel and IBM SPSS 12.0.1 for Windows were used for data collection and statistical analysis. Fisher exact test was used for comparing the prevalence of SFN in veterans with GWI vs without GWI. The independent samples <i>t</i> test was used for comparing COMPASS 31 scores for veterans with GWI by SFN status. α &lt; .05 was used for determining statistical significance. For those GWVs documented with SFN and GWI, potential explanations were documented in total and by condition. </p> <h2>Results</h2> <p>From March 1, 2015, to January 31, 2019, 141 GWVs received a comprehensive in person clinical evaluation at the WRIISC and 51 veterans (36%) received a skin biopsy and were included in this retrospective observational study (Figure). The mean age was 48.6 years, and the majority were male and served in the US Army. Skin biopsies met clinical criteria for GWI for 42 (82%) and 24 of 42 (57%) were determined to have SFN. Four of 9 (44%) veterans without GWI had positive SFN biopsies, though this difference was not statistically significant (Table 1). Veterans with SFN but no GWI were not included in the further analysis. </p> <p>Thirty-five veterans with GWI—18 with SFN and 17 without SFN—completed the COMPASS 31 (Table 2). COMPASS 31 data were not analyzed for veterans without GWI. Individual domain scores and the difference in COMPASS 31 scores for veterans with GWI and SFN vs GWI and no SFN (38.3 vs 37.8, respectively) were not statistically significant.<br/><br/>Sixteen of 24 veterans with GWI and SFN (67%) had ≥ 1 conditions that could potentially be responsible for SFN (Table 3), including 11 veterans (46%) with prediabetes/diabetes. Hyperlipidemia is only variably reported as a cause of SFN; when included, 19 (79%) of 24 of SFN cases were accounted for. We could not identify a medical explanation for SFN in 5 of 24 veterans (21%) with GWI, which were deemed to be idiopathic.</p> <h2>Discussion</h2> <p>Biopsy-confirmed SFN was present in more than half of our sample of veterans with GWI, which is broadly consistent with what has been reported in the literature.<sup>13,35-38</sup> In this clinical observation study, SFN was similarly prevalent in veterans with and without GWI; although it should be noted that biopsies only were obtained when there was a strong clinical suspicion for SFN. Almost half of patients with GWI did not have SFN, so our study does not support SFN as the underlying explanation for all GWI. Although our data cannot provide clinical guidance as to when skin biopsy may be indicated in GWI, work done in fibromyalgia found symptoms of dysautonomia and paresthesias are more specific for SFN and may be useful to help guide medical decision making.<sup>39</sup></p> <p>Veterans with GWI in our clinical sample reported a high burden of clinical symptoms conceivably attributable to ANS dysfunction. This symptom reporting is consistent with that seen in other GWI studies, as well as in other studies of SFN.<sup>4,5,7-9,14,15,34,38,40</sup> Our clinical sample of veterans with GWI found no differences in the ANS symptom reporting between those with and without SFN. Therefore, our study cannot support SFN alone as accounting for ANS symptom burden in patients with GWI. <br/><br/>Two-thirds of biopsy-confirmed SFN in our clinical sample of veterans with GWI could potentially be explained by established medical conditions. As in other studies of SFN, prediabetes and diabetes represented a plurality (46%). Even after considering hyperlipidemia as a potential explanation, about 21% of SFN cases in veterans with GWI still were deemed idiopathic.<br/><br/>Evidence supports certain environmental agents as causal factors for GWI. Neurotoxicants reportedly related to GWI include pesticides (particularly organophosphates and carbamates), pyridostigmine bromide (used during the Gulf War as a prophylactic agent against the use of chemical weapons), and low levels of the nerve agent sarin from environmental contamination due to chemical weapons detonations.<sup>1</sup> Some of these agents have been implicated in neuropathy as well.<sup>1,28-30</sup> It is biologically plausible that deployment-related exposures could trigger SFN, though the traditional consensus has been that remote exposure to neurotoxic substances is unlikely to produce neuropathy that presents many years after the exposure.<sup>41</sup> In the WRIISC clinical experience, however, veterans often report that their neuropathic symptoms predate the diagnosis of the associated medical conditions, sometimes by decades. It is conceivable that remote exposures may trigger the condition that is then potentiated by ongoing exposures, metabolic factors, and/or other medical conditions. These may perpetuate neuropathic symptoms and the illness experience of affected veterans. Our clinical observation study cannot clarify the extent to which this may be the case. Despite these findings and arguments, an environmental contribution to SFN cannot be discounted, and further research is needed to explore a potential relationship.</p> <h3>Limitations</h3> <p>This study’s conclusions are limited by its observational/retrospective design in a relatively small clinical sample of veterans evaluated at a tertiary referral center for postdeployment exposure-related health concerns. The WRIISC clinical sample is not representative of all GWVs or even of all veterans with GWI, as there is inherent selection bias as to who gets referred to and evaluated at the WRIISC. As with studies based on retrospective chart review, data are reliant on clinical documentation andaccuracy/consistency of the reviewer. Evaluation for SFN with skin biopsy is an invasive procedure and was performed when a high index of clinical suspicion for this condition existed, possibly representing confirmation bias. Therefore, the relatively high prevalence ofbiopsy-confirmed SFN seen in our clinical sample cannot be generalized to GWVs as a whole or even to veterans with GWI. </p> <p>Assessment of autonomic dysfunction was based on COMPASS 31 symptom reporting by an small subset of the clinical cohort. Symptom reporting may not be reflective of true abnormality in ANS function. Physiologic tests of the ANS were not performed; such studies could more objectively establish whether ANS dysfunction is more prevalent in GWI veterans with SFN.<br/><br/>Evaluation for all potential etiologic/contributory conditions to SFN was not exhaustive. For example, sodium channel gene mutations have been documented to account for up to one-third of all cases of idiopathic SFN.<sup>42</sup> For those cases in which no compelling etiology was identified, it is plausible that medical explanations for SFN may be found on further investigation. <br/><br/>Clinical assessments at the WRIISC were performed on GWVs ≥ 26 years after their deployment-related exposures. Other conditions/exposures may have occurred in the interim. What is not clear is whether the SFN predated the onset of any of these medical conditions or other putative contributors. This observational study is not able to tease out a temporal association to make a cause-and-effect assessment. </p> <h2>Conclusions</h2> <p>Retrospective analysis of clinical data of veterans evaluated at a specialized center for postdeployment health demonstrated that skin biopsy–confirmed SFN was prevalent, but not ubiquitous, in veterans with GWI. Symptom that may be attributed to ANS dysfunction in this clinical sample was consistent with literature on SFN and with GWI, but we could not definitively attribute ANS symptoms to SFN. Our study does not support the hypothesis that GWI symptoms are solely due to SFN, though it may still be relevant in a subset of veterans with GWI with strongly suggestive clinical features. We were able to identify a potential etiology for SFN in most veterans with GWI. Further investigations are recommended to explore any potential relationship between Gulf War exposures and SFN.</p> <p class="isub">Author affiliations</p> <p> <em><sup>a</sup>Veterans Affairs New Jersey Health Care Systems, East Orange<br/><br/><sup>b</sup>War Related Illness and Injury Study Center, East Orange, New Jersey<br/><br/><sup>c</sup>Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas</em> </p> <p class="isub">Author contributions</p> <p> <em><i>Concept</i>: Shadiack.<br/><br/><i>Data collection</i>: Shadiack, Osinubi, Bhate, Patrick-DeLuca, Cohen.<i>Data analysis</i>: Shadiack, Gruber-Fox, Helmer.<br/><br/><i>Drafting of manuscript</i>: Shadiack, Osinubi.<br/><br/><i>Critical review of manuscript</i>: Gruber-Fox, Patrick-DeLuca, Cohen, Helmer.</em> </p> <p class="isub">Author disclosures </p> <p> <em>The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.</em> </p> <p class="isub">Disclaimer</p> <p> <em>The opinions expressed herein are those of the authors and do not necessarily reflect those of <i>Federal Practitioner</i>,Frontline Medical Communications Inc., the US Government, or any of its agencies.</em> </p> <p class="isub">Funding and ethics</p> <p> <em>This study was reviewed and approved by the Veterans Affairs New Jersey Health Care System Institutional Review Board (IRB# 01497). Funding provided by Veterans Affairs War Related Illness and Injury Study Center-New Jersey clinical resources; US Department of Veterans Affairs Health Services Research &amp; Development (CIN 13-413). Poster presented virtually at the American Autonomic Society’s annual meeting, November 2020.</em> </p> <h2>References</h2> <p class="reference"> 1. White RF, Steele L, O’Callaghan JP, et al. Recent research on Gulf War illness and other health problems in veterans of the 1991 Gulf War: effects of toxicant exposures during deployment. <i>Cortex</i>. 2016;74:449-475. doi:10.1016/j.cortex.2015.08.022<br/><br/> 2. Committee on the Development of a Consensus Case Definition for Chronic Multisymptom Illness in 1990-1991 Gulf War Veterans, Board on the Health of Select Populations, Institute of Medicine. <i>Chronic Multisymptom Illness in Gulf War Veterans: Case Definitions Reexamined</i>. National Academies Press; 2014. <br/><br/> 3. Robbins R, Helmer D, Monahan P, et al. Management of chronic multisymptom illness: synopsis of the 2021 US Department of Veterans Affairs and US Department of Defense Clinical Practice Guideline. <i>Mayo Clin Proc</i>. 2022;97(5):991-1002. doi:10.1016/j.mayocp.2022.01.031<br/><br/> 4. Fox A, Helmer D, Tseng CL, Patrick-DeLuca L, Osinubi O. Report of autonomic symptoms in a clinical sample of veterans with Gulf War Illness. <i>Mil Med</i>. 2018;183(3-4):e179-e185. doi:10.1093/milmed/usx052<br/><br/> 5. Fox A, Helmer D, Tseng CL, McCarron K, Satcher S, Osinubi O. Autonomic symptoms in Gulf War veterans evaluated at the War Related Illness and Injury Study Center. <i>Mil Med</i>. 2019;184(3-4):e191-e196. doi:10.1093/milmed/usy227<br/><br/> 6. Reyes L, Falvo M, Blatt M, Ghobreal B, Acosta A, Serrador J. Autonomic dysfunction in veterans with Gulf War illness [abstract]. <i>FASEB J</i>. 2014;28(S1):1068.19. doi:10.1096/fasebj.28.1_supplement.1068.19<br/><br/> 7. Haley RW, Charuvastra E, Shell WE, et al. Cholinergic autonomic dysfunction in veterans with Gulf War illness: confirmation in a population-based sample. <i>JAMA Neurol</i>. 2013;70(2):191-200. doi:10.1001/jamaneurol.2013.596<br/><br/> 8. Haley RW, Vongpatanasin W, Wolfe GI, et al. Blunted circadian variation in autonomic regulation of sinus node function in veterans with Gulf War syndrome. <i>Am J Med</i>. 2004;117(7):469-478. doi:10.1016/j.amjmed.2004.03.041<br/><br/> 9. Avery TJ, Mathersul DC, Schulz-Heik RJ, Mahoney L, Bayley PJ. Self-reported autonomic dysregulation in Gulf War Illness. <i>Mil Med</i>. Published online December 30, 2021. doi:10.1093/milmed/usab546<br/><br/>10. Verne ZT, Fields JZ, Zhang BB, Zhou Q. Autonomic dysfunction and gastroparesis in Gulf War veterans. <i>J Investig Med</i>. 2023;71(1):7-10. doi:10.1136/jim-2021-002291<br/><br/>11. Levine TD. Small fiber neuropathy: disease classification beyond pain and burning. <i>J Cent Nerv Syst Dis</i>. 2018;10:1179573518771703. doi:10.1177/1179573518771703<br/><br/>12. Novak P. Autonomic disorders. <i>Am J Med</i>. 2019;132(4):420-436. doi:10.1016/j.amjmed.2018.09.027<br/><br/>13. Oaklander AL, Klein MM. Undiagnosed small-fiber polyneuropathy: is it a component of Gulf War Illness? Defense Technical Information Center. Accessed February 21, 2024. https://apps.dtic.mil/sti/citations/ADA613891</p> <p class="reference">14. Sène D. Small fiber neuropathy: diagnosis, causes, and treatment. <i>Joint Bone Spine</i>. 2018;85(5):553-559. doi:10.1016/j.jbspin.2017.11.002<br/><br/>15. Novak V, Freimer ML, Kissel JT, et al. Autonomic impairment in painful neuropathy. <i>Neurology</i>. 2001;56(7):861-868. doi:10.1212/wnl.56.7.861<br/><br/>16. Myers MI, Peltier AC. Uses of skin biopsy for sensory and autonomic nerve assessment. <i>Curr Neurol Neurosci Rep</i>. 2013;13(1):323. doi:10.1007/s11910-012-0323-2<br/><br/>17. Haroutounian S, Todorovic MS, Leinders M, et al. Diagnostic criteria for idiopathic small fiber neuropathy: a systematic review. <i>Muscle Nerve</i>. 2021;63(2):170-177. doi:10.1002/mus.27070<br/><br/>18. Levine TD, Saperstein DS. Routine use of punch biopsy to diagnose small fiber neuropathy in fibromyalgia patients. <i>Clin Rheumatol</i>. 2015;34(3):413-417. doi:10.1007/s10067-014-2850-5<br/><br/>19. England JD, Gronseth G S, Franklin G, et al. Practice parameter: the evaluation of distal symmetric polyneuropathy: the role of autonomic testing, nerve biopsy, and skin biopsy (an evidence-based review). Report of the American Academy of Neurology, the American Association of Neuromuscular and Electrodiagnostic Medicine, and the American Academy of Physical Medicine and Rehabilitation. <i>PM R</i>. 2009;1(1):14-22. doi:10.1016/j.pmrj.2008.11.011<br/><br/>20. de Greef BTA, Hoeijmakers JGJ, Gorissen-Brouwers CML, Geerts M, Faber CG, Merkies ISJ. Associated conditions in small fiber neuropathy - a large cohort study and review of the literature. <i>Eur J Neurol</i>. 2018;25(2):348-355. doi:10.1111/ene.13508<br/><br/>21. Morkavuk G, Leventoglu A. Small fiber neuropathy associated with hyperlipidemia: utility of cutaneous silent periods and autonomic tests. <i>ISRN Neurol</i>. 2014;2014:579242. doi:10.1155/2014/579242<br/><br/>22. Bednarik J, Vlckova-Moravcova E, Bursova S, Belobradkova J, Dusek L, Sommer C. Etiology of small-fiber neuropathy. <i>J Peripher Nerv Syst</i>. 2009;14(3):177-183. doi:10.1111/j.1529-8027.2009.00229.x<br/><br/>23. Kokotis P, Papantoniou M, Schmelz M, Buntziouka C, Tzavellas E, Paparrigopoulos T. Pure small fiber neuropathy in alcohol dependency detected by skin biopsy. <i>Alcohol Fayettev N</i>. 2023;111:67-73. doi:10.1016/j.alcohol.2023.05.006<br/><br/>24. Guimarães-Costa R, Schoindre Y, Metlaine A, et al. N-hexane exposure: a cause of small fiber neuropathy. <i>J Peripher Nerv Syst</i>. 2018;23(2):143-146. doi:10.1111/jns.12261<br/><br/>25. Koszewicz M, Markowska K, Waliszewska-Prosol M, et al. The impact of chronic co-exposure to different heavy metals on small fibers of peripheral nerves. A study of metal industry workers. <i>J Occup Med Toxicol</i>. 2021;16(1):12. doi:10.1186/s12995-021-00302-6<br/><br/>26. Johns Hopkins Medicine. Small nerve fibers defy neuropathy conventions. April 11, 2016. Accessed February 21, 2024. https://www.hopkinsmedicine.org/news/media/releases/small_nerve_fibers_defy_neuropathy_conventions27. Jett DA. Neurotoxic pesticides and neurologic effects. <i>Neurol Clin</i>. 2011;29(3):667-677. doi:10.1016/j.ncl.2011.06.002<br/><br/>28. Berger AR, Schaumburg HH. Human toxic neuropathy caused by industrial agents. In: Dyck PJ, Thomas PK, eds. <i>Peripheral Neuropathy</i>. 4th ed. Saunders; 2005:2505-2525. doi:10.1016/B978-0-7216-9491-7.50115-0<br/><br/>29. Herskovitz S, Schaumburg HH. Neuropathy caused by drugs. In: Dyck PJ, Thomas PK, eds. <i>Peripheral Neuropathy</i>. 4th ed. Saunders; 2005:2553-2583.<br/><br/>30. Katona I, Weis J. Chapter 31 - Diseases of the peripheral nerves. <i>Handb Clin Neurol</i>. 2017;145:453-474. doi:10.1016/B978-0-12-802395-2.00031-6<br/><br/>31. Matikainen E, Juntunen J. Autonomic nervous system dysfunction in workers exposed to organic solvents. <i>J Neurol Neurosurg Psychiatry</i>. 1985;48(10):1021-1024. doi:10.1136/jnnp.48.10.1021<br/><br/>32. Murata K, Araki S, Yokoyama K, Maeda K. Autonomic and peripheral nervous system dysfunction in workers exposed to mixed organic solvents. <i>Int Arch Occup Environ Health</i>. 1991;63(5):335-340. doi:10.1007/BF00381584<br/><br/>33. Sletten DM, Suarez GA, Low PA, Mandrekar J, Singer W. COMPASS 31: a refined and abbreviated Composite Autonomic Symptom Score. <i>Mayo Clin Proc</i>. 2012;87(12):1196-1201. doi:10.1016/j.mayocp.2012.10.013<br/><br/>34. Treister R, O’Neil K, Downs HM, Oaklander AL. Validation of the Composite Autonomic Symptom Scale-31 (COMPASS-31) in patients with and without small-fiber polyneuropathy. <i>Eur J Neurol</i>. 2015;22(7):1124-1130. doi:10.1111/ene.12717<br/><br/>35. Joseph P, Arevalo C, Oliveira RKF, et al. Insights from invasive cardiopulmonary exercise testing of patients with myalgic encephalomyelitis/chronic fatigue syndrome. <i>Chest</i>. 2021;160(2):642-651. doi:10.1016/j.chest.2021.01.082<br/><br/>36. Giannoccaro MP, Donadio V, Incensi A, Avoni P, Liguori R. Small nerve fiber involvement in patients referred for fibromyalgia. <i>Muscle Nerve</i>. 2014;49(5):757-759. doi:10.1002/mus.24156<br/><br/>37. Oaklander AL, Herzog ZD, Downs HM, Klein MM. Objective evidence that small-fiber polyneuropathy underlies some illnesses currently labeled as fibromyalgia. <i>Pain</i>. 2013;154(11):2310-2316. doi:10.1016/j.pain.2013.06.001<br/><br/>38. Serrador JM. Diagnosis of late-stage, early-onset, small-fiber polyneuropathy. Defense Technical Information Center. December 1, 2019. Accessed February 21, 2024. https://apps.dtic.mil/sti/citations/AD109483139. Lodahl M, Treister R, Oaklander AL. Specific symptoms may discriminate between fibromyalgia patients with vs without objective test evidence of small-fiber polyneuropathy. <i>Pain Rep</i>. 2018;3(1):e633. doi:10.1097/PR9.000000000000063340. Sastre A, Cook MR. Autonomic dysfunction in Gulf War veterans. Defense Technical Information Center. April 1, 2004. Accessed February 21, 2024. https://apps.dtic.mil/sti/citations/ADA42952541. Little AA, Albers JW. Clinical description of toxic neuropathies. <i>Handb Clin Neurol</i>. 2015;131:253-296. doi:10.1016/B978-0-444-62627-1.00015-9<br/><br/>42. Faber CG, Hoeijmakers JGJ, Ahn HS, et al. Gain of function NaV1.7 mutations in idiopathic small fiber neuropathy. <i>Ann Neurol</i>. 2012;71(1):26-39.</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
Disable zoom
Off

Impact of the COVID-19 Pandemic on Care for Patients With Skin Cancer

Article Type
Changed
Tue, 05/07/2024 - 14:07
Display Headline
Impact of the COVID-19 Pandemic on Care for Patients With Skin Cancer

To the Editor:

The most common malignancy in the United States is skin cancer, with melanoma accounting for the majority of skin cancer deaths.1 Despite the lack of established guidelines for routine total-body skin examinations, many patients regularly visit their dermatologist for assessment of pigmented skin lesions.2 During the COVID-19 pandemic, many patients were unable to attend in-person dermatology visits, which resulted in many high-risk individuals not receiving care or alternatively seeking virtual care for cutaneous lesions.3 There has been a lack of research in the United States exploring the utilization of teledermatology during the pandemic and its overall impact on the care of patients with a history of skin cancer. We explored the impact of the COVID-19 pandemic on care for patients with skin cancer in a large US population.

CT113005208_eTable1_part1.jpg

CT113005208_eTable1_part2.jpg

Using anonymous survey data from the 2020-2021 National Health Interview Survey,4 we conducted a ­population-based, cross-sectional study to evaluate access to care during the COVID-19 pandemic for patients with a self-reported history of skin cancer—melanoma, nonmelanoma skin cancer, or unknown skin cancer. The 3 outcome variables included having a virtual medical appointment in the past 12 months (yes/no), delaying medical care due to the COVID-19 pandemic (yes/no), and not receiving care due to the COVID-19 pandemic (yes/no). Multivariable logistic regression models evaluating the relationship between a history of skin cancer and access to care were constructed using Stata/MP 17.0 (StataCorp LLC). We controlled for patient age; education; race/ethnicity; received public assistance or welfare payments; sex; region; US citizenship status; health insurance status; comorbidities including history of hypertension, diabetes, and hypercholesterolemia; and birthplace in the United States in the logistic regression models.

CT113005208_eTable2.jpg

Our analysis included 46,679 patients aged 18 years or older, of whom 3.4% (weighted)(n=2204) reported a history of skin cancer (eTable 1). The weighted percentage was calculated using National Health Interview Survey design parameters (accounting for the multistage sampling design) to represent the general US population. Compared with those with no history of skin cancer, patients with a history of skin cancer were significantly more likely to delay medical care (adjusted odds ratio [AOR], 1.37; 95% CI, 1.21-1.54; P<.001) or not receive care (AOR, 1.35; 95% CI, 1.16-1.57; P<.001) due to the pandemic and were more likely to have had a virtual medical visit in the past 12 months (AOR, 1.12; 95% CI, 1.00-1.26; P=.05). Additionally, subgroup analysis revealed that females were more likely than males to forego medical care (eTable 2). β Coefficients for independent and dependent variables were further analyzed using logistic regression (eTable 3).

CT113005208_eTable3.jpg

After adjusting for various potential confounders including comorbidities, our results revealed that patients with a history of skin cancer reported that they were less likely to receive in-person medical care due to the COVID-19 pandemic, as high-risk individuals with a history of skin cancer may have stopped receiving total-body skin examinations and dermatology care during the pandemic. Our findings showed that patients with a history of skin cancer were more likely than those without skin cancer to delay or forego care due to the pandemic, which may contribute to a higher incidence of advanced-stage melanomas postpandemic. Trepanowski et al5 reported an increased incidence of patients presenting with more advanced melanomas during the pandemic. Telemedicine was more commonly utilized by patients with a history of skin cancer during the pandemic.

In the future, virtual care may help limit advanced stages of skin cancer by serving as a viable alternative to in-person care.6 It has been reported that telemedicine can serve as a useful triage service reducing patient wait times.7 Teledermatology should not replace in-person care, as there is no evidence of the diagnostic accuracy of this service and many patients still will need to be seen in-person for confirmation of their diagnosis and potential biopsy. Further studies are needed to assess for missed skin cancer diagnoses due to the utilization of telemedicine.

Limitations of this study included a self-reported history of skin cancer, β coefficients that may suggest a high degree of collinearity, and lack of specific survey questions regarding dermatologic care during the COVID-19 pandemic. Further long-term studies exploring the clinical applicability and diagnostic accuracy of virtual medicine visits for cutaneous malignancies are vital, as teledermatology may play an essential role in curbing rising skin cancer rates even beyond the pandemic.

References
  1. Guy GP Jr, Thomas CC, Thompson T, et al. Vital signs: melanoma incidence and mortality trends and projections—United States, 1982-2030. MMWR Morb Mortal Wkly Rep. 2015;64:591-596.
  2. Whiteman DC, Olsen CM, MacGregor S, et al; QSkin Study. The effect of screening on melanoma incidence and biopsy rates. Br J Dermatol. 2022;187:515-522. doi:10.1111/bjd.21649
  3. Jobbágy A, Kiss N, Meznerics FA, et al. Emergency use and efficacy of an asynchronous teledermatology system as a novel tool for early diagnosis of skin cancer during the first wave of COVID-19 pandemic. Int J Environ Res Public Health. 2022;19:2699. doi:10.3390/ijerph19052699
  4. National Center for Health Statistics. NHIS Data, Questionnaires and Related Documentation. Centers for Disease Control and Prevention website. Accessed April 19, 2023. https://www.cdc.gov/nchs/nhis/data-questionnaires-documentation.htm
  5. Trepanowski N, Chang MS, Zhou G, et al. Delays in melanoma presentation during the COVID-19 pandemic: a nationwide multi-institutional cohort study. J Am Acad Dermatol. 2022;87:1217-1219. doi:10.1016/j.jaad.2022.06.031
  6. Chiru MR, Hindocha S, Burova E, et al. Management of the two-week wait pathway for skin cancer patients, before and during the pandemic: is virtual consultation an option? J Pers Med. 2022;12:1258. doi:10.3390/jpm12081258
  7. Finnane A, Dallest K, Janda M, et al. Teledermatology for the diagnosis and management of skin cancer: a systematic review. JAMA ­Dermatol. 2017;153:319-327. doi:10.1001/jamadermatol.2016.4361
Article PDF
Author and Disclosure Information

Brandon Smith is from the Drexel University College of Medicine, Philadelphia, Pennsylvania. Priya Engel is from the California University of Science and Medicine, Colton. Sogol Stephanie Javadi is from the David Geffen School of Medicine at UCLA, Los Angeles, California. Dr. Egeberg is from the Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark, and the Department of Clinical Medicine, University of Copenhagen. Dr. Wu is from the University of Miami Leonard M. Miller School of Medicine, Florida.

Brandon Smith, Priya Engel, and Sogol Stephanie Javadi report no conflict of interest. Dr. Egeberg has received research funding from AbbVie, Boehringer Ingelheim, Bristol-Myers Squibb, the Danish National Psoriasis Foundation, Eli Lilly and Company, Janssen Pharmaceuticals, the Kgl Hofbundtmager Aage Bang Foundation, Novartis, Pfizer, and the Simon Spies Foundation. He also is a consultant and/or speaker for or is/has been an employee of AbbVie, Almirall, Boehringer Ingelheim, Bristol-Myers Squibb, Dermavant, Eli Lilly and Company, Galápagos NV, Galderma, Horizon Therapeutics, Janssen Pharmaceuticals, LEO Pharma, McNeil Consumer Healthcare, Mylan, Novartis, Pfizer, Samsung Bioepis Co Ltd, Sun Pharmaceuticals, UCB, Union Therapeutics, and Zuellig Pharma Ltd. Dr. Wu is or has been a consultant, investigator, or speaker for AbbVie, Almirall, Amgen, Arcutis, Aristea Therapeutics, Bausch Health, Boehringer Ingelheim, Bristol-Myers Squibb, Codex Labs, Dermavant, DermTech, Dr. Reddy’s Laboratories, Eli Lilly and Company, EPI Health, Galderma, Incyte, Janssen, LEO Pharma, Mindera, Novartis, Pfizer, Regeneron, Samsung Bioepis, Sanofi Genzyme, Solius, Sun Pharmaceuticals, UCB, and Zerigo Health.

The eTables are available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Jashin J. Wu, MD, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, RMSB, Room 2023-A, Miami, FL 33136 (jashinwu@gmail.com). ORCID: 0000-0002-1722-1892. Scopus Author ID: 14629788600

Issue
Cutis - 113(5)
Publications
Topics
Page Number
208-209,E1-E4
Sections
Author and Disclosure Information

Brandon Smith is from the Drexel University College of Medicine, Philadelphia, Pennsylvania. Priya Engel is from the California University of Science and Medicine, Colton. Sogol Stephanie Javadi is from the David Geffen School of Medicine at UCLA, Los Angeles, California. Dr. Egeberg is from the Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark, and the Department of Clinical Medicine, University of Copenhagen. Dr. Wu is from the University of Miami Leonard M. Miller School of Medicine, Florida.

Brandon Smith, Priya Engel, and Sogol Stephanie Javadi report no conflict of interest. Dr. Egeberg has received research funding from AbbVie, Boehringer Ingelheim, Bristol-Myers Squibb, the Danish National Psoriasis Foundation, Eli Lilly and Company, Janssen Pharmaceuticals, the Kgl Hofbundtmager Aage Bang Foundation, Novartis, Pfizer, and the Simon Spies Foundation. He also is a consultant and/or speaker for or is/has been an employee of AbbVie, Almirall, Boehringer Ingelheim, Bristol-Myers Squibb, Dermavant, Eli Lilly and Company, Galápagos NV, Galderma, Horizon Therapeutics, Janssen Pharmaceuticals, LEO Pharma, McNeil Consumer Healthcare, Mylan, Novartis, Pfizer, Samsung Bioepis Co Ltd, Sun Pharmaceuticals, UCB, Union Therapeutics, and Zuellig Pharma Ltd. Dr. Wu is or has been a consultant, investigator, or speaker for AbbVie, Almirall, Amgen, Arcutis, Aristea Therapeutics, Bausch Health, Boehringer Ingelheim, Bristol-Myers Squibb, Codex Labs, Dermavant, DermTech, Dr. Reddy’s Laboratories, Eli Lilly and Company, EPI Health, Galderma, Incyte, Janssen, LEO Pharma, Mindera, Novartis, Pfizer, Regeneron, Samsung Bioepis, Sanofi Genzyme, Solius, Sun Pharmaceuticals, UCB, and Zerigo Health.

The eTables are available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Jashin J. Wu, MD, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, RMSB, Room 2023-A, Miami, FL 33136 (jashinwu@gmail.com). ORCID: 0000-0002-1722-1892. Scopus Author ID: 14629788600

Author and Disclosure Information

Brandon Smith is from the Drexel University College of Medicine, Philadelphia, Pennsylvania. Priya Engel is from the California University of Science and Medicine, Colton. Sogol Stephanie Javadi is from the David Geffen School of Medicine at UCLA, Los Angeles, California. Dr. Egeberg is from the Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark, and the Department of Clinical Medicine, University of Copenhagen. Dr. Wu is from the University of Miami Leonard M. Miller School of Medicine, Florida.

Brandon Smith, Priya Engel, and Sogol Stephanie Javadi report no conflict of interest. Dr. Egeberg has received research funding from AbbVie, Boehringer Ingelheim, Bristol-Myers Squibb, the Danish National Psoriasis Foundation, Eli Lilly and Company, Janssen Pharmaceuticals, the Kgl Hofbundtmager Aage Bang Foundation, Novartis, Pfizer, and the Simon Spies Foundation. He also is a consultant and/or speaker for or is/has been an employee of AbbVie, Almirall, Boehringer Ingelheim, Bristol-Myers Squibb, Dermavant, Eli Lilly and Company, Galápagos NV, Galderma, Horizon Therapeutics, Janssen Pharmaceuticals, LEO Pharma, McNeil Consumer Healthcare, Mylan, Novartis, Pfizer, Samsung Bioepis Co Ltd, Sun Pharmaceuticals, UCB, Union Therapeutics, and Zuellig Pharma Ltd. Dr. Wu is or has been a consultant, investigator, or speaker for AbbVie, Almirall, Amgen, Arcutis, Aristea Therapeutics, Bausch Health, Boehringer Ingelheim, Bristol-Myers Squibb, Codex Labs, Dermavant, DermTech, Dr. Reddy’s Laboratories, Eli Lilly and Company, EPI Health, Galderma, Incyte, Janssen, LEO Pharma, Mindera, Novartis, Pfizer, Regeneron, Samsung Bioepis, Sanofi Genzyme, Solius, Sun Pharmaceuticals, UCB, and Zerigo Health.

The eTables are available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Jashin J. Wu, MD, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, RMSB, Room 2023-A, Miami, FL 33136 (jashinwu@gmail.com). ORCID: 0000-0002-1722-1892. Scopus Author ID: 14629788600

Article PDF
Article PDF

To the Editor:

The most common malignancy in the United States is skin cancer, with melanoma accounting for the majority of skin cancer deaths.1 Despite the lack of established guidelines for routine total-body skin examinations, many patients regularly visit their dermatologist for assessment of pigmented skin lesions.2 During the COVID-19 pandemic, many patients were unable to attend in-person dermatology visits, which resulted in many high-risk individuals not receiving care or alternatively seeking virtual care for cutaneous lesions.3 There has been a lack of research in the United States exploring the utilization of teledermatology during the pandemic and its overall impact on the care of patients with a history of skin cancer. We explored the impact of the COVID-19 pandemic on care for patients with skin cancer in a large US population.

CT113005208_eTable1_part1.jpg

CT113005208_eTable1_part2.jpg

Using anonymous survey data from the 2020-2021 National Health Interview Survey,4 we conducted a ­population-based, cross-sectional study to evaluate access to care during the COVID-19 pandemic for patients with a self-reported history of skin cancer—melanoma, nonmelanoma skin cancer, or unknown skin cancer. The 3 outcome variables included having a virtual medical appointment in the past 12 months (yes/no), delaying medical care due to the COVID-19 pandemic (yes/no), and not receiving care due to the COVID-19 pandemic (yes/no). Multivariable logistic regression models evaluating the relationship between a history of skin cancer and access to care were constructed using Stata/MP 17.0 (StataCorp LLC). We controlled for patient age; education; race/ethnicity; received public assistance or welfare payments; sex; region; US citizenship status; health insurance status; comorbidities including history of hypertension, diabetes, and hypercholesterolemia; and birthplace in the United States in the logistic regression models.

CT113005208_eTable2.jpg

Our analysis included 46,679 patients aged 18 years or older, of whom 3.4% (weighted)(n=2204) reported a history of skin cancer (eTable 1). The weighted percentage was calculated using National Health Interview Survey design parameters (accounting for the multistage sampling design) to represent the general US population. Compared with those with no history of skin cancer, patients with a history of skin cancer were significantly more likely to delay medical care (adjusted odds ratio [AOR], 1.37; 95% CI, 1.21-1.54; P<.001) or not receive care (AOR, 1.35; 95% CI, 1.16-1.57; P<.001) due to the pandemic and were more likely to have had a virtual medical visit in the past 12 months (AOR, 1.12; 95% CI, 1.00-1.26; P=.05). Additionally, subgroup analysis revealed that females were more likely than males to forego medical care (eTable 2). β Coefficients for independent and dependent variables were further analyzed using logistic regression (eTable 3).

CT113005208_eTable3.jpg

After adjusting for various potential confounders including comorbidities, our results revealed that patients with a history of skin cancer reported that they were less likely to receive in-person medical care due to the COVID-19 pandemic, as high-risk individuals with a history of skin cancer may have stopped receiving total-body skin examinations and dermatology care during the pandemic. Our findings showed that patients with a history of skin cancer were more likely than those without skin cancer to delay or forego care due to the pandemic, which may contribute to a higher incidence of advanced-stage melanomas postpandemic. Trepanowski et al5 reported an increased incidence of patients presenting with more advanced melanomas during the pandemic. Telemedicine was more commonly utilized by patients with a history of skin cancer during the pandemic.

In the future, virtual care may help limit advanced stages of skin cancer by serving as a viable alternative to in-person care.6 It has been reported that telemedicine can serve as a useful triage service reducing patient wait times.7 Teledermatology should not replace in-person care, as there is no evidence of the diagnostic accuracy of this service and many patients still will need to be seen in-person for confirmation of their diagnosis and potential biopsy. Further studies are needed to assess for missed skin cancer diagnoses due to the utilization of telemedicine.

Limitations of this study included a self-reported history of skin cancer, β coefficients that may suggest a high degree of collinearity, and lack of specific survey questions regarding dermatologic care during the COVID-19 pandemic. Further long-term studies exploring the clinical applicability and diagnostic accuracy of virtual medicine visits for cutaneous malignancies are vital, as teledermatology may play an essential role in curbing rising skin cancer rates even beyond the pandemic.

To the Editor:

The most common malignancy in the United States is skin cancer, with melanoma accounting for the majority of skin cancer deaths.1 Despite the lack of established guidelines for routine total-body skin examinations, many patients regularly visit their dermatologist for assessment of pigmented skin lesions.2 During the COVID-19 pandemic, many patients were unable to attend in-person dermatology visits, which resulted in many high-risk individuals not receiving care or alternatively seeking virtual care for cutaneous lesions.3 There has been a lack of research in the United States exploring the utilization of teledermatology during the pandemic and its overall impact on the care of patients with a history of skin cancer. We explored the impact of the COVID-19 pandemic on care for patients with skin cancer in a large US population.

CT113005208_eTable1_part1.jpg

CT113005208_eTable1_part2.jpg

Using anonymous survey data from the 2020-2021 National Health Interview Survey,4 we conducted a ­population-based, cross-sectional study to evaluate access to care during the COVID-19 pandemic for patients with a self-reported history of skin cancer—melanoma, nonmelanoma skin cancer, or unknown skin cancer. The 3 outcome variables included having a virtual medical appointment in the past 12 months (yes/no), delaying medical care due to the COVID-19 pandemic (yes/no), and not receiving care due to the COVID-19 pandemic (yes/no). Multivariable logistic regression models evaluating the relationship between a history of skin cancer and access to care were constructed using Stata/MP 17.0 (StataCorp LLC). We controlled for patient age; education; race/ethnicity; received public assistance or welfare payments; sex; region; US citizenship status; health insurance status; comorbidities including history of hypertension, diabetes, and hypercholesterolemia; and birthplace in the United States in the logistic regression models.

CT113005208_eTable2.jpg

Our analysis included 46,679 patients aged 18 years or older, of whom 3.4% (weighted)(n=2204) reported a history of skin cancer (eTable 1). The weighted percentage was calculated using National Health Interview Survey design parameters (accounting for the multistage sampling design) to represent the general US population. Compared with those with no history of skin cancer, patients with a history of skin cancer were significantly more likely to delay medical care (adjusted odds ratio [AOR], 1.37; 95% CI, 1.21-1.54; P<.001) or not receive care (AOR, 1.35; 95% CI, 1.16-1.57; P<.001) due to the pandemic and were more likely to have had a virtual medical visit in the past 12 months (AOR, 1.12; 95% CI, 1.00-1.26; P=.05). Additionally, subgroup analysis revealed that females were more likely than males to forego medical care (eTable 2). β Coefficients for independent and dependent variables were further analyzed using logistic regression (eTable 3).

CT113005208_eTable3.jpg

After adjusting for various potential confounders including comorbidities, our results revealed that patients with a history of skin cancer reported that they were less likely to receive in-person medical care due to the COVID-19 pandemic, as high-risk individuals with a history of skin cancer may have stopped receiving total-body skin examinations and dermatology care during the pandemic. Our findings showed that patients with a history of skin cancer were more likely than those without skin cancer to delay or forego care due to the pandemic, which may contribute to a higher incidence of advanced-stage melanomas postpandemic. Trepanowski et al5 reported an increased incidence of patients presenting with more advanced melanomas during the pandemic. Telemedicine was more commonly utilized by patients with a history of skin cancer during the pandemic.

In the future, virtual care may help limit advanced stages of skin cancer by serving as a viable alternative to in-person care.6 It has been reported that telemedicine can serve as a useful triage service reducing patient wait times.7 Teledermatology should not replace in-person care, as there is no evidence of the diagnostic accuracy of this service and many patients still will need to be seen in-person for confirmation of their diagnosis and potential biopsy. Further studies are needed to assess for missed skin cancer diagnoses due to the utilization of telemedicine.

Limitations of this study included a self-reported history of skin cancer, β coefficients that may suggest a high degree of collinearity, and lack of specific survey questions regarding dermatologic care during the COVID-19 pandemic. Further long-term studies exploring the clinical applicability and diagnostic accuracy of virtual medicine visits for cutaneous malignancies are vital, as teledermatology may play an essential role in curbing rising skin cancer rates even beyond the pandemic.

References
  1. Guy GP Jr, Thomas CC, Thompson T, et al. Vital signs: melanoma incidence and mortality trends and projections—United States, 1982-2030. MMWR Morb Mortal Wkly Rep. 2015;64:591-596.
  2. Whiteman DC, Olsen CM, MacGregor S, et al; QSkin Study. The effect of screening on melanoma incidence and biopsy rates. Br J Dermatol. 2022;187:515-522. doi:10.1111/bjd.21649
  3. Jobbágy A, Kiss N, Meznerics FA, et al. Emergency use and efficacy of an asynchronous teledermatology system as a novel tool for early diagnosis of skin cancer during the first wave of COVID-19 pandemic. Int J Environ Res Public Health. 2022;19:2699. doi:10.3390/ijerph19052699
  4. National Center for Health Statistics. NHIS Data, Questionnaires and Related Documentation. Centers for Disease Control and Prevention website. Accessed April 19, 2023. https://www.cdc.gov/nchs/nhis/data-questionnaires-documentation.htm
  5. Trepanowski N, Chang MS, Zhou G, et al. Delays in melanoma presentation during the COVID-19 pandemic: a nationwide multi-institutional cohort study. J Am Acad Dermatol. 2022;87:1217-1219. doi:10.1016/j.jaad.2022.06.031
  6. Chiru MR, Hindocha S, Burova E, et al. Management of the two-week wait pathway for skin cancer patients, before and during the pandemic: is virtual consultation an option? J Pers Med. 2022;12:1258. doi:10.3390/jpm12081258
  7. Finnane A, Dallest K, Janda M, et al. Teledermatology for the diagnosis and management of skin cancer: a systematic review. JAMA ­Dermatol. 2017;153:319-327. doi:10.1001/jamadermatol.2016.4361
References
  1. Guy GP Jr, Thomas CC, Thompson T, et al. Vital signs: melanoma incidence and mortality trends and projections—United States, 1982-2030. MMWR Morb Mortal Wkly Rep. 2015;64:591-596.
  2. Whiteman DC, Olsen CM, MacGregor S, et al; QSkin Study. The effect of screening on melanoma incidence and biopsy rates. Br J Dermatol. 2022;187:515-522. doi:10.1111/bjd.21649
  3. Jobbágy A, Kiss N, Meznerics FA, et al. Emergency use and efficacy of an asynchronous teledermatology system as a novel tool for early diagnosis of skin cancer during the first wave of COVID-19 pandemic. Int J Environ Res Public Health. 2022;19:2699. doi:10.3390/ijerph19052699
  4. National Center for Health Statistics. NHIS Data, Questionnaires and Related Documentation. Centers for Disease Control and Prevention website. Accessed April 19, 2023. https://www.cdc.gov/nchs/nhis/data-questionnaires-documentation.htm
  5. Trepanowski N, Chang MS, Zhou G, et al. Delays in melanoma presentation during the COVID-19 pandemic: a nationwide multi-institutional cohort study. J Am Acad Dermatol. 2022;87:1217-1219. doi:10.1016/j.jaad.2022.06.031
  6. Chiru MR, Hindocha S, Burova E, et al. Management of the two-week wait pathway for skin cancer patients, before and during the pandemic: is virtual consultation an option? J Pers Med. 2022;12:1258. doi:10.3390/jpm12081258
  7. Finnane A, Dallest K, Janda M, et al. Teledermatology for the diagnosis and management of skin cancer: a systematic review. JAMA ­Dermatol. 2017;153:319-327. doi:10.1001/jamadermatol.2016.4361
Issue
Cutis - 113(5)
Issue
Cutis - 113(5)
Page Number
208-209,E1-E4
Page Number
208-209,E1-E4
Publications
Publications
Topics
Article Type
Display Headline
Impact of the COVID-19 Pandemic on Care for Patients With Skin Cancer
Display Headline
Impact of the COVID-19 Pandemic on Care for Patients With Skin Cancer
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>Smith skin cancer</fileName> <TBEID>0C02F659.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F659</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Smith skin cancer</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240507T082921</firstPublished> <LastPublished>20240507T082921</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240507T082921</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Brandon Smith, BA; Priya Engel, MPH; Sogol Stephanie Javadi, BS</byline> <bylineText>Brandon Smith, BA; Priya Engel, MPH; Sogol Stephanie Javadi, BS; Alexander Egeberg, MD, PhD, DMSc; Jashin J. Wu, MD</bylineText> <bylineFull>Brandon Smith, BA; Priya Engel, MPH; Sogol Stephanie Javadi, BS</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>208-209,E1-E4</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>To the Editor:The most common malignancy in the United States is skin cancer, with melanoma accounting for the majority of skin cancer deaths.1 Despite the lack</metaDescription> <articlePDF>301294</articlePDF> <teaserImage/> <title>Impact of the COVID-19 Pandemic on Care for Patients With Skin Cancer</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>May</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>5</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2165</CMSID> </CMSIDs> <keywords> <keyword>melanoma</keyword> <keyword> nonmelanoma skin cancer</keyword> <keyword> COVID-19</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>May 2024</pubIssueName> <pubArticleType>Audio | 2165</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">104</term> </sections> <topics> <term canonical="true">244</term> <term>245</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002720.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Impact of the COVID-19 Pandemic on Care for Patients With Skin Cancer</title> <deck/> </itemMeta> <itemContent> <p>To the Editor:<br/><br/>The most common malignancy in the United States is skin cancer, with melanoma accounting for the majority of skin cancer deaths.<sup>1</sup> Despite the lack of established guidelines for routine total-body skin examinations, many patients regularly visit their dermatologist for assessment of pigmented skin lesions.<sup>2</sup> During the COVID-19 pandemic, many patients were unable to attend in-person dermatology visits, which resulted in many high-risk individuals not receiving care or alternatively seeking virtual care for cutaneous lesions.<sup>3</sup> There has been a lack of research in the United States exploring the utilization of teledermatology during the pandemic and its overall impact on the care of patients with a history of skin cancer. We explored the impact of the COVID-19 pandemic on care for patients with skin cancer in a large US population. </p> <p>Using anonymous survey data from the 2020-2021 National Health Interview Survey,<sup>4</sup> we conducted a ­population-based, cross-sectional study to evaluate access to care during the COVID-19 pandemic for patients with a self-reported history of skin cancer—melanoma, nonmelanoma skin cancer, or unknown skin cancer. The 3 outcome variables included having a virtual medical appointment in the past 12 months (yes/no), delaying medical care due to the COVID-19 pandemic (yes/no), and not receiving care due to the COVID-19 pandemic (yes/no). Multivariable logistic regression models evaluating the relationship between a history of skin cancer and access to care were constructed using Stata/MP 17.0 (StataCorp LLC). We controlled for patient age; education; race/ethnicity; received public assistance or welfare payments; sex; region; US citizenship status; health insurance status; comorbidities including history of hypertension, diabetes, and hypercholesterolemia; and birthplace in the United States in the logistic regression models. <br/><br/>Our analysis included 46,679 patients aged 18 years or older, of whom 3.4% (weighted)(n=2204) reported a history of skin cancer (eTable 1). The weighted percentage was calculated using National Health Interview Survey design parameters (accounting for the multistage sampling design) to represent the general US population. Compared with those with no history of skin cancer, patients with a history of skin cancer were significantly more likely to delay medical care (adjusted odds ratio [AOR], 1.37; 95% CI, 1.21-1.54; <i>P</i>&lt;.001) or not receive care (AOR, 1.35; 95% CI, 1.16-1.57; <i>P</i>&lt;.001) due to the pandemic and were more likely to have had a virtual medical visit in the past 12 months (AOR, 1.12; 95% CI, 1.00-1.26; <i>P</i>=.05). Additionally, subgroup analysis revealed that females were more likely than males to forego medical care (eTable 2). β Coefficients for independent and dependent variables were further analyzed using logistic regression (eTable 3).<br/><br/>After adjusting for various potential confounders including comorbidities, our results revealed that patients with a history of skin cancer reported that they were less likely to receive in-person medical care due to the COVID-19 pandemic, as high-risk individuals with a history of skin cancer may have stopped receiving total-body skin examinations and dermatology care during the pandemic. Our findings showed that patients with a history of skin cancer were more likely than those without skin cancer to delay or forego care due to the pandemic, which may contribute to a higher incidence of advanced-stage melanomas postpandemic. Trepanowski et al<sup>5</sup> reported an increased incidence of patients presenting with more advanced melanomas during the pandemic. Telemedicine was more commonly utilized by patients with a history of skin cancer during the pandemic. <br/><br/>In the future, virtual care may help limit advanced stages of skin cancer by serving as a viable alternative to in-person care.<sup>6</sup> It has been reported that telemedicine can serve as a useful triage service reducing patient wait times.<sup>7</sup> Teledermatology should not replace in-person care, as there is no evidence of the diagnostic accuracy of this service and many patients still will need to be seen in-person for confirmation of their diagnosis and potential biopsy. Further studies are needed to assess for missed skin cancer diagnoses due to the utilization of telemedicine. <br/><br/>Limitations of this study included a self-reported history of skin cancer, β coefficients that may suggest a high degree of collinearity, and lack of specific survey questions regarding dermatologic care during the COVID-19 pandemic. Further long-term studies exploring the clinical applicability and diagnostic accuracy of virtual medicine visits for cutaneous malignancies are vital, as teledermatology may play an essential role in curbing rising skin cancer rates even beyond the pandemic. </p> <h2>References</h2> <p class="reference"> 1. Guy GP Jr, Thomas CC, Thompson T, et al. Vital signs: melanoma incidence and mortality trends and projections—United States, 1982-2030.<span class="apple-converted-space"> </span><i>MMWR Morb Mortal Wkly Rep</i>. 2015;64:591-596.<br/><br/> 2. Whiteman DC, Olsen CM, MacGregor S, et al; QSkin Study. The effect of screening on melanoma incidence and biopsy rates. <i>Br J Dermatol.</i> 2022;187:515-522. doi:10.1111/bjd.21649<br/><br/><span class="apple-converted-space"> 3. </span>Jobbágy A, Kiss N, Meznerics FA, et al. Emergency use and efficacy of an asynchronous teledermatology system as a novel tool for early diagnosis of skin cancer during the first wave of COVID-19 pandemic.<span class="apple-converted-space"> </span><i>Int J Environ Res Public Health</i>. 2022;19:2699. doi:10.3390/ijerph19052699<br/><br/> 4. National Center for Health Statistics. <i>NHIS Data, Questionnaires and Related Documentation</i>. Centers for Disease Control and Prevention website. Accessed April 19, 2023. https://www.cdc.gov/nchs/nhis/data-questionnaires-documentation.htm<br/><br/> 5. Trepanowski N, Chang MS, Zhou G, et al. Delays in melanoma presentation during the COVID-19 pandemic: a nationwide multi-institutional cohort study.<span class="apple-converted-space"> </span><i>J Am Acad Dermatol</i>. 2022;87:1217-1219. doi:10.1016/j.jaad.2022.06.031<br/><br/> 6. Chiru MR, Hindocha S, Burova E, et al. Management of the two-week wait pathway for skin cancer patients, before and during the pandemic: is virtual consultation an option? <i>J Pers Med</i>. 2022;12:1258. doi:10.3390/jpm12081258<br/><br/> 7. Finnane A<span class="al-author-delim">,</span><span class="apple-converted-space"> </span>Dallest K<span class="al-author-delim">,</span><span class="apple-converted-space"> </span>Janda M<span class="al-author-delim">,</span><span class="apple-converted-space"> </span>et al. Teledermatology for the diagnosis and management of skin cancer<span class="colon-for-citation-subtitle">:</span><span class="apple-converted-space"> </span><span class="Subtitle1">a systematic review</span>.<span class="apple-converted-space"> </span><span class="Emphasis">JAMA ­Dermatol.</span><span class="apple-converted-space"> </span>2017;153:319-327. doi:10.1001/jamadermatol.2016.4361</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Brandon Smith is from the Drexel University College of Medicine, Philadelphia, Pennsylvania. Priya Engel is from the California University of Science and Medicine, Colton. Sogol Stephanie Javadi is from the David Geffen School of Medicine at UCLA, Los Angeles, California. Dr. Egeberg is from the Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark, and the Department of Clinical Medicine, University of Copenhagen. Dr. Wu is from the University of Miami Leonard M. Miller School of Medicine, Florida.</p> <p class="disclosure">Brandon Smith, Priya Engel, and Sogol Stephanie Javadi report no conflict of interest. Dr. Egeberg has received research funding from AbbVie, Boehringer Ingelheim, Bristol-Myers Squibb, the Danish National Psoriasis Foundation, Eli Lilly and Company, Janssen Pharmaceuticals, the Kgl Hofbundtmager Aage Bang Foundation, Novartis, Pfizer, and the Simon Spies Foundation. He also is a consultant and/or speaker for or is/has been an employee of AbbVie, Almirall, Boehringer Ingelheim, Bristol-Myers Squibb, Dermavant, Eli Lilly and Company, Galápagos NV, Galderma, Horizon Therapeutics, Janssen Pharmaceuticals, LEO Pharma, McNeil Consumer Healthcare, Mylan, Novartis, Pfizer, Samsung Bioepis Co Ltd, Sun Pharmaceuticals, UCB, Union Therapeutics, and Zuellig Pharma Ltd. Dr. Wu is or has been a consultant, investigator, or speaker for AbbVie, Almirall, Amgen, Arcutis, Aristea Therapeutics, Bausch Health, Boehringer Ingelheim, Bristol-Myers Squibb, Codex Labs, Dermavant, DermTech, Dr. Reddy’s Laboratories, Eli Lilly and Company, EPI Health, Galderma, Incyte, Janssen, LEO Pharma, Mindera, Novartis, Pfizer, Regeneron, Samsung Bioepis, Sanofi Genzyme, Solius, Sun Pharmaceuticals, UCB, and Zerigo Health.The eTables are available in the Appendix online at www.mdedge.com/dermatology.<br/><br/>Correspondence: Jashin J. Wu, MD, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, RMSB, Room 2023-A, Miami, FL 33136 (jashinwu@gmail.com). ORCID: 0000-0002-1722-1892. Scopus Author ID: 14629788600<br/><br/><i>Cutis</i><i>.</i> 2024 May;113(5):208-209, E1-E4. doi:10.12788/cutis.1011</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>pp</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>The COVID-19 pandemic has altered the landscape of medicine, as many individuals are now utilizing telemedicine to receive care.</li> <li>Many individuals will continue to receive telemedicine moving forward, making it crucial to understand access to care. </li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

PRACTICE POINTS

  • The COVID-19 pandemic has altered the landscape of medicine, as many individuals are now utilizing telemedicine to receive care.
  • Many individuals will continue to receive telemedicine moving forward, making it crucial to understand access to care.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Disable zoom
Off

Recurrence Rates of Mohs Micrographic Surgery vs Radiation Therapy for Basal Cell Carcinoma of the Ear

Article Type
Changed
Tue, 05/07/2024 - 13:50
Display Headline
Recurrence Rates of Mohs Micrographic Surgery vs Radiation Therapy for Basal Cell Carcinoma of the Ear

Basal cell carcinoma (BCC) of the ear may have aggressive histologic subtypes and a greater propensity for subclinical spread than BCC in other anatomic locations. In this retrospective analysis, we evaluated recurrence rates of BCC of the ear in 102 patients who underwent treatment with Mohs micrographic surgery (MMS) or radiation therapy (RT) at a single institution between January 2017 and December 2019. Data on patient demographics, tumor characteristics, treatment modality, and recurrence rates were collected from medical records. Recurrence rates were assessed over a mean follow-up time of 2.8 years. Although MMS is the gold standard for treatment of BCC of the ear, RT may be a suitable alternative for nonsurgical candidates.

Basal cell carcinoma (BCC) of the ear may have aggressive histologic subtypes and a greater propensity for subclinical spread than BCC in other anatomic locations. Given that these aggressive histologic subtypes—defined as morpheaform, basosquamous, sclerosing, infiltrative, or micronodular in any portion of the tumor—have been reported as independent predictors of recurrence,1,2 BCC of the ear may be more likely to recur.

Mohs micrographic surgery (MMS) is the gold standard for the treatment of BCC of the ear. For nonsurgical candidates—those with high bleeding risk, low life expectancy, or other medical or social factors—definitive radiation therapy (RT) may be an option. Our study sought to examine recurrence rates in patients with BCC of the ear treated with MMS vs RT.

Methods

A retrospective review of patients undergoing treatment of BCC of the ear at Bighorn Mohs Surgery and Dermatology Center (San Diego, California) between January 2017 and December 2019 was conducted. A total of 507 medical records were reviewed, and 102 patients were included in the study. Inclusion criteria consisted of biopsy-confirmed BCC of the ear that was treated with MMS, RT, or both. Data on patient demographics, tumor characteristics, treatment modality, and recurrence rates were collected from medical records. This retrospective review of medical records was exempt from institutional review board approval, as it did not involve direct human research subjects, solely entailing a retrospective examination of existing data.

Results

Of the 102 patients included, 82 were male and 20 were female, with an average age of 71 years. All patients were White with the exception of 1 patient whose race was unknown. Two patients were immunocompromised. The helix was identified as the most frequently involved site on the ear (Table). Most of the tumors (56/102) exhibited aggressive histologic subtypes; 36 tumors had nonaggressive histology, and 10 had no subtype listed. Two of the BCCs demonstrated perineural invasion on biopsy. Mohs micrographic surgery was used to treat 96 BCCs, definitive RT was used to treat 5 BCCs (all of which occurred in nonsurgical candidates), and MMS and adjuvant RT were used in 1 patient given multifocal perineural involvement. All 5 patients treated with definitive RT received electron beam radiation therapy; the total dose ranged from 5100 to 6000 cGy divided into 17 to 24 fractions. The final MMS defects ranged from 6 to 55 mm in size. The average follow-up time was 2.8 years. One of the BCCs on the helix that was treated with MMS recurred after 1.3 years. The overall recurrence rate was 0.98%. None of the patients treated with definitive RT experienced recurrence after the mean follow-up time of 2.8 years.

CT113005216_Table.jpg

Comment

Basal cell carcinoma is the most commonly diagnosed cancer in the United States, with approximately 2 million new cases each year.1 Treatment modalities for localized BCC include MMS, surgical excision, electrodesiccation and curettage, topical and intralesional medications, laser therapy, and RT. For high-risk BCCs, MMS is associated with the lowest recurrence rates4 and remains the gold standard for treatment. For patients with contraindications to surgery, definitive RT is an alternative treatment for high-risk BCC.1

Definitive RT can be employed for patients who are poor surgical candidates or when surgery would result in substantial morbidity, impaired function, and/or poor cosmesis.3 Radiation therapy for skin cancers of the ear commonly is administered using high-energy electrons that produce double-strand breaks in the DNA of malignant cells, leading to cell death.4 Disadvantages of RT compared to MMS include a longer treatment course (3 to 6 weeks), possible minimal long-term cosmetic sequelae (eg, color or texture mismatch), lack of pathologic confirmation of margin control, and small risk for secondary malignancy in the treatment field over 2 to 3 decades. For patients with incurable or metastatic disease, palliative RT can provide local control and/or symptomatic relief to improve quality of life.4 Adjuvant RT may be indicated if there is substantial perineural involvement or positive margins after MMS when margins are unable to be achieved or in patients who may not tolerate prolonged or extensive surgical procedures.3

 

 

Basal cell carcinoma of the ear is considered a high-risk anatomic location independent of other prognostic factors. Basal cell carcinomas of the ear have a higher propensity for more aggressive histologic subtypes and subclinical spread.5 Our study demonstrated a higher proportion of aggressive histologic subtypes (56/102 [54.9%]) compared with nonaggressive subtypes (36/102 [35.3%]). There was 1 recurrence of a nodular, sclerosing, and infiltrative BCC on the helix treated with MMS after 1.3 years.

Limitations of our study include that it was conducted at a single institution with a homogenous study population and with relatively short follow-up.

Conclusion

Our study further validates the well-known utility of MMS for the treatment of BCC of the ears. Definitive RT is a suitable alternative for patients who are not surgical candidates. Adjuvant RT may be considered for substantial perineural involvement or positive margins after MMS.3

References
  1. Lee CT, Lehrer EJ, Aphale A, et al. Surgical excision, Mohs micrographic surgery, external-beam radiotherapy, or brachytherapy for indolent skin cancer: an international meta-analysis of 58 studies with 21,000 patients. Cancer. 2019;125:3582-3594.
  2. Cameron MC, Lee E, Hibler BP, et al. Basal cell carcinoma: contemporary approaches to diagnosis, treatment, and prevention. J Am Acad Dermatol. 2019;80:321-339.
  3. Wilmas KM, Garner WB, Ballo MT, et al. The role of radiation therapy in the management of cutaneous malignancies. part II: when is radiation therapy indicated? J Am Acad Dermatol. 2021;85:551-562.
  4. Wilmas KM, Garner WB, Ballo MT, et al. The role of radiation therapy in the management of cutaneous malignancies. part I: diagnostic modalities and applications. J Am Acad Dermatol. 2021;85:539-548.
  5. Bichakjian CK, Olencki T, Aasi SZ, et al. Basal cell skin cancer, version 1.2016, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2016;14:574-597.
Article PDF
Author and Disclosure Information

Drs. Clements, Tripuraneni, Kelley, and Greenway are from Bighorn Mohs Surgery and Dermatology Center, Scripps Clinic, San Diego, California. Dr. Jeha is from the Department of Dermatology, Louisiana State University Health Sciences Center, New Orleans.

The authors report no conflict of interest.

Correspondence: George M. Jeha, MD, Department of Dermatology, Louisiana State University Health Sciences Center, 2021 Perdido St, Ste 7153, New Orleans, LA 70112 (gmjeha@gmail.com).

Issue
Cutis - 113(5)
Publications
Topics
Page Number
216-217
Sections
Author and Disclosure Information

Drs. Clements, Tripuraneni, Kelley, and Greenway are from Bighorn Mohs Surgery and Dermatology Center, Scripps Clinic, San Diego, California. Dr. Jeha is from the Department of Dermatology, Louisiana State University Health Sciences Center, New Orleans.

The authors report no conflict of interest.

Correspondence: George M. Jeha, MD, Department of Dermatology, Louisiana State University Health Sciences Center, 2021 Perdido St, Ste 7153, New Orleans, LA 70112 (gmjeha@gmail.com).

Author and Disclosure Information

Drs. Clements, Tripuraneni, Kelley, and Greenway are from Bighorn Mohs Surgery and Dermatology Center, Scripps Clinic, San Diego, California. Dr. Jeha is from the Department of Dermatology, Louisiana State University Health Sciences Center, New Orleans.

The authors report no conflict of interest.

Correspondence: George M. Jeha, MD, Department of Dermatology, Louisiana State University Health Sciences Center, 2021 Perdido St, Ste 7153, New Orleans, LA 70112 (gmjeha@gmail.com).

Article PDF
Article PDF

Basal cell carcinoma (BCC) of the ear may have aggressive histologic subtypes and a greater propensity for subclinical spread than BCC in other anatomic locations. In this retrospective analysis, we evaluated recurrence rates of BCC of the ear in 102 patients who underwent treatment with Mohs micrographic surgery (MMS) or radiation therapy (RT) at a single institution between January 2017 and December 2019. Data on patient demographics, tumor characteristics, treatment modality, and recurrence rates were collected from medical records. Recurrence rates were assessed over a mean follow-up time of 2.8 years. Although MMS is the gold standard for treatment of BCC of the ear, RT may be a suitable alternative for nonsurgical candidates.

Basal cell carcinoma (BCC) of the ear may have aggressive histologic subtypes and a greater propensity for subclinical spread than BCC in other anatomic locations. Given that these aggressive histologic subtypes—defined as morpheaform, basosquamous, sclerosing, infiltrative, or micronodular in any portion of the tumor—have been reported as independent predictors of recurrence,1,2 BCC of the ear may be more likely to recur.

Mohs micrographic surgery (MMS) is the gold standard for the treatment of BCC of the ear. For nonsurgical candidates—those with high bleeding risk, low life expectancy, or other medical or social factors—definitive radiation therapy (RT) may be an option. Our study sought to examine recurrence rates in patients with BCC of the ear treated with MMS vs RT.

Methods

A retrospective review of patients undergoing treatment of BCC of the ear at Bighorn Mohs Surgery and Dermatology Center (San Diego, California) between January 2017 and December 2019 was conducted. A total of 507 medical records were reviewed, and 102 patients were included in the study. Inclusion criteria consisted of biopsy-confirmed BCC of the ear that was treated with MMS, RT, or both. Data on patient demographics, tumor characteristics, treatment modality, and recurrence rates were collected from medical records. This retrospective review of medical records was exempt from institutional review board approval, as it did not involve direct human research subjects, solely entailing a retrospective examination of existing data.

Results

Of the 102 patients included, 82 were male and 20 were female, with an average age of 71 years. All patients were White with the exception of 1 patient whose race was unknown. Two patients were immunocompromised. The helix was identified as the most frequently involved site on the ear (Table). Most of the tumors (56/102) exhibited aggressive histologic subtypes; 36 tumors had nonaggressive histology, and 10 had no subtype listed. Two of the BCCs demonstrated perineural invasion on biopsy. Mohs micrographic surgery was used to treat 96 BCCs, definitive RT was used to treat 5 BCCs (all of which occurred in nonsurgical candidates), and MMS and adjuvant RT were used in 1 patient given multifocal perineural involvement. All 5 patients treated with definitive RT received electron beam radiation therapy; the total dose ranged from 5100 to 6000 cGy divided into 17 to 24 fractions. The final MMS defects ranged from 6 to 55 mm in size. The average follow-up time was 2.8 years. One of the BCCs on the helix that was treated with MMS recurred after 1.3 years. The overall recurrence rate was 0.98%. None of the patients treated with definitive RT experienced recurrence after the mean follow-up time of 2.8 years.

CT113005216_Table.jpg

Comment

Basal cell carcinoma is the most commonly diagnosed cancer in the United States, with approximately 2 million new cases each year.1 Treatment modalities for localized BCC include MMS, surgical excision, electrodesiccation and curettage, topical and intralesional medications, laser therapy, and RT. For high-risk BCCs, MMS is associated with the lowest recurrence rates4 and remains the gold standard for treatment. For patients with contraindications to surgery, definitive RT is an alternative treatment for high-risk BCC.1

Definitive RT can be employed for patients who are poor surgical candidates or when surgery would result in substantial morbidity, impaired function, and/or poor cosmesis.3 Radiation therapy for skin cancers of the ear commonly is administered using high-energy electrons that produce double-strand breaks in the DNA of malignant cells, leading to cell death.4 Disadvantages of RT compared to MMS include a longer treatment course (3 to 6 weeks), possible minimal long-term cosmetic sequelae (eg, color or texture mismatch), lack of pathologic confirmation of margin control, and small risk for secondary malignancy in the treatment field over 2 to 3 decades. For patients with incurable or metastatic disease, palliative RT can provide local control and/or symptomatic relief to improve quality of life.4 Adjuvant RT may be indicated if there is substantial perineural involvement or positive margins after MMS when margins are unable to be achieved or in patients who may not tolerate prolonged or extensive surgical procedures.3

 

 

Basal cell carcinoma of the ear is considered a high-risk anatomic location independent of other prognostic factors. Basal cell carcinomas of the ear have a higher propensity for more aggressive histologic subtypes and subclinical spread.5 Our study demonstrated a higher proportion of aggressive histologic subtypes (56/102 [54.9%]) compared with nonaggressive subtypes (36/102 [35.3%]). There was 1 recurrence of a nodular, sclerosing, and infiltrative BCC on the helix treated with MMS after 1.3 years.

Limitations of our study include that it was conducted at a single institution with a homogenous study population and with relatively short follow-up.

Conclusion

Our study further validates the well-known utility of MMS for the treatment of BCC of the ears. Definitive RT is a suitable alternative for patients who are not surgical candidates. Adjuvant RT may be considered for substantial perineural involvement or positive margins after MMS.3

Basal cell carcinoma (BCC) of the ear may have aggressive histologic subtypes and a greater propensity for subclinical spread than BCC in other anatomic locations. In this retrospective analysis, we evaluated recurrence rates of BCC of the ear in 102 patients who underwent treatment with Mohs micrographic surgery (MMS) or radiation therapy (RT) at a single institution between January 2017 and December 2019. Data on patient demographics, tumor characteristics, treatment modality, and recurrence rates were collected from medical records. Recurrence rates were assessed over a mean follow-up time of 2.8 years. Although MMS is the gold standard for treatment of BCC of the ear, RT may be a suitable alternative for nonsurgical candidates.

Basal cell carcinoma (BCC) of the ear may have aggressive histologic subtypes and a greater propensity for subclinical spread than BCC in other anatomic locations. Given that these aggressive histologic subtypes—defined as morpheaform, basosquamous, sclerosing, infiltrative, or micronodular in any portion of the tumor—have been reported as independent predictors of recurrence,1,2 BCC of the ear may be more likely to recur.

Mohs micrographic surgery (MMS) is the gold standard for the treatment of BCC of the ear. For nonsurgical candidates—those with high bleeding risk, low life expectancy, or other medical or social factors—definitive radiation therapy (RT) may be an option. Our study sought to examine recurrence rates in patients with BCC of the ear treated with MMS vs RT.

Methods

A retrospective review of patients undergoing treatment of BCC of the ear at Bighorn Mohs Surgery and Dermatology Center (San Diego, California) between January 2017 and December 2019 was conducted. A total of 507 medical records were reviewed, and 102 patients were included in the study. Inclusion criteria consisted of biopsy-confirmed BCC of the ear that was treated with MMS, RT, or both. Data on patient demographics, tumor characteristics, treatment modality, and recurrence rates were collected from medical records. This retrospective review of medical records was exempt from institutional review board approval, as it did not involve direct human research subjects, solely entailing a retrospective examination of existing data.

Results

Of the 102 patients included, 82 were male and 20 were female, with an average age of 71 years. All patients were White with the exception of 1 patient whose race was unknown. Two patients were immunocompromised. The helix was identified as the most frequently involved site on the ear (Table). Most of the tumors (56/102) exhibited aggressive histologic subtypes; 36 tumors had nonaggressive histology, and 10 had no subtype listed. Two of the BCCs demonstrated perineural invasion on biopsy. Mohs micrographic surgery was used to treat 96 BCCs, definitive RT was used to treat 5 BCCs (all of which occurred in nonsurgical candidates), and MMS and adjuvant RT were used in 1 patient given multifocal perineural involvement. All 5 patients treated with definitive RT received electron beam radiation therapy; the total dose ranged from 5100 to 6000 cGy divided into 17 to 24 fractions. The final MMS defects ranged from 6 to 55 mm in size. The average follow-up time was 2.8 years. One of the BCCs on the helix that was treated with MMS recurred after 1.3 years. The overall recurrence rate was 0.98%. None of the patients treated with definitive RT experienced recurrence after the mean follow-up time of 2.8 years.

CT113005216_Table.jpg

Comment

Basal cell carcinoma is the most commonly diagnosed cancer in the United States, with approximately 2 million new cases each year.1 Treatment modalities for localized BCC include MMS, surgical excision, electrodesiccation and curettage, topical and intralesional medications, laser therapy, and RT. For high-risk BCCs, MMS is associated with the lowest recurrence rates4 and remains the gold standard for treatment. For patients with contraindications to surgery, definitive RT is an alternative treatment for high-risk BCC.1

Definitive RT can be employed for patients who are poor surgical candidates or when surgery would result in substantial morbidity, impaired function, and/or poor cosmesis.3 Radiation therapy for skin cancers of the ear commonly is administered using high-energy electrons that produce double-strand breaks in the DNA of malignant cells, leading to cell death.4 Disadvantages of RT compared to MMS include a longer treatment course (3 to 6 weeks), possible minimal long-term cosmetic sequelae (eg, color or texture mismatch), lack of pathologic confirmation of margin control, and small risk for secondary malignancy in the treatment field over 2 to 3 decades. For patients with incurable or metastatic disease, palliative RT can provide local control and/or symptomatic relief to improve quality of life.4 Adjuvant RT may be indicated if there is substantial perineural involvement or positive margins after MMS when margins are unable to be achieved or in patients who may not tolerate prolonged or extensive surgical procedures.3

 

 

Basal cell carcinoma of the ear is considered a high-risk anatomic location independent of other prognostic factors. Basal cell carcinomas of the ear have a higher propensity for more aggressive histologic subtypes and subclinical spread.5 Our study demonstrated a higher proportion of aggressive histologic subtypes (56/102 [54.9%]) compared with nonaggressive subtypes (36/102 [35.3%]). There was 1 recurrence of a nodular, sclerosing, and infiltrative BCC on the helix treated with MMS after 1.3 years.

Limitations of our study include that it was conducted at a single institution with a homogenous study population and with relatively short follow-up.

Conclusion

Our study further validates the well-known utility of MMS for the treatment of BCC of the ears. Definitive RT is a suitable alternative for patients who are not surgical candidates. Adjuvant RT may be considered for substantial perineural involvement or positive margins after MMS.3

References
  1. Lee CT, Lehrer EJ, Aphale A, et al. Surgical excision, Mohs micrographic surgery, external-beam radiotherapy, or brachytherapy for indolent skin cancer: an international meta-analysis of 58 studies with 21,000 patients. Cancer. 2019;125:3582-3594.
  2. Cameron MC, Lee E, Hibler BP, et al. Basal cell carcinoma: contemporary approaches to diagnosis, treatment, and prevention. J Am Acad Dermatol. 2019;80:321-339.
  3. Wilmas KM, Garner WB, Ballo MT, et al. The role of radiation therapy in the management of cutaneous malignancies. part II: when is radiation therapy indicated? J Am Acad Dermatol. 2021;85:551-562.
  4. Wilmas KM, Garner WB, Ballo MT, et al. The role of radiation therapy in the management of cutaneous malignancies. part I: diagnostic modalities and applications. J Am Acad Dermatol. 2021;85:539-548.
  5. Bichakjian CK, Olencki T, Aasi SZ, et al. Basal cell skin cancer, version 1.2016, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2016;14:574-597.
References
  1. Lee CT, Lehrer EJ, Aphale A, et al. Surgical excision, Mohs micrographic surgery, external-beam radiotherapy, or brachytherapy for indolent skin cancer: an international meta-analysis of 58 studies with 21,000 patients. Cancer. 2019;125:3582-3594.
  2. Cameron MC, Lee E, Hibler BP, et al. Basal cell carcinoma: contemporary approaches to diagnosis, treatment, and prevention. J Am Acad Dermatol. 2019;80:321-339.
  3. Wilmas KM, Garner WB, Ballo MT, et al. The role of radiation therapy in the management of cutaneous malignancies. part II: when is radiation therapy indicated? J Am Acad Dermatol. 2021;85:551-562.
  4. Wilmas KM, Garner WB, Ballo MT, et al. The role of radiation therapy in the management of cutaneous malignancies. part I: diagnostic modalities and applications. J Am Acad Dermatol. 2021;85:539-548.
  5. Bichakjian CK, Olencki T, Aasi SZ, et al. Basal cell skin cancer, version 1.2016, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2016;14:574-597.
Issue
Cutis - 113(5)
Issue
Cutis - 113(5)
Page Number
216-217
Page Number
216-217
Publications
Publications
Topics
Article Type
Display Headline
Recurrence Rates of Mohs Micrographic Surgery vs Radiation Therapy for Basal Cell Carcinoma of the Ear
Display Headline
Recurrence Rates of Mohs Micrographic Surgery vs Radiation Therapy for Basal Cell Carcinoma of the Ear
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>Clements</fileName> <TBEID>0C02F5EF.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F5EF</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Clements</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240506T133453</firstPublished> <LastPublished>20240506T133453</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240506T133453</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Stephanie Clements, MD; George M. Jeha, MD</byline> <bylineText>Stephanie Clements, MD; George M. Jeha, MD; Prabhakar Tripuraneni, MD; Benjamin Kelley, MD; Hubert Greenway Jr, MD</bylineText> <bylineFull>Stephanie Clements, MD; George M. Jeha, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>216-217</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Basal cell carcinoma (BCC) of the ear may have aggressive histologic subtypes and a greater propensity for subclinical spread than BCC in other anatomic locatio</metaDescription> <articlePDF>301297</articlePDF> <teaserImage/> <title>Recurrence Rates of Mohs Micrographic Surgery vs Radiation Therapy for Basal Cell Carcinoma of the Ear</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>May</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>5</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2161</CMSID> </CMSIDs> <keywords> <keyword>nonmelanoma skin cancer</keyword> <keyword> mohs micrographic surgery</keyword> <keyword> basal cell carcinoma</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>May 2024</pubIssueName> <pubArticleType>Original Articles | 2161</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">104</term> </sections> <topics> <term canonical="true">245</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002723.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Recurrence Rates of Mohs Micrographic Surgery vs Radiation Therapy for Basal Cell Carcinoma of the Ear</title> <deck/> </itemMeta> <itemContent> <p class="abstract">Basal cell carcinoma (BCC) of the ear may have aggressive histologic subtypes and a greater propensity for subclinical spread than BCC in other anatomic locations. In this retrospective analysis, we evaluated recurrence rates of BCC of the ear in 102 patients who underwent treatment with Mohs micrographic surgery (MMS) or radiation therapy (RT) at a single institution between January 2017 and December 2019. Data on patient demographics, tumor characteristics, treatment modality, and recurrence rates were collected from medical records. Recurrence rates were assessed over a mean follow-up time of 2.8 years. Although MMS is the gold standard for treatment of BCC of the ear, RT may be a suitable alternative for nonsurgical candidates.</p> <p>Basal cell carcinoma (BCC) of the ear may have aggressive histologic subtypes and a greater propensity for subclinical spread than BCC in other anatomic locations. Given that these aggressive histologic subtypes—defined as morpheaform, basosquamous, sclerosing, infiltrative, or micronodular in any portion of the tumor—have been reported as independent predictors of recurrence,<sup>1,2</sup> BCC of the ear may be more likely to recur. </p> <p>Mohs micrographic surgery (MMS) is the gold standard for the treatment of BCC of the ear. For nonsurgical candidates—those with high bleeding risk, low life expectancy, or other medical or social factors—definitive radiation therapy (RT) may be an option. Our study sought to examine recurrence rates in patients with BCC of the ear treated with MMS vs RT. </p> <h3>Methods</h3> <p>A retrospective review of patients undergoing treatment of BCC of the ear at Bighorn Mohs Surgery and Dermatology Center (San Diego, California) between January 2017 and December 2019 was conducted. A total of 507 medical records were reviewed, and 102 patients were included in the study. Inclusion criteria consisted of biopsy-confirmed BCC of the ear that was treated with MMS, RT, or both. Data on patient demographics, tumor characteristics, treatment modality, and recurrence rates were collected from medical records. This retrospective review of medical records was exempt from institutional review board approval, as it did not involve direct human research subjects, solely entailing a retrospective examination of existing data.</p> <h3>Results</h3> <p>Of the 102 patients included, 82 were male and 20 were female, with an average age of 71 years. All patients were White with the exception of 1 patient whose race was unknown. Two patients were immunocompromised. The helix was identified as the most frequently involved site on the ear (Table). Most of the tumors (56/102) exhibited aggressive histologic subtypes; 36 tumors had nonaggressive histology, and 10 had no subtype listed. Two of the BCCs demonstrated perineural invasion on biopsy. Mohs micrographic surgery was used to treat 96 BCCs, definitive RT was used to treat 5 BCCs (all of which occurred in nonsurgical candidates), and MMS and adjuvant RT were used in 1 patient given multifocal perineural involvement. All 5 patients treated with definitive RT received electron beam radiation therapy; the total dose ranged from 5100 to 6000 cGy divided into 17 to 24 fractions. The final MMS defects ranged from 6 to 55 mm in size. The average follow-up time was 2.8 years. One of the BCCs on the helix that was treated with MMS recurred after 1.3 years. The overall recurrence rate was 0.98%. None of the patients treated with definitive RT experienced recurrence after the mean follow-up time of 2.8 years.</p> <h3>Comment</h3> <p>Basal cell carcinoma is the most commonly diagnosed cancer in the United States, with approximately 2 million new cases each year.<sup>1</sup> Treatment modalities for localized BCC include MMS, surgical excision, electrodesiccation and curettage, topical and intralesional medications, laser therapy, and RT. For high-risk BCCs, MMS is associated with the lowest recurrence rates<sup>4</sup> and remains the gold standard for treatment. For patients with contraindications to surgery, definitive RT is an alternative treatment for high-risk BCC.<sup>1</sup></p> <p>Definitive RT can be employed for patients who are poor surgical candidates or when surgery would result in substantial morbidity, impaired function, and/or poor cosmesis.<sup>3</sup> Radiation therapy for skin cancers of the ear commonly is administered using high-energy electrons that produce double-strand breaks in the DNA of malignant cells, leading to cell death.<sup>4</sup> Disadvantages of RT compared to MMS include a longer treatment course (3 to 6 weeks), possible minimal long-term cosmetic sequelae (eg, color or texture mismatch), lack of pathologic confirmation of margin control, and small risk for secondary malignancy in the treatment field over 2 to 3 decades. For patients with incurable or metastatic disease, palliative RT can provide local control and/or symptomatic relief to improve quality of life.<sup>4</sup> Adjuvant RT may be indicated if there is substantial perineural involvement or positive margins after MMS when margins are unable to be achieved or in patients who may not tolerate prolonged or extensive surgical procedures.<sup>3<br/><br/></sup>Basal cell carcinoma of the ear is considered a high-risk anatomic location independent of other prognostic factors. Basal cell carcinomas of the ear have a higher propensity for more aggressive histologic subtypes and subclinical spread.<sup>5</sup> Our study demonstrated a higher proportion of aggressive histologic subtypes (56/102 [54.9%]) compared with nonaggressive subtypes (36/102 [35.3%]). There was 1 recurrence of a nodular, sclerosing, and infiltrative BCC on the helix treated with MMS after 1.3 years.<br/><br/>Limitations of our study include that it was conducted at a single institution with a homogenous study population and with relatively short follow-up.</p> <h3>Conclusion</h3> <p>Our study further validates the well-known utility of MMS for the treatment of BCC of the ears. Definitive RT is a suitable alternative for patients who are not surgical candidates. Adjuvant RT may be considered for substantial perineural involvement or positive margins after MMS.<sup>3</sup> </p> <h2>References</h2> <p class="reference"> 1. Lee CT, Lehrer EJ, Aphale A, et al. Surgical excision, Mohs micrographic surgery, external-beam radiotherapy, or brachytherapy for indolent skin cancer: an international meta-analysis of 58 studies with 21,000 patients. <i>Cancer.</i> 2019;125:3582-3594. <br/><br/> 2. Cameron MC, Lee E, Hibler BP, et al. Basal cell carcinoma: contemporary approaches to diagnosis, treatment, and prevention. <i>J Am Acad Dermatol.</i> 2019;80:321-339.<br/><br/> 3. Wilmas KM, Garner WB, Ballo MT, et al. The role of radiation therapy in the management of cutaneous malignancies. part II: when is radiation therapy indicated? <i>J Am Acad Dermatol.</i> 2021;85:551-562.<br/><br/> 4. Wilmas KM, Garner WB, Ballo MT, et al. The role of radiation therapy in the management of cutaneous malignancies. part I: diagnostic modalities and applications. <i>J Am Acad Dermatol.</i> 2021;85:539-548.</p> <p class="reference"> 5. Bichakjian CK, Olencki T, Aasi SZ, et al. Basal cell skin cancer, version 1.2016, NCCN Clinical Practice Guidelines in Oncology. <i>J Natl Compr Canc Netw.</i> 2016;14:574-597.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Drs. Clements, Tripuraneni, Kelley, and Greenway are from Bighorn Mohs Surgery and Dermatology Center, Scripps Clinic, San Diego, California. Dr. Jeha is from the Department of Dermatology, Louisiana State University Health Sciences Center, New Orleans.</p> <p class="disclosure">The authors report no conflict of interest. <br/><br/>Correspondence: George M. Jeha, MD, Department of Dermatology, Louisiana State University Health Sciences Center, 2021 Perdido St, Ste 7153, New Orleans, LA 70112 (gmjeha@gmail.com).<br/><br/><i>Cutis</i>. 2024 May;113(5):216-217. doi:10.12788/cutis.1004</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>pp</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>Basal cell carcinoma (BCC) of the ear may have aggressive histologic subtypes and a greater propensity for subclinical spread than BCC in other anatomic locations, highlighting the importance of careful management and follow-up.</li> <li>Although Mohs micrographic surgery remains the gold standard for treating BCC of the ear, radiation therapy can be considered as a suitable alternative for nonsurgical candidates.</li> </ul> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>table</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="Normal"> Distribution of Anatomic Sites in Patients With Basal Cell Carcinoma of the Ear (N=102) </p> <p class="Normal"> <table> <tbody> <tr> <td> <p class="tablebody"> <strong>Site</strong> </p> </td> <td> <p class="tablebody"> <strong>Patients, n (%)</strong> </p> </td> </tr> <tr> <td> <p class="tablebody">Helix</p> </td> <td> <p class="tablebody">39 (38.2)</p> </td> </tr> <tr> <td> <p class="tablebody">Posterior ear</p> </td> <td> <p class="tablebody">21 (20.6)</p> </td> </tr> <tr> <td> <p class="tablebody">Antihelix</p> </td> <td> <p class="tablebody">9 (8.8)</p> </td> </tr> <tr> <td> <p class="tablebody">Concha</p> </td> <td> <p class="tablebody">8 (7.8)</p> </td> </tr> <tr> <td> <p class="tablebody">Tragus</p> </td> <td> <p class="tablebody">6 (5.9)</p> </td> </tr> <tr> <td> <p class="tablebody">Earlobe</p> </td> <td> <p class="tablebody">3 (2.9)</p> </td> </tr> <tr> <td> <p class="tablebody">Scapha/scaphoid fossa</p> </td> <td> <p class="tablebody">2 (2.0)</p> </td> </tr> <tr> <td> <p class="tablebody">Antitragus</p> </td> <td> <p class="tablebody">1 (1.0)</p> </td> </tr> <tr> <td> <p class="tablebody">Unspecified</p> </td> <td> <p class="tablebody">13 (12.8)</p> </td> </tr> </tbody> </table> </p> </itemContent> </newsItem> </itemSet></root>
Inside the Article

PRACTICE POINTS

  • Basal cell carcinoma (BCC) of the ear may have aggressive histologic subtypes and a greater propensity for subclinical spread than BCC in other anatomic locations, highlighting the importance of careful management and follow-up.
  • Although Mohs micrographic surgery remains the gold standard for treating BCC of the ear, radiation therapy can be considered as a suitable alternative for nonsurgical candidates.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Disable zoom
Off

Risk for COVID-19 Infection in Patients With Vitiligo

Article Type
Changed
Thu, 06/20/2024 - 12:35
Display Headline
Risk for COVID-19 Infection in Patients With Vitiligo

To the Editor:

Vitiligo is a depigmentation disorder that results from the loss of melanocytes in the epidermis.1 The most widely accepted pathophysiology for melanocyte destruction in vitiligo is an autoimmune process involving dysregulated cytokine production and autoreactive T-cell activation.1 Individuals with cutaneous autoinflammatory conditions currently are vital patient populations warranting research, as their susceptibility to COVID-19 infection may differ from the general population. We previously found a small increased risk for COVID-19 infection in patients with psoriasis,2 which suggests that other dermatologic conditions also may impact COVID-19 risk. The risk for COVID-19 infection in patients with vitiligo remains largely unknown. In this retrospective cohort study, we investigated the risk for COVID-19 infection in patients with vitiligo compared with those without vitiligo utilizing claims data from the COVID-19 Research Database (https://covid19researchdatabase.org/).

Claims were evaluated for patients aged 3 years and older with a vitiligo diagnosis (International Classification of Diseases, Tenth Revision [ICD-10] code L80) that was made between January 1, 2016, and January 1, 2020. Individuals without a vitiligo diagnosis during the same period were placed (4:1 ratio) in the control group and were matched with study group patients for age and sex. All comorbidity variables and vitiligo diagnoses were extracted from ICD-10 codes that were given prior to a diagnosis of COVID-19. We then constructed multivariable logistic regression models adjusting for measured confounders to evaluate if vitiligo was associated with higher risk for COVID-19 infection after January 1, 2020.

The vitiligo and nonvitiligo cohorts included 40,363 and 161,452 patients, respectively (Table 1). Logistic regression analysis with adjustment for confounding variables, including high comorbid risk factors (Table 2) revealed that patients with a diagnosis of vitiligo had significantly increased odds of COVID-19 infection compared with patients without vitiligo (adjusted odds ratio [AOR], 1.47; 95% CI, 1.37-1.57; P<.001)(Table 3). Additionally, subgroup logistic analyses for sex, age, and exclusion of patients who were HIV positive revealed that females with vitiligo had higher odds of contracting COVID-19 than males with vitiligo (Table 3).

CT113004030_Table1.jpg

Our results showed that patients with vitiligo had a higher relative risk for contracting COVID-19 than individuals without vitiligo. It has been reported that the prevalence of COVID-19 is higher among patients with autoimmune diseases compared to the general population.3 Additionally, a handful of vitiligo patients are managed with immunosuppressive agents that may further weaken their immune response.1 Moreover, survey results from dermatologists managing vitiligo patients revealed that physicians were fairly comfortable prescribing immunosuppressants and encouraging in-office phototherapy during the COVID-19 pandemic.4 As a result, more patients may have been attending in-office visits for their phototherapy, which may have increased their risk for COVID-19. Although these factors play a role in ­COVID-19 infection rates, the underlying immune dysregulation in vitiligo in relation to COVID-19 remains unknown and should be further explored.

CT113004030_Table2.jpg

Our findings are limited by the use of ICD-10 codes, the inability to control for all potential confounding variables, the lack of data regarding the stage of vitiligo, and the absence of data for undiagnosed COVID-19 infections. In addition, patients with vitiligo may be more likely to seek care, potentially increasing their rates of COVID-19 testing. The inability to identify the stage of vitiligo during enrollment in the database may have altered our results, as individuals with active disease have increased levels of IFN-γ. Increased secretion of IFN-γ also potentially helps in the clearance of COVID-19 infection.1 Future studies should investigate this relationship via planned ­COVID-19 testing, identification of vitiligo stage, and controlling for other associated comorbidities.

CT113004030_Table3.jpg

References
  1. Rashighi M, Harris JE. Vitiligo pathogenesis and emerging treatments. Dermatol Clin. 2017;35:257-265. doi:10.1016/j.det.2016.11.014
  2. Wu JJ, Liu J, Thatiparthi A, et al. The risk of COVID-19 in patients with psoriasis—a retrospective cohort study [published online September 20, 2022]. J Am Acad Dermatol. doi:10.1016/j.jaad.2022.07.040
  3. Zhong J, Shen G, Yang H, et al. COVID-19 in patients with rheumatic disease in Hubei province, China: a multicentre retrospective observational study. Lancet Rheumatol. 2020;2:E557-E564. doi:10.1016/S2665-9913(20)30227-7
  4. Chatterjee M, Das A. Management of vitiligo amidst the ­COVID-19 pandemic: a survey and resulting consensus. Indian J Dermatol. 2021;66:479-483. doi:10.4103/ijd.ijd_859_20
Article PDF
Author and Disclosure Information

Brandon Smith is from the Drexel University College of Medicine, Philadelphia, Pennsylvania. Shahin Shahsavari is from the Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire. Aislyn Oulee is from the University of California Riverside School of Medicine. Priya Engel is from the California University of Science and Medicine, Colton. Dr. Egeberg is from the Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark, and the Department of Clinical Medicine, University of Copenhagen. Dr. Wu is from the University of Miami Leonard M. Miller School of Medicine, Florida.

Brandon Smith, Shahin Shahsavari, Aislyn Oulee, and Priya Engel report no conflict of interest. Dr. Egeberg has received research funding from AbbVie, Boehringer Ingelheim, Bristol-Myers Squibb, the Danish National Psoriasis Foundation, Eli Lilly and Company, Janssen Pharmaceuticals, the Kgl Hofbundtmager Aage Bangs Foundation, Novartis, Pfizer, and the Simon Spies Foundation. He also is a consultant and/or speaker for or is/has been an employee of AbbVie, Almirall, Boehringer Ingelheim, Bristol-Myers Squibb, Dermavant, Eli Lilly and Company, Galápagos NV, Galderma, Horizon Therapeutics, Janssen Pharmaceuticals, LEO Pharma, McNeil Consumer Healthcare, Mylan, Novartis, Pfizer, Samsung Bioepis Co Ltd, Sun Pharmaceuticals, UCB, Union Therapeutics, and Zuellig Pharma Ltd. Dr. Wu is or has been a consultant, investigator, or speaker for AbbVie, Almirall, Amgen, Arcutis, Aristea Therapeutics, Bausch Health, Boehringer Ingelheim, Bristol-Myers Squibb, Codex Labs, Dermavant, DermTech, Dr. Reddy’s Laboratories, Eli Lilly and Company, EPI Health, Galderma, Incyte, Janssen, LEO Pharma, Mindera, Novartis, Pfizer, Regeneron, Samsung Bioepis, Sanofi Genzyme, Solius, Sun Pharmaceuticals, UCB, and Zerigo Health.

Correspondence: Jashin J. Wu, MD, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, RMSB, Room 2023-A, Miami, FL 33136 (jashinwu@gmail.com). ORCID: 0000-0002-1722-1892. Scopus Author ID: 14629788600

Issue
Cutis - 113(4)
Publications
Topics
Page Number
E30-E32
Sections
Author and Disclosure Information

Brandon Smith is from the Drexel University College of Medicine, Philadelphia, Pennsylvania. Shahin Shahsavari is from the Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire. Aislyn Oulee is from the University of California Riverside School of Medicine. Priya Engel is from the California University of Science and Medicine, Colton. Dr. Egeberg is from the Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark, and the Department of Clinical Medicine, University of Copenhagen. Dr. Wu is from the University of Miami Leonard M. Miller School of Medicine, Florida.

Brandon Smith, Shahin Shahsavari, Aislyn Oulee, and Priya Engel report no conflict of interest. Dr. Egeberg has received research funding from AbbVie, Boehringer Ingelheim, Bristol-Myers Squibb, the Danish National Psoriasis Foundation, Eli Lilly and Company, Janssen Pharmaceuticals, the Kgl Hofbundtmager Aage Bangs Foundation, Novartis, Pfizer, and the Simon Spies Foundation. He also is a consultant and/or speaker for or is/has been an employee of AbbVie, Almirall, Boehringer Ingelheim, Bristol-Myers Squibb, Dermavant, Eli Lilly and Company, Galápagos NV, Galderma, Horizon Therapeutics, Janssen Pharmaceuticals, LEO Pharma, McNeil Consumer Healthcare, Mylan, Novartis, Pfizer, Samsung Bioepis Co Ltd, Sun Pharmaceuticals, UCB, Union Therapeutics, and Zuellig Pharma Ltd. Dr. Wu is or has been a consultant, investigator, or speaker for AbbVie, Almirall, Amgen, Arcutis, Aristea Therapeutics, Bausch Health, Boehringer Ingelheim, Bristol-Myers Squibb, Codex Labs, Dermavant, DermTech, Dr. Reddy’s Laboratories, Eli Lilly and Company, EPI Health, Galderma, Incyte, Janssen, LEO Pharma, Mindera, Novartis, Pfizer, Regeneron, Samsung Bioepis, Sanofi Genzyme, Solius, Sun Pharmaceuticals, UCB, and Zerigo Health.

Correspondence: Jashin J. Wu, MD, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, RMSB, Room 2023-A, Miami, FL 33136 (jashinwu@gmail.com). ORCID: 0000-0002-1722-1892. Scopus Author ID: 14629788600

Author and Disclosure Information

Brandon Smith is from the Drexel University College of Medicine, Philadelphia, Pennsylvania. Shahin Shahsavari is from the Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire. Aislyn Oulee is from the University of California Riverside School of Medicine. Priya Engel is from the California University of Science and Medicine, Colton. Dr. Egeberg is from the Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark, and the Department of Clinical Medicine, University of Copenhagen. Dr. Wu is from the University of Miami Leonard M. Miller School of Medicine, Florida.

Brandon Smith, Shahin Shahsavari, Aislyn Oulee, and Priya Engel report no conflict of interest. Dr. Egeberg has received research funding from AbbVie, Boehringer Ingelheim, Bristol-Myers Squibb, the Danish National Psoriasis Foundation, Eli Lilly and Company, Janssen Pharmaceuticals, the Kgl Hofbundtmager Aage Bangs Foundation, Novartis, Pfizer, and the Simon Spies Foundation. He also is a consultant and/or speaker for or is/has been an employee of AbbVie, Almirall, Boehringer Ingelheim, Bristol-Myers Squibb, Dermavant, Eli Lilly and Company, Galápagos NV, Galderma, Horizon Therapeutics, Janssen Pharmaceuticals, LEO Pharma, McNeil Consumer Healthcare, Mylan, Novartis, Pfizer, Samsung Bioepis Co Ltd, Sun Pharmaceuticals, UCB, Union Therapeutics, and Zuellig Pharma Ltd. Dr. Wu is or has been a consultant, investigator, or speaker for AbbVie, Almirall, Amgen, Arcutis, Aristea Therapeutics, Bausch Health, Boehringer Ingelheim, Bristol-Myers Squibb, Codex Labs, Dermavant, DermTech, Dr. Reddy’s Laboratories, Eli Lilly and Company, EPI Health, Galderma, Incyte, Janssen, LEO Pharma, Mindera, Novartis, Pfizer, Regeneron, Samsung Bioepis, Sanofi Genzyme, Solius, Sun Pharmaceuticals, UCB, and Zerigo Health.

Correspondence: Jashin J. Wu, MD, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, RMSB, Room 2023-A, Miami, FL 33136 (jashinwu@gmail.com). ORCID: 0000-0002-1722-1892. Scopus Author ID: 14629788600

Article PDF
Article PDF

To the Editor:

Vitiligo is a depigmentation disorder that results from the loss of melanocytes in the epidermis.1 The most widely accepted pathophysiology for melanocyte destruction in vitiligo is an autoimmune process involving dysregulated cytokine production and autoreactive T-cell activation.1 Individuals with cutaneous autoinflammatory conditions currently are vital patient populations warranting research, as their susceptibility to COVID-19 infection may differ from the general population. We previously found a small increased risk for COVID-19 infection in patients with psoriasis,2 which suggests that other dermatologic conditions also may impact COVID-19 risk. The risk for COVID-19 infection in patients with vitiligo remains largely unknown. In this retrospective cohort study, we investigated the risk for COVID-19 infection in patients with vitiligo compared with those without vitiligo utilizing claims data from the COVID-19 Research Database (https://covid19researchdatabase.org/).

Claims were evaluated for patients aged 3 years and older with a vitiligo diagnosis (International Classification of Diseases, Tenth Revision [ICD-10] code L80) that was made between January 1, 2016, and January 1, 2020. Individuals without a vitiligo diagnosis during the same period were placed (4:1 ratio) in the control group and were matched with study group patients for age and sex. All comorbidity variables and vitiligo diagnoses were extracted from ICD-10 codes that were given prior to a diagnosis of COVID-19. We then constructed multivariable logistic regression models adjusting for measured confounders to evaluate if vitiligo was associated with higher risk for COVID-19 infection after January 1, 2020.

The vitiligo and nonvitiligo cohorts included 40,363 and 161,452 patients, respectively (Table 1). Logistic regression analysis with adjustment for confounding variables, including high comorbid risk factors (Table 2) revealed that patients with a diagnosis of vitiligo had significantly increased odds of COVID-19 infection compared with patients without vitiligo (adjusted odds ratio [AOR], 1.47; 95% CI, 1.37-1.57; P<.001)(Table 3). Additionally, subgroup logistic analyses for sex, age, and exclusion of patients who were HIV positive revealed that females with vitiligo had higher odds of contracting COVID-19 than males with vitiligo (Table 3).

CT113004030_Table1.jpg

Our results showed that patients with vitiligo had a higher relative risk for contracting COVID-19 than individuals without vitiligo. It has been reported that the prevalence of COVID-19 is higher among patients with autoimmune diseases compared to the general population.3 Additionally, a handful of vitiligo patients are managed with immunosuppressive agents that may further weaken their immune response.1 Moreover, survey results from dermatologists managing vitiligo patients revealed that physicians were fairly comfortable prescribing immunosuppressants and encouraging in-office phototherapy during the COVID-19 pandemic.4 As a result, more patients may have been attending in-office visits for their phototherapy, which may have increased their risk for COVID-19. Although these factors play a role in ­COVID-19 infection rates, the underlying immune dysregulation in vitiligo in relation to COVID-19 remains unknown and should be further explored.

CT113004030_Table2.jpg

Our findings are limited by the use of ICD-10 codes, the inability to control for all potential confounding variables, the lack of data regarding the stage of vitiligo, and the absence of data for undiagnosed COVID-19 infections. In addition, patients with vitiligo may be more likely to seek care, potentially increasing their rates of COVID-19 testing. The inability to identify the stage of vitiligo during enrollment in the database may have altered our results, as individuals with active disease have increased levels of IFN-γ. Increased secretion of IFN-γ also potentially helps in the clearance of COVID-19 infection.1 Future studies should investigate this relationship via planned ­COVID-19 testing, identification of vitiligo stage, and controlling for other associated comorbidities.

CT113004030_Table3.jpg

To the Editor:

Vitiligo is a depigmentation disorder that results from the loss of melanocytes in the epidermis.1 The most widely accepted pathophysiology for melanocyte destruction in vitiligo is an autoimmune process involving dysregulated cytokine production and autoreactive T-cell activation.1 Individuals with cutaneous autoinflammatory conditions currently are vital patient populations warranting research, as their susceptibility to COVID-19 infection may differ from the general population. We previously found a small increased risk for COVID-19 infection in patients with psoriasis,2 which suggests that other dermatologic conditions also may impact COVID-19 risk. The risk for COVID-19 infection in patients with vitiligo remains largely unknown. In this retrospective cohort study, we investigated the risk for COVID-19 infection in patients with vitiligo compared with those without vitiligo utilizing claims data from the COVID-19 Research Database (https://covid19researchdatabase.org/).

Claims were evaluated for patients aged 3 years and older with a vitiligo diagnosis (International Classification of Diseases, Tenth Revision [ICD-10] code L80) that was made between January 1, 2016, and January 1, 2020. Individuals without a vitiligo diagnosis during the same period were placed (4:1 ratio) in the control group and were matched with study group patients for age and sex. All comorbidity variables and vitiligo diagnoses were extracted from ICD-10 codes that were given prior to a diagnosis of COVID-19. We then constructed multivariable logistic regression models adjusting for measured confounders to evaluate if vitiligo was associated with higher risk for COVID-19 infection after January 1, 2020.

The vitiligo and nonvitiligo cohorts included 40,363 and 161,452 patients, respectively (Table 1). Logistic regression analysis with adjustment for confounding variables, including high comorbid risk factors (Table 2) revealed that patients with a diagnosis of vitiligo had significantly increased odds of COVID-19 infection compared with patients without vitiligo (adjusted odds ratio [AOR], 1.47; 95% CI, 1.37-1.57; P<.001)(Table 3). Additionally, subgroup logistic analyses for sex, age, and exclusion of patients who were HIV positive revealed that females with vitiligo had higher odds of contracting COVID-19 than males with vitiligo (Table 3).

CT113004030_Table1.jpg

Our results showed that patients with vitiligo had a higher relative risk for contracting COVID-19 than individuals without vitiligo. It has been reported that the prevalence of COVID-19 is higher among patients with autoimmune diseases compared to the general population.3 Additionally, a handful of vitiligo patients are managed with immunosuppressive agents that may further weaken their immune response.1 Moreover, survey results from dermatologists managing vitiligo patients revealed that physicians were fairly comfortable prescribing immunosuppressants and encouraging in-office phototherapy during the COVID-19 pandemic.4 As a result, more patients may have been attending in-office visits for their phototherapy, which may have increased their risk for COVID-19. Although these factors play a role in ­COVID-19 infection rates, the underlying immune dysregulation in vitiligo in relation to COVID-19 remains unknown and should be further explored.

CT113004030_Table2.jpg

Our findings are limited by the use of ICD-10 codes, the inability to control for all potential confounding variables, the lack of data regarding the stage of vitiligo, and the absence of data for undiagnosed COVID-19 infections. In addition, patients with vitiligo may be more likely to seek care, potentially increasing their rates of COVID-19 testing. The inability to identify the stage of vitiligo during enrollment in the database may have altered our results, as individuals with active disease have increased levels of IFN-γ. Increased secretion of IFN-γ also potentially helps in the clearance of COVID-19 infection.1 Future studies should investigate this relationship via planned ­COVID-19 testing, identification of vitiligo stage, and controlling for other associated comorbidities.

CT113004030_Table3.jpg

References
  1. Rashighi M, Harris JE. Vitiligo pathogenesis and emerging treatments. Dermatol Clin. 2017;35:257-265. doi:10.1016/j.det.2016.11.014
  2. Wu JJ, Liu J, Thatiparthi A, et al. The risk of COVID-19 in patients with psoriasis—a retrospective cohort study [published online September 20, 2022]. J Am Acad Dermatol. doi:10.1016/j.jaad.2022.07.040
  3. Zhong J, Shen G, Yang H, et al. COVID-19 in patients with rheumatic disease in Hubei province, China: a multicentre retrospective observational study. Lancet Rheumatol. 2020;2:E557-E564. doi:10.1016/S2665-9913(20)30227-7
  4. Chatterjee M, Das A. Management of vitiligo amidst the ­COVID-19 pandemic: a survey and resulting consensus. Indian J Dermatol. 2021;66:479-483. doi:10.4103/ijd.ijd_859_20
References
  1. Rashighi M, Harris JE. Vitiligo pathogenesis and emerging treatments. Dermatol Clin. 2017;35:257-265. doi:10.1016/j.det.2016.11.014
  2. Wu JJ, Liu J, Thatiparthi A, et al. The risk of COVID-19 in patients with psoriasis—a retrospective cohort study [published online September 20, 2022]. J Am Acad Dermatol. doi:10.1016/j.jaad.2022.07.040
  3. Zhong J, Shen G, Yang H, et al. COVID-19 in patients with rheumatic disease in Hubei province, China: a multicentre retrospective observational study. Lancet Rheumatol. 2020;2:E557-E564. doi:10.1016/S2665-9913(20)30227-7
  4. Chatterjee M, Das A. Management of vitiligo amidst the ­COVID-19 pandemic: a survey and resulting consensus. Indian J Dermatol. 2021;66:479-483. doi:10.4103/ijd.ijd_859_20
Issue
Cutis - 113(4)
Issue
Cutis - 113(4)
Page Number
E30-E32
Page Number
E30-E32
Publications
Publications
Topics
Article Type
Display Headline
Risk for COVID-19 Infection in Patients With Vitiligo
Display Headline
Risk for COVID-19 Infection in Patients With Vitiligo
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>Smith vitiligo</fileName> <TBEID>0C02F614.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F614</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Smith vitiligo</storyname> <articleType>1</articleType> <TBLocation>Ready for Layout-CT</TBLocation> <QCDate/> <firstPublished>20240426T152037</firstPublished> <LastPublished>20240426T152038</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240426T152037</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Brandon Smith, BA; Shahin Shahsavari, BS; Aislyn Oulee, MD</byline> <bylineText>Brandon Smith, BA; Shahin Shahsavari, BS; Aislyn Oulee, MD; Priya Engel, MPH; Alexander Egeberg, MD, PhD, DMSc; Jashin J. Wu, MD</bylineText> <bylineFull>Brandon Smith, BA; Shahin Shahsavari, BS; Aislyn Oulee, MD</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>E30-E32</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>To the Editor:Vitiligo is a depigmentation disorder that results from the loss of melanocytes in the epidermis.1 The most widely accepted pathophysiology for me</metaDescription> <articlePDF>301207</articlePDF> <teaserImage/> <title>Risk for COVID-19 Infection in Patients With Vitiligo</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>April</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>4</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2163</CMSID> </CMSIDs> <keywords> <keyword>pigmentation disorder</keyword> <keyword> COVID-19</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>April 2024</pubIssueName> <pubArticleType>Online Exclusive | 2163</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">104</term> </sections> <topics> <term>63993</term> <term canonical="true">276</term> </topics> <links> <link> <itemClass qcode="ninat:composite"/> <altRep contenttype="application/pdf">images/18002719.pdf</altRep> <description role="drol:caption"/> <description role="drol:credit"/> </link> </links> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Risk for COVID-19 Infection in Patients With Vitiligo</title> <deck/> </itemMeta> <itemContent> <p>To the Editor:<br/><br/>Vitiligo is a depigmentation disorder that results from the loss of melanocytes in the epidermis.<sup>1</sup> The most widely accepted pathophysiology for melanocyte destruction in vitiligo is an autoimmune process involving dysregulated cytokine production and autoreactive T-cell activation.<sup>1</sup> Individuals with cutaneous autoinflammatory conditions currently are vital patient populations warranting research, as their susceptibility to COVID-19 infection may differ from the general population. We previously found a small increased risk for COVID-19 infection in patients with psoriasis,<sup>2</sup> which suggests that other dermatologic conditions also may impact COVID-19 risk. The risk for COVID-19 infection in patients with vitiligo remains largely unknown. In this retrospective cohort study, we investigated the risk for COVID-19 infection in patients with vitiligo compared with those without vitiligo utilizing claims data from the COVID-19 Research Database (https://covid19researchdatabase.org/).</p> <p>Claims were evaluated for patients aged 3 years and older with a vitiligo diagnosis (<i>International Classification of Diseases, Tenth Revision</i> [<i>ICD-10</i>] code L80) that was made between January 1, 2016, and January 1, 2020. Individuals without a vitiligo diagnosis during the same period were placed (4:1 ratio) in the control group and were matched with study group patients for age and sex. All comorbidity variables and vitiligo diagnoses were extracted from <i>ICD-10</i> codes that were given prior to a diagnosis of COVID-19. We then constructed multivariable logistic regression models adjusting for measured confounders to evaluate if vitiligo was associated with higher risk for COVID-19 infection after January 1, 2020. <br/><br/>The vitiligo and nonvitiligo cohorts included 40,363 and 161,452 patients, respectively (Table 1). Logistic regression analysis with adjustment for confounding variables, including high comorbid risk factors (Table 2) revealed that patients with a diagnosis of vitiligo had significantly increased odds of COVID-19 infection compared with patients without vitiligo (adjusted odds ratio [AOR], 1.47; 95% CI, 1.37-1.57; <i>P</i>&lt;.001)(Table 3). Additionally, subgroup logistic analyses for sex, age, and exclusion of patients who were HIV positive revealed that females with vitiligo had higher odds of contracting COVID-19 than males with vitiligo (Table 3).<br/><br/>Our results showed that patients with vitiligo had a higher relative risk for contracting COVID-19 than individuals without vitiligo. It has been reported that the prevalence of COVID-19 is higher among patients with autoimmune diseases compared to the general population.<sup>3</sup> Additionally, a handful of vitiligo patients are managed with immunosuppressive agents that may further weaken their immune response.<sup>1</sup> Moreover, survey results from dermatologists managing vitiligo patients revealed that physicians were fairly comfortable prescribing immunosuppressants and encouraging in-office phototherapy during the COVID-19 pandemic.<sup>4</sup> As a result, more patients may have been attending in-office visits for their phototherapy, which may have increased their risk for COVID-19. Although these factors play a role in ­COVID-19 infection rates, the underlying immune dysregulation in vitiligo in relation to COVID-19 remains unknown and should be further explored.<br/><br/>Our findings are limited by the use of <i>ICD-10</i> codes, the inability to control for all potential confounding variables, the lack of data regarding the stage of vitiligo, and the absence of data for undiagnosed COVID-19 infections. In addition, patients with vitiligo may be more likely to seek care, potentially increasing their rates of COVID-19 testing. The inability to identify the stage of vitiligo during enrollment in the database may have altered our results, as individuals with active disease have increased levels of IFN-<span class="hgkelc">γ</span>. Increased secretion of IFN-<span class="hgkelc">γ</span><span class="hgkelc"> also</span> potentially helps in the clearance of COVID-19 infection.<sup>1</sup> Future studies should investigate this relationship via planned ­COVID-19 testing, identification of vitiligo stage, and controlling for other associated comorbidities.</p> <h2>References</h2> <p class="reference"> 1. Rashighi M, Harris JE. Vitiligo pathogenesis and emerging treatments.<span class="apple-converted-space"> </span><i>Dermatol Clin</i>. 2017;35:257-265. doi:10.1016/j.det.2016.11.014<br/><br/> 2. Wu JJ, Liu J, Thatiparthi A, et al. The risk of COVID-19 in patients with psoriasis—a retrospective cohort study [published online September 20, 2022].<span class="apple-converted-space"> </span><i>J Am Acad Dermatol</i>. doi:10.1016/j.jaad.2022.07.040<br/><br/> 3. Zhong J, Shen G, Yang H, et al. COVID-19 in patients with rheumatic disease in Hubei province, China: a multicentre retrospective observational study.<span class="apple-converted-space"> </span><i>Lancet Rheumatol</i>. 2020;2:E557-E564. doi:10.1016/S2665-9913(20)30227-7<br/><br/> 4. Chatterjee M, Das A. Management of vitiligo amidst the ­COVID-19 pandemic: a survey and resulting consensus.<span class="apple-converted-space"> </span><i>Indian J Dermatol</i>. 2021;66:479-483. doi:10.4103/ijd.ijd_859_20</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">Brandon Smith is from the Drexel University College of Medicine, Philadelphia, Pennsylvania. Shahin Shahsavari is from the Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire. Aislyn Oulee is from the University of California Riverside School of Medicine. Priya Engel is from the California University of Science and Medicine, Colton. Dr. Egeberg is from the Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark, and the Department of Clinical Medicine, University of Copenhagen. Dr. Wu is from the University of Miami Leonard M. Miller School of Medicine, Florida.</p> <p class="disclosure">Brandon Smith, Shahin Shahsavari, Aislyn Oulee, and Priya Engel report no conflict of interest. Dr. Egeberg has received research funding from AbbVie, Boehringer Ingelheim, Bristol-Myers Squibb, the Danish National Psoriasis Foundation, Eli Lilly and Company, Janssen Pharmaceuticals, the Kgl Hofbundtmager Aage Bangs Foundation, Novartis, Pfizer, and the Simon Spies Foundation. He also is a consultant and/or speaker for or is/has been an employee of AbbVie, Almirall, Boehringer Ingelheim, Bristol-Myers Squibb, Dermavant, Eli Lilly and Company, Galápagos NV, Galderma, Horizon Therapeutics, Janssen Pharmaceuticals, LEO Pharma, McNeil Consumer Healthcare, Mylan, Novartis, Pfizer, Samsung Bioepis Co Ltd, Sun Pharmaceuticals, UCB, Union Therapeutics, and Zuellig Pharma Ltd. Dr. Wu is or has been a consultant, investigator, or speaker for AbbVie, Almirall, Amgen, Arcutis, Aristea Therapeutics, Bausch Health, Boehringer Ingelheim, Bristol-Myers Squibb, Codex Labs, Dermavant, DermTech, Dr. Reddy’s Laboratories, Eli Lilly and Company, EPI Health, Galderma, Incyte, Janssen, LEO Pharma, Mindera, Novartis, Pfizer, Regeneron, Samsung Bioepis, Sanofi Genzyme, Solius, Sun Pharmaceuticals, UCB, and Zerigo Health.<br/><br/>Correspondence: Jashin J. Wu, MD, University of Miami Leonard M. Miller School of Medicine, 1600 NW 10th Ave, RMSB, Room 2023-A, Miami, FL 33136 (jashinwu@gmail.com). ORCID: 0000-0002-1722-1892. Scopus Author ID: 14629788600 <br/><br/><i>Cutis.</i> 2024 April;113(4):E30-E32. doi:10.12788/cutis.1006</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>pp</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>The underlying autoimmune process in vitiligo can result in various changes to the immune system.</li> <li>A diagnosis of vitiligo may alter the body’s immune response to COVID-19 infection.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • The underlying autoimmune process in vitiligo can result in various changes to the immune system.
  • A diagnosis of vitiligo may alter the body’s immune response to COVID-19 infection.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media
Image
Disable zoom
Off

Association of Atrial Fibrillation and/or Flutter With Adverse Cardiac Outcomes and Mortality in Patients With Wolff-Parkinson-White Syndrome

Article Type
Changed
Thu, 04/11/2024 - 12:04

Wolff-Parkinson-White (WPW) syndrome is characterized by the presence of ≥ 1 accessory pathways and the development of both recurrent paroxysmal atrial fibrillation (AF) and supraventricular tachycardia that can lead to further malignant arrhythmias resulting in sudden cardiac death (SCD).1-7 Historically, incidental, ventricular pre-excitation on electrocardiogram has conferred a relatively low SCD risk in adults; however, newer WPW syndrome data suggest the endpoint may not be as benign as previously thought.7 The current literature has defined atrioventricular reentrant tachycardia triggering AF, rather than symptoms, as an independent risk factor for malignant arrhythmias. Still, long-term data detailing the association of AF with serious cardiac events and death in patients with WPW syndrome are still limited.1-7

While previous guidelines for the treatment of WPW syndrome only recommended routine electrophysiology testing (EPT) with liberal catheter ablation for symptomatic individuals, the 2015 American College of Cardiology/American Heart Association/Heart Rhythm Society guidelines now suggest its potential benefit for risk stratification in the asymptomatic population.8-12 Given the limited existing data, more long-term studies are needed to corroborate the latest EPT recommendations before routinely applying them in practice. Furthermore, since concomitant AF can lead to adverse cardiac outcomes in patients with WPW syndrome, additional data evaluating this association are also necessary. In this study, we aimed to determine the impact of atrial fibrillation and/or flutter (AF/AFL) on adverse cardiac outcomes and mortality in patients with WPW syndrome.

METHODS

This study used data from the Military Health System (MHS) Database Repository. The MHS is one of the largest health care systems in the country and includes information on about 10 million active duty and retired military service members and their families (51% male; 49% female).13,14 Data were fully anonymized and complied in accordance with federal and state laws, including the Health Insurance Portability and Accountability Act of 1996. The Naval Medical Center Portsmouth Institutional Review Board approved this study.

 

Study Design

This retrospective, observational cohort study identified MHS patients with WPW syndrome from January 1, 2014, to December 31, 2019. Patients were included if they had ≥ 2 International Classification of Diseases, Ninth Revision (ICD-9) or International Classification of Diseases, Tenth Revision (ICD-10) diagnosis codes for WPW syndrome (ICD-9, 426.7; ICD-10, I45.6) on separate dates; were aged ≥ 18 years at index date; and had ≥ 1 year of continuous eligibility prior to the index date (enrollment gaps ≤ 30 days were considered continuous). Patients were then divided into 2 subgroups by the presence or absence of AF/AFL using diagnostic codes. Patients were excluded if they had evidence of an implantable cardioverter-defibrillator, permanent pacemaker or were missing age or sex data. Patients were followed from index date until the first occurrence of the outcome of interest, MHS disenrollment, or the end of the study period.

Cardiac composite outcomes comprised of sudden cardiac arrest (SCA), ventricular fibrillation (VF), ventricular tachycardia and death, as well as death specifically, were the outcomes of interest and assessed after index date using ICD-9 and ICD-10 codes. Death was defined as all-cause mortality. Time to event was calculated based on the date of the initial component from the composite outcome and date of death specifically for mortality. Those not experiencing an outcome were followed until MHS disenrollment or the end of the study period.

Various patient characteristics were assessed at index including age, sex, military sponsor (the patient’s active or retired duty member through which their dependent receives TRICARE benefits) rank and branch, geographic region, type of US Department of Defense beneficiary, and index year. Clinical characteristics were assessed over a 1-year baseline period prior to index date and included the number of cardiologist and clinical visits for WPW syndrome, Charlson Comorbidity Index (CCI) scores calculated from diagnostic codes outlined in the Quan coding method, and preindex time.15 Comorbidities were assessed at baseline and defined as having ≥ 1 ICD-9 or ICD-10 code for a corresponding condition within 1 year prior to index.

 

 

Statistical Analysis

Baseline characteristics were assessed and descriptive statistics for categorical and continuous variables were presented accordingly. To assess bivariate association with exposure, χ2 tests were used to compare categorical variables, while t tests were used to compare continuous variables by exposure status. Incidence proportions and rates were reported for each outcome of interest. Kaplan-Meier curves were constructed to assess the bivariate association between exposure and study outcomes. Cox proportional hazard modeling was performed to estimate the association between AF/AFL and time to each of the outcomes. Multiple models were designed to assess cardiac and metabolic covariates, in addition to baseline characteristics. This included a base model adjusted for age, sex, military sponsor rank and branch, geographic region, and duty status.

Additional models adjusted for cardiac and metabolic confounders and CCI score. A comprehensive model included the base, cardiac, and metabolic covariates. Multicollinearity between covariates was assessed. Variables with a variance inflation factor > 4 or a tolerance level < 0.1 were added to the models. Cox proportional hazard models were used to estimate the unadjusted and adjusted hazard ratios (HRs) and 95% CIs of the association between AF/AFL and the study outcomes. Data were analyzed using SAS, version 9.4 for Windows.

RESULTS

table_1.png

From 2014 through 2019, 35,539 patients with WPW syndrome were identified in the MHS, 5291 had AF/AFL (14.9%); 19,961 were female (56.2%), the mean (SD) age was 62.9 (18.0) years, and 11,742 were aged ≥ 75 years (33.0%) (Table 1).

figure_1.png

There were 4121 (11.6%), 322 (0.9%), and 848 (2.4%) patients with AF, AFL, and both arrhythmias, respectively. The mean (SD) number of cardiology visits was 3.9 (3.0). The mean (SD) baseline CCI score for the AF/AFL subgroup was 5.9 (3.5) vs 3.7 (2.2) for the non-AF/AFL subgroup (P < .001). The most prevalent comorbid conditions were hypertension, hyperlipidemia, chronic obstructive pulmonary disease, and diabetes (P < .001) (Figure 1).

 

Composite Outcomes

figure_2.png

In the overall cohort, during a mean (SD) follow-up time of 3.4 (2.0) years comprising 119,682 total person-years, the components of the composite outcome occurred 6506 times with an incidence rate of 5.44 per 100 person-years. Ventricular tachycardia was the most common event, occurring 3281 times with an incidence rate of 2.74 per 100 person-years. SCA and VF occurred 841 and 135 times with incidence rates of 0.70 and 0.11 per 100 person-years, respectively. Death was the initial event 2249 times with an incidence rate of 1.88 per 100 person-years. Figure 2 shows the Kaplan-Meier curve of cardiac composite outcome by AF/AFL status.

table_2.png

The subgroup with AF/AFL comprised 17,412 total person-years and 1424 cardiac composite incidences compared with 102,270 person years and 5082 incidences in the no AF/AFL group (Table 2). Comparing AF/AFL vs no AF/AFL incidence rates were 8.18 vs 4.97 per 100 person-years, respectively (P < .001). SCA and VF occurred 233 and 38 times and respectively had incidence rates of 1.34 and 0.22 per 100 person-years in the AF/AFL group vs 0.59 and 0.09 per 100 person-years in the no AF/AFL group (P < .001). There were 549 deaths and a 3.15 per 100 person-years incidence rate in the AF/AFL group vs 1700 deaths and a 1.66 incidence rate in the no AF/AFL group (P < .001).

table_3.png

The HR for the composite outcome in the base model was 1.33 (95% CI, 1.26-1.42, P < .001) (Table 3). The association between AF/AFL and the composite outcome remained significant after adjusting for additional metabolic and cardiac covariates. The HRs for the metabolic and cardiac models were 1.30 (95% CI, 1.23-1.38, P < .001) and 1.11 (95% CI, 1.05-1.18, P < .001), respectively. After adjusting for the full model, the HR was 1.12 (95% CI, 1.05-1.19, P < .001).

 

 

Mortality

figure_3.png

Over the 6-year study period, there was a lower survival probability for patients with AF/AFL. In the overall cohort, during a mean (SD) follow-up time of 3.7 (1.9) years comprising 129,391 total person-years, there were 3130 (8.8%) deaths and an incidence rate of 2.42 per 100 person-years. Death occurred 786 times with a 4.09 incidence rate per 100 person-years in the AF/AFL vs 2344 deaths and a 2.13 incidence rate per 100 person-years in the no AF/AFL group (P < .001). In the non-AF/AFL subgroup, death occurred 2344 times during a mean (SD) follow-up of 3.7 (1.9) years comprising 110,151 total person-years. Figure 3 shows the Kaplan-Meier curve of mortality by AF/AFL status.

table_4.png

After adjusting for the base, metabolic and cardiac covariates, the HRs for mortality were 1.45 (95% CI, 1.33-1.57, P < .001), 1.40 (95% CI, 1.29-1.51, P < .001) and 1.15 (95% CI, 1.06-1.25, P = .001), respectively (Table 4). The HR after adjusting for the full model was 1.16 (95% CI, 1.07-1.26, P < .001).

DISCUSSION

In this large retrospective cohort study, patients with WPW syndrome and comorbid AF/AFL had a significantly higher association with the cardiac composite outcome and death during a 3-year follow-up period when compared with patients without AF/AFL. After adjusting for confounding variables, the AF/AFL subgroup maintained a 12% and 16% higher association with the composite outcome and mortality, respectively. There was minimal difference in confounding effects between demographic data and metabolic profiles, suggesting one may serve as a proxy for the other.

To our knowledge, this is the largest WPW syndrome cohort study evaluating cardiac outcomes and mortality to date. Although previous research has shown the relatively low and mostly anecdotal SCD incidence within this population,our results demonstrate a higher association of adverse cardiac outcomes and death in an AF/AFL subgroup.16-18 Notably, in this study the AF/AFL cohort was older and had higher CCI scores than their counterparts (P < .001), thus inferring an inherently greater degree of morbidity and 10-year mortality risk. Our study is also unique in that the mean patient age was significantly older than previously reported (63 vs 27 years), which may suggest a longer living history of both ventricular pre-excitation and the comorbidities outlined in Figure 1.19 Given these age discrepancies, it is possible that our overall study population was still relatively low risk and that not all reported deaths were necessarily related to WPW syndrome. Despite these assumptions, when comparing the WPW syndrome subgroups, we still found the AF/AFL cohort maintained a statistically significant higher association with the 2 study outcomes, even after adjusting for the greater presence of comorbidities. This suggests that the presence of AF/AFL may still portend a worse prognosis in patients with WPW syndrome.

Although the association of AF and development of VF in patients with WPW syndrome—due to rapid conduction over the accessory pathway(s)—was first reported > 40 years ago, there has still been few large, long-term data studies exploring mortality in this cohort.19-25 Furthermore, even though the current literature attributes the development of AF with the electrophysiologic properties of the accessory pathway, as well as intrinsic atrial architecture and muscle vulnerability, there is still equivocal consensus regarding EPT screening and ablation indications for asymptomatic patients with WPW syndrome.26-28 Notably, Pappone and colleagues demonstrated the potential benefit of liberal ablation indications for asymptomatic patients, arguing that the intrinsic electrophysiologic properties of the accessory pathway—ie, short accessory-pathway antegrade effective refractory period, inducibility of atrioventricular reentrant tachycardia triggering AF, and multiple accessory pathway—rather than symptoms, are independent predictors of developing malignant arrhythmia.1-5

These findings contradict those reported by Obeyesekere and colleagues, who concluded that the low SCD incidence rates in patients with WPW syndrome precluded routine invasive screening.19,28 They argued that Pappone and colleagues used malignant arrhythmia as a surrogate marker for death, and that the positive predictive value of a short accessory-pathway antegrade effective refractory period for developing malignant arrhythmia was lower than reported (15% vs 82%, respectively) and that its negative predictive value was 100%.1,19,28 Given these conflicting recommendations, we hope our data elucidates the higher association of adverse outcomes and support considerations for more intensive EPT indications in patients with WPW syndrome.

While our study does not report SCD incidence, it does provide robust and reliable mortality data that suggests a greater association of death within an AF/AFL subgroup. Our findings would support more liberal EPT recommendations in patients with WPW syndrome.1-5,8,9 In this study, the SCA incidence rate was more than double the rate in the AF/AFL cohort (P < .001) and is commonly the initial presenting event in WPW syndrome.9 Even though the reported SCD incidence rate is low in WPW syndrome, our data demonstrated an increased association of death within the AF/AFL cohort. Physicians should consider early risk stratification and ablation to prevent potential recurrent malignant arrhythmia leading to death.1-5,8,9,12,19,20

 

 

Limitations

As a retrospective study and without access to the National Death Index, we were unable to determine the exact cause or events leading to death and instead utilized all-cause mortality data. Subsequently, our observations may only demonstrate association, rather than causality, between AF/AFL and death in patients with WPW syndrome. Additionally, we could not distinguish between AF and AFL as the arrhythmia leading to death. However, since overall survivability was the outcome of interest, our adjusted HR models were still able to demonstrate the increased association of the composite outcome and death within an AF/AFL cohort.

Although a large cohort was analyzed, due to the constraints of utilizing diagnostic codes to determine study outcomes, we could not distinguish between symptomatic and asymptomatic patients, nor how they were managed prior to the outcome event. However, as recent literature demonstrates, updated predictors of malignant arrhythmia and decisions for early EPT are similar for both symptomatic and asymptomatic patients and should be driven by the intrinsic electrophysiologic properties of the accessory pathway, rather than symptomatology;thus, our inability to discern this should have negligible consequence in determining when to perform risk stratification and ablation.1

MHS eligible patients have direct access to care; the generalizability of our data may not necessarily correspond to a community population with lower socioeconomic status (we did adjust for military sponsor rank which has been used as a proxy), reduced access to care, or uninsured individuals. However, the prevalence of WPW syndrome within our cohort was comparable to the general population, 0.4% vs 0.1%-0.3%, respectively.13,14,19 Similarly, the incidence of AF within our population was comparable to the general population, 15% vs 16%-26%, respectively.23 These similar data points suggest our results may apply beyond MHS patients.

CONCLUSIONS

Patients with WPW syndrome and AF/AFL have a higher association with adverse cardiac outcomes and death. Despite previously reported low SCD incidence rates in this population, our study demonstrates the increased association of mortality in an AF/AFL cohort. The limitations of utilizing all-cause mortality data necessitate further investigation into the etiology behind the deaths in our study population. Since ventricular pre-excitation can predispose patients to AF and potentially lead to malignant arrhythmia and SCD, understanding the cause of mortality will allow physicians to determine the appropriate monitoring and intervention strategies to improve outcomes in this population. Our results suggest consideration for more aggressive EPT screening and ablation recommendations in patients with WPW syndrome may be warranted.

References

1. Pappone C, Vicedomini G, Manguso F, et al. The natural history of WPW syndrome. Eur Heart J Suppl. 2015; 17 (Supplement A):A8-A11.doi:10.1093/eurheartj/suv004

2. Pappone C, Vicedomini G, Manguso F, et al. Risk of malignant arrhythmias in initially symptomatic patients with Wolff-Parkinson-White syndrome: results of a prospective long-term electrophysiological follow-up study. Circulation. 2012;125(5):661-668. doi:10.1161/CIRCULATIONAHA.111.065722

3. Pappone C, Santinelli V, Rosanio S, et al. Usefulness of invasive electrophysiologic testing to stratify the risk of arrhythmic events in asymptomatic patients with Wolff-Parkinson-White pattern: results from a large prospective long-term follow-up study. J Am Coll Cardiol. 2003;41(2):239-244. doi:10.1016/s0735-1097(02)02706-7

4. Pappone C, Vicedomini G, Manguso F, et al. Wolff-Parkinson-White syndrome in the era of catheter ablation: insights from a registry study of 2169 patients. Circulation. 2014;130(10):811-819. doi:10.1161/CIRCULATIONAHA.114.011154

5. Pappone C, Santinelli V, Manguso F, et al. A randomized study of prophylactic catheter ablation in asymptomatic patients with the Wolff-Parkinson-White syndrome. N Engl J Med. 2003;349(19):1803-1811. doi:10.1056/NEJMoa035345

6. Santinelli V, Radinovic A, Manguso F, et al. Asymptomatic ventricular preexcitation: a long-term prospective follow-up study of 293 adult patients. Circ Arrhythm Electrophysiol. 2009;2(2):102-107. doi:10.1161/CIRCEP.108.827550

7. Santinelli V, Radinovic A, Manguso F, et al. The natural history of asymptomatic ventricular pre-excitation a long-term prospective follow-up study of 184 asymptomatic children. J Am Coll Cardiol. 2009;53(3):275-280. doi:10.1016/j.jacc.2008.09.037

8. Al-Khatib SM, Arshad A, Balk EM, et al. Risk Stratification for Arrhythmic Events in Patients With Asymptomatic Pre-Excitation: A Systematic Review for the 2015 ACC/AHA/HRS Guideline for the Management of Adult Patients With Supraventricular Tachycardia: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. J Am Coll Cardiol. 2016;67(13):1624-1638. doi:10.1016/j.jacc.2015.09.018

9. Blomström-Lundqvist C, Scheinman MM, Aliot EM, et al. ACC/AHA/ESC guidelines for the management of patients with supraventricular arrhythmias--executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the European Society of Cardiology Committee for Practice Guidelines (Writing Committee to Develop Guidelines for the Management of Patients With Supraventricular Arrhythmias). Circulation. 2003;108(15):1871-1909.doi:10.1161/01.CIR.0000091380.04100.84

10. Pediatric and Congenital Electrophysiology Society (PACES); Heart Rhythm Society (HRS); American College of Cardiology Foundation (ACCF); PACES/HRS expert consensus statement on the management of the asymptomatic young patient with a Wolff-Parkinson-White (WPW, ventricular preexcitation) electrocardiographic pattern: developed in partnership between the Pediatric and Congenital Electrophysiology Society (PACES) and the Heart Rhythm Society (HRS). Endorsed by the governing bodies of PACES, HRS, the American College of Cardiology Foundation (ACCF), the American Heart Association (AHA), the American Academy of Pediatrics (AAP), and the Canadian Heart Rhythm Society (CHRS). Heart Rhythm. 2012;9(6):1006-1024. doi:10.1016/j.hrthm.2012.03.050

11. Cohen M, Triedman J. Guidelines for management of asymptomatic ventricular pre-excitation: brave new world or Pandora’s box?. Circ Arrhythm Electrophysiol. 2014;7(2):187-189. doi:10.1161/CIRCEP.114.001528

12. Svendsen JH, Dagres N, Dobreanu D, et al. Current strategy for treatment of patients with Wolff-Parkinson-White syndrome and asymptomatic preexcitation in Europe: European Heart Rhythm Association survey. Europace. 2013;15(5):750-753. doi:10.1093/europace/eut094

13. Gimbel RW, Pangaro L, Barbour G. America’s “undiscovered” laboratory for health services research. Med Care. 2010;48(8):751-756. doi:10.1097/MLR.0b013e3181e35be8

14. Dorrance KA, Ramchandani S, Neil N, Fisher H. Leveraging the military health system as a laboratory for health care reform. Mil Med. 2013;178(2):142-145. doi:10.7205/milmed-d-12-00168

15. Quan H, Sundararajan V, Halfon P, et al. Coding algorithms for defining comorbidities in ICD-9-CM and ICD-10 administrative data. Med Care. 2005;43(11):1130-1139. doi:10.1097/01.mlr.0000182534.19832.83

16. Finocchiaro G, Papadakis M, Behr ER, Sharma S, Sheppard M. Sudden Cardiac Death in Pre-Excitation and Wolff-Parkinson-White: Demographic and Clinical Features. J Am Coll Cardiol. 2017;69(12):1644-1645. doi:10.1016/j.jacc.2017.01.023

17. Munger TM, Packer DL, Hammill SC, et al. A population study of the natural history of Wolff-Parkinson-White syndrome in Olmsted County, Minnesota, 1953-1989. Circulation. 1993;87(3):866-873. doi:10.1161/01.cir.87.3.866

18. Fitzsimmons PJ, McWhirter PD, Peterson DW, Kruyer WB. The natural history of Wolff-Parkinson-White syndrome in 228 military aviators: a long-term follow-up of 22 years. Am Heart J. 2001;142(3):530-536. doi:10.1067/mhj.2001.117779

19. Obeyesekere MN, Leong-Sit P, Massel D, et al. Risk of arrhythmia and sudden death in patients with asymptomatic preexcitation: a meta-analysis. Circulation. 2012;125(19):2308-2315. doi:10.1161/CIRCULATIONAHA.111.055350

20. Waspe LE, Brodman R, Kim SG, Fisher JD. Susceptibility to atrial fibrillation and ventricular tachyarrhythmia in the Wolff-Parkinson-White syndrome: role of the accessory pathway. Am Heart J. 1986;112(6):1141-1152. doi:10.1016/0002-8703(86)90342-x

21. Pietersen AH, Andersen ED, Sandøe E. Atrial fibrillation in the Wolff-Parkinson-White syndrome. Am J Cardiol. 1992;70(5):38A-43A. doi:10.1016/0002-9149(92)91076-g

22. Della Bella P, Brugada P, Talajic M, et al. Atrial fibrillation in patients with an accessory pathway: importance of the conduction properties of the accessory pathway. J Am Coll Cardiol. 1991;17(6):1352-1356. doi:10.1016/s0735-1097(10)80146-9

23. Fujimura O, Klein GJ, Yee R, Sharma AD. Mode of onset of atrial fibrillation in the Wolff-Parkinson-White syndrome: how important is the accessory pathway?. J Am Coll Cardiol. 1990;15(5):1082-1086. doi:10.1016/0735-1097(90)90244-j

24. Montoya PT, Brugada P, Smeets J, et al. Ventricular fibrillation in the Wolff-Parkinson-White syndrome. Eur Heart J. 1991;12(2):144-150. doi:10.1093/oxfordjournals.eurheartj.a059860

25. Klein GJ, Bashore TM, Sellers TD, Pritchett EL, Smith WM, Gallagher JJ. Ventricular fibrillation in the Wolff-Parkinson-White syndrome. N Engl J Med. 1979;301(20):1080-1085. doi:10.1056/NEJM197911153012003

26. Centurion OA. Atrial Fibrillation in the Wolff-Parkinson-White Syndrome. J Atr Fibrillation. 2011;4(1):287. Published 2011 May 4. doi:10.4022/jafib.287

27. Song C, Guo Y, Zheng X, et al. Prognostic Significance and Risk of Atrial Fibrillation of Wolff-Parkinson-White Syndrome in Patients With Hypertrophic Cardiomyopathy. Am J Cardiol. 2018;122(9):1546-1550. doi:10.1016/j.amjcard.2018.07.021

28. Obeyesekere M, Gula LJ, Skanes AC, Leong-Sit P, Klein GJ. Risk of sudden death in Wolff-Parkinson-White syndrome: how high is the risk?. Circulation. 2012;125(5):659-660. doi:10.1161/CIRCULATIONAHA.111.085159

Article PDF
Author and Disclosure Information

LCDR John C. Chin, MD, MC, USNa; CAPT Andrew H. Lin, MD, MC, USNa; Nicholas M. Sicignano, MPHb; Toni M. Rush, PhD, MPHb

Correspondence:  John Chin   (chinjoh@gmail.com)

aNaval Medical Center Portsmouth, Virginia

bHealth ResearchTx LLC, Trevose, Pennsylvania

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Ethics and consent

Research and data from this study were reviewed andapproved by the Naval Medical Center PortsmouthInstitutional Review Board.

Issue
Federal Practitioner - 41(4)a
Publications
Topics
Page Number
108
Sections
Author and Disclosure Information

LCDR John C. Chin, MD, MC, USNa; CAPT Andrew H. Lin, MD, MC, USNa; Nicholas M. Sicignano, MPHb; Toni M. Rush, PhD, MPHb

Correspondence:  John Chin   (chinjoh@gmail.com)

aNaval Medical Center Portsmouth, Virginia

bHealth ResearchTx LLC, Trevose, Pennsylvania

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Ethics and consent

Research and data from this study were reviewed andapproved by the Naval Medical Center PortsmouthInstitutional Review Board.

Author and Disclosure Information

LCDR John C. Chin, MD, MC, USNa; CAPT Andrew H. Lin, MD, MC, USNa; Nicholas M. Sicignano, MPHb; Toni M. Rush, PhD, MPHb

Correspondence:  John Chin   (chinjoh@gmail.com)

aNaval Medical Center Portsmouth, Virginia

bHealth ResearchTx LLC, Trevose, Pennsylvania

Author disclosures

The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Ethics and consent

Research and data from this study were reviewed andapproved by the Naval Medical Center PortsmouthInstitutional Review Board.

Article PDF
Article PDF

Wolff-Parkinson-White (WPW) syndrome is characterized by the presence of ≥ 1 accessory pathways and the development of both recurrent paroxysmal atrial fibrillation (AF) and supraventricular tachycardia that can lead to further malignant arrhythmias resulting in sudden cardiac death (SCD).1-7 Historically, incidental, ventricular pre-excitation on electrocardiogram has conferred a relatively low SCD risk in adults; however, newer WPW syndrome data suggest the endpoint may not be as benign as previously thought.7 The current literature has defined atrioventricular reentrant tachycardia triggering AF, rather than symptoms, as an independent risk factor for malignant arrhythmias. Still, long-term data detailing the association of AF with serious cardiac events and death in patients with WPW syndrome are still limited.1-7

While previous guidelines for the treatment of WPW syndrome only recommended routine electrophysiology testing (EPT) with liberal catheter ablation for symptomatic individuals, the 2015 American College of Cardiology/American Heart Association/Heart Rhythm Society guidelines now suggest its potential benefit for risk stratification in the asymptomatic population.8-12 Given the limited existing data, more long-term studies are needed to corroborate the latest EPT recommendations before routinely applying them in practice. Furthermore, since concomitant AF can lead to adverse cardiac outcomes in patients with WPW syndrome, additional data evaluating this association are also necessary. In this study, we aimed to determine the impact of atrial fibrillation and/or flutter (AF/AFL) on adverse cardiac outcomes and mortality in patients with WPW syndrome.

METHODS

This study used data from the Military Health System (MHS) Database Repository. The MHS is one of the largest health care systems in the country and includes information on about 10 million active duty and retired military service members and their families (51% male; 49% female).13,14 Data were fully anonymized and complied in accordance with federal and state laws, including the Health Insurance Portability and Accountability Act of 1996. The Naval Medical Center Portsmouth Institutional Review Board approved this study.

 

Study Design

This retrospective, observational cohort study identified MHS patients with WPW syndrome from January 1, 2014, to December 31, 2019. Patients were included if they had ≥ 2 International Classification of Diseases, Ninth Revision (ICD-9) or International Classification of Diseases, Tenth Revision (ICD-10) diagnosis codes for WPW syndrome (ICD-9, 426.7; ICD-10, I45.6) on separate dates; were aged ≥ 18 years at index date; and had ≥ 1 year of continuous eligibility prior to the index date (enrollment gaps ≤ 30 days were considered continuous). Patients were then divided into 2 subgroups by the presence or absence of AF/AFL using diagnostic codes. Patients were excluded if they had evidence of an implantable cardioverter-defibrillator, permanent pacemaker or were missing age or sex data. Patients were followed from index date until the first occurrence of the outcome of interest, MHS disenrollment, or the end of the study period.

Cardiac composite outcomes comprised of sudden cardiac arrest (SCA), ventricular fibrillation (VF), ventricular tachycardia and death, as well as death specifically, were the outcomes of interest and assessed after index date using ICD-9 and ICD-10 codes. Death was defined as all-cause mortality. Time to event was calculated based on the date of the initial component from the composite outcome and date of death specifically for mortality. Those not experiencing an outcome were followed until MHS disenrollment or the end of the study period.

Various patient characteristics were assessed at index including age, sex, military sponsor (the patient’s active or retired duty member through which their dependent receives TRICARE benefits) rank and branch, geographic region, type of US Department of Defense beneficiary, and index year. Clinical characteristics were assessed over a 1-year baseline period prior to index date and included the number of cardiologist and clinical visits for WPW syndrome, Charlson Comorbidity Index (CCI) scores calculated from diagnostic codes outlined in the Quan coding method, and preindex time.15 Comorbidities were assessed at baseline and defined as having ≥ 1 ICD-9 or ICD-10 code for a corresponding condition within 1 year prior to index.

 

 

Statistical Analysis

Baseline characteristics were assessed and descriptive statistics for categorical and continuous variables were presented accordingly. To assess bivariate association with exposure, χ2 tests were used to compare categorical variables, while t tests were used to compare continuous variables by exposure status. Incidence proportions and rates were reported for each outcome of interest. Kaplan-Meier curves were constructed to assess the bivariate association between exposure and study outcomes. Cox proportional hazard modeling was performed to estimate the association between AF/AFL and time to each of the outcomes. Multiple models were designed to assess cardiac and metabolic covariates, in addition to baseline characteristics. This included a base model adjusted for age, sex, military sponsor rank and branch, geographic region, and duty status.

Additional models adjusted for cardiac and metabolic confounders and CCI score. A comprehensive model included the base, cardiac, and metabolic covariates. Multicollinearity between covariates was assessed. Variables with a variance inflation factor > 4 or a tolerance level < 0.1 were added to the models. Cox proportional hazard models were used to estimate the unadjusted and adjusted hazard ratios (HRs) and 95% CIs of the association between AF/AFL and the study outcomes. Data were analyzed using SAS, version 9.4 for Windows.

RESULTS

table_1.png

From 2014 through 2019, 35,539 patients with WPW syndrome were identified in the MHS, 5291 had AF/AFL (14.9%); 19,961 were female (56.2%), the mean (SD) age was 62.9 (18.0) years, and 11,742 were aged ≥ 75 years (33.0%) (Table 1).

figure_1.png

There were 4121 (11.6%), 322 (0.9%), and 848 (2.4%) patients with AF, AFL, and both arrhythmias, respectively. The mean (SD) number of cardiology visits was 3.9 (3.0). The mean (SD) baseline CCI score for the AF/AFL subgroup was 5.9 (3.5) vs 3.7 (2.2) for the non-AF/AFL subgroup (P < .001). The most prevalent comorbid conditions were hypertension, hyperlipidemia, chronic obstructive pulmonary disease, and diabetes (P < .001) (Figure 1).

 

Composite Outcomes

figure_2.png

In the overall cohort, during a mean (SD) follow-up time of 3.4 (2.0) years comprising 119,682 total person-years, the components of the composite outcome occurred 6506 times with an incidence rate of 5.44 per 100 person-years. Ventricular tachycardia was the most common event, occurring 3281 times with an incidence rate of 2.74 per 100 person-years. SCA and VF occurred 841 and 135 times with incidence rates of 0.70 and 0.11 per 100 person-years, respectively. Death was the initial event 2249 times with an incidence rate of 1.88 per 100 person-years. Figure 2 shows the Kaplan-Meier curve of cardiac composite outcome by AF/AFL status.

table_2.png

The subgroup with AF/AFL comprised 17,412 total person-years and 1424 cardiac composite incidences compared with 102,270 person years and 5082 incidences in the no AF/AFL group (Table 2). Comparing AF/AFL vs no AF/AFL incidence rates were 8.18 vs 4.97 per 100 person-years, respectively (P < .001). SCA and VF occurred 233 and 38 times and respectively had incidence rates of 1.34 and 0.22 per 100 person-years in the AF/AFL group vs 0.59 and 0.09 per 100 person-years in the no AF/AFL group (P < .001). There were 549 deaths and a 3.15 per 100 person-years incidence rate in the AF/AFL group vs 1700 deaths and a 1.66 incidence rate in the no AF/AFL group (P < .001).

table_3.png

The HR for the composite outcome in the base model was 1.33 (95% CI, 1.26-1.42, P < .001) (Table 3). The association between AF/AFL and the composite outcome remained significant after adjusting for additional metabolic and cardiac covariates. The HRs for the metabolic and cardiac models were 1.30 (95% CI, 1.23-1.38, P < .001) and 1.11 (95% CI, 1.05-1.18, P < .001), respectively. After adjusting for the full model, the HR was 1.12 (95% CI, 1.05-1.19, P < .001).

 

 

Mortality

figure_3.png

Over the 6-year study period, there was a lower survival probability for patients with AF/AFL. In the overall cohort, during a mean (SD) follow-up time of 3.7 (1.9) years comprising 129,391 total person-years, there were 3130 (8.8%) deaths and an incidence rate of 2.42 per 100 person-years. Death occurred 786 times with a 4.09 incidence rate per 100 person-years in the AF/AFL vs 2344 deaths and a 2.13 incidence rate per 100 person-years in the no AF/AFL group (P < .001). In the non-AF/AFL subgroup, death occurred 2344 times during a mean (SD) follow-up of 3.7 (1.9) years comprising 110,151 total person-years. Figure 3 shows the Kaplan-Meier curve of mortality by AF/AFL status.

table_4.png

After adjusting for the base, metabolic and cardiac covariates, the HRs for mortality were 1.45 (95% CI, 1.33-1.57, P < .001), 1.40 (95% CI, 1.29-1.51, P < .001) and 1.15 (95% CI, 1.06-1.25, P = .001), respectively (Table 4). The HR after adjusting for the full model was 1.16 (95% CI, 1.07-1.26, P < .001).

DISCUSSION

In this large retrospective cohort study, patients with WPW syndrome and comorbid AF/AFL had a significantly higher association with the cardiac composite outcome and death during a 3-year follow-up period when compared with patients without AF/AFL. After adjusting for confounding variables, the AF/AFL subgroup maintained a 12% and 16% higher association with the composite outcome and mortality, respectively. There was minimal difference in confounding effects between demographic data and metabolic profiles, suggesting one may serve as a proxy for the other.

To our knowledge, this is the largest WPW syndrome cohort study evaluating cardiac outcomes and mortality to date. Although previous research has shown the relatively low and mostly anecdotal SCD incidence within this population,our results demonstrate a higher association of adverse cardiac outcomes and death in an AF/AFL subgroup.16-18 Notably, in this study the AF/AFL cohort was older and had higher CCI scores than their counterparts (P < .001), thus inferring an inherently greater degree of morbidity and 10-year mortality risk. Our study is also unique in that the mean patient age was significantly older than previously reported (63 vs 27 years), which may suggest a longer living history of both ventricular pre-excitation and the comorbidities outlined in Figure 1.19 Given these age discrepancies, it is possible that our overall study population was still relatively low risk and that not all reported deaths were necessarily related to WPW syndrome. Despite these assumptions, when comparing the WPW syndrome subgroups, we still found the AF/AFL cohort maintained a statistically significant higher association with the 2 study outcomes, even after adjusting for the greater presence of comorbidities. This suggests that the presence of AF/AFL may still portend a worse prognosis in patients with WPW syndrome.

Although the association of AF and development of VF in patients with WPW syndrome—due to rapid conduction over the accessory pathway(s)—was first reported > 40 years ago, there has still been few large, long-term data studies exploring mortality in this cohort.19-25 Furthermore, even though the current literature attributes the development of AF with the electrophysiologic properties of the accessory pathway, as well as intrinsic atrial architecture and muscle vulnerability, there is still equivocal consensus regarding EPT screening and ablation indications for asymptomatic patients with WPW syndrome.26-28 Notably, Pappone and colleagues demonstrated the potential benefit of liberal ablation indications for asymptomatic patients, arguing that the intrinsic electrophysiologic properties of the accessory pathway—ie, short accessory-pathway antegrade effective refractory period, inducibility of atrioventricular reentrant tachycardia triggering AF, and multiple accessory pathway—rather than symptoms, are independent predictors of developing malignant arrhythmia.1-5

These findings contradict those reported by Obeyesekere and colleagues, who concluded that the low SCD incidence rates in patients with WPW syndrome precluded routine invasive screening.19,28 They argued that Pappone and colleagues used malignant arrhythmia as a surrogate marker for death, and that the positive predictive value of a short accessory-pathway antegrade effective refractory period for developing malignant arrhythmia was lower than reported (15% vs 82%, respectively) and that its negative predictive value was 100%.1,19,28 Given these conflicting recommendations, we hope our data elucidates the higher association of adverse outcomes and support considerations for more intensive EPT indications in patients with WPW syndrome.

While our study does not report SCD incidence, it does provide robust and reliable mortality data that suggests a greater association of death within an AF/AFL subgroup. Our findings would support more liberal EPT recommendations in patients with WPW syndrome.1-5,8,9 In this study, the SCA incidence rate was more than double the rate in the AF/AFL cohort (P < .001) and is commonly the initial presenting event in WPW syndrome.9 Even though the reported SCD incidence rate is low in WPW syndrome, our data demonstrated an increased association of death within the AF/AFL cohort. Physicians should consider early risk stratification and ablation to prevent potential recurrent malignant arrhythmia leading to death.1-5,8,9,12,19,20

 

 

Limitations

As a retrospective study and without access to the National Death Index, we were unable to determine the exact cause or events leading to death and instead utilized all-cause mortality data. Subsequently, our observations may only demonstrate association, rather than causality, between AF/AFL and death in patients with WPW syndrome. Additionally, we could not distinguish between AF and AFL as the arrhythmia leading to death. However, since overall survivability was the outcome of interest, our adjusted HR models were still able to demonstrate the increased association of the composite outcome and death within an AF/AFL cohort.

Although a large cohort was analyzed, due to the constraints of utilizing diagnostic codes to determine study outcomes, we could not distinguish between symptomatic and asymptomatic patients, nor how they were managed prior to the outcome event. However, as recent literature demonstrates, updated predictors of malignant arrhythmia and decisions for early EPT are similar for both symptomatic and asymptomatic patients and should be driven by the intrinsic electrophysiologic properties of the accessory pathway, rather than symptomatology;thus, our inability to discern this should have negligible consequence in determining when to perform risk stratification and ablation.1

MHS eligible patients have direct access to care; the generalizability of our data may not necessarily correspond to a community population with lower socioeconomic status (we did adjust for military sponsor rank which has been used as a proxy), reduced access to care, or uninsured individuals. However, the prevalence of WPW syndrome within our cohort was comparable to the general population, 0.4% vs 0.1%-0.3%, respectively.13,14,19 Similarly, the incidence of AF within our population was comparable to the general population, 15% vs 16%-26%, respectively.23 These similar data points suggest our results may apply beyond MHS patients.

CONCLUSIONS

Patients with WPW syndrome and AF/AFL have a higher association with adverse cardiac outcomes and death. Despite previously reported low SCD incidence rates in this population, our study demonstrates the increased association of mortality in an AF/AFL cohort. The limitations of utilizing all-cause mortality data necessitate further investigation into the etiology behind the deaths in our study population. Since ventricular pre-excitation can predispose patients to AF and potentially lead to malignant arrhythmia and SCD, understanding the cause of mortality will allow physicians to determine the appropriate monitoring and intervention strategies to improve outcomes in this population. Our results suggest consideration for more aggressive EPT screening and ablation recommendations in patients with WPW syndrome may be warranted.

Wolff-Parkinson-White (WPW) syndrome is characterized by the presence of ≥ 1 accessory pathways and the development of both recurrent paroxysmal atrial fibrillation (AF) and supraventricular tachycardia that can lead to further malignant arrhythmias resulting in sudden cardiac death (SCD).1-7 Historically, incidental, ventricular pre-excitation on electrocardiogram has conferred a relatively low SCD risk in adults; however, newer WPW syndrome data suggest the endpoint may not be as benign as previously thought.7 The current literature has defined atrioventricular reentrant tachycardia triggering AF, rather than symptoms, as an independent risk factor for malignant arrhythmias. Still, long-term data detailing the association of AF with serious cardiac events and death in patients with WPW syndrome are still limited.1-7

While previous guidelines for the treatment of WPW syndrome only recommended routine electrophysiology testing (EPT) with liberal catheter ablation for symptomatic individuals, the 2015 American College of Cardiology/American Heart Association/Heart Rhythm Society guidelines now suggest its potential benefit for risk stratification in the asymptomatic population.8-12 Given the limited existing data, more long-term studies are needed to corroborate the latest EPT recommendations before routinely applying them in practice. Furthermore, since concomitant AF can lead to adverse cardiac outcomes in patients with WPW syndrome, additional data evaluating this association are also necessary. In this study, we aimed to determine the impact of atrial fibrillation and/or flutter (AF/AFL) on adverse cardiac outcomes and mortality in patients with WPW syndrome.

METHODS

This study used data from the Military Health System (MHS) Database Repository. The MHS is one of the largest health care systems in the country and includes information on about 10 million active duty and retired military service members and their families (51% male; 49% female).13,14 Data were fully anonymized and complied in accordance with federal and state laws, including the Health Insurance Portability and Accountability Act of 1996. The Naval Medical Center Portsmouth Institutional Review Board approved this study.

 

Study Design

This retrospective, observational cohort study identified MHS patients with WPW syndrome from January 1, 2014, to December 31, 2019. Patients were included if they had ≥ 2 International Classification of Diseases, Ninth Revision (ICD-9) or International Classification of Diseases, Tenth Revision (ICD-10) diagnosis codes for WPW syndrome (ICD-9, 426.7; ICD-10, I45.6) on separate dates; were aged ≥ 18 years at index date; and had ≥ 1 year of continuous eligibility prior to the index date (enrollment gaps ≤ 30 days were considered continuous). Patients were then divided into 2 subgroups by the presence or absence of AF/AFL using diagnostic codes. Patients were excluded if they had evidence of an implantable cardioverter-defibrillator, permanent pacemaker or were missing age or sex data. Patients were followed from index date until the first occurrence of the outcome of interest, MHS disenrollment, or the end of the study period.

Cardiac composite outcomes comprised of sudden cardiac arrest (SCA), ventricular fibrillation (VF), ventricular tachycardia and death, as well as death specifically, were the outcomes of interest and assessed after index date using ICD-9 and ICD-10 codes. Death was defined as all-cause mortality. Time to event was calculated based on the date of the initial component from the composite outcome and date of death specifically for mortality. Those not experiencing an outcome were followed until MHS disenrollment or the end of the study period.

Various patient characteristics were assessed at index including age, sex, military sponsor (the patient’s active or retired duty member through which their dependent receives TRICARE benefits) rank and branch, geographic region, type of US Department of Defense beneficiary, and index year. Clinical characteristics were assessed over a 1-year baseline period prior to index date and included the number of cardiologist and clinical visits for WPW syndrome, Charlson Comorbidity Index (CCI) scores calculated from diagnostic codes outlined in the Quan coding method, and preindex time.15 Comorbidities were assessed at baseline and defined as having ≥ 1 ICD-9 or ICD-10 code for a corresponding condition within 1 year prior to index.

 

 

Statistical Analysis

Baseline characteristics were assessed and descriptive statistics for categorical and continuous variables were presented accordingly. To assess bivariate association with exposure, χ2 tests were used to compare categorical variables, while t tests were used to compare continuous variables by exposure status. Incidence proportions and rates were reported for each outcome of interest. Kaplan-Meier curves were constructed to assess the bivariate association between exposure and study outcomes. Cox proportional hazard modeling was performed to estimate the association between AF/AFL and time to each of the outcomes. Multiple models were designed to assess cardiac and metabolic covariates, in addition to baseline characteristics. This included a base model adjusted for age, sex, military sponsor rank and branch, geographic region, and duty status.

Additional models adjusted for cardiac and metabolic confounders and CCI score. A comprehensive model included the base, cardiac, and metabolic covariates. Multicollinearity between covariates was assessed. Variables with a variance inflation factor > 4 or a tolerance level < 0.1 were added to the models. Cox proportional hazard models were used to estimate the unadjusted and adjusted hazard ratios (HRs) and 95% CIs of the association between AF/AFL and the study outcomes. Data were analyzed using SAS, version 9.4 for Windows.

RESULTS

table_1.png

From 2014 through 2019, 35,539 patients with WPW syndrome were identified in the MHS, 5291 had AF/AFL (14.9%); 19,961 were female (56.2%), the mean (SD) age was 62.9 (18.0) years, and 11,742 were aged ≥ 75 years (33.0%) (Table 1).

figure_1.png

There were 4121 (11.6%), 322 (0.9%), and 848 (2.4%) patients with AF, AFL, and both arrhythmias, respectively. The mean (SD) number of cardiology visits was 3.9 (3.0). The mean (SD) baseline CCI score for the AF/AFL subgroup was 5.9 (3.5) vs 3.7 (2.2) for the non-AF/AFL subgroup (P < .001). The most prevalent comorbid conditions were hypertension, hyperlipidemia, chronic obstructive pulmonary disease, and diabetes (P < .001) (Figure 1).

 

Composite Outcomes

figure_2.png

In the overall cohort, during a mean (SD) follow-up time of 3.4 (2.0) years comprising 119,682 total person-years, the components of the composite outcome occurred 6506 times with an incidence rate of 5.44 per 100 person-years. Ventricular tachycardia was the most common event, occurring 3281 times with an incidence rate of 2.74 per 100 person-years. SCA and VF occurred 841 and 135 times with incidence rates of 0.70 and 0.11 per 100 person-years, respectively. Death was the initial event 2249 times with an incidence rate of 1.88 per 100 person-years. Figure 2 shows the Kaplan-Meier curve of cardiac composite outcome by AF/AFL status.

table_2.png

The subgroup with AF/AFL comprised 17,412 total person-years and 1424 cardiac composite incidences compared with 102,270 person years and 5082 incidences in the no AF/AFL group (Table 2). Comparing AF/AFL vs no AF/AFL incidence rates were 8.18 vs 4.97 per 100 person-years, respectively (P < .001). SCA and VF occurred 233 and 38 times and respectively had incidence rates of 1.34 and 0.22 per 100 person-years in the AF/AFL group vs 0.59 and 0.09 per 100 person-years in the no AF/AFL group (P < .001). There were 549 deaths and a 3.15 per 100 person-years incidence rate in the AF/AFL group vs 1700 deaths and a 1.66 incidence rate in the no AF/AFL group (P < .001).

table_3.png

The HR for the composite outcome in the base model was 1.33 (95% CI, 1.26-1.42, P < .001) (Table 3). The association between AF/AFL and the composite outcome remained significant after adjusting for additional metabolic and cardiac covariates. The HRs for the metabolic and cardiac models were 1.30 (95% CI, 1.23-1.38, P < .001) and 1.11 (95% CI, 1.05-1.18, P < .001), respectively. After adjusting for the full model, the HR was 1.12 (95% CI, 1.05-1.19, P < .001).

 

 

Mortality

figure_3.png

Over the 6-year study period, there was a lower survival probability for patients with AF/AFL. In the overall cohort, during a mean (SD) follow-up time of 3.7 (1.9) years comprising 129,391 total person-years, there were 3130 (8.8%) deaths and an incidence rate of 2.42 per 100 person-years. Death occurred 786 times with a 4.09 incidence rate per 100 person-years in the AF/AFL vs 2344 deaths and a 2.13 incidence rate per 100 person-years in the no AF/AFL group (P < .001). In the non-AF/AFL subgroup, death occurred 2344 times during a mean (SD) follow-up of 3.7 (1.9) years comprising 110,151 total person-years. Figure 3 shows the Kaplan-Meier curve of mortality by AF/AFL status.

table_4.png

After adjusting for the base, metabolic and cardiac covariates, the HRs for mortality were 1.45 (95% CI, 1.33-1.57, P < .001), 1.40 (95% CI, 1.29-1.51, P < .001) and 1.15 (95% CI, 1.06-1.25, P = .001), respectively (Table 4). The HR after adjusting for the full model was 1.16 (95% CI, 1.07-1.26, P < .001).

DISCUSSION

In this large retrospective cohort study, patients with WPW syndrome and comorbid AF/AFL had a significantly higher association with the cardiac composite outcome and death during a 3-year follow-up period when compared with patients without AF/AFL. After adjusting for confounding variables, the AF/AFL subgroup maintained a 12% and 16% higher association with the composite outcome and mortality, respectively. There was minimal difference in confounding effects between demographic data and metabolic profiles, suggesting one may serve as a proxy for the other.

To our knowledge, this is the largest WPW syndrome cohort study evaluating cardiac outcomes and mortality to date. Although previous research has shown the relatively low and mostly anecdotal SCD incidence within this population,our results demonstrate a higher association of adverse cardiac outcomes and death in an AF/AFL subgroup.16-18 Notably, in this study the AF/AFL cohort was older and had higher CCI scores than their counterparts (P < .001), thus inferring an inherently greater degree of morbidity and 10-year mortality risk. Our study is also unique in that the mean patient age was significantly older than previously reported (63 vs 27 years), which may suggest a longer living history of both ventricular pre-excitation and the comorbidities outlined in Figure 1.19 Given these age discrepancies, it is possible that our overall study population was still relatively low risk and that not all reported deaths were necessarily related to WPW syndrome. Despite these assumptions, when comparing the WPW syndrome subgroups, we still found the AF/AFL cohort maintained a statistically significant higher association with the 2 study outcomes, even after adjusting for the greater presence of comorbidities. This suggests that the presence of AF/AFL may still portend a worse prognosis in patients with WPW syndrome.

Although the association of AF and development of VF in patients with WPW syndrome—due to rapid conduction over the accessory pathway(s)—was first reported > 40 years ago, there has still been few large, long-term data studies exploring mortality in this cohort.19-25 Furthermore, even though the current literature attributes the development of AF with the electrophysiologic properties of the accessory pathway, as well as intrinsic atrial architecture and muscle vulnerability, there is still equivocal consensus regarding EPT screening and ablation indications for asymptomatic patients with WPW syndrome.26-28 Notably, Pappone and colleagues demonstrated the potential benefit of liberal ablation indications for asymptomatic patients, arguing that the intrinsic electrophysiologic properties of the accessory pathway—ie, short accessory-pathway antegrade effective refractory period, inducibility of atrioventricular reentrant tachycardia triggering AF, and multiple accessory pathway—rather than symptoms, are independent predictors of developing malignant arrhythmia.1-5

These findings contradict those reported by Obeyesekere and colleagues, who concluded that the low SCD incidence rates in patients with WPW syndrome precluded routine invasive screening.19,28 They argued that Pappone and colleagues used malignant arrhythmia as a surrogate marker for death, and that the positive predictive value of a short accessory-pathway antegrade effective refractory period for developing malignant arrhythmia was lower than reported (15% vs 82%, respectively) and that its negative predictive value was 100%.1,19,28 Given these conflicting recommendations, we hope our data elucidates the higher association of adverse outcomes and support considerations for more intensive EPT indications in patients with WPW syndrome.

While our study does not report SCD incidence, it does provide robust and reliable mortality data that suggests a greater association of death within an AF/AFL subgroup. Our findings would support more liberal EPT recommendations in patients with WPW syndrome.1-5,8,9 In this study, the SCA incidence rate was more than double the rate in the AF/AFL cohort (P < .001) and is commonly the initial presenting event in WPW syndrome.9 Even though the reported SCD incidence rate is low in WPW syndrome, our data demonstrated an increased association of death within the AF/AFL cohort. Physicians should consider early risk stratification and ablation to prevent potential recurrent malignant arrhythmia leading to death.1-5,8,9,12,19,20

 

 

Limitations

As a retrospective study and without access to the National Death Index, we were unable to determine the exact cause or events leading to death and instead utilized all-cause mortality data. Subsequently, our observations may only demonstrate association, rather than causality, between AF/AFL and death in patients with WPW syndrome. Additionally, we could not distinguish between AF and AFL as the arrhythmia leading to death. However, since overall survivability was the outcome of interest, our adjusted HR models were still able to demonstrate the increased association of the composite outcome and death within an AF/AFL cohort.

Although a large cohort was analyzed, due to the constraints of utilizing diagnostic codes to determine study outcomes, we could not distinguish between symptomatic and asymptomatic patients, nor how they were managed prior to the outcome event. However, as recent literature demonstrates, updated predictors of malignant arrhythmia and decisions for early EPT are similar for both symptomatic and asymptomatic patients and should be driven by the intrinsic electrophysiologic properties of the accessory pathway, rather than symptomatology;thus, our inability to discern this should have negligible consequence in determining when to perform risk stratification and ablation.1

MHS eligible patients have direct access to care; the generalizability of our data may not necessarily correspond to a community population with lower socioeconomic status (we did adjust for military sponsor rank which has been used as a proxy), reduced access to care, or uninsured individuals. However, the prevalence of WPW syndrome within our cohort was comparable to the general population, 0.4% vs 0.1%-0.3%, respectively.13,14,19 Similarly, the incidence of AF within our population was comparable to the general population, 15% vs 16%-26%, respectively.23 These similar data points suggest our results may apply beyond MHS patients.

CONCLUSIONS

Patients with WPW syndrome and AF/AFL have a higher association with adverse cardiac outcomes and death. Despite previously reported low SCD incidence rates in this population, our study demonstrates the increased association of mortality in an AF/AFL cohort. The limitations of utilizing all-cause mortality data necessitate further investigation into the etiology behind the deaths in our study population. Since ventricular pre-excitation can predispose patients to AF and potentially lead to malignant arrhythmia and SCD, understanding the cause of mortality will allow physicians to determine the appropriate monitoring and intervention strategies to improve outcomes in this population. Our results suggest consideration for more aggressive EPT screening and ablation recommendations in patients with WPW syndrome may be warranted.

References

1. Pappone C, Vicedomini G, Manguso F, et al. The natural history of WPW syndrome. Eur Heart J Suppl. 2015; 17 (Supplement A):A8-A11.doi:10.1093/eurheartj/suv004

2. Pappone C, Vicedomini G, Manguso F, et al. Risk of malignant arrhythmias in initially symptomatic patients with Wolff-Parkinson-White syndrome: results of a prospective long-term electrophysiological follow-up study. Circulation. 2012;125(5):661-668. doi:10.1161/CIRCULATIONAHA.111.065722

3. Pappone C, Santinelli V, Rosanio S, et al. Usefulness of invasive electrophysiologic testing to stratify the risk of arrhythmic events in asymptomatic patients with Wolff-Parkinson-White pattern: results from a large prospective long-term follow-up study. J Am Coll Cardiol. 2003;41(2):239-244. doi:10.1016/s0735-1097(02)02706-7

4. Pappone C, Vicedomini G, Manguso F, et al. Wolff-Parkinson-White syndrome in the era of catheter ablation: insights from a registry study of 2169 patients. Circulation. 2014;130(10):811-819. doi:10.1161/CIRCULATIONAHA.114.011154

5. Pappone C, Santinelli V, Manguso F, et al. A randomized study of prophylactic catheter ablation in asymptomatic patients with the Wolff-Parkinson-White syndrome. N Engl J Med. 2003;349(19):1803-1811. doi:10.1056/NEJMoa035345

6. Santinelli V, Radinovic A, Manguso F, et al. Asymptomatic ventricular preexcitation: a long-term prospective follow-up study of 293 adult patients. Circ Arrhythm Electrophysiol. 2009;2(2):102-107. doi:10.1161/CIRCEP.108.827550

7. Santinelli V, Radinovic A, Manguso F, et al. The natural history of asymptomatic ventricular pre-excitation a long-term prospective follow-up study of 184 asymptomatic children. J Am Coll Cardiol. 2009;53(3):275-280. doi:10.1016/j.jacc.2008.09.037

8. Al-Khatib SM, Arshad A, Balk EM, et al. Risk Stratification for Arrhythmic Events in Patients With Asymptomatic Pre-Excitation: A Systematic Review for the 2015 ACC/AHA/HRS Guideline for the Management of Adult Patients With Supraventricular Tachycardia: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. J Am Coll Cardiol. 2016;67(13):1624-1638. doi:10.1016/j.jacc.2015.09.018

9. Blomström-Lundqvist C, Scheinman MM, Aliot EM, et al. ACC/AHA/ESC guidelines for the management of patients with supraventricular arrhythmias--executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the European Society of Cardiology Committee for Practice Guidelines (Writing Committee to Develop Guidelines for the Management of Patients With Supraventricular Arrhythmias). Circulation. 2003;108(15):1871-1909.doi:10.1161/01.CIR.0000091380.04100.84

10. Pediatric and Congenital Electrophysiology Society (PACES); Heart Rhythm Society (HRS); American College of Cardiology Foundation (ACCF); PACES/HRS expert consensus statement on the management of the asymptomatic young patient with a Wolff-Parkinson-White (WPW, ventricular preexcitation) electrocardiographic pattern: developed in partnership between the Pediatric and Congenital Electrophysiology Society (PACES) and the Heart Rhythm Society (HRS). Endorsed by the governing bodies of PACES, HRS, the American College of Cardiology Foundation (ACCF), the American Heart Association (AHA), the American Academy of Pediatrics (AAP), and the Canadian Heart Rhythm Society (CHRS). Heart Rhythm. 2012;9(6):1006-1024. doi:10.1016/j.hrthm.2012.03.050

11. Cohen M, Triedman J. Guidelines for management of asymptomatic ventricular pre-excitation: brave new world or Pandora’s box?. Circ Arrhythm Electrophysiol. 2014;7(2):187-189. doi:10.1161/CIRCEP.114.001528

12. Svendsen JH, Dagres N, Dobreanu D, et al. Current strategy for treatment of patients with Wolff-Parkinson-White syndrome and asymptomatic preexcitation in Europe: European Heart Rhythm Association survey. Europace. 2013;15(5):750-753. doi:10.1093/europace/eut094

13. Gimbel RW, Pangaro L, Barbour G. America’s “undiscovered” laboratory for health services research. Med Care. 2010;48(8):751-756. doi:10.1097/MLR.0b013e3181e35be8

14. Dorrance KA, Ramchandani S, Neil N, Fisher H. Leveraging the military health system as a laboratory for health care reform. Mil Med. 2013;178(2):142-145. doi:10.7205/milmed-d-12-00168

15. Quan H, Sundararajan V, Halfon P, et al. Coding algorithms for defining comorbidities in ICD-9-CM and ICD-10 administrative data. Med Care. 2005;43(11):1130-1139. doi:10.1097/01.mlr.0000182534.19832.83

16. Finocchiaro G, Papadakis M, Behr ER, Sharma S, Sheppard M. Sudden Cardiac Death in Pre-Excitation and Wolff-Parkinson-White: Demographic and Clinical Features. J Am Coll Cardiol. 2017;69(12):1644-1645. doi:10.1016/j.jacc.2017.01.023

17. Munger TM, Packer DL, Hammill SC, et al. A population study of the natural history of Wolff-Parkinson-White syndrome in Olmsted County, Minnesota, 1953-1989. Circulation. 1993;87(3):866-873. doi:10.1161/01.cir.87.3.866

18. Fitzsimmons PJ, McWhirter PD, Peterson DW, Kruyer WB. The natural history of Wolff-Parkinson-White syndrome in 228 military aviators: a long-term follow-up of 22 years. Am Heart J. 2001;142(3):530-536. doi:10.1067/mhj.2001.117779

19. Obeyesekere MN, Leong-Sit P, Massel D, et al. Risk of arrhythmia and sudden death in patients with asymptomatic preexcitation: a meta-analysis. Circulation. 2012;125(19):2308-2315. doi:10.1161/CIRCULATIONAHA.111.055350

20. Waspe LE, Brodman R, Kim SG, Fisher JD. Susceptibility to atrial fibrillation and ventricular tachyarrhythmia in the Wolff-Parkinson-White syndrome: role of the accessory pathway. Am Heart J. 1986;112(6):1141-1152. doi:10.1016/0002-8703(86)90342-x

21. Pietersen AH, Andersen ED, Sandøe E. Atrial fibrillation in the Wolff-Parkinson-White syndrome. Am J Cardiol. 1992;70(5):38A-43A. doi:10.1016/0002-9149(92)91076-g

22. Della Bella P, Brugada P, Talajic M, et al. Atrial fibrillation in patients with an accessory pathway: importance of the conduction properties of the accessory pathway. J Am Coll Cardiol. 1991;17(6):1352-1356. doi:10.1016/s0735-1097(10)80146-9

23. Fujimura O, Klein GJ, Yee R, Sharma AD. Mode of onset of atrial fibrillation in the Wolff-Parkinson-White syndrome: how important is the accessory pathway?. J Am Coll Cardiol. 1990;15(5):1082-1086. doi:10.1016/0735-1097(90)90244-j

24. Montoya PT, Brugada P, Smeets J, et al. Ventricular fibrillation in the Wolff-Parkinson-White syndrome. Eur Heart J. 1991;12(2):144-150. doi:10.1093/oxfordjournals.eurheartj.a059860

25. Klein GJ, Bashore TM, Sellers TD, Pritchett EL, Smith WM, Gallagher JJ. Ventricular fibrillation in the Wolff-Parkinson-White syndrome. N Engl J Med. 1979;301(20):1080-1085. doi:10.1056/NEJM197911153012003

26. Centurion OA. Atrial Fibrillation in the Wolff-Parkinson-White Syndrome. J Atr Fibrillation. 2011;4(1):287. Published 2011 May 4. doi:10.4022/jafib.287

27. Song C, Guo Y, Zheng X, et al. Prognostic Significance and Risk of Atrial Fibrillation of Wolff-Parkinson-White Syndrome in Patients With Hypertrophic Cardiomyopathy. Am J Cardiol. 2018;122(9):1546-1550. doi:10.1016/j.amjcard.2018.07.021

28. Obeyesekere M, Gula LJ, Skanes AC, Leong-Sit P, Klein GJ. Risk of sudden death in Wolff-Parkinson-White syndrome: how high is the risk?. Circulation. 2012;125(5):659-660. doi:10.1161/CIRCULATIONAHA.111.085159

References

1. Pappone C, Vicedomini G, Manguso F, et al. The natural history of WPW syndrome. Eur Heart J Suppl. 2015; 17 (Supplement A):A8-A11.doi:10.1093/eurheartj/suv004

2. Pappone C, Vicedomini G, Manguso F, et al. Risk of malignant arrhythmias in initially symptomatic patients with Wolff-Parkinson-White syndrome: results of a prospective long-term electrophysiological follow-up study. Circulation. 2012;125(5):661-668. doi:10.1161/CIRCULATIONAHA.111.065722

3. Pappone C, Santinelli V, Rosanio S, et al. Usefulness of invasive electrophysiologic testing to stratify the risk of arrhythmic events in asymptomatic patients with Wolff-Parkinson-White pattern: results from a large prospective long-term follow-up study. J Am Coll Cardiol. 2003;41(2):239-244. doi:10.1016/s0735-1097(02)02706-7

4. Pappone C, Vicedomini G, Manguso F, et al. Wolff-Parkinson-White syndrome in the era of catheter ablation: insights from a registry study of 2169 patients. Circulation. 2014;130(10):811-819. doi:10.1161/CIRCULATIONAHA.114.011154

5. Pappone C, Santinelli V, Manguso F, et al. A randomized study of prophylactic catheter ablation in asymptomatic patients with the Wolff-Parkinson-White syndrome. N Engl J Med. 2003;349(19):1803-1811. doi:10.1056/NEJMoa035345

6. Santinelli V, Radinovic A, Manguso F, et al. Asymptomatic ventricular preexcitation: a long-term prospective follow-up study of 293 adult patients. Circ Arrhythm Electrophysiol. 2009;2(2):102-107. doi:10.1161/CIRCEP.108.827550

7. Santinelli V, Radinovic A, Manguso F, et al. The natural history of asymptomatic ventricular pre-excitation a long-term prospective follow-up study of 184 asymptomatic children. J Am Coll Cardiol. 2009;53(3):275-280. doi:10.1016/j.jacc.2008.09.037

8. Al-Khatib SM, Arshad A, Balk EM, et al. Risk Stratification for Arrhythmic Events in Patients With Asymptomatic Pre-Excitation: A Systematic Review for the 2015 ACC/AHA/HRS Guideline for the Management of Adult Patients With Supraventricular Tachycardia: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. J Am Coll Cardiol. 2016;67(13):1624-1638. doi:10.1016/j.jacc.2015.09.018

9. Blomström-Lundqvist C, Scheinman MM, Aliot EM, et al. ACC/AHA/ESC guidelines for the management of patients with supraventricular arrhythmias--executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the European Society of Cardiology Committee for Practice Guidelines (Writing Committee to Develop Guidelines for the Management of Patients With Supraventricular Arrhythmias). Circulation. 2003;108(15):1871-1909.doi:10.1161/01.CIR.0000091380.04100.84

10. Pediatric and Congenital Electrophysiology Society (PACES); Heart Rhythm Society (HRS); American College of Cardiology Foundation (ACCF); PACES/HRS expert consensus statement on the management of the asymptomatic young patient with a Wolff-Parkinson-White (WPW, ventricular preexcitation) electrocardiographic pattern: developed in partnership between the Pediatric and Congenital Electrophysiology Society (PACES) and the Heart Rhythm Society (HRS). Endorsed by the governing bodies of PACES, HRS, the American College of Cardiology Foundation (ACCF), the American Heart Association (AHA), the American Academy of Pediatrics (AAP), and the Canadian Heart Rhythm Society (CHRS). Heart Rhythm. 2012;9(6):1006-1024. doi:10.1016/j.hrthm.2012.03.050

11. Cohen M, Triedman J. Guidelines for management of asymptomatic ventricular pre-excitation: brave new world or Pandora’s box?. Circ Arrhythm Electrophysiol. 2014;7(2):187-189. doi:10.1161/CIRCEP.114.001528

12. Svendsen JH, Dagres N, Dobreanu D, et al. Current strategy for treatment of patients with Wolff-Parkinson-White syndrome and asymptomatic preexcitation in Europe: European Heart Rhythm Association survey. Europace. 2013;15(5):750-753. doi:10.1093/europace/eut094

13. Gimbel RW, Pangaro L, Barbour G. America’s “undiscovered” laboratory for health services research. Med Care. 2010;48(8):751-756. doi:10.1097/MLR.0b013e3181e35be8

14. Dorrance KA, Ramchandani S, Neil N, Fisher H. Leveraging the military health system as a laboratory for health care reform. Mil Med. 2013;178(2):142-145. doi:10.7205/milmed-d-12-00168

15. Quan H, Sundararajan V, Halfon P, et al. Coding algorithms for defining comorbidities in ICD-9-CM and ICD-10 administrative data. Med Care. 2005;43(11):1130-1139. doi:10.1097/01.mlr.0000182534.19832.83

16. Finocchiaro G, Papadakis M, Behr ER, Sharma S, Sheppard M. Sudden Cardiac Death in Pre-Excitation and Wolff-Parkinson-White: Demographic and Clinical Features. J Am Coll Cardiol. 2017;69(12):1644-1645. doi:10.1016/j.jacc.2017.01.023

17. Munger TM, Packer DL, Hammill SC, et al. A population study of the natural history of Wolff-Parkinson-White syndrome in Olmsted County, Minnesota, 1953-1989. Circulation. 1993;87(3):866-873. doi:10.1161/01.cir.87.3.866

18. Fitzsimmons PJ, McWhirter PD, Peterson DW, Kruyer WB. The natural history of Wolff-Parkinson-White syndrome in 228 military aviators: a long-term follow-up of 22 years. Am Heart J. 2001;142(3):530-536. doi:10.1067/mhj.2001.117779

19. Obeyesekere MN, Leong-Sit P, Massel D, et al. Risk of arrhythmia and sudden death in patients with asymptomatic preexcitation: a meta-analysis. Circulation. 2012;125(19):2308-2315. doi:10.1161/CIRCULATIONAHA.111.055350

20. Waspe LE, Brodman R, Kim SG, Fisher JD. Susceptibility to atrial fibrillation and ventricular tachyarrhythmia in the Wolff-Parkinson-White syndrome: role of the accessory pathway. Am Heart J. 1986;112(6):1141-1152. doi:10.1016/0002-8703(86)90342-x

21. Pietersen AH, Andersen ED, Sandøe E. Atrial fibrillation in the Wolff-Parkinson-White syndrome. Am J Cardiol. 1992;70(5):38A-43A. doi:10.1016/0002-9149(92)91076-g

22. Della Bella P, Brugada P, Talajic M, et al. Atrial fibrillation in patients with an accessory pathway: importance of the conduction properties of the accessory pathway. J Am Coll Cardiol. 1991;17(6):1352-1356. doi:10.1016/s0735-1097(10)80146-9

23. Fujimura O, Klein GJ, Yee R, Sharma AD. Mode of onset of atrial fibrillation in the Wolff-Parkinson-White syndrome: how important is the accessory pathway?. J Am Coll Cardiol. 1990;15(5):1082-1086. doi:10.1016/0735-1097(90)90244-j

24. Montoya PT, Brugada P, Smeets J, et al. Ventricular fibrillation in the Wolff-Parkinson-White syndrome. Eur Heart J. 1991;12(2):144-150. doi:10.1093/oxfordjournals.eurheartj.a059860

25. Klein GJ, Bashore TM, Sellers TD, Pritchett EL, Smith WM, Gallagher JJ. Ventricular fibrillation in the Wolff-Parkinson-White syndrome. N Engl J Med. 1979;301(20):1080-1085. doi:10.1056/NEJM197911153012003

26. Centurion OA. Atrial Fibrillation in the Wolff-Parkinson-White Syndrome. J Atr Fibrillation. 2011;4(1):287. Published 2011 May 4. doi:10.4022/jafib.287

27. Song C, Guo Y, Zheng X, et al. Prognostic Significance and Risk of Atrial Fibrillation of Wolff-Parkinson-White Syndrome in Patients With Hypertrophic Cardiomyopathy. Am J Cardiol. 2018;122(9):1546-1550. doi:10.1016/j.amjcard.2018.07.021

28. Obeyesekere M, Gula LJ, Skanes AC, Leong-Sit P, Klein GJ. Risk of sudden death in Wolff-Parkinson-White syndrome: how high is the risk?. Circulation. 2012;125(5):659-660. doi:10.1161/CIRCULATIONAHA.111.085159

Issue
Federal Practitioner - 41(4)a
Issue
Federal Practitioner - 41(4)a
Page Number
108
Page Number
108
Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>0424 FED WPW</fileName> <TBEID>0C02F397.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F397</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname/> <articleType>1</articleType> <TBLocation>Copyfitting-FED</TBLocation> <QCDate/> <firstPublished>20240408T155519</firstPublished> <LastPublished>20240408T155519</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240408T155519</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline/> <bylineText>LCDR John C. Chin, MD, MC, USNa; CAPT Andrew H. Lin, MD, MC, USNa; Nicholas M. Sicignano, MPHb; Toni M. Rush, PhD, MPHb</bylineText> <bylineFull/> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>Wolff-Parkinson-White (WPW) syndrome is characterized by the presence of ≥ 1 accessory pathways and the development of both recurrent paroxysmal atrial fibrilla</metaDescription> <articlePDF/> <teaserImage/> <title>Association of Atrial Fibrillation and/or Flutter With Adverse Cardiac Outcomes and Mortality in Patients With Wolff-Parkinson-White Syndrome</title> <deck/> <eyebrow>Original Research</eyebrow> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>April</pubPubdateMonth> <pubPubdateDay/> <pubVolume>41</pubVolume> <pubNumber>4</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2951</CMSID> <CMSID>3639</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>FED</publicationCode> <pubIssueName>April 2024</pubIssueName> <pubArticleType>Feature Articles | 3639</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Feature | 2951<pubSubsection/></pubSection> </pubSections> <journalTitle>Fed Pract</journalTitle> <journalFullTitle>Federal Practitioner</journalFullTitle> <copyrightStatement>Copyright 2017 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">16</term> </publications> <sections> <term canonical="true">104</term> </sections> <topics> <term canonical="true">194</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Association of Atrial Fibrillation and/or Flutter With Adverse Cardiac Outcomes and Mortality in Patients With Wolff-Parkinson-White Syndrome</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><b>Background: </b>Wolff-Parkinson-White (WPW) syndrome is characterized by the presence of accessory pathways and development of potentially malignant arrhythmias that can lead to sudden cardiac death.<sup> </sup>We aimed to determine the impact of atrial fibrillation and/or flutter (AF/AFL) on adverse cardiac outcomes and mortality in patients with WPW syndrome.<b>Methods:</b> This study identified a cohort of Military Health System patients with WPW syndrome from January 1, 2014, to December 31, 2019. The cohort was divided into 2 subgroups by the presence or absence of AF/AFL. Cardiac composite outcome and mortality were assessed. Kaplan-Meier curves were constructed to assess the bivariate association between exposure and these 2 study outcomes. Cox proportional models were used to estimate the hazard ratios and 95% CIs associated with the cardiac composite outcome and mortality. <br/><br/><b>Results: </b>Of 35,539 patients included in the study, 19,961<b> </b>were female (56.2%), the mean (SD) age was 62.9 (18.0) years, and 5291 patients (14.9%) had AF/AFL. The cardiac composite outcome and mortality incidence rates per 100 person-years in the AF/AFL vs non-AF/AFL subgroups were 8.18 vs 4.90, and 4.09 vs 2.13, respectively (<i>P</i> &lt; .001). There were 3130 (8.8%) deaths. After adjusting for confounding variables, the AF/AFL subgroup maintained a 12% and 16% higher association with the composite outcome and mortality, respectively.<br/><br/><b>Conclusions: </b>Patients with WPW syndrome and AF/AFL have a higher association with adverse cardiac outcomes and death. Consideration for more aggressive electrophysiology screening and ablation strategies may be warranted in this population.</p> <p><span class="Drop">W</span>olff-Parkinson-White (WPW) syndrome is characterized by the presence of ≥ 1 accessory pathways and the development of both recurrent paroxysmal atrial fibrillation (AF) and supraventricular tachycardia that can lead to further malignant arrhythmias resulting in sudden cardiac death (SCD).<sup>1-7</sup> Historically, incidental, ventricular pre-excitation on electrocardiogram has conferred a relatively low SCD risk in adults; however, newer WPW syndrome data suggest the endpoint may not be as benign as previously thought.<sup>7</sup> The current literature has defined atrioventricular reentrant tachycardia triggering AF, rather than symptoms, as an independent risk factor for malignant arrhythmias. Still, long-term data detailing the association of AF with serious cardiac events and death in patients with WPW syndrome are still limited.<sup>1-7 </sup></p> <p>While previous guidelines for the treatment of WPW syndrome only recommended routine electrophysiology testing (EPT) with liberal catheter ablation for symptomatic individuals, the 2015 American College of Cardiology/American Heart Association/Heart Rhythm Society guidelines now suggest its potential benefit for risk stratification in the asymptomatic population.<sup>8-12</sup> Given the limited existing data, more long-term studies are needed to corroborate the latest EPT recommendations before routinely applying them in practice. Furthermore, since concomitant AF can lead to adverse cardiac outcomes in patients with WPW syndrome, additional data evaluating this association are also necessary. In this study, we aimed to determine the impact of atrial fibrillation and/or flutter (AF/AFL) on adverse cardiac outcomes and mortality in patients with WPW syndrome.</p> <h2>METHODS</h2> <p>This study used data from the Military Health System (MHS) Database Repository. The MHS is one of the largest health care systems in the country and includes information on about 10 million active duty and retired military service members and their families (51% male; 49% female).<sup>13,14</sup> Data were fully anonymized and complied in accordance with federal and state laws, including the Health Insurance Portability and Accountability Act of 1996. The Naval Medical Center Portsmouth Institutional Review Board approved this study. </p> <h3>Study Design</h3> <p>This retrospective, observational cohort study identified MHS patients with WPW syndrome from January 1, 2014, to December 31, 2019. Patients were included if they had ≥ 2 <i>International Classification of Diseases, Ninth Revision</i> (ICD-9) or <i>International Classification of Diseases, Tenth Revision</i> (ICD-10) diagnosis codes for WPW syndrome (ICD-9, 426.7; ICD-10, I45.6) on separate dates; were aged ≥ 18 years at index date; and had ≥ 1 year of continuous eligibility prior to the index date (enrollment gaps ≤ 30 days were considered continuous). Patients were then divided into 2 subgroups by the presence or absence of AF/AFL using diagnostic codes. Patients were excluded if they had evidence of an implantable cardioverter-defibrillator, permanent pacemaker or were missing age or sex data. Patients were followed from index date until the first occurrence of the outcome of interest, MHS disenrollment, or the end of the study period.</p> <p>Cardiac composite outcomes comprised of sudden cardiac arrest (SCA), ventricular fibrillation (VF), ventricular tachycardia and death, as well as death specifically, were the outcomes of interest and assessed after index date using ICD-9 and ICD-10 codes. Death was defined as all-cause mortality. Time to event was calculated based on the date of the initial component from the composite outcome and date of death specifically for mortality. Those not experiencing an outcome were followed until MHS disenrollment or the end of the study period.Various patient characteristics were assessed at index including age, sex, military sponsor (the patient’s active or retired duty member through which their dependent receives TRICARE benefits) rank and branch, geographic region, type of US Department of Defense beneficiary, and index year. Clinical characteristics were assessed over a 1-year baseline period prior to index date and included the number of cardiologist and clinical visits for WPW syndrome, Charlson Comorbidity Index (CCI) scores calculated from diagnostic codes outlined in the Quan coding method, and preindex time.<sup>15</sup> Comorbidities were assessed at baseline and defined as having ≥ 1 ICD-9 or ICD-10 code for a corresponding condition within 1 year prior to index.</p> <h3>Statistical Analysis</h3> <p>Baseline characteristics were assessed and descriptive statistics for categorical and continuous variables were presented accordingly. To assess bivariate association with exposure, <span class="Strong">χ</span><sup>2</sup> tests were used to compare categorical variables, while <i>t</i> tests were used to compare continuous variables by exposure status. Incidence proportions and rates were reported for each outcome of interest. Kaplan-Meier curves were constructed to assess the bivariate association between exposure and study outcomes. Cox proportional hazard modeling was performed to estimate the association between AF/AFL and time to each of the outcomes. Multiple models were designed to assess cardiac and metabolic covariates, in addition to baseline characteristics. This included a base model adjusted for age, sex, military sponsor rank and branch, geographic region, and duty status. </p> <p>Additional models adjusted for cardiac and metabolic confounders and CCI score. A comprehensive model included the base, cardiac, and metabolic covariates. Multicollinearity between covariates was assessed. Variables with a variance inflation factor &gt; 4 or a tolerance level &lt; 0.1 were added to the models. Cox proportional hazard models were used to estimate the unadjusted and adjusted hazard ratios (HRs) and 95% CIs of the association between AF/AFL and the study outcomes. Data were analyzed using SAS, version 9.4 for Windows. </p> <h2>RESULTS</h2> <p>From 2014 through 2019, 35,539 patients with WPW syndrome were identified in the MHS, 5291 had AF/AFL (14.9%); 19,961 were female (56.2%), the mean (SD) age was 62.9 (18.0) years, and 11,742 were aged ≥ 75 years (33.0%) (Table 1). </p> <p>There were 4121 (11.6%), 322 (0.9%), and 848 (2.4%) patients with AF, AFL, and both arrhythmias, respectively. The mean (SD) number of cardiology visits was 3.9 (3.0). The mean (SD) baseline CCI score for the AF/AFL subgroup was 5.9 (3.5) vs 3.7 (2.2) for the non-AF/AFL subgroup (<i>P</i> &lt; .001). The most prevalent comorbid conditions were hypertension, hyperlipidemia, chronic obstructive pulmonary disease, and diabetes (<i>P</i> &lt; .001) (Figure 1).</p> <h3>Composite Outcomes</h3> <p>In the overall cohort, during a mean (SD) follow-up time of 3.4 (2.0) years comprising 119,682 total person-years, the components of the composite outcome occurred 6506 times with an incidence rate of 5.44 per 100 person-years. Ventricular tachycardia was the most common event, occurring 3281 times with an incidence rate of 2.74 per 100 person-years. SCA and VF occurred 841 and 135 times with incidence rates of 0.70 and 0.11 per 100 person-years, respectively. Death was the initial event 2249 times with an incidence rate of 1.88 per 100 person-years. Figure 2 shows the Kaplan-Meier curve of cardiac composite outcome by AF/AFL status. </p> <p>The subgroup with AF/AFL comprised 17,412 total person-years and 1424 cardiac composite incidences compared with 102,270 person years and 5082 incidences in the no AF/AFL group (Table 2). Comparing AF/AFL vs no AF/AFL incidence rates were 8.18 vs 4.97 per 100 person-years, respectively (<i>P</i> &lt; .001). SCA and VF occurred 233 and 38 times and respectively had incidence rates of 1.34 and 0.22 per 100 person-years in the AF/AFL group vs 0.59 and 0.09 per 100 person-years in the no AF/AFL group (<i>P</i> &lt; .001). There were 549 deaths and a 3.15 per 100 person-years incidence rate in the AF/AFL group vs 1700 deaths and a 1.66 incidence rate in the no AF/AFL group (<i>P</i> &lt; .001). <br/><br/>The HR for the composite outcome in the base model was 1.33 (95% CI, 1.26-1.42, <i>P</i> &lt; .001) (Table 3). The association between AF/AFL and the composite outcome remained significant after adjusting for additional metabolic and cardiac covariates. The HRs for the metabolic and cardiac models were 1.30 (95% CI, 1.23-1.38, <i>P</i> &lt; .001) and 1.11 (95% CI, 1.05-1.18, <i>P</i> &lt; .001), respectively. After adjusting for the full model, the HR was 1.12 (95% CI, 1.05-1.19, <i>P</i> &lt; .001). </p> <h3>Mortality</h3> <p>Over the 6-year study period, there was a lower survival probability for patients with AF/AFL. In the overall cohort, during a mean (SD) follow-up time of 3.7 (1.9) years comprising 129,391 total person-years, there were 3130 (8.8%) deaths and an incidence rate of 2.42 per 100 person-years. Death occurred 786 times with a 4.09 incidence rate per 100 person-years in the AF/AFL vs 2344 deaths and a 2.13 incidence rate per 100 person-years in the no AF/AFL group (<i>P</i> &lt; .001). In the non-AF/AFL subgroup, death occurred 2344 times during a mean (SD) follow-up of 3.7 (1.9) years comprising 110,151 total person-years. Figure 3 shows the Kaplan-Meier curve of mortality by AF/AFL status. </p> <p>After adjusting for the base, metabolic and cardiac covariates, the HRs for mortality were 1.45 (95% CI, 1.33-1.57, <i>P</i> &lt; .001), 1.40 (95% CI, 1.29-1.51, <i>P</i> &lt; .001) and 1.15 (95% CI, 1.06-1.25, <i>P</i> = .001), respectively (Table 4). The HR after adjusting for the full model was 1.16 (95% CI, 1.07-1.26, <i>P</i> &lt; .001). </p> <h2>DISCUSSION</h2> <p>In this large retrospective cohort study, patients with WPW syndrome and comorbid AF/AFL had a significantly higher association with the cardiac composite outcome and death during a 3-year follow-up period when compared with patients without AF/AFL. After adjusting for confounding variables, the AF/AFL subgroup maintained a 12% and 16% higher association with the composite outcome and mortality, respectively. There was minimal difference in confounding effects between demographic data and metabolic profiles, suggesting one may serve as a proxy for the other. </p> <p>To our knowledge, this is the largest WPW syndrome cohort study evaluating cardiac outcomes and mortality to date. Although previous research has shown the relatively low and mostly anecdotal SCD incidence within this population,<sup> </sup>our results demonstrate a higher association of adverse cardiac outcomes and death in an AF/AFL subgroup.<sup>16-18</sup> Notably, in this study the AF/AFL cohort was older and had higher CCI scores than their counterparts (<i>P</i> &lt; .001), thus inferring an inherently greater degree of morbidity and 10-year mortality risk. Our study is also unique in that the mean patient age was significantly older than previously reported (63 vs 27 years), which may suggest a longer living history of both ventricular pre-excitation and the comorbidities outlined in Figure 1.<sup>19</sup> Given these age discrepancies, it is possible that our overall study population was still relatively low risk and that not all reported deaths were necessarily related to WPW syndrome. Despite these assumptions, when comparing the WPW syndrome subgroups, we still found the AF/AFL cohort maintained a statistically significant higher association with the 2 study outcomes, even after adjusting for the greater presence of comorbidities. This suggests that the presence of AF/AFL may still portend a worse prognosis in patients with WPW syndrome.<br/><br/>Although the association of AF and development of VF in patients with WPW syndrome—due to rapid conduction over the accessory pathway(s)—was first reported &gt; 40 years ago, there has still been few large, long-term data studies exploring mortality in this cohort.<sup>19-25</sup> Furthermore, even though the current literature attributes the development of AF with the electrophysiologic properties of the accessory pathway, as well as intrinsic atrial architecture and muscle vulnerability, there is still equivocal consensus regarding EPT screening and ablation indications for asymptomatic patients with WPW syndrome.<sup>26-28</sup> Notably, Pappone and colleagues demonstrated the potential benefit of liberal ablation indications for asymptomatic patients, arguing that the intrinsic electrophysiologic properties of the accessory pathway—ie, short accessory-pathway antegrade effective refractory period, inducibility of atrioventricular reentrant tachycardia triggering AF, and multiple accessory pathway—rather than symptoms, are independent predictors of developing malignant arrhythmia.<sup>1-5</sup><br/><br/>These findings contradict those reported by Obeyesekere and colleagues, who concluded that the low SCD incidence rates in patients with WPW syndrome precluded routine invasive screening.<sup>19,28</sup> They argued that Pappone and colleagues used malignant arrhythmia as a surrogate marker for death, and that the positive predictive value of a short accessory-pathway antegrade effective refractory period for developing malignant arrhythmia was lower than reported (15% vs 82%, respectively) and that its negative predictive value was 100%.<sup>1,19,28</sup> Given these conflicting recommendations, we hope our data elucidates the higher association of adverse outcomes and support considerations for more intensive EPT indications in patients with WPW syndrome. <br/><br/>While our study does not report SCD incidence, it does provide robust and reliable mortality data that suggests a greater association of death within an AF/AFL subgroup. Our findings would support more liberal EPT recommendations in patients with WPW syndrome.<sup>1-5,8,9</sup> In this study, the SCA incidence rate was more than double the rate in the AF/AFL cohort (<i>P</i> &lt; .001) and is commonly the initial presenting event in WPW syndrome.<sup>9 </sup>Even though the reported SCD incidence rate is low in WPW syndrome, our data demonstrated an increased association of death within the AF/AFL cohort. Physicians should consider early risk stratification and ablation to prevent potential recurrent malignant arrhythmia leading to death.<sup>1-5,8,9,12,19,20</sup></p> <h3>Limitations</h3> <p>As a retrospective study and without access to the National Death Index, we were unable to determine the exact cause or events leading to death and instead utilized all-cause mortality data. Subsequently, our observations may only demonstrate association, rather than causality, between AF/AFL and death in patients with WPW syndrome. Additionally, we could not distinguish between AF and AFL as the arrhythmia leading to death. However, since overall survivability was the outcome of interest, our adjusted HR models were still able to demonstrate the increased association of the composite outcome and death within an AF/AFL cohort. </p> <p>Although a large cohort was analyzed, due to the constraints of utilizing diagnostic codes to determine study outcomes, we could not distinguish between symptomatic and asymptomatic patients, nor how they were managed prior to the outcome event. However, as recent literature demonstrates, updated predictors of malignant arrhythmia and decisions for early EPT are similar for both symptomatic and asymptomatic patients and should be driven by the intrinsic electrophysiologic properties of the accessory pathway, rather than symptomatology;<sup> </sup>thus, our inability to discern this should have negligible consequence in determining when to perform risk stratification and ablation.<sup>1</sup><br/><br/>MHS eligible patients have direct access to care; the generalizability of our data may not necessarily correspond to a community population with lower socioeconomic status (we did adjust for military sponsor rank which has been used as a proxy), reduced access to care, or uninsured individuals. However, the prevalence of WPW syndrome within our cohort was comparable to the general population, 0.4% vs 0.1%-0.3%, respectively.<sup>13,14,19</sup> Similarly, the incidence of AF within our population was comparable to the general population, 15% vs 16%-26%, respectively.<sup>23</sup> These similar data points suggest our results may apply beyond MHS patients. </p> <h2>CONCLUSIONS</h2> <p>Patients with WPW syndrome and AF/AFL have a higher association with adverse cardiac outcomes and death. Despite previously reported low SCD incidence rates in this population, our study demonstrates the increased association of mortality in an AF/AFL cohort. The limitations of utilizing all-cause mortality data necessitate further investigation into the etiology behind the deaths in our study population. Since ventricular pre-excitation can predispose patients to AF and potentially lead to malignant arrhythmia and SCD, understanding the cause of mortality will allow physicians to determine the appropriate monitoring and intervention strategies to improve outcomes in this population. Our results suggest consideration for more aggressive EPT screening and ablation recommendations in patients with WPW syndrome may be warranted. </p> <p class="isub">Author affiliations</p> <p> <em><sup>a</sup>Naval Medical Center Portsmouth, Virginia<br/><br/><sup>b</sup>Health ResearchTx LLC, Trevose, Pennsylvania</em> </p> <p class="isub">Author disclosures </p> <p> <em>The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article. </em> </p> <p class="isub">Disclaimer </p> <p> <em>The opinions expressed herein are those of the authors and do not necessarily reflect those of <i>Federal Practitioner,</i> Frontline Medical Communications Inc., the US Government, or any of its agencies.</em> </p> <p class="isub">Ethics and consent</p> <p> <em>Research and data from this study were reviewed andapproved by the Naval Medical Center PortsmouthInstitutional Review Board.</em> </p> <h2>References</h2> <p class="reference"> 1. Pappone C, Vicedomini G, Manguso F, et al. The natural history of WPW syndrome. <i>Eur Heart J Suppl</i>. 2015; 17 (Supplement A):A8-A11.doi:10.1093/eurheartj/suv004 <br/><br/> 2. Pappone C, Vicedomini G, Manguso F, et al. Risk of malignant arrhythmias in initially symptomatic patients with Wolff-Parkinson-White syndrome: results of a prospective long-term electrophysiological follow-up study. <i>Circulation</i>. 2012;125(5):661-668. doi:10.1161/CIRCULATIONAHA.111.065722<br/><br/> 3. Pappone C, Santinelli V, Rosanio S, et al. Usefulness of invasive electrophysiologic testing to stratify the risk of arrhythmic events in asymptomatic patients with Wolff-Parkinson-White pattern: results from a large prospective long-term follow-up study. <i>J Am Coll Cardiol</i>. 2003;41(2):239-244. doi:10.1016/s0735-1097(02)02706-7<br/><br/> 4. Pappone C, Vicedomini G, Manguso F, et al. Wolff-Parkinson-White syndrome in the era of catheter ablation: insights from a registry study of 2169 patients. <i>Circulation</i>. 2014;130(10):811-819. doi:10.1161/CIRCULATIONAHA.114.011154<br/><br/> 5. Pappone C, Santinelli V, Manguso F, et al. A randomized study of prophylactic catheter ablation in asymptomatic patients with the Wolff-Parkinson-White syndrome. <i>N Engl J Med</i>. 2003;349(19):1803-1811. doi:10.1056/NEJMoa035345<br/><br/> 6. Santinelli V, Radinovic A, Manguso F, et al. Asymptomatic ventricular preexcitation: a long-term prospective follow-up study of 293 adult patients. <i>Circ Arrhythm Electrophysiol</i>. 2009;2(2):102-107. doi:10.1161/CIRCEP.108.827550<br/><br/> 7. Santinelli V, Radinovic A, Manguso F, et al. The natural history of asymptomatic ventricular pre-excitation a long-term prospective follow-up study of 184 asymptomatic children. <i>J Am Coll Cardiol</i>. 2009;53(3):275-280. doi:10.1016/j.jacc.2008.09.037<br/><br/> 8. Al-Khatib SM, Arshad A, Balk EM, et al. Risk Stratification for Arrhythmic Events in Patients With Asymptomatic Pre-Excitation: A Systematic Review for the 2015 ACC/AHA/HRS Guideline for the Management of Adult Patients With Supraventricular Tachycardia: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. <i>J Am Coll Cardiol</i>. 2016;67(13):1624-1638. doi:10.1016/j.jacc.2015.09.018<br/><br/> 9. Blomström-Lundqvist C, Scheinman MM, Aliot EM, et al. ACC/AHA/ESC guidelines for the management of patients with supraventricular arrhythmias--executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the European Society of Cardiology Committee for Practice Guidelines (Writing Committee to Develop Guidelines for the Management of Patients With Supraventricular Arrhythmias). <i>Circulation</i>. 2003;108(15):1871-1909.doi:10.1161/01.CIR.0000091380.04100.84<br/><br/>10. Pediatric and Congenital Electrophysiology Society (PACES); Heart Rhythm Society (HRS); American College of Cardiology Foundation (ACCF); PACES/HRS expert consensus statement on the management of the asymptomatic young patient with a Wolff-Parkinson-White (WPW, ventricular preexcitation) electrocardiographic pattern: developed in partnership between the Pediatric and Congenital Electrophysiology Society (PACES) and the Heart Rhythm Society (HRS). Endorsed by the governing bodies of PACES, HRS, the American College of Cardiology Foundation (ACCF), the American Heart Association (AHA), the American Academy of Pediatrics (AAP), and the Canadian Heart Rhythm Society (CHRS). <i>Heart Rhythm</i>. 2012;9(6):1006-1024. doi:10.1016/j.hrthm.2012.03.050<br/><br/>11. Cohen M, Triedman J. Guidelines for management of asymptomatic ventricular pre-excitation: brave new world or Pandora’s box?. <i>Circ Arrhythm Electrophysiol</i>. 2014;7(2):187-189. doi:10.1161/CIRCEP.114.001528<br/><br/>12. Svendsen JH, Dagres N, Dobreanu D, et al. Current strategy for treatment of patients with Wolff-Parkinson-White syndrome and asymptomatic preexcitation in Europe: European Heart Rhythm Association survey. <i>Europace</i>. 2013;15(5):750-753. doi:10.1093/europace/eut094<br/><br/>13. Gimbel RW, Pangaro L, Barbour G. America’s “undiscovered” laboratory for health services research. <i>Med Care</i>. 2010;48(8):751-756. doi:10.1097/MLR.0b013e3181e35be8<br/><br/>14. Dorrance KA, Ramchandani S, Neil N, Fisher H. Leveraging the military health system as a laboratory for health care reform. <i>Mil Med</i>. 2013;178(2):142-145. doi:10.7205/milmed-d-12-00168<br/><br/>15. Quan H, Sundararajan V, Halfon P, et al. Coding algorithms for defining comorbidities in ICD-9-CM and ICD-10 administrative data. <i>Med Care</i>. 2005;43(11):1130-1139. doi:10.1097/01.mlr.0000182534.19832.83<br/><br/>16. Finocchiaro G, Papadakis M, Behr ER, Sharma S, Sheppard M. Sudden Cardiac Death in Pre-Excitation and Wolff-Parkinson-White: Demographic and Clinical Features. <i>J Am Coll Cardiol</i>. 2017;69(12):1644-1645. doi:10.1016/j.jacc.2017.01.023<br/><br/>17. Munger TM, Packer DL, Hammill SC, et al. A population study of the natural history of Wolff-Parkinson-White syndrome in Olmsted County, Minnesota, 1953-1989. <i>Circulation</i>. 1993;87(3):866-873. doi:10.1161/01.cir.87.3.866<br/><br/>18. Fitzsimmons PJ, McWhirter PD, Peterson DW, Kruyer WB. The natural history of Wolff-Parkinson-White syndrome in 228 military aviators: a long-term follow-up of 22 years. <i>Am Heart J</i>. 2001;142(3):530-536. doi:10.1067/mhj.2001.117779<br/><br/>19. Obeyesekere MN, Leong-Sit P, Massel D, et al. Risk of arrhythmia and sudden death in patients with asymptomatic preexcitation: a meta-analysis. <i>Circulation</i>. 2012;125(19):2308-2315. doi:10.1161/CIRCULATIONAHA.111.055350<br/><br/>20. Waspe LE, Brodman R, Kim SG, Fisher JD. Susceptibility to atrial fibrillation and ventricular tachyarrhythmia in the Wolff-Parkinson-White syndrome: role of the accessory pathway. <i>Am Heart J</i>. 1986;112(6):1141-1152. doi:10.1016/0002-8703(86)90342-x<br/><br/>21. Pietersen AH, Andersen ED, Sandøe E. Atrial fibrillation in the Wolff-Parkinson-White syndrome. <i>Am J Cardiol</i>. 1992;70(5):38A-43A. doi:10.1016/0002-9149(92)91076-g<br/><br/>22. Della Bella P, Brugada P, Talajic M, et al. Atrial fibrillation in patients with an accessory pathway: importance of the conduction properties of the accessory pathway. <i>J Am Coll Cardiol</i>. 1991;17(6):1352-1356. doi:10.1016/s0735-1097(10)80146-9<br/><br/>23. Fujimura O, Klein GJ, Yee R, Sharma AD. Mode of onset of atrial fibrillation in the Wolff-Parkinson-White syndrome: how important is the accessory pathway?. <i>J Am Coll Cardiol</i>. 1990;15(5):1082-1086. doi:10.1016/0735-1097(90)90244-j<br/><br/>24. Montoya PT, Brugada P, Smeets J, et al. Ventricular fibrillation in the Wolff-Parkinson-White syndrome. <i>Eur Heart J</i>. 1991;12(2):144-150. doi:10.1093/oxfordjournals.eurheartj.a059860<br/><br/>25. Klein GJ, Bashore TM, Sellers TD, Pritchett EL, Smith WM, Gallagher JJ. Ventricular fibrillation in the Wolff-Parkinson-White syndrome. <i>N Engl J Med</i>. 1979;301(20):1080-1085. doi:10.1056/NEJM197911153012003<br/><br/>26. Centurion OA. Atrial Fibrillation in the Wolff-Parkinson-White Syndrome. <i>J Atr Fibrillation</i>. 2011;4(1):287. Published 2011 May 4. doi:10.4022/jafib.287<br/><br/>27. Song C, Guo Y, Zheng X, et al. Prognostic Significance and Risk of Atrial Fibrillation of Wolff-Parkinson-White Syndrome in Patients With Hypertrophic Cardiomyopathy. <i>Am J Cardiol</i>. 2018;122(9):1546-1550. doi:10.1016/j.amjcard.2018.07.021<br/><br/>28. Obeyesekere M, Gula LJ, Skanes AC, Leong-Sit P, Klein GJ. Risk of sudden death in Wolff-Parkinson-White syndrome: how high is the risk?. <i>Circulation</i>. 2012;125(5):659-660. doi:10.1161/CIRCULATIONAHA.111.085159</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

Nasal Cannula Dislodgement During Sleep in Veterans Receiving Long-term Oxygen Therapy for Hypoxemic Chronic Respiratory Failure

Article Type
Changed
Wed, 04/10/2024 - 16:42

The prevalence of chronic obstructive pulmonary disease (COPD) among male US veterans is higher than in the general population.1 Veterans with COPD have higher rates of comorbidities and increased respiratory-related and all-cause health care use, including the use of long-term oxygen therapy (LTOT).2-5 It has been well established that LTOT reduces all-cause mortality in patients with COPD and resting hypoxemic chronic respiratory failure (CRF) when used for ≥ 15 hours per day.6-8

Delivery of domiciliary LTOT entails placing a nasal cannula into both nostrils and loosely securing it around both ears throughout the wake-sleep cycle. Several veterans with hypoxemic CRF due to COPD at the Jesse Brown Veterans Affairs Medical Center (JBVAMC) in Chicago, Illinois, who were receiving LTOT reported nasal cannula dislodgement (NCD) while they slept. However, the clinical significance and impact of these repeated episodes on respiratory-related health care utilization, such as frequent COPD exacerbations with hospitalization, were not recognized. Moreover, we found no published reports or clinical practice guidelines alluding to similar events reported by patients with hypoxemic CRF due to COPD receiving LTOT either at home or in an acute care setting.8,9 Nonetheless, frequent COPD exacerbations are associated with increased hospital admissions and account for a large portion of health care costs attributed to COPD.10-13

The purpose of this study was to determine whether veterans with hypoxemic CRF due to COPD and receiving 24-hour LTOT at JBVAMC were experiencing NCD during sleep and, if so, its impact on their hospitalizations for COPD exacerbations.

METHODS

We reviewed electronic health records (EHRs) of veterans with hypoxemic CRF from COPD who received 24-hour LTOT administered through nasal cannula and were followed in the JBVAMC pulmonary outpatient clinic between February 1, 2022, and December 31, 2022. In each case, LTOT was prescribed by a board-certified pulmonologist based on Veterans Health Administration clinical practice guidelines.14 A licensed durable medical equipment company contracted by the JBVAMC delivered and established home oxygen equipment at each veteran’s residence.

Pertinent patient demographics, clinical and physiologic variables, and hospitalizations with length of JBVAMC stay for each physician-diagnosed COPD exacerbation in the preceding year from the date last seen in the clinic were abstracted from EHRs. Overall hospital cost, defined as a veteran overnight stay in either the medical intensive care unit (MICU) or a general acute medicine bed in a US Department of Veterans Affairs (VA) facility, was calculated for each hospitalization for physician-diagnosed COPD exacerbation using VA Managerial Cost Accounting System National Cost Extracts for inpatient encounters.15 We then contacted each veteran by telephone and asked whether they had experienced NCD and, if so, its weekly frequency ranging from once to nightly.

Data Analysis

Data were reported as mean (SD) where appropriate. The t test and Fisher exact test were used as indicated. P < .05 was considered statistically significant. The study protocol was determined to be exempt by the JBVAMC Institutional Review Board (Protocol #1725748).

 

 

RESULTS

table_1.png

During the study period, 75 patients with hypoxemic CRF from COPD received LTOT and were followed at the JBVAMC. No patients were hospitalized outside the JBVAMC for COPD exacerbation during this time frame. We also found no documentation in the EHRs indicating that the clinicians managing these patients at the JBVAMC inquired about NCD during sleep.

figure.png

Of the 75 patients, 66 (88%) responded to the telephone survey and 22 patients (33%) reported weekly episodes of NCD while they slept (median, 4 dislodgments per week). (Table 1). Eight patients (36%) reported nightly NCDs (Figure). All 66 respondents were male and 14 of 22 in the NCD group as well as 21 of 44 in the no NCD group were Black veterans. The mean age was similar in both groups: 71 years in the NCD group and 72 years in the no NCD group. There were no statistically significant differences in demographics, including prevalence of obstructive sleep apnea (OSA), supplemental oxygen flow rate, and duration of LTOT, or in pulmonary function test results between patients who did and did not experience NCD while sleeping (Table 2).

table_3.png
table_2.png

Ten of 22 patients (45%) with NCD and 9 of 44 patients (20%) without NCD were hospitalized at the JBVAMC for ≥ 1 COPD exacerbation in the preceding year that was diagnosed by a physician (P = .045). Three of 22 patients (14%) with NCD and no patients in the no NCD group were admitted to the MICU. No patients required intubation and mechanical ventilation during hospitalization, and no patients died. Overall hospital costs were 25% ($64,342) higher in NCD group compared with the no NCD group and were attributed to the MICU admissions in the NCD group (Table 3). Nine veterans did not respond to repeated telephone calls. One physician-diagnosed COPD exacerbation requiring hospitalization was documented in the nonresponder group; the patient was hospitalized for 2 days. One veteran died before being contacted.

 

 

DISCUSSION

There are 3 new findings in this study. First, health care practitioners at JBVAMC did not document the presence of NCD during sleep in patients with hypoxemic CRF due to COPD and receiving LTOT. Second, one-third of these patients reported frequent NCD during sleep when interviewed. Third, the nocturnal events were associated with a higher hospitalization rate for physician-diagnosed COPD exacerbation and higher overall hospital costs. These findings are unlikely to be explained by differences in COPD severity and/or known triggers that lead to COPD exacerbation and require hospitalization because baseline physiologic and LTOT parameters were similar in both groups. Conceivably, patients with untreated OSA could be restless while asleep, leading to NCD. However, this explanation seems unlikely because the frequency of OSA was similar in both groups.

Nocturnal arterial oxygen desaturation in patients with COPD without evidence of OSA may contribute to the frequency of exacerbations.16 Although the mechanism(s) underlying this phenomenon is uncertain, we posit that prolonged nocturnal airway wall hypoxia could amplify underlying chronic inflammation through local generation of reactive oxygen species, thereby predisposing patients to exacerbations. Frequent COPD exacerbations promote disease progression and health status decline and are associated with increased mortality.11,13 Moreover, hospitalization of patients with COPD is the largest contributor to the annual direct cost of COPD per patient.10,12 The higher hospitalization rate observed in the NCD group in our study suggests that interruption of supplemental oxygen delivery while asleep may be a risk factor for COPD exacerbation. Alternatively, an independent factor or factors may have contributed to both NCD during sleep and COPD exacerbation in these patients or an impending exacerbation resulted in sleep disturbances that led to NCD. Additional research is warranted on veterans with hypoxemic CRF from COPD who are receiving LTOT and report frequent NCD during sleep that may support or refute these hypotheses.

To the best of our knowledge, NCD during sleep has not been previously reported in patients with hypoxemic CRF due to COPD who are receiving LTOT at home or in an acute care setting.17-20 Several layperson proposals to secure nasal cannulas to the face while sleeping are posted online. These include wearing a commercially available headband with 2 Velcro loops that fix the cannula tube, using fabric medical tape on both cheeks, and wearing a sleep mask. Conceivably, the efficacy and safety of these inexpensive interventions to mitigate NCD during sleep in patients receiving LTOT with hypoxemic CRF from COPD could be tested in clinical trials.

Limitations

This was a small, single-site study, comprised entirely of male patients who are predominantly Black veterans. The telephone interviews with veterans self-reporting NCD during their sleep are prone to recall bias. In addition, the validity and reproducibility of NCD during sleep were not addressed in this study. Missing data from 9 nonresponders may have introduced a nonresponse bias in data analysis and interpretation. The overall hospital cost for a COPD exacerbation at JBVAMC was derived from VA data; US Centers for Medicare & Medicaid Services or commercial carrier data may be different.15,21 Lastly, access to LTOT for veterans with hypoxemic CRF from COPD is regulated and supervised at VA medical facilities.14 This process may be different for patients outside the VA. Taken together, it is difficult to generalize our initial observations to non-VA patients with hypoxemic CRF from COPD who are receiving LTOT. We suggest a large, prospective study of veterans be conducted to determine the prevalence of NCD during sleep and its relationship with COPD exacerbations in veterans receiving LTOT with hypoxemic CRF due to COPD.

CONCLUSIONS

Clinicians at the JBVAMC did not document the presence of NCD during sleep in patients with hypoxemic CRF from COPD who received LTOT. However, self-reported, weekly nocturnal NCD episodes were associated with a higher hospitalization rate for COPD exacerbation and higher hospital costs. Accordingly, user-friendly devices to mitigate NCD during sleep should be developed.

Acknowledgments

We thank Yolanda Davis, RRT, and George Adam for their assistance with this project.

References

1. Boersma P, Cohen RA, Zelaya CE, Moy E. Multiple chronic conditions among veterans and nonveterans: United States, 2015-2018. Natl Health Stat Report. 2021;(153):1-13. doi:10.15620/cdc:101659

2. Sharafkhaneh A, Petersen NJ, Yu H-J, Dalal AA, Johnson ML, Hanania NA. Burden of COPD in a government health care system: a retrospective observational study using data from the US Veterans Affairs population. Int J Chron Obstruct Pulmon Dis. 2010;5:125-132. doi:10.2147/copd.s8047

3. LaBedz SL, Krishnan JA, Chung Y-C, et al. Chronic obstructive pulmonary disease outcomes at Veterans Affairs versus non-Veterans Affairs hospitals. Chronic Obstr Pulm Dis. 2021;8(3):306-313. doi:10.15326/jcopdf.2021.0201

4. Darnell K, Dwivedi AK, Weng Z, Panos RJ. Disproportionate utilization of healthcare resources among veterans with COPD: a retrospective analysis of factors associated with COPD healthcare cost. Cost Eff Resour Alloc. 2013;11:13. doi:10.1186/1478-7547-11-13

5. Bamonti PM, Robinson SA, Wan ES, Moy ML. Improving physiological, physical, and psychological health outcomes: a narrative review in US Veterans with COPD. Int J Chron Obstruct Pulmon Dis. 2022;17:1269-1283. doi:10.2147/COPD.S339323

6. Cranston JM, Crockett AJ, Moss JR, Alpers JH. Domiciliary oxygen for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2005;2005(4):CD001744. doi:10.1002/14651858.CD001744.pub2

7. Lacasse Y, Tan AM, Maltais F, Krishnan JA. Home oxygen in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2018;197(10):1254-1264. doi:10.1164/rccm.201802-0382CI

8. Jacobs SS, Krishnan JA, Lederer DJ, et al. Home oxygen therapy for adults with chronic lung disease. An official American Thoracic Society Clinical Practice Guideline. Am J Respir Crit Care Med. 2020;202(10):e121-e141. doi:10.1164/rccm.202009-3608ST

9. AARC. AARC clinical practice guideline. Oxygen therapy in the home or alternate site health care facility--2007 revision & update. Respir Care. 2007;52(8):1063-1068.

10. Foo J, Landis SH, Maskell J, et al. Continuing to confront COPD international patient survey: economic impact of COPD in 12 countries. PLoS One. 2016;11(4):e0152618. doi:10.1371/journal.pone.0152618

11. Rothnie KJ, Müllerová H, Smeeth L, Quint JK. Natural history of chronic obstructive pulmonary disease exacerbations in a general practice-based population with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2018;198(4):464-471. doi:10.1164/rccm.201710-2029OC

12. Stanford RH, Engel-Nitz NM, Bancroft T, Essoi B. The identification and cost of acute chronic obstructive pulmonary disease exacerbations in a United States population healthcare claims database. COPD. 2020;17(5):499-508. doi:10.1080/15412555.2020.1817357

13. Hurst JR, Han MK, Singh B, et al. Prognostic risk factors for moderate-to-severe exacerbations in patients with chronic obstructive pulmonary disease: a systematic literature review. Respir Res. 2022;23(1):213. doi:10.1186/s12931-022-02123-5

14. US Department of Veterans Affairs, Veterans Health Administration. Home oxygen program. VHA Directive 1173.13(1). Published August 5, 2020. Accessed February 28, 2024. https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=8947

15. Phibbs CS, Barnett PG, Fan A, Harden C, King SS, Scott JY. Research guide to decision support system national cost extracts. Health Economics Resource Center of Health Service R&D Services, US Department of Veterans Affairs. September 2010. Accessed February 14, 2024. https://www.herc.research.va.gov/files/book_621.pdf

16. Agusti A, Hedner J, Marin JM, Barbé F, Cazzola M, Rennard S. Night-time symptoms: a forgotten dimension of COPD. Eur Respir Rev. 2011;20(121):183-194. doi:10.1183/09059180.00004311

17. Croxton TL, Bailey WC. Long-term oxygen treatment in chronic obstructive pulmonary disease: recommendations for future research: an NHLBI workshop report. Am J Respir Crit Care Med. 2006;174(4):373-378. doi:10.1164/rccm.200507-1161WS

18. Melani AS, Sestini P, Rottoli P. Home oxygen therapy: re-thinking the role of devices. Expert Rev Clin Pharmacol. 2018;11(3):279-289. doi:10.1080/17512433.2018.1421457

19. Sculley JA, Corbridge SJ, Prieto-Centurion V, et al. Home oxygen therapy for patients with COPD: time for a reboot. Respir Care. 2019;64(12):1574-1585. doi:10.4187/respcare.07135

20. Jacobs SS, Lindell KO, Collins EG, et al. Patient perceptions of the adequacy of supplemental oxygen therapy. Results of the American Thoracic Society Nursing Assembly Oxygen Working Group Survey. Ann Am Thorac Soc. 2018;15:24-32. doi:10.1513/AnnalsATS.201703-209OC

21. US Centers for Medicare & Medicaid Services. Home use of oxygen. Publication number 100-3. January 3, 2023. Accessed February 14, 2024. https://www.cms.gov/medicare-coverage-database/view/ncd.aspx?NCDId=169

Article PDF
Author and Disclosure Information

Zane Elfessi, PharmD, BCPS, BCCCPa,b; Ahmed Mahgoub, MDa; Emily Tran, PharmDa,b; Grace McGeeb; Israel Rubinstein, MDa,b

Correspondence:  Israel Rubinstein (irubinst@uic.edu)

aJesse Brown Veterans Affairs Medical Center, Chicago, Illinois

bUniversity of Illinois Chicago College of Pharmacy

Author roles

All authors were involved in the conception, study design, data acquisition, analysis, writing, and editing of the manuscript.

Disclosures

This material is the result of work supported with resources and the use of facilities at the Jesse Brown Veterans Affairs Medical Center. The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Ethics and consent

The study protocol was determined to be exempt by Jesse Brown Veterans Affairs Medical Center Institutional Review Board (Protocol #1725748).

Issue
Federal Practitioner - 41(4)a
Publications
Topics
Page Number
117
Sections
Author and Disclosure Information

Zane Elfessi, PharmD, BCPS, BCCCPa,b; Ahmed Mahgoub, MDa; Emily Tran, PharmDa,b; Grace McGeeb; Israel Rubinstein, MDa,b

Correspondence:  Israel Rubinstein (irubinst@uic.edu)

aJesse Brown Veterans Affairs Medical Center, Chicago, Illinois

bUniversity of Illinois Chicago College of Pharmacy

Author roles

All authors were involved in the conception, study design, data acquisition, analysis, writing, and editing of the manuscript.

Disclosures

This material is the result of work supported with resources and the use of facilities at the Jesse Brown Veterans Affairs Medical Center. The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Ethics and consent

The study protocol was determined to be exempt by Jesse Brown Veterans Affairs Medical Center Institutional Review Board (Protocol #1725748).

Author and Disclosure Information

Zane Elfessi, PharmD, BCPS, BCCCPa,b; Ahmed Mahgoub, MDa; Emily Tran, PharmDa,b; Grace McGeeb; Israel Rubinstein, MDa,b

Correspondence:  Israel Rubinstein (irubinst@uic.edu)

aJesse Brown Veterans Affairs Medical Center, Chicago, Illinois

bUniversity of Illinois Chicago College of Pharmacy

Author roles

All authors were involved in the conception, study design, data acquisition, analysis, writing, and editing of the manuscript.

Disclosures

This material is the result of work supported with resources and the use of facilities at the Jesse Brown Veterans Affairs Medical Center. The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.

Disclaimer

The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Ethics and consent

The study protocol was determined to be exempt by Jesse Brown Veterans Affairs Medical Center Institutional Review Board (Protocol #1725748).

Article PDF
Article PDF

The prevalence of chronic obstructive pulmonary disease (COPD) among male US veterans is higher than in the general population.1 Veterans with COPD have higher rates of comorbidities and increased respiratory-related and all-cause health care use, including the use of long-term oxygen therapy (LTOT).2-5 It has been well established that LTOT reduces all-cause mortality in patients with COPD and resting hypoxemic chronic respiratory failure (CRF) when used for ≥ 15 hours per day.6-8

Delivery of domiciliary LTOT entails placing a nasal cannula into both nostrils and loosely securing it around both ears throughout the wake-sleep cycle. Several veterans with hypoxemic CRF due to COPD at the Jesse Brown Veterans Affairs Medical Center (JBVAMC) in Chicago, Illinois, who were receiving LTOT reported nasal cannula dislodgement (NCD) while they slept. However, the clinical significance and impact of these repeated episodes on respiratory-related health care utilization, such as frequent COPD exacerbations with hospitalization, were not recognized. Moreover, we found no published reports or clinical practice guidelines alluding to similar events reported by patients with hypoxemic CRF due to COPD receiving LTOT either at home or in an acute care setting.8,9 Nonetheless, frequent COPD exacerbations are associated with increased hospital admissions and account for a large portion of health care costs attributed to COPD.10-13

The purpose of this study was to determine whether veterans with hypoxemic CRF due to COPD and receiving 24-hour LTOT at JBVAMC were experiencing NCD during sleep and, if so, its impact on their hospitalizations for COPD exacerbations.

METHODS

We reviewed electronic health records (EHRs) of veterans with hypoxemic CRF from COPD who received 24-hour LTOT administered through nasal cannula and were followed in the JBVAMC pulmonary outpatient clinic between February 1, 2022, and December 31, 2022. In each case, LTOT was prescribed by a board-certified pulmonologist based on Veterans Health Administration clinical practice guidelines.14 A licensed durable medical equipment company contracted by the JBVAMC delivered and established home oxygen equipment at each veteran’s residence.

Pertinent patient demographics, clinical and physiologic variables, and hospitalizations with length of JBVAMC stay for each physician-diagnosed COPD exacerbation in the preceding year from the date last seen in the clinic were abstracted from EHRs. Overall hospital cost, defined as a veteran overnight stay in either the medical intensive care unit (MICU) or a general acute medicine bed in a US Department of Veterans Affairs (VA) facility, was calculated for each hospitalization for physician-diagnosed COPD exacerbation using VA Managerial Cost Accounting System National Cost Extracts for inpatient encounters.15 We then contacted each veteran by telephone and asked whether they had experienced NCD and, if so, its weekly frequency ranging from once to nightly.

Data Analysis

Data were reported as mean (SD) where appropriate. The t test and Fisher exact test were used as indicated. P < .05 was considered statistically significant. The study protocol was determined to be exempt by the JBVAMC Institutional Review Board (Protocol #1725748).

 

 

RESULTS

table_1.png

During the study period, 75 patients with hypoxemic CRF from COPD received LTOT and were followed at the JBVAMC. No patients were hospitalized outside the JBVAMC for COPD exacerbation during this time frame. We also found no documentation in the EHRs indicating that the clinicians managing these patients at the JBVAMC inquired about NCD during sleep.

figure.png

Of the 75 patients, 66 (88%) responded to the telephone survey and 22 patients (33%) reported weekly episodes of NCD while they slept (median, 4 dislodgments per week). (Table 1). Eight patients (36%) reported nightly NCDs (Figure). All 66 respondents were male and 14 of 22 in the NCD group as well as 21 of 44 in the no NCD group were Black veterans. The mean age was similar in both groups: 71 years in the NCD group and 72 years in the no NCD group. There were no statistically significant differences in demographics, including prevalence of obstructive sleep apnea (OSA), supplemental oxygen flow rate, and duration of LTOT, or in pulmonary function test results between patients who did and did not experience NCD while sleeping (Table 2).

table_3.png
table_2.png

Ten of 22 patients (45%) with NCD and 9 of 44 patients (20%) without NCD were hospitalized at the JBVAMC for ≥ 1 COPD exacerbation in the preceding year that was diagnosed by a physician (P = .045). Three of 22 patients (14%) with NCD and no patients in the no NCD group were admitted to the MICU. No patients required intubation and mechanical ventilation during hospitalization, and no patients died. Overall hospital costs were 25% ($64,342) higher in NCD group compared with the no NCD group and were attributed to the MICU admissions in the NCD group (Table 3). Nine veterans did not respond to repeated telephone calls. One physician-diagnosed COPD exacerbation requiring hospitalization was documented in the nonresponder group; the patient was hospitalized for 2 days. One veteran died before being contacted.

 

 

DISCUSSION

There are 3 new findings in this study. First, health care practitioners at JBVAMC did not document the presence of NCD during sleep in patients with hypoxemic CRF due to COPD and receiving LTOT. Second, one-third of these patients reported frequent NCD during sleep when interviewed. Third, the nocturnal events were associated with a higher hospitalization rate for physician-diagnosed COPD exacerbation and higher overall hospital costs. These findings are unlikely to be explained by differences in COPD severity and/or known triggers that lead to COPD exacerbation and require hospitalization because baseline physiologic and LTOT parameters were similar in both groups. Conceivably, patients with untreated OSA could be restless while asleep, leading to NCD. However, this explanation seems unlikely because the frequency of OSA was similar in both groups.

Nocturnal arterial oxygen desaturation in patients with COPD without evidence of OSA may contribute to the frequency of exacerbations.16 Although the mechanism(s) underlying this phenomenon is uncertain, we posit that prolonged nocturnal airway wall hypoxia could amplify underlying chronic inflammation through local generation of reactive oxygen species, thereby predisposing patients to exacerbations. Frequent COPD exacerbations promote disease progression and health status decline and are associated with increased mortality.11,13 Moreover, hospitalization of patients with COPD is the largest contributor to the annual direct cost of COPD per patient.10,12 The higher hospitalization rate observed in the NCD group in our study suggests that interruption of supplemental oxygen delivery while asleep may be a risk factor for COPD exacerbation. Alternatively, an independent factor or factors may have contributed to both NCD during sleep and COPD exacerbation in these patients or an impending exacerbation resulted in sleep disturbances that led to NCD. Additional research is warranted on veterans with hypoxemic CRF from COPD who are receiving LTOT and report frequent NCD during sleep that may support or refute these hypotheses.

To the best of our knowledge, NCD during sleep has not been previously reported in patients with hypoxemic CRF due to COPD who are receiving LTOT at home or in an acute care setting.17-20 Several layperson proposals to secure nasal cannulas to the face while sleeping are posted online. These include wearing a commercially available headband with 2 Velcro loops that fix the cannula tube, using fabric medical tape on both cheeks, and wearing a sleep mask. Conceivably, the efficacy and safety of these inexpensive interventions to mitigate NCD during sleep in patients receiving LTOT with hypoxemic CRF from COPD could be tested in clinical trials.

Limitations

This was a small, single-site study, comprised entirely of male patients who are predominantly Black veterans. The telephone interviews with veterans self-reporting NCD during their sleep are prone to recall bias. In addition, the validity and reproducibility of NCD during sleep were not addressed in this study. Missing data from 9 nonresponders may have introduced a nonresponse bias in data analysis and interpretation. The overall hospital cost for a COPD exacerbation at JBVAMC was derived from VA data; US Centers for Medicare & Medicaid Services or commercial carrier data may be different.15,21 Lastly, access to LTOT for veterans with hypoxemic CRF from COPD is regulated and supervised at VA medical facilities.14 This process may be different for patients outside the VA. Taken together, it is difficult to generalize our initial observations to non-VA patients with hypoxemic CRF from COPD who are receiving LTOT. We suggest a large, prospective study of veterans be conducted to determine the prevalence of NCD during sleep and its relationship with COPD exacerbations in veterans receiving LTOT with hypoxemic CRF due to COPD.

CONCLUSIONS

Clinicians at the JBVAMC did not document the presence of NCD during sleep in patients with hypoxemic CRF from COPD who received LTOT. However, self-reported, weekly nocturnal NCD episodes were associated with a higher hospitalization rate for COPD exacerbation and higher hospital costs. Accordingly, user-friendly devices to mitigate NCD during sleep should be developed.

Acknowledgments

We thank Yolanda Davis, RRT, and George Adam for their assistance with this project.

The prevalence of chronic obstructive pulmonary disease (COPD) among male US veterans is higher than in the general population.1 Veterans with COPD have higher rates of comorbidities and increased respiratory-related and all-cause health care use, including the use of long-term oxygen therapy (LTOT).2-5 It has been well established that LTOT reduces all-cause mortality in patients with COPD and resting hypoxemic chronic respiratory failure (CRF) when used for ≥ 15 hours per day.6-8

Delivery of domiciliary LTOT entails placing a nasal cannula into both nostrils and loosely securing it around both ears throughout the wake-sleep cycle. Several veterans with hypoxemic CRF due to COPD at the Jesse Brown Veterans Affairs Medical Center (JBVAMC) in Chicago, Illinois, who were receiving LTOT reported nasal cannula dislodgement (NCD) while they slept. However, the clinical significance and impact of these repeated episodes on respiratory-related health care utilization, such as frequent COPD exacerbations with hospitalization, were not recognized. Moreover, we found no published reports or clinical practice guidelines alluding to similar events reported by patients with hypoxemic CRF due to COPD receiving LTOT either at home or in an acute care setting.8,9 Nonetheless, frequent COPD exacerbations are associated with increased hospital admissions and account for a large portion of health care costs attributed to COPD.10-13

The purpose of this study was to determine whether veterans with hypoxemic CRF due to COPD and receiving 24-hour LTOT at JBVAMC were experiencing NCD during sleep and, if so, its impact on their hospitalizations for COPD exacerbations.

METHODS

We reviewed electronic health records (EHRs) of veterans with hypoxemic CRF from COPD who received 24-hour LTOT administered through nasal cannula and were followed in the JBVAMC pulmonary outpatient clinic between February 1, 2022, and December 31, 2022. In each case, LTOT was prescribed by a board-certified pulmonologist based on Veterans Health Administration clinical practice guidelines.14 A licensed durable medical equipment company contracted by the JBVAMC delivered and established home oxygen equipment at each veteran’s residence.

Pertinent patient demographics, clinical and physiologic variables, and hospitalizations with length of JBVAMC stay for each physician-diagnosed COPD exacerbation in the preceding year from the date last seen in the clinic were abstracted from EHRs. Overall hospital cost, defined as a veteran overnight stay in either the medical intensive care unit (MICU) or a general acute medicine bed in a US Department of Veterans Affairs (VA) facility, was calculated for each hospitalization for physician-diagnosed COPD exacerbation using VA Managerial Cost Accounting System National Cost Extracts for inpatient encounters.15 We then contacted each veteran by telephone and asked whether they had experienced NCD and, if so, its weekly frequency ranging from once to nightly.

Data Analysis

Data were reported as mean (SD) where appropriate. The t test and Fisher exact test were used as indicated. P < .05 was considered statistically significant. The study protocol was determined to be exempt by the JBVAMC Institutional Review Board (Protocol #1725748).

 

 

RESULTS

table_1.png

During the study period, 75 patients with hypoxemic CRF from COPD received LTOT and were followed at the JBVAMC. No patients were hospitalized outside the JBVAMC for COPD exacerbation during this time frame. We also found no documentation in the EHRs indicating that the clinicians managing these patients at the JBVAMC inquired about NCD during sleep.

figure.png

Of the 75 patients, 66 (88%) responded to the telephone survey and 22 patients (33%) reported weekly episodes of NCD while they slept (median, 4 dislodgments per week). (Table 1). Eight patients (36%) reported nightly NCDs (Figure). All 66 respondents were male and 14 of 22 in the NCD group as well as 21 of 44 in the no NCD group were Black veterans. The mean age was similar in both groups: 71 years in the NCD group and 72 years in the no NCD group. There were no statistically significant differences in demographics, including prevalence of obstructive sleep apnea (OSA), supplemental oxygen flow rate, and duration of LTOT, or in pulmonary function test results between patients who did and did not experience NCD while sleeping (Table 2).

table_3.png
table_2.png

Ten of 22 patients (45%) with NCD and 9 of 44 patients (20%) without NCD were hospitalized at the JBVAMC for ≥ 1 COPD exacerbation in the preceding year that was diagnosed by a physician (P = .045). Three of 22 patients (14%) with NCD and no patients in the no NCD group were admitted to the MICU. No patients required intubation and mechanical ventilation during hospitalization, and no patients died. Overall hospital costs were 25% ($64,342) higher in NCD group compared with the no NCD group and were attributed to the MICU admissions in the NCD group (Table 3). Nine veterans did not respond to repeated telephone calls. One physician-diagnosed COPD exacerbation requiring hospitalization was documented in the nonresponder group; the patient was hospitalized for 2 days. One veteran died before being contacted.

 

 

DISCUSSION

There are 3 new findings in this study. First, health care practitioners at JBVAMC did not document the presence of NCD during sleep in patients with hypoxemic CRF due to COPD and receiving LTOT. Second, one-third of these patients reported frequent NCD during sleep when interviewed. Third, the nocturnal events were associated with a higher hospitalization rate for physician-diagnosed COPD exacerbation and higher overall hospital costs. These findings are unlikely to be explained by differences in COPD severity and/or known triggers that lead to COPD exacerbation and require hospitalization because baseline physiologic and LTOT parameters were similar in both groups. Conceivably, patients with untreated OSA could be restless while asleep, leading to NCD. However, this explanation seems unlikely because the frequency of OSA was similar in both groups.

Nocturnal arterial oxygen desaturation in patients with COPD without evidence of OSA may contribute to the frequency of exacerbations.16 Although the mechanism(s) underlying this phenomenon is uncertain, we posit that prolonged nocturnal airway wall hypoxia could amplify underlying chronic inflammation through local generation of reactive oxygen species, thereby predisposing patients to exacerbations. Frequent COPD exacerbations promote disease progression and health status decline and are associated with increased mortality.11,13 Moreover, hospitalization of patients with COPD is the largest contributor to the annual direct cost of COPD per patient.10,12 The higher hospitalization rate observed in the NCD group in our study suggests that interruption of supplemental oxygen delivery while asleep may be a risk factor for COPD exacerbation. Alternatively, an independent factor or factors may have contributed to both NCD during sleep and COPD exacerbation in these patients or an impending exacerbation resulted in sleep disturbances that led to NCD. Additional research is warranted on veterans with hypoxemic CRF from COPD who are receiving LTOT and report frequent NCD during sleep that may support or refute these hypotheses.

To the best of our knowledge, NCD during sleep has not been previously reported in patients with hypoxemic CRF due to COPD who are receiving LTOT at home or in an acute care setting.17-20 Several layperson proposals to secure nasal cannulas to the face while sleeping are posted online. These include wearing a commercially available headband with 2 Velcro loops that fix the cannula tube, using fabric medical tape on both cheeks, and wearing a sleep mask. Conceivably, the efficacy and safety of these inexpensive interventions to mitigate NCD during sleep in patients receiving LTOT with hypoxemic CRF from COPD could be tested in clinical trials.

Limitations

This was a small, single-site study, comprised entirely of male patients who are predominantly Black veterans. The telephone interviews with veterans self-reporting NCD during their sleep are prone to recall bias. In addition, the validity and reproducibility of NCD during sleep were not addressed in this study. Missing data from 9 nonresponders may have introduced a nonresponse bias in data analysis and interpretation. The overall hospital cost for a COPD exacerbation at JBVAMC was derived from VA data; US Centers for Medicare & Medicaid Services or commercial carrier data may be different.15,21 Lastly, access to LTOT for veterans with hypoxemic CRF from COPD is regulated and supervised at VA medical facilities.14 This process may be different for patients outside the VA. Taken together, it is difficult to generalize our initial observations to non-VA patients with hypoxemic CRF from COPD who are receiving LTOT. We suggest a large, prospective study of veterans be conducted to determine the prevalence of NCD during sleep and its relationship with COPD exacerbations in veterans receiving LTOT with hypoxemic CRF due to COPD.

CONCLUSIONS

Clinicians at the JBVAMC did not document the presence of NCD during sleep in patients with hypoxemic CRF from COPD who received LTOT. However, self-reported, weekly nocturnal NCD episodes were associated with a higher hospitalization rate for COPD exacerbation and higher hospital costs. Accordingly, user-friendly devices to mitigate NCD during sleep should be developed.

Acknowledgments

We thank Yolanda Davis, RRT, and George Adam for their assistance with this project.

References

1. Boersma P, Cohen RA, Zelaya CE, Moy E. Multiple chronic conditions among veterans and nonveterans: United States, 2015-2018. Natl Health Stat Report. 2021;(153):1-13. doi:10.15620/cdc:101659

2. Sharafkhaneh A, Petersen NJ, Yu H-J, Dalal AA, Johnson ML, Hanania NA. Burden of COPD in a government health care system: a retrospective observational study using data from the US Veterans Affairs population. Int J Chron Obstruct Pulmon Dis. 2010;5:125-132. doi:10.2147/copd.s8047

3. LaBedz SL, Krishnan JA, Chung Y-C, et al. Chronic obstructive pulmonary disease outcomes at Veterans Affairs versus non-Veterans Affairs hospitals. Chronic Obstr Pulm Dis. 2021;8(3):306-313. doi:10.15326/jcopdf.2021.0201

4. Darnell K, Dwivedi AK, Weng Z, Panos RJ. Disproportionate utilization of healthcare resources among veterans with COPD: a retrospective analysis of factors associated with COPD healthcare cost. Cost Eff Resour Alloc. 2013;11:13. doi:10.1186/1478-7547-11-13

5. Bamonti PM, Robinson SA, Wan ES, Moy ML. Improving physiological, physical, and psychological health outcomes: a narrative review in US Veterans with COPD. Int J Chron Obstruct Pulmon Dis. 2022;17:1269-1283. doi:10.2147/COPD.S339323

6. Cranston JM, Crockett AJ, Moss JR, Alpers JH. Domiciliary oxygen for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2005;2005(4):CD001744. doi:10.1002/14651858.CD001744.pub2

7. Lacasse Y, Tan AM, Maltais F, Krishnan JA. Home oxygen in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2018;197(10):1254-1264. doi:10.1164/rccm.201802-0382CI

8. Jacobs SS, Krishnan JA, Lederer DJ, et al. Home oxygen therapy for adults with chronic lung disease. An official American Thoracic Society Clinical Practice Guideline. Am J Respir Crit Care Med. 2020;202(10):e121-e141. doi:10.1164/rccm.202009-3608ST

9. AARC. AARC clinical practice guideline. Oxygen therapy in the home or alternate site health care facility--2007 revision & update. Respir Care. 2007;52(8):1063-1068.

10. Foo J, Landis SH, Maskell J, et al. Continuing to confront COPD international patient survey: economic impact of COPD in 12 countries. PLoS One. 2016;11(4):e0152618. doi:10.1371/journal.pone.0152618

11. Rothnie KJ, Müllerová H, Smeeth L, Quint JK. Natural history of chronic obstructive pulmonary disease exacerbations in a general practice-based population with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2018;198(4):464-471. doi:10.1164/rccm.201710-2029OC

12. Stanford RH, Engel-Nitz NM, Bancroft T, Essoi B. The identification and cost of acute chronic obstructive pulmonary disease exacerbations in a United States population healthcare claims database. COPD. 2020;17(5):499-508. doi:10.1080/15412555.2020.1817357

13. Hurst JR, Han MK, Singh B, et al. Prognostic risk factors for moderate-to-severe exacerbations in patients with chronic obstructive pulmonary disease: a systematic literature review. Respir Res. 2022;23(1):213. doi:10.1186/s12931-022-02123-5

14. US Department of Veterans Affairs, Veterans Health Administration. Home oxygen program. VHA Directive 1173.13(1). Published August 5, 2020. Accessed February 28, 2024. https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=8947

15. Phibbs CS, Barnett PG, Fan A, Harden C, King SS, Scott JY. Research guide to decision support system national cost extracts. Health Economics Resource Center of Health Service R&D Services, US Department of Veterans Affairs. September 2010. Accessed February 14, 2024. https://www.herc.research.va.gov/files/book_621.pdf

16. Agusti A, Hedner J, Marin JM, Barbé F, Cazzola M, Rennard S. Night-time symptoms: a forgotten dimension of COPD. Eur Respir Rev. 2011;20(121):183-194. doi:10.1183/09059180.00004311

17. Croxton TL, Bailey WC. Long-term oxygen treatment in chronic obstructive pulmonary disease: recommendations for future research: an NHLBI workshop report. Am J Respir Crit Care Med. 2006;174(4):373-378. doi:10.1164/rccm.200507-1161WS

18. Melani AS, Sestini P, Rottoli P. Home oxygen therapy: re-thinking the role of devices. Expert Rev Clin Pharmacol. 2018;11(3):279-289. doi:10.1080/17512433.2018.1421457

19. Sculley JA, Corbridge SJ, Prieto-Centurion V, et al. Home oxygen therapy for patients with COPD: time for a reboot. Respir Care. 2019;64(12):1574-1585. doi:10.4187/respcare.07135

20. Jacobs SS, Lindell KO, Collins EG, et al. Patient perceptions of the adequacy of supplemental oxygen therapy. Results of the American Thoracic Society Nursing Assembly Oxygen Working Group Survey. Ann Am Thorac Soc. 2018;15:24-32. doi:10.1513/AnnalsATS.201703-209OC

21. US Centers for Medicare & Medicaid Services. Home use of oxygen. Publication number 100-3. January 3, 2023. Accessed February 14, 2024. https://www.cms.gov/medicare-coverage-database/view/ncd.aspx?NCDId=169

References

1. Boersma P, Cohen RA, Zelaya CE, Moy E. Multiple chronic conditions among veterans and nonveterans: United States, 2015-2018. Natl Health Stat Report. 2021;(153):1-13. doi:10.15620/cdc:101659

2. Sharafkhaneh A, Petersen NJ, Yu H-J, Dalal AA, Johnson ML, Hanania NA. Burden of COPD in a government health care system: a retrospective observational study using data from the US Veterans Affairs population. Int J Chron Obstruct Pulmon Dis. 2010;5:125-132. doi:10.2147/copd.s8047

3. LaBedz SL, Krishnan JA, Chung Y-C, et al. Chronic obstructive pulmonary disease outcomes at Veterans Affairs versus non-Veterans Affairs hospitals. Chronic Obstr Pulm Dis. 2021;8(3):306-313. doi:10.15326/jcopdf.2021.0201

4. Darnell K, Dwivedi AK, Weng Z, Panos RJ. Disproportionate utilization of healthcare resources among veterans with COPD: a retrospective analysis of factors associated with COPD healthcare cost. Cost Eff Resour Alloc. 2013;11:13. doi:10.1186/1478-7547-11-13

5. Bamonti PM, Robinson SA, Wan ES, Moy ML. Improving physiological, physical, and psychological health outcomes: a narrative review in US Veterans with COPD. Int J Chron Obstruct Pulmon Dis. 2022;17:1269-1283. doi:10.2147/COPD.S339323

6. Cranston JM, Crockett AJ, Moss JR, Alpers JH. Domiciliary oxygen for chronic obstructive pulmonary disease. Cochrane Database Syst Rev. 2005;2005(4):CD001744. doi:10.1002/14651858.CD001744.pub2

7. Lacasse Y, Tan AM, Maltais F, Krishnan JA. Home oxygen in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2018;197(10):1254-1264. doi:10.1164/rccm.201802-0382CI

8. Jacobs SS, Krishnan JA, Lederer DJ, et al. Home oxygen therapy for adults with chronic lung disease. An official American Thoracic Society Clinical Practice Guideline. Am J Respir Crit Care Med. 2020;202(10):e121-e141. doi:10.1164/rccm.202009-3608ST

9. AARC. AARC clinical practice guideline. Oxygen therapy in the home or alternate site health care facility--2007 revision & update. Respir Care. 2007;52(8):1063-1068.

10. Foo J, Landis SH, Maskell J, et al. Continuing to confront COPD international patient survey: economic impact of COPD in 12 countries. PLoS One. 2016;11(4):e0152618. doi:10.1371/journal.pone.0152618

11. Rothnie KJ, Müllerová H, Smeeth L, Quint JK. Natural history of chronic obstructive pulmonary disease exacerbations in a general practice-based population with chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2018;198(4):464-471. doi:10.1164/rccm.201710-2029OC

12. Stanford RH, Engel-Nitz NM, Bancroft T, Essoi B. The identification and cost of acute chronic obstructive pulmonary disease exacerbations in a United States population healthcare claims database. COPD. 2020;17(5):499-508. doi:10.1080/15412555.2020.1817357

13. Hurst JR, Han MK, Singh B, et al. Prognostic risk factors for moderate-to-severe exacerbations in patients with chronic obstructive pulmonary disease: a systematic literature review. Respir Res. 2022;23(1):213. doi:10.1186/s12931-022-02123-5

14. US Department of Veterans Affairs, Veterans Health Administration. Home oxygen program. VHA Directive 1173.13(1). Published August 5, 2020. Accessed February 28, 2024. https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=8947

15. Phibbs CS, Barnett PG, Fan A, Harden C, King SS, Scott JY. Research guide to decision support system national cost extracts. Health Economics Resource Center of Health Service R&D Services, US Department of Veterans Affairs. September 2010. Accessed February 14, 2024. https://www.herc.research.va.gov/files/book_621.pdf

16. Agusti A, Hedner J, Marin JM, Barbé F, Cazzola M, Rennard S. Night-time symptoms: a forgotten dimension of COPD. Eur Respir Rev. 2011;20(121):183-194. doi:10.1183/09059180.00004311

17. Croxton TL, Bailey WC. Long-term oxygen treatment in chronic obstructive pulmonary disease: recommendations for future research: an NHLBI workshop report. Am J Respir Crit Care Med. 2006;174(4):373-378. doi:10.1164/rccm.200507-1161WS

18. Melani AS, Sestini P, Rottoli P. Home oxygen therapy: re-thinking the role of devices. Expert Rev Clin Pharmacol. 2018;11(3):279-289. doi:10.1080/17512433.2018.1421457

19. Sculley JA, Corbridge SJ, Prieto-Centurion V, et al. Home oxygen therapy for patients with COPD: time for a reboot. Respir Care. 2019;64(12):1574-1585. doi:10.4187/respcare.07135

20. Jacobs SS, Lindell KO, Collins EG, et al. Patient perceptions of the adequacy of supplemental oxygen therapy. Results of the American Thoracic Society Nursing Assembly Oxygen Working Group Survey. Ann Am Thorac Soc. 2018;15:24-32. doi:10.1513/AnnalsATS.201703-209OC

21. US Centers for Medicare & Medicaid Services. Home use of oxygen. Publication number 100-3. January 3, 2023. Accessed February 14, 2024. https://www.cms.gov/medicare-coverage-database/view/ncd.aspx?NCDId=169

Issue
Federal Practitioner - 41(4)a
Issue
Federal Practitioner - 41(4)a
Page Number
117
Page Number
117
Publications
Publications
Topics
Article Type
Sections
Teambase XML
<?xml version="1.0" encoding="UTF-8"?>
<!--$RCSfile: InCopy_agile.xsl,v $ $Revision: 1.35 $-->
<!--$RCSfile: drupal.xsl,v $ $Revision: 1.7 $-->
<root generator="drupal.xsl" gversion="1.7"> <header> <fileName>0424 FED O2 Therapy</fileName> <TBEID>0C02F3C0.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F3C0</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname/> <articleType>1</articleType> <TBLocation>Copyfitting-FED</TBLocation> <QCDate/> <firstPublished>20240408T155151</firstPublished> <LastPublished>20240408T155151</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240408T155151</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline/> <bylineText>Zane Elfessi, PharmD, BCPS, BCCCPa,b; Ahmed Mahgoub, MDa; Emily Tran, PharmDa,b; Grace McGeeb; Israel Rubinstein, MDa,b</bylineText> <bylineFull/> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange/> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>The prevalence of chronic obstructive pulmonary disease (COPD) among male US veterans is higher than in the general population.1 Veterans with COPD have higher </metaDescription> <articlePDF/> <teaserImage/> <title>Nasal Cannula Dislodgement During Sleep in Veterans Receiving Long-term Oxygen Therapy for Hypoxemic Chronic Respiratory Failure</title> <deck/> <eyebrow>Original Study</eyebrow> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>April</pubPubdateMonth> <pubPubdateDay/> <pubVolume>41</pubVolume> <pubNumber>4</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2951</CMSID> <CMSID>3639</CMSID> </CMSIDs> <keywords/> <seeAlsos/> <publications_g> <publicationData> <publicationCode>FED</publicationCode> <pubIssueName>April 2024</pubIssueName> <pubArticleType>Feature Articles | 3639</pubArticleType> <pubTopics/> <pubCategories/> <pubSections> <pubSection>Feature | 2951<pubSubsection/></pubSection> </pubSections> <journalTitle>Fed Pract</journalTitle> <journalFullTitle>Federal Practitioner</journalFullTitle> <copyrightStatement>Copyright 2017 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">16</term> </publications> <sections> <term canonical="true">104</term> </sections> <topics> <term canonical="true">284</term> <term>27442</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Nasal Cannula Dislodgement During Sleep in Veterans Receiving Long-term Oxygen Therapy for Hypoxemic Chronic Respiratory Failure</title> <deck/> </itemMeta> <itemContent> <p class="abstract"><b>B</b><b>ackground: </b>Chronic obstructive pulmonary disease (COPD) is highly prevalent in male veterans. Long-term oxygen therapy (LTOT) can effectively reduce all-cause mortality in these patients, but the effects of nasal cannula dislodgement (NCD) during sleep have not been well studied. <hl name="33679"/><br/><br/><b>Methods: </b>This study sought<b> </b>to determine whether veterans receiving LTOT for hypoxemic chronic respiratory failure (CRF) due to COPD reported NCD while they slept and, if so, its impact on hospitalizations for COPD exacerbations. Electronic health records were reviewed of veterans with hypoxemic CRF due to COPD who received LTOT and were followed in the pulmonary clinic at the Jesse Brown Veterans Affairs Medical Center between February 1, 2022, and December 31, 2022. Overall hospital cost was calculated for each hospitalized veteran with diagnosed COPD exacerbation. Veterans reported whether they experienced NCD during sleep and, if so, its frequency. <br/><br/><b>Results<hl name="33680"/>:</b> <hl name="33681"/>Interviews were conducted with 66 veterans with a mean age of 71 years. Twenty-two veterans (33%) reported NCD ≥ 1 weekly (median, 4). There were no statistically significant differences in demographics, supplemental oxygen flow rate, duration of LTOT, and pulmonary function tests between patients with and without NCD. Ten patients (45%) with NCD and 9 patients (20%) without NCD were hospitalized for ≥ 1 COPD exacerbation (<i>P</i> = .045). Three patients (14%) with NCD were admitted to the medical intensive care unit. Overall hospital costs were 25% higher in the NCD group compared with the no NCD group.<br/><br/><b>Conclusions<hl name="33682"/>:</b> Veterans receiving LTOT for hypoxemic CRF due to COPD who report frequent NCD during sleep have higher hospitalization rates for COPD exacerbation and higher hospital costs.</p> <p><span class="Drop">T</span>he prevalence of chronic obstructive pulmonary disease (COPD) among male US veterans is higher than in the general population.<sup>1</sup> Veterans with COPD have higher rates of comorbidities and increased respiratory-related and all-cause health care use, including the use of long-term oxygen therapy (LTOT).<sup>2-5</sup> It has been well established that LTOT reduces all-cause mortality in patients with COPD and <hl name="33683"/>resting hypoxemic chronic respiratory failure (CRF) when used for ≥ 15 hours per day.<sup>6-8</sup> </p> <p>Delivery of domiciliary LTOT entails placing a nasal cannula into both nostrils and loosely securing it around both ears throughout the wake-sleep cycle. Several veterans with hypoxemic CRF due to COPD at the Jesse Brown Veterans Affairs Medical Center (JBVAMC) in Chicago, Illinois, who were receiving LTOT reported nasal cannula dislodgement (NCD) while they slept. However, the clinical significance and impact of these repeated episodes on respiratory-related health care utilization, such as frequent COPD exacerbations with hospitalization, were not recognized. <hl name="7"/>Moreover, we found no published reports or clinical practice guidelines alluding to similar events reported by patients with hypoxemic CRF due to COPD receiving LTOT either at home or in an acute care setting.<sup>8,9</sup> Nonetheless, frequent COPD exacerbations are associated with increased hospital admissions and account for a large portion of health care costs attributed to COPD.<sup>10-13</sup>The purpose of this study was to determine whether veterans with hypoxemic CRF due to COPD and receiving 24-hour LTOT at JBVAMC were experiencing NCD during sleep and, if so, its impact on <hl name="8"/>their hospitalizations for COPD exacerbations. </p> <h2>METHODS</h2> <p>We reviewed electronic health records (EHRs) of veterans with hypoxemic CRF from COPD who received 24-hour LTOT administered through nasal cannula and were followed <hl name="9"/>in the JBVAMC pulmonary outpatient clinic between February 1, 2022, and December 31, 2022. In each case, LTOT was prescribed by a board-certified pulmonologist based on Veterans Health Administration clinical practice guidelines.<sup>14</sup> A licensed durable medical equipment company contracted by the JBVAMC delivered and established home oxygen equipment at each veteran’s residence. </p> <p>Pertinent patient demographics, clinical and physiologic variables, and hospitalizations with length of JBVAMC stay for each physician-diagnosed COPD exacerbation in the preceding year from the date last seen in the clinic were abstracted from EHRs. Overall hospital cost, defined as a veteran overnight stay in either the medical intensive care unit (MICU) or a general acute medicine bed in a US Department of Veterans Affairs (VA) facility, was calculated for each hospitalization for physician-diagnosed COPD exacerbation using VA Managerial Cost Accounting System National Cost Extracts for inpatient encounters.<sup>15</sup> We then contacted each veteran by telephone and asked whether they had experienced NCD and, if so, its weekly frequency ranging from once to nightly.</p> <h3>Data Analysis</h3> <p>Data were reported as mean (SD) where appropriate. The<i> t</i> test and Fisher exact test were used as indicated. <i>P</i> &lt; .05 was considered statistically significant.<b> </b>The study protocol <hl name="10"/>was determined to be exempt by the JBVAMC Institutional Review Board (Protocol #1725748).</p> <h2>RESULTS</h2> <p>During the study period, <hl name="11"/><hl name="12"/>75 patients with hypoxemic CRF from COPD received LTOT and were followed at the JBVAMC. No patients were hospitalized outside the JBVAMC for COPD exacerbation during this time frame. We also found no documentation in the EHRs indicating that the clinicians managing these patients at the JBVAMC inquired about NCD during sleep.</p> <p>Of the 75 patients, 66 (88%) responded to the telephone survey and 22 patients (33%) reported weekly episodes of NCD while they slept (median, 4 dislodgments per week). (Table 1). Eight patients (36%) reported nightly NCDs (Figure). All 66 respondents were male and 14 of 22 in the NCD group as well as 21 of 44 in the no NCD group were Black veterans. The mean age was similar in both groups: 71 years in the NCD group and 72 years in the no NCD group. There were no statistically significant differences in demographics, including prevalence of obstructive sleep apnea (OSA), supplemental oxygen flow rate, and duration of LTOT, or in pulmonary function test results between patients who did and did not experience NCD while sleeping (Table 2). <br/><br/>Ten of 22 patients (45%) with NCD and 9 of 44 patients (20%) without NCD were hospitalized at the JBVAMC for ≥ 1 COPD exacerbation in the preceding year that was diagnosed by a physician (<i>P</i> = .045). Three of 22 patients (14%) with NCD and no patients in the no NCD group were admitted to the MICU. No patients required intubation and mechanical ventilation during hospitalization, and no patients died. Overall hospital costs were 25% ($64,342) higher in NCD group compared with the no NCD group and were attributed to the MICU admissions in the NCD group (Table 3). Nine veterans did not respond to repeated telephone calls. One physician-diagnosed COPD exacerbation requiring hospitalization was documented in the nonresponder group; the patient was hospitalized for 2 days. One veteran died before being contacted.</p> <h2>DISCUSSION</h2> <p>There are 3 new findings in this study. <hl name="13"/><hl name="14"/>First, health care practitioners at JBVAMC did not document the presence of NCD during sleep in patients with <hl name="15"/>hypoxemic CRF due to COPD and receiving LTOT. Second, one-third of these patients reported frequent NCD during sleep when interviewed. Third, the nocturnal events were associated with a higher hospitalization rate for physician-diagnosed COPD exacerbation and higher overall hospital costs. These findings are unlikely to be explained by differences in COPD severity and/or known triggers that lead to COPD exacerbation and require hospitalization because baseline physiologic and LTOT parameters were similar in both groups. Conceivably, patients with untreated OSA could be restless while asleep, leading to NCD. However, this explanation seems unlikely because the frequency of OSA was similar in both groups. </p> <p>Nocturnal arterial oxygen desaturation in patients with COPD without evidence of OSA may contribute to the frequency of exacerbations.<sup>16</sup> Although the mechanism(s) underlying this phenomenon is uncertain, we posit that prolonged nocturnal airway wall hypoxia could amplify underlying chronic inflammation through local generation of reactive oxygen species, thereby predisposing patients to exacerbations. Frequent COPD exacerbations promote disease progression and health status decline and are associated with increased mortality.<sup>11,13</sup> Moreover, hospitalization of patients with COPD is the largest contributor to the annual direct cost of COPD per patient.<sup>10,12</sup> The higher hospitalization rate observed in the NCD group in our study suggests that interruption of supplemental oxygen delivery while asleep may be a risk factor for COPD exacerbation. Alternatively, an independent factor or factors may have contributed to both NCD during sleep and COPD exacerbation in these patients or an impending exacerbation resulted in sleep disturbances that led to NCD. Additional research is warranted on veterans with hypoxemic CRF from COPD who are receiving LTOT and report frequent NCD during sleep that may support or refute these hypotheses.<br/><br/>To the best of our knowledge, NCD during sleep has not been previously reported in patients <hl name="16"/>with hypoxemic CRF due to COPD who are receiving LTOT at home or in an acute care setting.<sup>17-20</sup> Several layperson proposals to secure nasal cannulas to the face while sleeping are posted online. These include wearing a commercially available headband with 2 Velcro loops that fix the cannula tube, using fabric medical tape on both cheeks, and wearing a sleep mask. Conceivably, the efficacy and safety of these inexpensive interventions to mitigate NCD during sleep in patients receiving LTOT with hypoxemic CRF from COPD could be tested in clinical trials.</p> <h3>Limitations</h3> <p>This was a small, single-site study, comprised entirely of male patients who are predominantly Black veterans. The telephone interviews with veterans self-reporting NCD during their sleep are prone to recall bias. In addition, the validity and reproducibility of NCD during sleep were not addressed in this study. Missing data from 9 nonresponders may have introduced a nonresponse bias in data analysis and interpretation. The overall hospital cost for a COPD exacerbation at JBVAMC was derived from VA data; US Centers for Medicare &amp; Medicaid Services or commercial carrier data may be different.<sup>15,21</sup> Lastly, access to LTOT for veterans with hypoxemic CRF from COPD is regulated and supervised at VA medical facilities.<sup>14</sup> This process may be different for patients outside the VA. Taken together, it is difficult to generalize our initial observations to non-VA patients with hypoxemic CRF from COPD who are receiving LTOT. We suggest a large, prospective study of veterans be conducted to determine the prevalence of NCD during sleep and its relationship with COPD exacerbations in veterans receiving LTOT with hypoxemic CRF due to COPD. </p> <h2>CONCLUSIONS</h2> <p><hl name="17"/>Clinicians at the JBVAMC did not document the presence of NCD during sleep in patients with hypoxemic CRF from COPD who received LTOT. However, self-reported, weekly nocturnal NCD episodes were associated with a higher hospitalization rate for COPD exacerbation and higher hospital costs. Accordingly, user-friendly devices to mitigate NCD during sleep should be developed.</p> <p class="isub">Acknowledgments</p> <p> <em>We thank Yolanda Davis, RRT, and George Adam for their assistance with this project. </em> </p> <p class="isub">Author affiliations</p> <p> <em><sup>a</sup>Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois<br/><br/><sup>b</sup>University of Illinois Chicago College of Pharmacy</em> </p> <p class="isub">Author roles</p> <p> <em>All authors were involved in the conception, study design, data acquisition, analysis, writing, and editing of the manuscript. </em> </p> <p class="isub">Disclosures</p> <p> <em>This material is the result of work supported with resources and the use of facilities at the Jesse Brown Veterans Affairs Medical Center. The authors report no actual or potential conflicts of interest or outside sources of funding with regard to this article.</em> </p> <p class="isub">Disclaimer</p> <p> <em>The opinions expressed herein are those of the authors and do not necessarily reflect those of <i>Federal Practitioner</i>, Frontline Medical Communications Inc., the US Government, or any of its agencies.</em> </p> <p class="isub">Ethics and consent</p> <p> <em>The study protocol was determined to be exempt by Jesse Brown Veterans Affairs Medical Center Institutional Review Board (Protocol #1725748).</em> </p> <h2><hl name="18"/>References</h2> <p class="reference"> 1. Boersma P, Cohen RA, Zelaya CE, Moy E. Multiple chronic conditions among veterans and nonveterans: United States, 2015-2018. <i>Natl Health Stat Report</i>. 2021;(153):1-13. doi:10.15620/cdc:101659<br/><br/> 2. Sharafkhaneh A, Petersen NJ, Yu H-J, Dalal AA, Johnson ML, Hanania NA. Burden of COPD in a government health care system: a retrospective observational study using data from the US Veterans Affairs population. <i>Int J Chron Obstruct Pulmon Dis</i>. 2010;5:125-132. doi:10.2147/copd.s8047<br/><br/> 3. LaBedz SL, Krishnan JA, Chung Y-C, et al. Chronic obstructive pulmonary disease outcomes at Veterans Affairs versus non-Veterans Affairs hospitals. <i>Chronic Obstr Pulm Dis</i>. 2021;8(3):306-313. doi:10.15326/jcopdf.2021.0201<br/><br/> 4. Darnell K, Dwivedi AK, Weng Z, Panos RJ. Disproportionate utilization of healthcare resources among veterans with COPD: a retrospective analysis of factors associated with COPD healthcare cost. <i>Cost Eff Resour Alloc</i>. 2013;11:13. doi:10.1186/1478-7547-11-13<br/><br/> 5. Bamonti PM, Robinson SA, Wan ES, Moy ML. Improving physiological, physical, and psychological health outcomes: a narrative review in US Veterans with COPD. <i>Int J Chron Obstruct Pulmon Dis</i>. 2022;17:1269-1283. doi:10.2147/COPD.S339323<br/><br/> 6. Cranston JM, Crockett AJ, Moss JR, Alpers JH. Domiciliary oxygen for chronic obstructive pulmonary disease. <i>Cochrane Database Syst Rev</i>. 2005;2005(4):CD001744. doi:10.1002/14651858.CD001744.pub2<br/><br/> 7. Lacasse Y, Tan AM, Maltais F, Krishnan JA. Home oxygen in chronic obstructive pulmonary disease. <i>Am J Respir Crit Care Med</i>. 2018;197(10):1254-1264. doi:10.1164/rccm.201802-0382CI<br/><br/> 8. Jacobs SS, Krishnan JA, Lederer DJ, et al. Home oxygen therapy for adults with chronic lung disease. An official American Thoracic Society Clinical Practice Guideline. <i>Am J Respir Crit Care Med</i>. 2020;202(10):e121-e141. doi:10.1164/rccm.202009-3608ST<br/><br/> 9. AARC. AARC clinical practice guideline. Oxygen therapy in the home or alternate site health care facility--2007 revision &amp; update. <i>Respir Care</i>. 2007;52(8):1063-1068. <br/><br/>10. Foo J, Landis SH, Maskell J, et al. Continuing to confront COPD international patient survey: economic impact of COPD in 12 countries. <i>PLoS One.</i> 2016;11(4):e0152618. doi:10.1371/journal.pone.0152618<br/><br/>11. Rothnie KJ, Müllerová H, Smeeth L, Quint JK. Natural history of chronic obstructive pulmonary disease exacerbations in a general practice-based population with chronic obstructive pulmonary disease. <i>Am J Respir Crit Care Med</i>. 2018;198(4):464-471. doi:10.1164/rccm.201710-2029OC<br/><br/>12. Stanford RH, Engel-Nitz NM, Bancroft T, Essoi B. The identification and cost of acute chronic obstructive pulmonary disease exacerbations in a United States population healthcare claims database. <i>COPD</i>. 2020;17(5):499-508. doi:10.1080/15412555.2020.1817357<br/><br/>13. Hurst JR, Han MK, Singh B, et al. Prognostic risk factors for moderate-to-severe exacerbations in patients with chronic obstructive pulmonary disease: a systematic literature review. <i>Respir Res</i>. 2022;23(1):213. doi:10.1186/s12931-022-02123-5<br/><br/>14. US Department of Veterans Affairs, Veterans Health Administration. Home oxygen program. VHA Directive 1173.13(1). Published August 5, 2020. Accessed February 28, 2024. https://www.va.gov/vhapublications/ViewPublication.asp?pub_ID=8947<br/><br/>15. Phibbs CS, Barnett PG, Fan A, Harden C, King SS, Scott JY. Research guide to decision support system national cost extracts. Health Economics Resource Center of Health Service R&amp;D Services, US Department of Veterans Affairs. September 2010. Accessed February 14, 2024. https://www.herc.research.va.gov/files/book_621.pdf<br/><br/>16. Agusti A, Hedner J, Marin JM, Barbé F, Cazzola M, Rennard S. Night-time symptoms: a forgotten dimension of COPD. <i>Eur Respir Rev</i>. 2011;20(121):183-194. doi:10.1183/09059180.00004311<br/><br/>17. Croxton TL, Bailey WC. Long-term oxygen treatment in chronic obstructive pulmonary disease: recommendations for future research: an NHLBI workshop report. <i>Am J Respir Crit Care Med</i>. 2006;174(4):373-378. doi:10.1164/rccm.200507-1161WS<br/><br/>18. Melani AS, Sestini P, Rottoli P. Home oxygen therapy: re-thinking the role of devices. <i>Expert Rev Clin Pharmacol</i>. 2018;11(3):279-289. doi:10.1080/17512433.2018.1421457<br/><br/>19. Sculley JA, Corbridge SJ, Prieto-Centurion V, et al. Home oxygen therapy for patients with COPD: time for a reboot. <i>Respir Care</i>. 2019;64(12):1574-1585. doi:10.4187/respcare.07135<br/><br/>20. Jacobs SS, Lindell KO, Collins EG, et al. Patient perceptions of the adequacy of supplemental oxygen therapy. Results of the American Thoracic Society Nursing Assembly Oxygen Working Group Survey. <i>Ann Am Thorac Soc</i>. 2018;15:24-32. doi:10.1513/AnnalsATS.201703-209OC<br/><br/>21. US Centers for Medicare &amp; Medicaid Services. Home use of oxygen. Publication number 100-3. January 3, 2023. Accessed February 14, 2024. https://www.cms.gov/medicare-coverage-database/view/ncd.aspx?NCDId=169</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

Recurrent Aphthous Stomatitis: Clinical Experience From a University Hospital in Brazil

Article Type
Changed
Thu, 06/06/2024 - 11:24
Display Headline
Recurrent Aphthous Stomatitis: Clinical Experience From a University Hospital in Brazil

To the Editor:

Recurrent aphthous stomatitis (RAS) is a mucocutaneous condition characterized by single or multiple, painful,1,2 round ulcerations of variable sizes with a tendency for recurrence, most commonly located in nonkeratinized areas of the oral mucosa. Pathergy commonly is observed.3 Although many authors consider the terms RAS andaphtha to be synonymous,4,5 differentiating the clinical lesion (aphthous ulceration) from the disease (aphtha or RAS) can be useful, as several other diseases can at times manifest with similar ulcers (called aphthoid lesions), such as pemphigus vulgaris, mucous membrane pemphigoid, and erythema multiforme.6

It is estimated that approximately 20% of individuals worldwide have at least one episode of aphtha during their lifetime,7 and it is considered the most common disease of the oral mucosa.8,9 However, only patients presenting with severe acute outbreaks or frequent relapses typically seek medical treatment. Clinically, aphthous ulcers are classified as aphtha minor (small number of small lesions), aphtha major (large deep lesions that also can affect the minor salivary glands with intense necrosis, difficulty in healing, and mucosal scarring), and aphtha herpetiformis (innumerous tiny lesions that reappear in recurring outbreaks).1-3 The term complex aphthosis was introduced in 198510 and is defined as recurrent oral and genital aphthous ulcerations or recurring multiple oral aphthous ulcers in the absence of systemic manifestations or Behçet disease11,12; however, complex aphthosis also has been reported as frequent episodes of ulcerations that may be associated with systemic diseases including Behçet disease.13,14

Currently, RAS is considered an immunologically mediated alteration in cutaneous mucosal reactivity with a multifactorial systemic cause. Underlying conditions such as Behçet disease, inflammatory bowel disease (IBD), iatrogenic immunosuppression (eg, following solid organ transplantation), AIDS, and cyclic neutropenia may or may not be detected.11-13

Our retrospective study explored the systemic nature of RAS. We reviewed patient records to evaluate underlying systemic conditions associated with the diagnosis of RAS and the use of oral medications in managing the disease. Medical records from the Department of Dermatology of the University of São Paulo, Brazil, from 2003 to 2017 were reviewed to identify patients with a diagnosis of RAS. Clinical classification of RAS—minor, major, or herpetiform—as well as the presence of aphthous lesions in other locations and the presence of other associated inflammatory cutaneous manifestations also were noted. Associated systemic diseases and treatments for RAS were recorded. Patients for whom the diagnosis of RAS was changed during follow-up were excluded. Because this was a retrospective analysis of medical records and without any patient risk, informed consent was not needed.

Medical records for 125 patients were reviewed; 63 were male (50.4%), and 62 were female (49.6%). The age at onset of symptoms, which ranged from a few months after birth to 74 years, was reported in only 92 (73.6%) patient medical records. Of these, 30 (32.6%) reported onset before 20 years of age, 39 (42.4%) between 20 and 39 years, 17 (18.5%) between 40 and 59 years, and 6 (6.5%) at 60 years or older. Morphologically, 72 (57.6%) had minor, 42 (33.6%) had major, and 11 (8.8%) had herpetiform aphthous ulcers. None of the patients presented with sporadic lesions; the disease was long-standing and persistent in all cases (complex aphthosis).

Regarding the location of the ulcers, 92 (73.6%) patients had lesions on the oral mucosa only. Some patients had lesions in more than one site in addition to the oral mucosa: 32 (25.6%) had aphthae in the genital/groin region and 4 (3.2%) presented with perianal/anal aphthae. Nineteen patients (19.2%) presented other cutaneous manifestations in addition to aphthae: 11 (45.8%) had folliculitis/pseudofolliculitis, and 8 (33.3%) had erythema nodosum (EN). Eight patients (33.3%) presented with uveitis, and 6 (25%) presented with concomitant arthralgia/arthritis. Fifty-four patients (43.2%) had confirmed or suspected associated disease: Behçet disease (21 [38.9%]), IBD (10 [18.5%]), solid organ transplantation (7 [13.0%])(kidney, 4 [57.1%]; heart, 2 [28.6%]; liver, 1 [14.3%]), HIV infection (6 [11.1%]), lymphoma (1 [1.9%]), aplastic anemia (1 [1.9%]), or myelodysplastic syndrome (1 [1.9%]). Ten patients (18.5%) presented with other diseases under investigation (eg, unidentified rheumatologic disease, unexplained neutropenia, undiagnosed immunodeficiencies, autoinflammatory syndromes, possible cyclic neutropenia).

Biopsies of the oral mucosa were performed in 31 patients. Histopathologic findings will be discussed in a future publication (unpublished data).

 

 

Five patients (4.0%) were lost to follow-up and did not receive treatment; 10 (8.0%) received only topical treatment (analgesics and/or corticosteroids). All 9 (7.2%) patients undergoing intralesional corticosteroid injections also were on a systemic treatment. One hundred ten (88.0%) patients were treated systemically—with colchicine (84/110 [76.4%]), thalidomide (43/110 [39.1%]), small pulses of oral corticosteroids (26 [23.6%]), dapsone (12/110 [10.9%]), or pentoxifylline (3 [2.7%]). Furthermore, in patients with associated diseases, treatment of the underlying condition was conducted when available, and follow-up was carried out in conjunction with the appropriate specialists. For treatment of the associated disease, patients received other medications such as methotrexate, azathioprine, cyclophosphamide, intravenous corticosteroid pulse, and immunobiologics.

The prevalence of RAS between sexes in our study population was similar (50.4% male; 49.6% female). Results from prior studies have been mixed; some reported a higher prevalence in females,15-18 while others found no predilection for sex among patients diagnosed with RAS.19,20 In our analysis, 75% of patients experienced symptoms of RAS before 40 years of age; in prior studies, up to 56% of patients experienced symptoms between the ages of 20 and 40 years.21,22

In our study, 26.4% of patients had extraoral aphthae. Genital lesions have been described as infrequent,23 and lesions manifesting in other mucous membranes or on the skin are rare.24 A study reported genital involvement in 8% to 13% of patients with oral aphtha.25 We observed genital involvement in 25.6% of patients. Likewise, this higher value may be due to our study population of patients referred to our university hospital. In our study, 19.2% of patients presented with other inflammatory manifestations in addition to aphthous ulcerations (eg, folliculitis, EN, uveitis, arthritis). As dermatologists in a tertiary reference hospital, we actively look for such associations in every aphtha patient, which may not be the case in many nondermatologic oral care services.

In our study population, 43.2% of patients were diagnosed with or were under investigation for systemic diseases known to be associated with RAS. We found associations with Behçet disease most frequently, followed by IBD,26 solid organ transplantation, and HIV. In this group of patients, the respective systemic disease was active or poorly controlled. In transplant recipients, aphtha major was the most common type, similar to other studies.27 We observed no notable difference in the clinical picture of the oral ulcers in patients with a well-established systemic disease vs those without.

Most of our cases did not present findings other than aphtha, indicating that the intrinsic defect that predisposes to RAS is always systemic. Even mild and sporadic cases may be attributable to a systemic disorder of cutaneous-mucosal reactivity. The predisposition to RAS never originates in the oral cavity, hence the confusion caused and the uselessness of studies that relate aphthae to factors such as local food allergies, pH changes, or local infection with microorganisms.5,28 The disease course (reducing the frequency of lesion appearance and accelerating the healing of extensive lesions) is only modified with systemic treatment, with local measures proving to be only moderately useful to relieve pain. We believe that RAS can in many ways be compared to EN and pyoderma gangrenosum (PG): some systemic conditions that predispose patients to EN and PG also may predispose them to RAS (eg, IBD, hematologic disorders). Similar to RAS, many cases of EN and PG are idiopathic. In addition, pathergy also occurs in PG.11,13

We were unable to observe or establish any predictive clinical element that could indicate a better or worse response to the prescribed treatments, which also has been noted by other authors.3,4 Treatment of RAS is empiric, generally starting with drugs that are easier to prescribe and with fewer adverse effects, then progressing to more complex drugs when a good response is not obtained. Colchicine was the most commonly prescribed medication (76.4% [84/110]). It has been proposed by several authors3,4 as a first-line systemic medication for the treatment of recurrent aphthae, as it has been shown to be effective and safe. The dosage ranged from 0.5 mg twice daily to 0.5 mg 4 times daily. Dapsone is an established drug for aphtha29,30 and was used in 12 of our patients. The dosage used in our patients ranged from 50 to 100 mg/d. Adverse effects such as hemolytic anemia frequently are seen, and one of the patients in our study developed DRESS (drug reaction with eosinophilia and systemic symptoms) syndrome in response to dapsone. In 7 cases, colchicine and dapsone were used together, which is believed to potentiate the therapeutic effects. This combination may be useful in patients for whom thalidomide cannot be used or those who have not improved with monotherapy.29 Thalidomide is considered one of the most effective drugs for RAS.30,31 Forty-three patients in our analysis were treated with thalidomide,usually as a first choice. The dosage ranged from 100 to 200 mg/d. It was mainly chosen in disabling pediatric cases, adult men with aphthous major, and women with no risk for pregnancy. Due to its potential adverse effects, thalidomide has been recommended when there is no response with other medications that are dose dependent; severe adverse effects such as thromboembolism and peripheral neuropathy are rare.31 Oral corticosteroids were used in 26 patients, aiming at rapid improvement in very symptomatic cases; however, due to the potential for long-term adverse effects, in all cases they were prescribed in combination with another medication that was maintained after the corticosteroid was discontinued.

We highlight the systemic nature of RAS as well as its frequent association with systemic diseases and other correlated manifestations (pustules, EN, arthralgia). We also emphasize the importance of using oral medications to adequately control the disease and do not recommend topical medications aimed at treating local causes. Dermatologists should be consulted in managing severe cases of RAS.

References
  1. Buño IJ, Huff JC, Weston WL, et al. Elevated levels of interferon gamma, tumor necrosis factor alpha, interleukins 2, 4, and 5, but not interleukin 10, are present in recurrent aphthous stomatitis. Arch Dermatol. 1998;134:827-831.
  2. Femiano F, Lanza A, Buonaiuto C, et al. Guidelines for diagnosis and management of aphthous stomatitis. Pediatr Infect Dis J. 2007;26:728- 732.
  3. Natah SS, Konttinen YTEnattah NS, et al. Recurrent aphthous ulcers today: a review of the growing knowledge. Int J Oral Maxillofac Surg. 2004;33:221-234.
  4. Zunt SL. Recurrent aphthous stomatitis. Dermatol Clin. 2003;21:33-39.
  5. Jurge S, Kuffer R, Scully C, et al. Mucosal disease series. number VI. recurrent aphthous stomatitis. Oral Dis. 2006;12:1-21.
  6. Chams-Davatchi C, Shizarpour M, Davatchi F, et al. Comparison of oral aphthae in Behçet’s disease and idiopathic recurrent aphthous stomatitis. Adv Exp Med Biol. 2003;528:317-320.
  7. Schemel-Suárez M, López-López J, Chimenos-Küstner E. Oral ulcers: differential diagnosis and treatment [in Spanish]. Med Clin (Barc). 2015;145:499-503.
  8. S´lebioda Z, Szponar E, Kowalska A. Etiopathogenesis of recurrent aphthous stomatitis and the role of immunologic aspects: literature review. Arch Immunol Ther Exp (Warsz). 2014;62:205-215.
  9. Edgar NR, Saleh D, Miller RA. Recurrent aphthous stomatitis: a review. J Clin Aesthet Dermatol. 2017;10:26-36.
  10. Jorizzo JL, Taylor RS, Schmalstieg FC, et al. Complex aphthosis: a forme fruste of Behçet’s syndrome? J Am Acad Dermatol. 1985;13:80-84.
  11. McCarty MA, Garton RA, Jorizzo JL. Complex aphthosis and Behçet’s disease. Dermatol Clin. 2003;21:41-48.
  12. Bulur I, Melrem O. Behçet disease: new aspects. Clin Dermatol. 2017;35:421-434.
  13. Cui RZ, Rogers RS 3rd. Recurrent aphthous stomatitis. Clin Dermatol. 2016;34:475-481.
  14. Femiano F, Lanza A, Buonaiuto C, et al. Guidelines for diagnosis and management of aphthous stomatitis. Pediatr Infect Dis J. 2007;26:728-732.
  15. Ship II. Epidemiologic aspects of recurrent aphthous ulcerations. Oral Surg Oral Med Oral Pathol. 1972;33:400-406.
  16. Ship JA. Recurrent aphthous stomatitis. an update. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 1996;81:141-147.
  17. Wilhelmsen NS, Weber R, Monteiro F, et al. Correlation between histocompatibility antigens and recurrent aphthous stomatitis in the Brazilian population. Braz J Otorhinolaryngol. 2009;75:426-431.
  18. S´lebioda Z, Dorocka-Bobkowska B. Systemic and environmental risk factors for recurrent aphthous stomatitis in a Polish cohort of patients. Postepy Dermatol Alergol. 2019;36:196-201.
  19. Ship JA, Chavez EM, Doerr PA, et al. Recurrent aphthous stomatitis. Quintessence Int. 2000;31:95-112.
  20. Brocklehurst P, Tickle M, Glenny AM, et al. Systemic interventions for recurrent aphthous stomatitis (mouth ulcers). Cochrane Database Syst Rev. 2012;12:CD005411.
  21. Belenguer-Guallar I, Jiménez-Soriano Y, Ariadna Claramunt-Lozano A. Treatment of recurrent aphthous stomatitis. a literature review. J Clin Exp Dent. 2014;6:E168-E174.
  22. Bagán JV, Sanchis JM, Milián MA, et al. Recurrent aphthous stomatitis. a study of the clinical characteristics of lesions in 93 cases. J Oral Pathol Med. 1991;20:395-397.
  23. Huppert JS, Gerber MA, Deitch HR, et al. Vulvar ulcers in young females: a manifestation of aphthosis. J Pediatr Adolesc Gynecol. 2006;19:195-204. 
  24. Scully C, Porter S. Recurrent aphthous stomatitis: current concepts of etiology, pathogenesis and management. J Oral Pathol Med. 1989;18:21-27
  25. Chapel TA. Origins of penile ulcerations. Arch Androl. 1979; 3: 351-357.
  26. Lourenço SV, Hussein TP, Bologna SB, et al. Oral manifestations of inflammatory bowel disease: a review based on the observation of six cases. J Eur Acad Dermatol Venereol. 2010;24:204-207.
  27. Nico MM, Brito AE, Martins LE, et al. Oral ulcers in an immunosuppressed 5-year-old boy. Clin Exp Dermatol. 2008;33:367-368.
  28. Trakji B, Baroudi K, Kharma Y. The effect of dietary habits on the development of the recurrent aphthous stomatitis. Niger Med J. 2012;53:9-11.
  29. Lynde CB, Bruce AJ, Rogers RS 3rd. Successful treatment of complex aphthosis with colchicine and dapsone. Arch Dermatol. 2009;145:273-276. 
  30. Letsinger JAMcCarty MAJorizzo JL. Complex aphthosis: a large case series with evaluation algorithm and therapeutic ladder from topicals to thalidomide. J Am Acad Dermatol. 2005(3 pt 1);52:500-508.
  31. Hello M, Barbarot S, Bastuji-Garin S, et al. Use of thalidomide for severe recurrent aphthous stomatitis: a multicenter cohort analysis. Medicine (Baltimore). 2010;89:176-182.
Article PDF
Author and Disclosure Information

From the Universidade de São Paulo, Brazil. Drs. Pinto and Nico are from the Department of Dermatology, Faculdade de Medicina, and Dr. Lourenço is from the Department of Pathology, Faculdade de Odontologia.

The authors report no conflict of interest.

Correspondence: Marcello Menta Simonsen Nico, MD, Departamento de Dermatologia, Faculdade de Medicina da Universidade de São Paulo, Brasil, R. Itapeva 500-3A. CEP-01332-000, São Paulo, Brasil (mentanico@hotmail.com).

Issue
Cutis - 113(4)
Publications
Topics
Page Number
171-173
Sections
Author and Disclosure Information

From the Universidade de São Paulo, Brazil. Drs. Pinto and Nico are from the Department of Dermatology, Faculdade de Medicina, and Dr. Lourenço is from the Department of Pathology, Faculdade de Odontologia.

The authors report no conflict of interest.

Correspondence: Marcello Menta Simonsen Nico, MD, Departamento de Dermatologia, Faculdade de Medicina da Universidade de São Paulo, Brasil, R. Itapeva 500-3A. CEP-01332-000, São Paulo, Brasil (mentanico@hotmail.com).

Author and Disclosure Information

From the Universidade de São Paulo, Brazil. Drs. Pinto and Nico are from the Department of Dermatology, Faculdade de Medicina, and Dr. Lourenço is from the Department of Pathology, Faculdade de Odontologia.

The authors report no conflict of interest.

Correspondence: Marcello Menta Simonsen Nico, MD, Departamento de Dermatologia, Faculdade de Medicina da Universidade de São Paulo, Brasil, R. Itapeva 500-3A. CEP-01332-000, São Paulo, Brasil (mentanico@hotmail.com).

Article PDF
Article PDF

To the Editor:

Recurrent aphthous stomatitis (RAS) is a mucocutaneous condition characterized by single or multiple, painful,1,2 round ulcerations of variable sizes with a tendency for recurrence, most commonly located in nonkeratinized areas of the oral mucosa. Pathergy commonly is observed.3 Although many authors consider the terms RAS andaphtha to be synonymous,4,5 differentiating the clinical lesion (aphthous ulceration) from the disease (aphtha or RAS) can be useful, as several other diseases can at times manifest with similar ulcers (called aphthoid lesions), such as pemphigus vulgaris, mucous membrane pemphigoid, and erythema multiforme.6

It is estimated that approximately 20% of individuals worldwide have at least one episode of aphtha during their lifetime,7 and it is considered the most common disease of the oral mucosa.8,9 However, only patients presenting with severe acute outbreaks or frequent relapses typically seek medical treatment. Clinically, aphthous ulcers are classified as aphtha minor (small number of small lesions), aphtha major (large deep lesions that also can affect the minor salivary glands with intense necrosis, difficulty in healing, and mucosal scarring), and aphtha herpetiformis (innumerous tiny lesions that reappear in recurring outbreaks).1-3 The term complex aphthosis was introduced in 198510 and is defined as recurrent oral and genital aphthous ulcerations or recurring multiple oral aphthous ulcers in the absence of systemic manifestations or Behçet disease11,12; however, complex aphthosis also has been reported as frequent episodes of ulcerations that may be associated with systemic diseases including Behçet disease.13,14

Currently, RAS is considered an immunologically mediated alteration in cutaneous mucosal reactivity with a multifactorial systemic cause. Underlying conditions such as Behçet disease, inflammatory bowel disease (IBD), iatrogenic immunosuppression (eg, following solid organ transplantation), AIDS, and cyclic neutropenia may or may not be detected.11-13

Our retrospective study explored the systemic nature of RAS. We reviewed patient records to evaluate underlying systemic conditions associated with the diagnosis of RAS and the use of oral medications in managing the disease. Medical records from the Department of Dermatology of the University of São Paulo, Brazil, from 2003 to 2017 were reviewed to identify patients with a diagnosis of RAS. Clinical classification of RAS—minor, major, or herpetiform—as well as the presence of aphthous lesions in other locations and the presence of other associated inflammatory cutaneous manifestations also were noted. Associated systemic diseases and treatments for RAS were recorded. Patients for whom the diagnosis of RAS was changed during follow-up were excluded. Because this was a retrospective analysis of medical records and without any patient risk, informed consent was not needed.

Medical records for 125 patients were reviewed; 63 were male (50.4%), and 62 were female (49.6%). The age at onset of symptoms, which ranged from a few months after birth to 74 years, was reported in only 92 (73.6%) patient medical records. Of these, 30 (32.6%) reported onset before 20 years of age, 39 (42.4%) between 20 and 39 years, 17 (18.5%) between 40 and 59 years, and 6 (6.5%) at 60 years or older. Morphologically, 72 (57.6%) had minor, 42 (33.6%) had major, and 11 (8.8%) had herpetiform aphthous ulcers. None of the patients presented with sporadic lesions; the disease was long-standing and persistent in all cases (complex aphthosis).

Regarding the location of the ulcers, 92 (73.6%) patients had lesions on the oral mucosa only. Some patients had lesions in more than one site in addition to the oral mucosa: 32 (25.6%) had aphthae in the genital/groin region and 4 (3.2%) presented with perianal/anal aphthae. Nineteen patients (19.2%) presented other cutaneous manifestations in addition to aphthae: 11 (45.8%) had folliculitis/pseudofolliculitis, and 8 (33.3%) had erythema nodosum (EN). Eight patients (33.3%) presented with uveitis, and 6 (25%) presented with concomitant arthralgia/arthritis. Fifty-four patients (43.2%) had confirmed or suspected associated disease: Behçet disease (21 [38.9%]), IBD (10 [18.5%]), solid organ transplantation (7 [13.0%])(kidney, 4 [57.1%]; heart, 2 [28.6%]; liver, 1 [14.3%]), HIV infection (6 [11.1%]), lymphoma (1 [1.9%]), aplastic anemia (1 [1.9%]), or myelodysplastic syndrome (1 [1.9%]). Ten patients (18.5%) presented with other diseases under investigation (eg, unidentified rheumatologic disease, unexplained neutropenia, undiagnosed immunodeficiencies, autoinflammatory syndromes, possible cyclic neutropenia).

Biopsies of the oral mucosa were performed in 31 patients. Histopathologic findings will be discussed in a future publication (unpublished data).

 

 

Five patients (4.0%) were lost to follow-up and did not receive treatment; 10 (8.0%) received only topical treatment (analgesics and/or corticosteroids). All 9 (7.2%) patients undergoing intralesional corticosteroid injections also were on a systemic treatment. One hundred ten (88.0%) patients were treated systemically—with colchicine (84/110 [76.4%]), thalidomide (43/110 [39.1%]), small pulses of oral corticosteroids (26 [23.6%]), dapsone (12/110 [10.9%]), or pentoxifylline (3 [2.7%]). Furthermore, in patients with associated diseases, treatment of the underlying condition was conducted when available, and follow-up was carried out in conjunction with the appropriate specialists. For treatment of the associated disease, patients received other medications such as methotrexate, azathioprine, cyclophosphamide, intravenous corticosteroid pulse, and immunobiologics.

The prevalence of RAS between sexes in our study population was similar (50.4% male; 49.6% female). Results from prior studies have been mixed; some reported a higher prevalence in females,15-18 while others found no predilection for sex among patients diagnosed with RAS.19,20 In our analysis, 75% of patients experienced symptoms of RAS before 40 years of age; in prior studies, up to 56% of patients experienced symptoms between the ages of 20 and 40 years.21,22

In our study, 26.4% of patients had extraoral aphthae. Genital lesions have been described as infrequent,23 and lesions manifesting in other mucous membranes or on the skin are rare.24 A study reported genital involvement in 8% to 13% of patients with oral aphtha.25 We observed genital involvement in 25.6% of patients. Likewise, this higher value may be due to our study population of patients referred to our university hospital. In our study, 19.2% of patients presented with other inflammatory manifestations in addition to aphthous ulcerations (eg, folliculitis, EN, uveitis, arthritis). As dermatologists in a tertiary reference hospital, we actively look for such associations in every aphtha patient, which may not be the case in many nondermatologic oral care services.

In our study population, 43.2% of patients were diagnosed with or were under investigation for systemic diseases known to be associated with RAS. We found associations with Behçet disease most frequently, followed by IBD,26 solid organ transplantation, and HIV. In this group of patients, the respective systemic disease was active or poorly controlled. In transplant recipients, aphtha major was the most common type, similar to other studies.27 We observed no notable difference in the clinical picture of the oral ulcers in patients with a well-established systemic disease vs those without.

Most of our cases did not present findings other than aphtha, indicating that the intrinsic defect that predisposes to RAS is always systemic. Even mild and sporadic cases may be attributable to a systemic disorder of cutaneous-mucosal reactivity. The predisposition to RAS never originates in the oral cavity, hence the confusion caused and the uselessness of studies that relate aphthae to factors such as local food allergies, pH changes, or local infection with microorganisms.5,28 The disease course (reducing the frequency of lesion appearance and accelerating the healing of extensive lesions) is only modified with systemic treatment, with local measures proving to be only moderately useful to relieve pain. We believe that RAS can in many ways be compared to EN and pyoderma gangrenosum (PG): some systemic conditions that predispose patients to EN and PG also may predispose them to RAS (eg, IBD, hematologic disorders). Similar to RAS, many cases of EN and PG are idiopathic. In addition, pathergy also occurs in PG.11,13

We were unable to observe or establish any predictive clinical element that could indicate a better or worse response to the prescribed treatments, which also has been noted by other authors.3,4 Treatment of RAS is empiric, generally starting with drugs that are easier to prescribe and with fewer adverse effects, then progressing to more complex drugs when a good response is not obtained. Colchicine was the most commonly prescribed medication (76.4% [84/110]). It has been proposed by several authors3,4 as a first-line systemic medication for the treatment of recurrent aphthae, as it has been shown to be effective and safe. The dosage ranged from 0.5 mg twice daily to 0.5 mg 4 times daily. Dapsone is an established drug for aphtha29,30 and was used in 12 of our patients. The dosage used in our patients ranged from 50 to 100 mg/d. Adverse effects such as hemolytic anemia frequently are seen, and one of the patients in our study developed DRESS (drug reaction with eosinophilia and systemic symptoms) syndrome in response to dapsone. In 7 cases, colchicine and dapsone were used together, which is believed to potentiate the therapeutic effects. This combination may be useful in patients for whom thalidomide cannot be used or those who have not improved with monotherapy.29 Thalidomide is considered one of the most effective drugs for RAS.30,31 Forty-three patients in our analysis were treated with thalidomide,usually as a first choice. The dosage ranged from 100 to 200 mg/d. It was mainly chosen in disabling pediatric cases, adult men with aphthous major, and women with no risk for pregnancy. Due to its potential adverse effects, thalidomide has been recommended when there is no response with other medications that are dose dependent; severe adverse effects such as thromboembolism and peripheral neuropathy are rare.31 Oral corticosteroids were used in 26 patients, aiming at rapid improvement in very symptomatic cases; however, due to the potential for long-term adverse effects, in all cases they were prescribed in combination with another medication that was maintained after the corticosteroid was discontinued.

We highlight the systemic nature of RAS as well as its frequent association with systemic diseases and other correlated manifestations (pustules, EN, arthralgia). We also emphasize the importance of using oral medications to adequately control the disease and do not recommend topical medications aimed at treating local causes. Dermatologists should be consulted in managing severe cases of RAS.

To the Editor:

Recurrent aphthous stomatitis (RAS) is a mucocutaneous condition characterized by single or multiple, painful,1,2 round ulcerations of variable sizes with a tendency for recurrence, most commonly located in nonkeratinized areas of the oral mucosa. Pathergy commonly is observed.3 Although many authors consider the terms RAS andaphtha to be synonymous,4,5 differentiating the clinical lesion (aphthous ulceration) from the disease (aphtha or RAS) can be useful, as several other diseases can at times manifest with similar ulcers (called aphthoid lesions), such as pemphigus vulgaris, mucous membrane pemphigoid, and erythema multiforme.6

It is estimated that approximately 20% of individuals worldwide have at least one episode of aphtha during their lifetime,7 and it is considered the most common disease of the oral mucosa.8,9 However, only patients presenting with severe acute outbreaks or frequent relapses typically seek medical treatment. Clinically, aphthous ulcers are classified as aphtha minor (small number of small lesions), aphtha major (large deep lesions that also can affect the minor salivary glands with intense necrosis, difficulty in healing, and mucosal scarring), and aphtha herpetiformis (innumerous tiny lesions that reappear in recurring outbreaks).1-3 The term complex aphthosis was introduced in 198510 and is defined as recurrent oral and genital aphthous ulcerations or recurring multiple oral aphthous ulcers in the absence of systemic manifestations or Behçet disease11,12; however, complex aphthosis also has been reported as frequent episodes of ulcerations that may be associated with systemic diseases including Behçet disease.13,14

Currently, RAS is considered an immunologically mediated alteration in cutaneous mucosal reactivity with a multifactorial systemic cause. Underlying conditions such as Behçet disease, inflammatory bowel disease (IBD), iatrogenic immunosuppression (eg, following solid organ transplantation), AIDS, and cyclic neutropenia may or may not be detected.11-13

Our retrospective study explored the systemic nature of RAS. We reviewed patient records to evaluate underlying systemic conditions associated with the diagnosis of RAS and the use of oral medications in managing the disease. Medical records from the Department of Dermatology of the University of São Paulo, Brazil, from 2003 to 2017 were reviewed to identify patients with a diagnosis of RAS. Clinical classification of RAS—minor, major, or herpetiform—as well as the presence of aphthous lesions in other locations and the presence of other associated inflammatory cutaneous manifestations also were noted. Associated systemic diseases and treatments for RAS were recorded. Patients for whom the diagnosis of RAS was changed during follow-up were excluded. Because this was a retrospective analysis of medical records and without any patient risk, informed consent was not needed.

Medical records for 125 patients were reviewed; 63 were male (50.4%), and 62 were female (49.6%). The age at onset of symptoms, which ranged from a few months after birth to 74 years, was reported in only 92 (73.6%) patient medical records. Of these, 30 (32.6%) reported onset before 20 years of age, 39 (42.4%) between 20 and 39 years, 17 (18.5%) between 40 and 59 years, and 6 (6.5%) at 60 years or older. Morphologically, 72 (57.6%) had minor, 42 (33.6%) had major, and 11 (8.8%) had herpetiform aphthous ulcers. None of the patients presented with sporadic lesions; the disease was long-standing and persistent in all cases (complex aphthosis).

Regarding the location of the ulcers, 92 (73.6%) patients had lesions on the oral mucosa only. Some patients had lesions in more than one site in addition to the oral mucosa: 32 (25.6%) had aphthae in the genital/groin region and 4 (3.2%) presented with perianal/anal aphthae. Nineteen patients (19.2%) presented other cutaneous manifestations in addition to aphthae: 11 (45.8%) had folliculitis/pseudofolliculitis, and 8 (33.3%) had erythema nodosum (EN). Eight patients (33.3%) presented with uveitis, and 6 (25%) presented with concomitant arthralgia/arthritis. Fifty-four patients (43.2%) had confirmed or suspected associated disease: Behçet disease (21 [38.9%]), IBD (10 [18.5%]), solid organ transplantation (7 [13.0%])(kidney, 4 [57.1%]; heart, 2 [28.6%]; liver, 1 [14.3%]), HIV infection (6 [11.1%]), lymphoma (1 [1.9%]), aplastic anemia (1 [1.9%]), or myelodysplastic syndrome (1 [1.9%]). Ten patients (18.5%) presented with other diseases under investigation (eg, unidentified rheumatologic disease, unexplained neutropenia, undiagnosed immunodeficiencies, autoinflammatory syndromes, possible cyclic neutropenia).

Biopsies of the oral mucosa were performed in 31 patients. Histopathologic findings will be discussed in a future publication (unpublished data).

 

 

Five patients (4.0%) were lost to follow-up and did not receive treatment; 10 (8.0%) received only topical treatment (analgesics and/or corticosteroids). All 9 (7.2%) patients undergoing intralesional corticosteroid injections also were on a systemic treatment. One hundred ten (88.0%) patients were treated systemically—with colchicine (84/110 [76.4%]), thalidomide (43/110 [39.1%]), small pulses of oral corticosteroids (26 [23.6%]), dapsone (12/110 [10.9%]), or pentoxifylline (3 [2.7%]). Furthermore, in patients with associated diseases, treatment of the underlying condition was conducted when available, and follow-up was carried out in conjunction with the appropriate specialists. For treatment of the associated disease, patients received other medications such as methotrexate, azathioprine, cyclophosphamide, intravenous corticosteroid pulse, and immunobiologics.

The prevalence of RAS between sexes in our study population was similar (50.4% male; 49.6% female). Results from prior studies have been mixed; some reported a higher prevalence in females,15-18 while others found no predilection for sex among patients diagnosed with RAS.19,20 In our analysis, 75% of patients experienced symptoms of RAS before 40 years of age; in prior studies, up to 56% of patients experienced symptoms between the ages of 20 and 40 years.21,22

In our study, 26.4% of patients had extraoral aphthae. Genital lesions have been described as infrequent,23 and lesions manifesting in other mucous membranes or on the skin are rare.24 A study reported genital involvement in 8% to 13% of patients with oral aphtha.25 We observed genital involvement in 25.6% of patients. Likewise, this higher value may be due to our study population of patients referred to our university hospital. In our study, 19.2% of patients presented with other inflammatory manifestations in addition to aphthous ulcerations (eg, folliculitis, EN, uveitis, arthritis). As dermatologists in a tertiary reference hospital, we actively look for such associations in every aphtha patient, which may not be the case in many nondermatologic oral care services.

In our study population, 43.2% of patients were diagnosed with or were under investigation for systemic diseases known to be associated with RAS. We found associations with Behçet disease most frequently, followed by IBD,26 solid organ transplantation, and HIV. In this group of patients, the respective systemic disease was active or poorly controlled. In transplant recipients, aphtha major was the most common type, similar to other studies.27 We observed no notable difference in the clinical picture of the oral ulcers in patients with a well-established systemic disease vs those without.

Most of our cases did not present findings other than aphtha, indicating that the intrinsic defect that predisposes to RAS is always systemic. Even mild and sporadic cases may be attributable to a systemic disorder of cutaneous-mucosal reactivity. The predisposition to RAS never originates in the oral cavity, hence the confusion caused and the uselessness of studies that relate aphthae to factors such as local food allergies, pH changes, or local infection with microorganisms.5,28 The disease course (reducing the frequency of lesion appearance and accelerating the healing of extensive lesions) is only modified with systemic treatment, with local measures proving to be only moderately useful to relieve pain. We believe that RAS can in many ways be compared to EN and pyoderma gangrenosum (PG): some systemic conditions that predispose patients to EN and PG also may predispose them to RAS (eg, IBD, hematologic disorders). Similar to RAS, many cases of EN and PG are idiopathic. In addition, pathergy also occurs in PG.11,13

We were unable to observe or establish any predictive clinical element that could indicate a better or worse response to the prescribed treatments, which also has been noted by other authors.3,4 Treatment of RAS is empiric, generally starting with drugs that are easier to prescribe and with fewer adverse effects, then progressing to more complex drugs when a good response is not obtained. Colchicine was the most commonly prescribed medication (76.4% [84/110]). It has been proposed by several authors3,4 as a first-line systemic medication for the treatment of recurrent aphthae, as it has been shown to be effective and safe. The dosage ranged from 0.5 mg twice daily to 0.5 mg 4 times daily. Dapsone is an established drug for aphtha29,30 and was used in 12 of our patients. The dosage used in our patients ranged from 50 to 100 mg/d. Adverse effects such as hemolytic anemia frequently are seen, and one of the patients in our study developed DRESS (drug reaction with eosinophilia and systemic symptoms) syndrome in response to dapsone. In 7 cases, colchicine and dapsone were used together, which is believed to potentiate the therapeutic effects. This combination may be useful in patients for whom thalidomide cannot be used or those who have not improved with monotherapy.29 Thalidomide is considered one of the most effective drugs for RAS.30,31 Forty-three patients in our analysis were treated with thalidomide,usually as a first choice. The dosage ranged from 100 to 200 mg/d. It was mainly chosen in disabling pediatric cases, adult men with aphthous major, and women with no risk for pregnancy. Due to its potential adverse effects, thalidomide has been recommended when there is no response with other medications that are dose dependent; severe adverse effects such as thromboembolism and peripheral neuropathy are rare.31 Oral corticosteroids were used in 26 patients, aiming at rapid improvement in very symptomatic cases; however, due to the potential for long-term adverse effects, in all cases they were prescribed in combination with another medication that was maintained after the corticosteroid was discontinued.

We highlight the systemic nature of RAS as well as its frequent association with systemic diseases and other correlated manifestations (pustules, EN, arthralgia). We also emphasize the importance of using oral medications to adequately control the disease and do not recommend topical medications aimed at treating local causes. Dermatologists should be consulted in managing severe cases of RAS.

References
  1. Buño IJ, Huff JC, Weston WL, et al. Elevated levels of interferon gamma, tumor necrosis factor alpha, interleukins 2, 4, and 5, but not interleukin 10, are present in recurrent aphthous stomatitis. Arch Dermatol. 1998;134:827-831.
  2. Femiano F, Lanza A, Buonaiuto C, et al. Guidelines for diagnosis and management of aphthous stomatitis. Pediatr Infect Dis J. 2007;26:728- 732.
  3. Natah SS, Konttinen YTEnattah NS, et al. Recurrent aphthous ulcers today: a review of the growing knowledge. Int J Oral Maxillofac Surg. 2004;33:221-234.
  4. Zunt SL. Recurrent aphthous stomatitis. Dermatol Clin. 2003;21:33-39.
  5. Jurge S, Kuffer R, Scully C, et al. Mucosal disease series. number VI. recurrent aphthous stomatitis. Oral Dis. 2006;12:1-21.
  6. Chams-Davatchi C, Shizarpour M, Davatchi F, et al. Comparison of oral aphthae in Behçet’s disease and idiopathic recurrent aphthous stomatitis. Adv Exp Med Biol. 2003;528:317-320.
  7. Schemel-Suárez M, López-López J, Chimenos-Küstner E. Oral ulcers: differential diagnosis and treatment [in Spanish]. Med Clin (Barc). 2015;145:499-503.
  8. S´lebioda Z, Szponar E, Kowalska A. Etiopathogenesis of recurrent aphthous stomatitis and the role of immunologic aspects: literature review. Arch Immunol Ther Exp (Warsz). 2014;62:205-215.
  9. Edgar NR, Saleh D, Miller RA. Recurrent aphthous stomatitis: a review. J Clin Aesthet Dermatol. 2017;10:26-36.
  10. Jorizzo JL, Taylor RS, Schmalstieg FC, et al. Complex aphthosis: a forme fruste of Behçet’s syndrome? J Am Acad Dermatol. 1985;13:80-84.
  11. McCarty MA, Garton RA, Jorizzo JL. Complex aphthosis and Behçet’s disease. Dermatol Clin. 2003;21:41-48.
  12. Bulur I, Melrem O. Behçet disease: new aspects. Clin Dermatol. 2017;35:421-434.
  13. Cui RZ, Rogers RS 3rd. Recurrent aphthous stomatitis. Clin Dermatol. 2016;34:475-481.
  14. Femiano F, Lanza A, Buonaiuto C, et al. Guidelines for diagnosis and management of aphthous stomatitis. Pediatr Infect Dis J. 2007;26:728-732.
  15. Ship II. Epidemiologic aspects of recurrent aphthous ulcerations. Oral Surg Oral Med Oral Pathol. 1972;33:400-406.
  16. Ship JA. Recurrent aphthous stomatitis. an update. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 1996;81:141-147.
  17. Wilhelmsen NS, Weber R, Monteiro F, et al. Correlation between histocompatibility antigens and recurrent aphthous stomatitis in the Brazilian population. Braz J Otorhinolaryngol. 2009;75:426-431.
  18. S´lebioda Z, Dorocka-Bobkowska B. Systemic and environmental risk factors for recurrent aphthous stomatitis in a Polish cohort of patients. Postepy Dermatol Alergol. 2019;36:196-201.
  19. Ship JA, Chavez EM, Doerr PA, et al. Recurrent aphthous stomatitis. Quintessence Int. 2000;31:95-112.
  20. Brocklehurst P, Tickle M, Glenny AM, et al. Systemic interventions for recurrent aphthous stomatitis (mouth ulcers). Cochrane Database Syst Rev. 2012;12:CD005411.
  21. Belenguer-Guallar I, Jiménez-Soriano Y, Ariadna Claramunt-Lozano A. Treatment of recurrent aphthous stomatitis. a literature review. J Clin Exp Dent. 2014;6:E168-E174.
  22. Bagán JV, Sanchis JM, Milián MA, et al. Recurrent aphthous stomatitis. a study of the clinical characteristics of lesions in 93 cases. J Oral Pathol Med. 1991;20:395-397.
  23. Huppert JS, Gerber MA, Deitch HR, et al. Vulvar ulcers in young females: a manifestation of aphthosis. J Pediatr Adolesc Gynecol. 2006;19:195-204. 
  24. Scully C, Porter S. Recurrent aphthous stomatitis: current concepts of etiology, pathogenesis and management. J Oral Pathol Med. 1989;18:21-27
  25. Chapel TA. Origins of penile ulcerations. Arch Androl. 1979; 3: 351-357.
  26. Lourenço SV, Hussein TP, Bologna SB, et al. Oral manifestations of inflammatory bowel disease: a review based on the observation of six cases. J Eur Acad Dermatol Venereol. 2010;24:204-207.
  27. Nico MM, Brito AE, Martins LE, et al. Oral ulcers in an immunosuppressed 5-year-old boy. Clin Exp Dermatol. 2008;33:367-368.
  28. Trakji B, Baroudi K, Kharma Y. The effect of dietary habits on the development of the recurrent aphthous stomatitis. Niger Med J. 2012;53:9-11.
  29. Lynde CB, Bruce AJ, Rogers RS 3rd. Successful treatment of complex aphthosis with colchicine and dapsone. Arch Dermatol. 2009;145:273-276. 
  30. Letsinger JAMcCarty MAJorizzo JL. Complex aphthosis: a large case series with evaluation algorithm and therapeutic ladder from topicals to thalidomide. J Am Acad Dermatol. 2005(3 pt 1);52:500-508.
  31. Hello M, Barbarot S, Bastuji-Garin S, et al. Use of thalidomide for severe recurrent aphthous stomatitis: a multicenter cohort analysis. Medicine (Baltimore). 2010;89:176-182.
References
  1. Buño IJ, Huff JC, Weston WL, et al. Elevated levels of interferon gamma, tumor necrosis factor alpha, interleukins 2, 4, and 5, but not interleukin 10, are present in recurrent aphthous stomatitis. Arch Dermatol. 1998;134:827-831.
  2. Femiano F, Lanza A, Buonaiuto C, et al. Guidelines for diagnosis and management of aphthous stomatitis. Pediatr Infect Dis J. 2007;26:728- 732.
  3. Natah SS, Konttinen YTEnattah NS, et al. Recurrent aphthous ulcers today: a review of the growing knowledge. Int J Oral Maxillofac Surg. 2004;33:221-234.
  4. Zunt SL. Recurrent aphthous stomatitis. Dermatol Clin. 2003;21:33-39.
  5. Jurge S, Kuffer R, Scully C, et al. Mucosal disease series. number VI. recurrent aphthous stomatitis. Oral Dis. 2006;12:1-21.
  6. Chams-Davatchi C, Shizarpour M, Davatchi F, et al. Comparison of oral aphthae in Behçet’s disease and idiopathic recurrent aphthous stomatitis. Adv Exp Med Biol. 2003;528:317-320.
  7. Schemel-Suárez M, López-López J, Chimenos-Küstner E. Oral ulcers: differential diagnosis and treatment [in Spanish]. Med Clin (Barc). 2015;145:499-503.
  8. S´lebioda Z, Szponar E, Kowalska A. Etiopathogenesis of recurrent aphthous stomatitis and the role of immunologic aspects: literature review. Arch Immunol Ther Exp (Warsz). 2014;62:205-215.
  9. Edgar NR, Saleh D, Miller RA. Recurrent aphthous stomatitis: a review. J Clin Aesthet Dermatol. 2017;10:26-36.
  10. Jorizzo JL, Taylor RS, Schmalstieg FC, et al. Complex aphthosis: a forme fruste of Behçet’s syndrome? J Am Acad Dermatol. 1985;13:80-84.
  11. McCarty MA, Garton RA, Jorizzo JL. Complex aphthosis and Behçet’s disease. Dermatol Clin. 2003;21:41-48.
  12. Bulur I, Melrem O. Behçet disease: new aspects. Clin Dermatol. 2017;35:421-434.
  13. Cui RZ, Rogers RS 3rd. Recurrent aphthous stomatitis. Clin Dermatol. 2016;34:475-481.
  14. Femiano F, Lanza A, Buonaiuto C, et al. Guidelines for diagnosis and management of aphthous stomatitis. Pediatr Infect Dis J. 2007;26:728-732.
  15. Ship II. Epidemiologic aspects of recurrent aphthous ulcerations. Oral Surg Oral Med Oral Pathol. 1972;33:400-406.
  16. Ship JA. Recurrent aphthous stomatitis. an update. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 1996;81:141-147.
  17. Wilhelmsen NS, Weber R, Monteiro F, et al. Correlation between histocompatibility antigens and recurrent aphthous stomatitis in the Brazilian population. Braz J Otorhinolaryngol. 2009;75:426-431.
  18. S´lebioda Z, Dorocka-Bobkowska B. Systemic and environmental risk factors for recurrent aphthous stomatitis in a Polish cohort of patients. Postepy Dermatol Alergol. 2019;36:196-201.
  19. Ship JA, Chavez EM, Doerr PA, et al. Recurrent aphthous stomatitis. Quintessence Int. 2000;31:95-112.
  20. Brocklehurst P, Tickle M, Glenny AM, et al. Systemic interventions for recurrent aphthous stomatitis (mouth ulcers). Cochrane Database Syst Rev. 2012;12:CD005411.
  21. Belenguer-Guallar I, Jiménez-Soriano Y, Ariadna Claramunt-Lozano A. Treatment of recurrent aphthous stomatitis. a literature review. J Clin Exp Dent. 2014;6:E168-E174.
  22. Bagán JV, Sanchis JM, Milián MA, et al. Recurrent aphthous stomatitis. a study of the clinical characteristics of lesions in 93 cases. J Oral Pathol Med. 1991;20:395-397.
  23. Huppert JS, Gerber MA, Deitch HR, et al. Vulvar ulcers in young females: a manifestation of aphthosis. J Pediatr Adolesc Gynecol. 2006;19:195-204. 
  24. Scully C, Porter S. Recurrent aphthous stomatitis: current concepts of etiology, pathogenesis and management. J Oral Pathol Med. 1989;18:21-27
  25. Chapel TA. Origins of penile ulcerations. Arch Androl. 1979; 3: 351-357.
  26. Lourenço SV, Hussein TP, Bologna SB, et al. Oral manifestations of inflammatory bowel disease: a review based on the observation of six cases. J Eur Acad Dermatol Venereol. 2010;24:204-207.
  27. Nico MM, Brito AE, Martins LE, et al. Oral ulcers in an immunosuppressed 5-year-old boy. Clin Exp Dermatol. 2008;33:367-368.
  28. Trakji B, Baroudi K, Kharma Y. The effect of dietary habits on the development of the recurrent aphthous stomatitis. Niger Med J. 2012;53:9-11.
  29. Lynde CB, Bruce AJ, Rogers RS 3rd. Successful treatment of complex aphthosis with colchicine and dapsone. Arch Dermatol. 2009;145:273-276. 
  30. Letsinger JAMcCarty MAJorizzo JL. Complex aphthosis: a large case series with evaluation algorithm and therapeutic ladder from topicals to thalidomide. J Am Acad Dermatol. 2005(3 pt 1);52:500-508.
  31. Hello M, Barbarot S, Bastuji-Garin S, et al. Use of thalidomide for severe recurrent aphthous stomatitis: a multicenter cohort analysis. Medicine (Baltimore). 2010;89:176-182.
Issue
Cutis - 113(4)
Issue
Cutis - 113(4)
Page Number
171-173
Page Number
171-173
Publications
Publications
Topics
Article Type
Display Headline
Recurrent Aphthous Stomatitis: Clinical Experience From a University Hospital in Brazil
Display Headline
Recurrent Aphthous Stomatitis: Clinical Experience From a University Hospital in Brazil
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>Pinto</fileName> <TBEID>0C02F48F.SIG</TBEID> <TBUniqueIdentifier>NJ_0C02F48F</TBUniqueIdentifier> <newsOrJournal>Journal</newsOrJournal> <publisherName>Frontline Medical Communications Inc.</publisherName> <storyname>Pinto</storyname> <articleType>1</articleType> <TBLocation>Copyfitting-CT</TBLocation> <QCDate/> <firstPublished>20240408T080710</firstPublished> <LastPublished>20240408T080710</LastPublished> <pubStatus qcode="stat:"/> <embargoDate/> <killDate/> <CMSDate>20240408T080710</CMSDate> <articleSource/> <facebookInfo/> <meetingNumber/> <byline>Nathalia Targa Pinto, MD; Silvia Vanessa Lourenço, DDS; Marcello</byline> <bylineText>Nathalia Targa Pinto, MD; Silvia Vanessa Lourenço, DDS; Marcello Menta Simonsen Nico, MD </bylineText> <bylineFull>Nathalia Targa Pinto, MD; Silvia Vanessa Lourenço, DDS; Marcello</bylineFull> <bylineTitleText/> <USOrGlobal/> <wireDocType/> <newsDocType/> <journalDocType/> <linkLabel/> <pageRange>171-173</pageRange> <citation/> <quizID/> <indexIssueDate/> <itemClass qcode="ninat:text"/> <provider qcode="provider:"> <name/> <rightsInfo> <copyrightHolder> <name/> </copyrightHolder> <copyrightNotice/> </rightsInfo> </provider> <abstract/> <metaDescription>To the Editor:Recurrent aphthous stomatitis (RAS) is a mucocutaneous condition characterized by single or multiple, painful,1,2 round ulcerations of variable si</metaDescription> <articlePDF/> <teaserImage/> <title>Recurrent Aphthous Stomatitis: Clinical Experience From a University Hospital in Brazil</title> <deck/> <disclaimer/> <AuthorList/> <articleURL/> <doi/> <pubMedID/> <publishXMLStatus/> <publishXMLVersion>1</publishXMLVersion> <useEISSN>0</useEISSN> <urgency/> <pubPubdateYear>2024</pubPubdateYear> <pubPubdateMonth>April</pubPubdateMonth> <pubPubdateDay/> <pubVolume>113</pubVolume> <pubNumber>4</pubNumber> <wireChannels/> <primaryCMSID/> <CMSIDs> <CMSID>2161</CMSID> </CMSIDs> <keywords> <keyword>recurrent aphthouse stomatitis</keyword> </keywords> <seeAlsos/> <publications_g> <publicationData> <publicationCode>CT</publicationCode> <pubIssueName>April 2024</pubIssueName> <pubArticleType>Original Articles | 2161</pubArticleType> <pubTopics/> <pubCategories/> <pubSections/> <journalTitle>Cutis</journalTitle> <journalFullTitle>Cutis</journalFullTitle> <copyrightStatement>Copyright 2015 Frontline Medical Communications Inc., Parsippany, NJ, USA. All rights reserved.</copyrightStatement> </publicationData> </publications_g> <publications> <term canonical="true">12</term> </publications> <sections> <term canonical="true">104</term> </sections> <topics> <term canonical="true">27442</term> </topics> <links/> </header> <itemSet> <newsItem> <itemMeta> <itemRole>Main</itemRole> <itemClass>text</itemClass> <title>Recurrent Aphthous Stomatitis: Clinical Experience From a University Hospital in Brazil</title> <deck/> </itemMeta> <itemContent> <p>To the Editor:<br/><br/>Recurrent aphthous stomatitis (RAS) is a mucocutaneous condition characterized by single or multiple, painful,<sup>1,2</sup> round ulcerations of variable sizes with a tendency for recurrence, most commonly located in nonkeratinized areas of the oral mucosa. Pathergy commonly is observed.<sup>3</sup> Although many authors consider the terms <i>RAS</i> and<i>aphtha</i> to be synonymous,<sup>4,5</sup> differentiating the clinical lesion (aphthous ulceration) from the disease (aphtha or RAS) can be useful, as several other diseases can at times manifest with similar ulcers (called <i>aphthoid lesions</i>), such as pemphigus vulgaris, mucous membrane pemphigoid, and erythema multiforme.<sup>6</sup></p> <p>It is estimated that approximately 20% of individuals worldwide have at least one episode of aphtha during their lifetime,<sup>7</sup> and it is considered the most common disease of the oral mucosa.<sup>8,9</sup> However, only patients presenting with severe acute outbreaks or frequent relapses typically seek medical treatment. Clinically, aphthous ulcers are classified as <i>aphtha minor</i> (small number of small lesions), <i>aphtha major</i> (large deep lesions that also can affect the minor salivary glands with intense necrosis, difficulty in healing, and mucosal scarring), and <i>aphtha herpetiformis</i> (innumerous tiny lesions that reappear in recurring outbreaks).<sup>1-3</sup> The term <i>complex aphthosis</i> was introduced in 1985<sup>10</sup> and is defined as recurrent oral and genital aphthous ulcerations or recurring multiple oral aphthous ulcers in the absence of systemic manifestations or Behçet disease<sup>11,12</sup>; however, complex aphthosis also has been reported as frequent episodes of ulcerations that may be associated with systemic diseases including Behçet disease.<sup>13,14<br/><br/></sup>Currently, RAS is considered an immunologically mediated alteration in cutaneous mucosal reactivity with a multifactorial systemic cause. Underlying conditions such as Behçet disease, inflammatory bowel disease (IBD), iatrogenic immunosuppression (eg, following solid organ transplantation), AIDS, and cyclic neutropenia may or may not be detected.<sup>11-13</sup> <br/><br/>Our retrospective study explored the systemic nature of RAS. We reviewed patient records to evaluate underlying systemic conditions associated with the diagnosis of RAS and the use of oral medications in managing the disease. Medical records from the Department of Dermatology of the University of São Paulo, Brazil, from 2003 to 2017 were reviewed to identify patients with a diagnosis of RAS. Clinical classification of RAS—minor, major, or herpetiform—as well as the presence of aphthous lesions in other locations and the presence of other associated inflammatory cutaneous manifestations also were noted. Associated systemic diseases and treatments for RAS were recorded. Patients for whom the diagnosis of RAS was changed during follow-up were excluded. Because this was a retrospective analysis of medical records and without any patient risk, informed consent was not needed. <br/><br/>Medical records for 125 patients were reviewed; 63 were male (50.4%), and 62 were female (49.6%). The age at onset of symptoms, which ranged from a few months after birth to 74 years, was reported in only 92 (73.6%) patient medical records. Of these, 30 (32.6%) reported onset before 20 years of age, 39 (42.4%) between 20 and 39 years, 17 (18.5%) between 40 and 59 years, and 6 (6.5%) at 60 years or older. Morphologically, 72 (57.6%) had minor, 42 (33.6%) had major, and 11 (8.8%) had herpetiform aphthous ulcers. None of the patients presented with sporadic lesions; the disease was long-standing and persistent in all cases (complex aphthosis). <br/><br/>Regarding the location of the ulcers, 92 (73.6%) patients had lesions on the oral mucosa only. Some patients had lesions in more than one site in addition to the oral mucosa: 32 (25.6%) had aphthae in the genital/groin region and 4 (3.2%) presented with perianal/anal aphthae. Nineteen patients (19.2%) presented other cutaneous manifestations in addition to aphthae: 11 (45.8%) had folliculitis/pseudofolliculitis, and 8 (33.3%) had erythema nodosum (EN). Eight patients (33.3%) presented with uveitis, and 6 (25%) presented with concomitant arthralgia/arthritis. Fifty-four patients (43.2%) had confirmed or suspected associated disease: Behçet disease (21 [38.9%]), IBD (10 [18.5%]), solid organ transplantation (7 [13.0%])(kidney, 4 [57.1%]; heart, 2 [28.6%]; liver, 1 [14.3%]), HIV infection (6 [11.1%]), lymphoma (1 [1.9%]), aplastic anemia (1 [1.9%]), or myelodysplastic syndrome (1 [1.9%]). Ten patients (18.5%) presented with other diseases under investigation (eg, unidentified rheumatologic disease, unexplained neutropenia, undiagnosed immunodeficiencies, autoinflammatory syndromes, possible cyclic neutropenia). <br/><br/>Biopsies of the oral mucosa were performed in 31 patients. Histopathologic findings will be discussed in a future publication (unpublished data).<br/><br/>Five patients (4.0%) were lost to follow-up and did not receive treatment; 10 (8.0%) received only topical treatment (analgesics and/or corticosteroids). All 9 (7.2%) patients undergoing intralesional corticosteroid injections also were on a systemic treatment. One hundred ten (88.0%) patients were treated systemically—with colchicine (84/110 [76.4%]), thalidomide (43/110 [39.1%]), small pulses of oral corticosteroids (26 [23.6%]), dapsone (12/110 [10.9%]), or pentoxifylline (3 [2.7%]). Furthermore, in patients with associated diseases, treatment of the underlying condition was conducted when available, and follow-up was carried out in conjunction with the appropriate specialists. For treatment of the associated disease, patients received other medications such as methotrexate, azathioprine, cyclophosphamide, intravenous corticosteroid pulse, and immunobiologics.<br/><br/>The prevalence of RAS between sexes in our study population was similar (50.4% male; 49.6% female). Results from prior studies have been mixed; some reported a higher prevalence in females,<sup>15-18</sup> while others found no predilection for sex among patients diagnosed with RAS.<sup>19,20</sup> In our analysis, 75% of patients experienced symptoms of RAS before 40 years of age; in prior studies, up to 56% of patients experienced symptoms between the ages of 20 and 40 years.<sup>21,22<br/><br/></sup>In our study, 26.4% of patients had extraoral aphthae. Genital lesions have been described as infrequent,<sup>23</sup> and lesions manifesting in other mucous membranes or on the skin are rare.<sup>24</sup> A study reported genital involvement in 8% to 13% of patients with oral aphtha.<sup>25</sup> We observed genital involvement in 25.6% of patients. Likewise, this higher value may be due to our study population of patients referred to our university hospital. In our study, 19.2% of patients presented with other inflammatory manifestations in addition to aphthous ulcerations (eg, folliculitis, EN, uveitis, arthritis). As dermatologists in a tertiary reference hospital, we actively look for such associations in every aphtha patient, which may not be the case in many nondermatologic oral care services. <br/><br/>In our study population, 43.2% of patients were diagnosed with or were under investigation for systemic diseases known to be associated with RAS. We found associations with Behçet disease most frequently, followed by IBD,<sup>26</sup> solid organ transplantation, and HIV. In this group of patients, the respective systemic disease was active or poorly controlled. In transplant recipients, aphtha major was the most common type, similar to other studies.<sup>27</sup> We observed no notable difference in the clinical picture of the oral ulcers in patients with a well-established systemic disease vs those without. <br/><br/>Most of our cases did not present findings other than aphtha, indicating that the intrinsic defect that predisposes to RAS is always systemic. Even mild and sporadic cases may be attributable to a systemic disorder of cutaneous-mucosal reactivity. The predisposition to RAS never originates in the oral cavity, hence the confusion caused and the uselessness of studies that relate aphthae to factors such as local food allergies, pH changes, or local infection with microorganisms.<sup>5,28</sup> The disease course (reducing the frequency of lesion appearance and accelerating the healing of extensive lesions) is only modified with systemic treatment, with local measures proving to be only moderately useful to relieve pain. We believe that RAS can in many ways be compared to EN and pyoderma gangrenosum (PG): some systemic conditions that predispose patients to EN and PG also may predispose them to RAS (eg, IBD, hematologic disorders). Similar to RAS, many cases of EN and PG are idiopathic. In addition, pathergy also occurs in PG.<sup>11,13</sup> <br/><br/>We were unable to observe or establish any predictive clinical element that could indicate a better or worse response to the prescribed treatments, which also has been noted by other authors.<sup>3,4</sup> Treatment of RAS is empiric, generally starting with drugs that are easier to prescribe and with fewer adverse effects, then progressing to more complex drugs when a good response is not obtained. Colchicine was the most commonly prescribed medication (76.4% [84/110]). It has been proposed by several authors<sup>3,4</sup> as a first-line systemic medication for the treatment of recurrent aphthae, as it has been shown to be effective and safe. The dosage ranged from 0.5 mg twice daily to 0.5 mg 4 times daily. Dapsone is an established drug for aphtha<sup>29,30</sup> and was used in 12 of our patients. The dosage used in our patients ranged from 50 to 100 mg/d. Adverse effects such as hemolytic anemia frequently are seen, and one of the patients in our study developed DRESS (drug reaction with eosinophilia and systemic symptoms) syndrome in response to dapsone. In 7 cases, colchicine and dapsone were used together, which is believed to potentiate the therapeutic effects. This combination may be useful in patients for whom thalidomide cannot be used or those who have not improved with monotherapy.<sup>29</sup> Thalidomide is considered one of the most effective drugs for RAS.<sup>30,31</sup> Forty-three patients in our analysis were treated with thalidomide,<sup> </sup>usually as a first choice. The dosage ranged from 100 to 200 mg/d. It was mainly chosen in disabling pediatric cases, adult men with aphthous major, and women with no risk for pregnancy. Due to its potential adverse effects, thalidomide has been recommended when there is no response with other medications that are dose dependent; severe adverse effects such as thromboembolism and peripheral neuropathy are rare.<sup>31</sup> Oral corticosteroids were used in 26 patients, aiming at rapid improvement in very symptomatic cases; however, due to the potential for long-term adverse effects, in all cases they were prescribed in combination with another medication that was maintained after the corticosteroid was discontinued.<br/><br/>We highlight the systemic nature of RAS as well as its frequent association with systemic diseases and other correlated manifestations (pustules, EN, arthralgia). We also emphasize the importance of using oral medications to adequately control the disease and do not recommend topical medications aimed at treating local causes. Dermatologists should be consulted in managing severe cases of RAS.</p> <h2>References</h2> <p class="reference"> 1. Buño IJ, Huff JC, Weston WL, et al. Elevated levels of interferon gamma, tumor necrosis factor alpha, interleukins 2, 4, and 5, but not interleukin 10, are present in recurrent aphthous stomatitis. <i>Arch Dermatol.</i> 1998;134:827-831. <br/><br/> 2. Femiano F, Lanza A, Buonaiuto C, et al. Guidelines for diagnosis and management of aphthous stomatitis. <i>Pediatr Infect Dis J.</i> 2007;26:728- 732. <br/><br/> 3. Natah SS, <span class="Hyperlink"><a href="https://www.ncbi.nlm.nih.gov/pubmed/?term=Konttinen%20YT%5BAuthor%5D&amp;cauthor=true&amp;cauthor_uid=15287304">Konttinen YT</a></span>, <span class="Hyperlink"><a href="https://www.ncbi.nlm.nih.gov/pubmed/?term=Enattah%20NS%5BAuthor%5D&amp;cauthor=true&amp;cauthor_uid=15287304">Enattah NS</a></span>, et al. Recurrent aphthous ulcers today: a review of the growing knowledge. <span class="Hyperlink"><i><a href="https://www.ncbi.nlm.nih.gov/pubmed/?term=.+Recurrent+aphthous+ulcers+today%3A+a+review+of+the+growing+knowledge">Int J Oral Maxillofac Surg.</a></i></span><i> </i>2004;33:221-234.<br/><br/> 4. Zunt SL. Recurrent aphthous stomatitis. <i>Dermatol Clin.</i> 2003;21:33-39. <br/><br/> 5. Jurge S, Kuffer R, Scully C, et al. Mucosal disease series. number VI. recurrent aphthous stomatitis. <i>Oral Dis.</i> 2006;12:1-21.<br/><br/> 6. Chams-Davatchi C, Shizarpour M, Davatchi F, et al. Comparison of oral aphthae in Behçet’s disease and idiopathic recurrent aphthous stomatitis. <i>Adv Exp Med Biol.</i> 2003;528:317-320.</p> <p class="reference"> 7. Schemel-Suárez M, López-López J, Chimenos-Küstner E. Oral ulcers: differential diagnosis and treatment [in Spanish]. <i>Med Clin (Barc).</i> 2015;145:499-503.<br/><br/> 8. S´lebioda Z, Szponar E, Kowalska A. <span class="highlight">Etiopathogenesis</span> of <span class="highlight">recurrent</span> <span class="highlight">aphthous stomatitis</span> and the <span class="highlight">role</span> of <span class="highlight">immunologic</span> <span class="highlight">aspects</span>: literature review. <span class="Hyperlink"><i><a href="https://www.ncbi.nlm.nih.gov/pubmed/24217985">Arch Immunol Ther Exp (Warsz).</a></i></span> 2014;62:205-215.<br/><br/> 9. Edgar NR, Saleh D, Miller RA. Recurrent aphthous stomatitis: a review. <i>J Clin Aesthet Dermatol.</i> 2017;10:26-36.<br/><br/>10. Jorizzo JL, Taylor RS, Schmalstieg FC, et al. Complex aphthosis: a forme fruste of Behçet’s syndrome? <i>J Am Acad Dermatol.</i> 1985;13:80-84.<br/><br/>11. McCarty MA, Garton RA, Jorizzo JL. Complex aphthosis and Behçet’s disease. <i>Dermatol Clin.</i> 2003;21:41-48.<br/><br/>12. Bulur I, Melrem O. Behçet disease: new aspects. <i>Clin Dermatol.</i> 2017;35:421-434.<br/><br/>13. Cui RZ, Rogers RS 3rd. Recurrent aphthous stomatitis. <i>Clin Dermatol.</i> 2016;34:475-481.<br/><br/>14. Femiano F, Lanza A, Buonaiuto C, et al. Guidelines for diagnosis and management of aphthous stomatitis. <i>Pediatr Infect Dis J.</i> 2007;26:728-732. <br/><br/>15. Ship II. Epidemiologic aspects of recurrent aphthous ulcerations. <i>Oral Surg Oral Med Oral Pathol. </i>1972;33:400-406. <br/><br/>16. Ship JA. Recurrent aphthous stomatitis. an update. <i>Oral Surg Oral Med Oral Pathol Oral Radiol Endod.</i> 1996;81:141-147.<br/><br/>17. Wilhelmsen NS, <span class="Hyperlink"><a href="https://www.ncbi.nlm.nih.gov/pubmed/?term=Weber%20R%5BAuthor%5D&amp;cauthor=true&amp;cauthor_uid=19649495">Weber R</a></span>, Monteiro F, et al. Correlation between histocompatibility antigens and <span class="highlight">recurrent</span> <span class="highlight">aphthous stomatitis</span> in the Brazilian population. <i>Braz J Otorhinolaryngol.</i> 2009;75:426-431.<br/><br/>18. S´lebioda Z, <span class="Hyperlink"><a href="https://www.ncbi.nlm.nih.gov/pubmed/?term=Dorocka-Bobkowska%20B%5BAuthor%5D&amp;cauthor=true&amp;cauthor_uid=31320854">Dorocka-Bobkowska B</a></span>. <span class="highlight">Systemic</span> and environmental risk factors for recurrent <span class="highlight">aphthous</span> stomatitis in a Polish cohort of patients. <i>Postepy Dermatol Alergol.</i> 2019;36:196-201.<br/><br/>19. Ship JA, Chavez EM, Doerr PA, et al. Recurrent aphthous stomatitis. <i>Quintessence Int.</i> 2000;31:95-112.<br/><br/>20. Brocklehurst P, Tickle M, Glenny AM, et al. Systemic interventions for recurrent aphthous stomatitis (mouth ulcers). <i>Cochrane Database Syst Rev.</i> 2012;12:CD005411.<br/><br/>21. Belenguer-Guallar I, Jiménez-Soriano Y, Ariadna Claramunt-Lozano A. Treatment of recurrent aphthous stomatitis. a literature review. <i>J Clin Exp Dent.</i> 2014;6:E168-E174.<br/><br/>22. Bagán JV, Sanchis JM, Milián MA, et al. Recurrent aphthous stomatitis. a study of the clinical characteristics of lesions in 93 cases. <i>J Oral Pathol Med.</i> 1991;20:395-397. <br/><br/>23. Huppert JS, Gerber MA, Deitch HR, et al. Vulvar ulcers in young females: a manifestation of aphthosis. <i>J Pediatr Adolesc Gynecol.</i> 2006;19:195-204. <br/><br/>24. Scully C, Porter S. Recurrent aphthous stomatitis: current concepts of etiology, pathogenesis and management. <i>J Oral Pathol Med. </i>1989;18:21-27<br/><br/>25. Chapel TA. Origins of penile ulcerations. <i>Arch Androl.</i> 1979; 3: 351-357.<br/><br/>26. Lourenço SV, Hussein TP, Bologna SB, et al. Oral manifestations of inflammatory bowel disease: a review based on the observation of six cases. <i>J Eur Acad Dermatol Venereol.</i> 2010;24:204-207.<br/><br/>27. Nico MM, Brito AE, Martins LE, et al. Oral ulcers in an immunosuppressed 5-year-old boy. <i>Clin Exp Dermatol.</i> 2008;33:367-368.<br/><br/>28. Trakji B, Baroudi K, Kharma Y. The effect of dietary habits on the development of the recurrent aphthous stomatitis. <i>Niger Med J.</i> 2012;53:9-11. <br/><br/>29. Lynde CB, Bruce AJ, Rogers RS 3rd. Successful treatment of complex aphthosis with colchicine and dapsone. <i>Arch Dermatol.</i> 2009;145:273-276. <br/><br/>30. <span class="Hyperlink"><a href="https://www.ncbi.nlm.nih.gov/pubmed/?term=Letsinger%20JA%5BAuthor%5D&amp;cauthor=true&amp;cauthor_uid=15761429">Letsinger JA</a></span>, <span class="Hyperlink"><a href="https://www.ncbi.nlm.nih.gov/pubmed/?term=McCarty%20MA%5BAuthor%5D&amp;cauthor=true&amp;cauthor_uid=15761429">McCarty MA</a></span>, <span class="Hyperlink"><a href="https://www.ncbi.nlm.nih.gov/pubmed/?term=Jorizzo%20JL%5BAuthor%5D&amp;cauthor=true&amp;cauthor_uid=15761429">Jorizzo JL</a></span>. Complex aphthosis: a large case series with evaluation algorithm and therapeutic ladder from topicals to thalidomide. <span class="Hyperlink"><i><a href="https://www.ncbi.nlm.nih.gov/pubmed/15761429">J Am Acad Dermatol.</a></i></span> 2005(3 pt 1);52:500-508.<br/><br/>31. Hello M, Barbarot S, Bastuji-Garin S, et al. Use of thalidomide for severe recurrent aphthous stomatitis: a multicenter cohort analysis. <i>Medicine (Baltimore).</i> 2010;89:176-182.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>bio</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="disclosure">From the Universidade de São Paulo, Brazil. Drs. Pinto and Nico are from the Department of Dermatology, Faculdade de Medicina, and Dr. Lourenço is from the Department of Pathology, Faculdade de Odontologia.</p> <p class="disclosure">The authors report no conflict of interest. <br/><br/>Correspondence: Marcello Menta Simonsen Nico, MD, Departamento de Dermatologia, Faculdade de Medicina da Universidade de São Paulo, Brasil, R. Itapeva 500-3A. CEP-01332-000, São Paulo, Brasil (mentanico@hotmail.com).<br/><br/><em>Cutis.</em> 2024 April;113(4):171-173. doi:10.12788/cutis.0992.</p> </itemContent> </newsItem> <newsItem> <itemMeta> <itemRole>in</itemRole> <itemClass>text</itemClass> <title/> <deck/> </itemMeta> <itemContent> <p class="insidehead">Practice <strong>Points</strong></p> <ul class="insidebody"> <li>The process that leads to the formation of aphthous ulcerations is always systemic, not local, even in the absence of a diagnosable systemic disease.</li> <li>Relapsing cases of aphthae should be treated with systemic medication.</li> </ul> </itemContent> </newsItem> </itemSet></root>
Inside the Article

Practice Points

  • The process that leads to the formation of aphthous ulcerations is always systemic, not local, even in the absence of a diagnosable systemic disease.
  • Relapsing cases of aphthae should be treated with systemic medication.
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