Do scare tactics work?

Article Type
Changed
Thu, 11/10/2022 - 10:40

I suspect that you have heard about or maybe read the recent Associated Press story reporting that four daycare workers in Hamilton, Miss., have been charged with felony child abuse for intentionally scaring the children “who didn’t clean up or act good” by wearing a Halloween mask and yelling in their faces. I can have some sympathy for those among us who choose to spend their days tending a flock of sometimes unruly and mischievous toddlers and preschoolers. But, I think one would be hard pressed to find very many adults who would condone the strategy of these misguided daycare providers. Not surprisingly, the parents of some of these children describe their children as traumatized and having disordered sleep.

Dr. William G. Wilkoff practiced primary care pediatrics in Brunswick, Maine, for nearly 40 years.
Dr. William G. Wilkoff

The news report of this incident in Mississippi doesn’t tell us if these daycare providers had used this tactic in the past. One wonders whether they had found less dramatic verbal threats just weren’t as effective as they had hoped and so decided to go all out.

How effective is fear in changing behavior? Certainly, we have all experienced situations in which a frightening experience has caused us to avoid places, people, and activities. But, is a fear-focused strategy one that health care providers should include in their quiver as we try to mold patient behavior? As luck would have it, 2 weeks before this news story broke I encountered a global study from 84 countries that sought to answer this question (Affect Sci. 2022 Sep. doi: 10.1007/s42761-022-00128-3).

Using the WHO four-point advice about COVID prevention (stay home/avoid shops/use face covering/isolate if exposed) as a model the researchers around the world reviewed the responses of 16,000 individuals. They found that there was no difference in the effectiveness of the message whether it was framed as a negative (“you have so much to lose”) or a positive (“you have so much to gain”). However, investigators observed that the negatively framed presentations generated significantly more anxiety in the respondents. The authors of the paper conclude that if there is no significant difference in the effectiveness, why would we chose a negatively framed presentation that is likely to generate anxiety that we know is associated with increased morbidity and mortality. From a purely public health perspective, it doesn’t make sense and is counterproductive.

I guess if we look back to the old carrot and stick metaphor we shouldn’t be surprised by the findings in this paper. If one’s only goal is to get a group of young preschoolers to behave by scaring the b’geezes out of them with a mask or a threat of bodily punishment, then go for it. Scare tactics will probably work just as well as offering a well-chosen reward system. However, the devil is in the side effects. It’s the same argument that I give to parents who argue that spanking works. Of course it does, but it has a narrow margin for safety and can set up ripples of negative side effects that can destroy healthy parent-child relationships.

The bottom line of this story is the sad truth that somewhere along the line someone failed to effectively train these four daycare workers. But, do we as health care providers need to rethink our training? Have we forgotten our commitment to “First do no harm?” As we craft our messaging have we thought enough about the potential side effects of our attempts at scaring the public into following our suggestions?

Dr. Wilkoff practiced primary care pediatrics in Brunswick, Maine, for nearly 40 years. He has authored several books on behavioral pediatrics, including “How to Say No to Your Toddler.” Other than a Littman stethoscope he accepted as a first-year medical student in 1966, Dr. Wilkoff reports having nothing to disclose. Email him at [email protected].

Publications
Topics
Sections

I suspect that you have heard about or maybe read the recent Associated Press story reporting that four daycare workers in Hamilton, Miss., have been charged with felony child abuse for intentionally scaring the children “who didn’t clean up or act good” by wearing a Halloween mask and yelling in their faces. I can have some sympathy for those among us who choose to spend their days tending a flock of sometimes unruly and mischievous toddlers and preschoolers. But, I think one would be hard pressed to find very many adults who would condone the strategy of these misguided daycare providers. Not surprisingly, the parents of some of these children describe their children as traumatized and having disordered sleep.

Dr. William G. Wilkoff practiced primary care pediatrics in Brunswick, Maine, for nearly 40 years.
Dr. William G. Wilkoff

The news report of this incident in Mississippi doesn’t tell us if these daycare providers had used this tactic in the past. One wonders whether they had found less dramatic verbal threats just weren’t as effective as they had hoped and so decided to go all out.

How effective is fear in changing behavior? Certainly, we have all experienced situations in which a frightening experience has caused us to avoid places, people, and activities. But, is a fear-focused strategy one that health care providers should include in their quiver as we try to mold patient behavior? As luck would have it, 2 weeks before this news story broke I encountered a global study from 84 countries that sought to answer this question (Affect Sci. 2022 Sep. doi: 10.1007/s42761-022-00128-3).

Using the WHO four-point advice about COVID prevention (stay home/avoid shops/use face covering/isolate if exposed) as a model the researchers around the world reviewed the responses of 16,000 individuals. They found that there was no difference in the effectiveness of the message whether it was framed as a negative (“you have so much to lose”) or a positive (“you have so much to gain”). However, investigators observed that the negatively framed presentations generated significantly more anxiety in the respondents. The authors of the paper conclude that if there is no significant difference in the effectiveness, why would we chose a negatively framed presentation that is likely to generate anxiety that we know is associated with increased morbidity and mortality. From a purely public health perspective, it doesn’t make sense and is counterproductive.

I guess if we look back to the old carrot and stick metaphor we shouldn’t be surprised by the findings in this paper. If one’s only goal is to get a group of young preschoolers to behave by scaring the b’geezes out of them with a mask or a threat of bodily punishment, then go for it. Scare tactics will probably work just as well as offering a well-chosen reward system. However, the devil is in the side effects. It’s the same argument that I give to parents who argue that spanking works. Of course it does, but it has a narrow margin for safety and can set up ripples of negative side effects that can destroy healthy parent-child relationships.

The bottom line of this story is the sad truth that somewhere along the line someone failed to effectively train these four daycare workers. But, do we as health care providers need to rethink our training? Have we forgotten our commitment to “First do no harm?” As we craft our messaging have we thought enough about the potential side effects of our attempts at scaring the public into following our suggestions?

Dr. Wilkoff practiced primary care pediatrics in Brunswick, Maine, for nearly 40 years. He has authored several books on behavioral pediatrics, including “How to Say No to Your Toddler.” Other than a Littman stethoscope he accepted as a first-year medical student in 1966, Dr. Wilkoff reports having nothing to disclose. Email him at [email protected].

I suspect that you have heard about or maybe read the recent Associated Press story reporting that four daycare workers in Hamilton, Miss., have been charged with felony child abuse for intentionally scaring the children “who didn’t clean up or act good” by wearing a Halloween mask and yelling in their faces. I can have some sympathy for those among us who choose to spend their days tending a flock of sometimes unruly and mischievous toddlers and preschoolers. But, I think one would be hard pressed to find very many adults who would condone the strategy of these misguided daycare providers. Not surprisingly, the parents of some of these children describe their children as traumatized and having disordered sleep.

Dr. William G. Wilkoff practiced primary care pediatrics in Brunswick, Maine, for nearly 40 years.
Dr. William G. Wilkoff

The news report of this incident in Mississippi doesn’t tell us if these daycare providers had used this tactic in the past. One wonders whether they had found less dramatic verbal threats just weren’t as effective as they had hoped and so decided to go all out.

How effective is fear in changing behavior? Certainly, we have all experienced situations in which a frightening experience has caused us to avoid places, people, and activities. But, is a fear-focused strategy one that health care providers should include in their quiver as we try to mold patient behavior? As luck would have it, 2 weeks before this news story broke I encountered a global study from 84 countries that sought to answer this question (Affect Sci. 2022 Sep. doi: 10.1007/s42761-022-00128-3).

Using the WHO four-point advice about COVID prevention (stay home/avoid shops/use face covering/isolate if exposed) as a model the researchers around the world reviewed the responses of 16,000 individuals. They found that there was no difference in the effectiveness of the message whether it was framed as a negative (“you have so much to lose”) or a positive (“you have so much to gain”). However, investigators observed that the negatively framed presentations generated significantly more anxiety in the respondents. The authors of the paper conclude that if there is no significant difference in the effectiveness, why would we chose a negatively framed presentation that is likely to generate anxiety that we know is associated with increased morbidity and mortality. From a purely public health perspective, it doesn’t make sense and is counterproductive.

I guess if we look back to the old carrot and stick metaphor we shouldn’t be surprised by the findings in this paper. If one’s only goal is to get a group of young preschoolers to behave by scaring the b’geezes out of them with a mask or a threat of bodily punishment, then go for it. Scare tactics will probably work just as well as offering a well-chosen reward system. However, the devil is in the side effects. It’s the same argument that I give to parents who argue that spanking works. Of course it does, but it has a narrow margin for safety and can set up ripples of negative side effects that can destroy healthy parent-child relationships.

The bottom line of this story is the sad truth that somewhere along the line someone failed to effectively train these four daycare workers. But, do we as health care providers need to rethink our training? Have we forgotten our commitment to “First do no harm?” As we craft our messaging have we thought enough about the potential side effects of our attempts at scaring the public into following our suggestions?

Dr. Wilkoff practiced primary care pediatrics in Brunswick, Maine, for nearly 40 years. He has authored several books on behavioral pediatrics, including “How to Say No to Your Toddler.” Other than a Littman stethoscope he accepted as a first-year medical student in 1966, Dr. Wilkoff reports having nothing to disclose. Email him at [email protected].

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

Cutaneous and Subcutaneous Perineuriomas in 2 Pediatric Patients

Article Type
Changed
Wed, 11/09/2022 - 10:00
Display Headline
Cutaneous and Subcutaneous Perineuriomas in 2 Pediatric Patients

Perineuriomas are benign, slow-growing tumors derived from perineurial cells,1 which form the structurally supportive perineurium that surrounds individual nerve fascicles.2,3 Perineuriomas are classified into 2 main forms: intraneural or extraneural.4 Intraneural perineuriomas are found within the border of the peripheral nerve,5 while extraneural perineuriomas usually are found in soft tissue and skin. Extraneural perineuriomas can be further classified into variants based on their histologic appearance, including reticular, sclerosing, and plexiform subtypes. Extraneural perineuriomas usually present on the extremities or trunk of young to middle-aged adults as a well-circumscribed, painless, subcutaneous masses.1 These tumors are especially unusual in children.4 We present 2 extraneural perineurioma cases in children, and we review the pertinent diagnostic features of perineurioma as well as the presentation in the pediatric population.

Proliferation of spindle cells involving the reticular dermis (H&E, original magnification ×200). Reference bar indicates 500 µm.
FIGURE 1. Proliferation of spindle cells involving the reticular dermis (H&E, original magnification ×200). Reference bar indicates 500 µm.

Case Reports

Patient 1—A 10-year-old boy with a history of cerebral palsy and related comorbidities presented to the clinic for evaluation of a lesion on the thigh with no associated pain, irritation, erythema, or drainage. Physical examination revealed a soft, pedunculated, mobile nodule on the right medial thigh. An elliptical excision was performed. Gross examination demonstrated a 2.0×2.0×1.8-cm polypoid nodule. Histologic examination showed a dermal-based proliferation of bland spindle cells (Figure 1). The cytomorphology was characterized by elongated tapering nuclei and many areas with delicate bipolar cytoplasmic processes. The constituent cells were arranged in a whorled pattern in a variably myxoid to collagenous stroma. The tumor cells were multifocally positive for CD34; focally positive for smooth muscle actin (SMA); and negative for S-100, epithelial membrane antigen (EMA), GLUT1, claudin-1, STAT6, and desmin. Rb protein was intact. The CD34 immunostain highlighted the cytoplasmic processes. Electron microscopy was performed because the immunohistochemical results were nonspecific despite the favorable histologic features for perineurioma and showed pinocytic vesicles with delicate cytoplasmic processes, characteristic of perineurioma (Figure 2). Follow-up visits were related to the management of multiple comorbidities; no known recurrence of the lesion was documented.

Electron microscopy showed long, slender, cytoplasmic processes coated by discontinuous basal lamina and the presence of many pinocytic vesicles (black arrow)(original magnification ×15,000).
FIGURE 2. Electron microscopy showed long, slender, cytoplasmic processes coated by discontinuous basal lamina and the presence of many pinocytic vesicles (black arrow)(original magnification ×15,000).

Patient 2—A 15-year-old adolescent boy with no notable medical history presented to the pediatric clinic for a bump on the right upper arm of 4 to 5 months’ duration. He did not recall an injury to the area and denied change in size, redness, bruising, or pain of the lesion. Ultrasonography demonstrated a 2.6×2.3×1.3-cm hypoechoic and slightly heterogeneous, well-circumscribed, subcutaneous mass with internal vascularity. The patient was then referred to a pediatric surgeon. The clinical differential included a lipoma, lymphadenopathy, or sebaceous cyst. An excision was performed. Gross inspection demonstrated a 7-g, 2.8×2.6×1.8-cm, homogeneous, tan-pink, rubbery nodule with minimal surrounding soft tissue. Histologic examination showed a bland proliferation of spindle cells with storiform and whorled patterns (Figure 3). No notable nuclear atypia or necrosis was identified. The tumor cells were focally positive for EMA (Figure 4), claudin-1, and CD34 and negative for S-100, SOX10, GLUT1, desmin, STAT6, pankeratin AE1/AE3, and SMA. The diagnosis of perineurioma was rendered. No recurrence of the lesion was appreciated clinically on a 6-month follow-up examination.

Spindle cell proliferation with storiform and whorled patterns (H&E, original magnification ×100). Reference bar indicates 100 µm.
FIGURE 3. Spindle cell proliferation with storiform and whorled patterns (H&E, original magnification ×100). Reference bar indicates 100 µm.

Comment

Characteristics of Perineuriomas—On gross evaluation, perineuriomas are firm, gray-white, and well circumscribed but not encapsulated. Histologically, perineuriomas can have a storiform, whorled, or lamellar pattern of spindle cells. Perivascular whorls can be a histologic clue. The spindle cells are bland appearing and typically are elongated and slender but can appear slightly ovoid and plump. The background stroma can be myxoid, collagenous, or mixed. There usually is no atypia, and mitotic figures are rare.2,3,6,7 Intraneural perineuriomas vary architecturally in that they display a unique onion bulb–like appearance in which whorls of cytoplasmic material of variable sizes surround central axons.3

Immunohistochemistry revealed epithelial membrane antigen positivity in spindle cells (original magnification ×100). Reference bar indicates 100 µm.
FIGURE 4. Immunohistochemistry revealed epithelial membrane antigen positivity in spindle cells (original magnification ×100). Reference bar indicates 100 µm.

Diagnosis—The diagnosis of perineuriomas usually requires characteristic immunohistochemical and sometimes ultrastructural features. Perineuriomas are positive for EMA and GLUT1 and variable for CD34.6 Approximately 20% to 91% will be positive for claudin-1, a tight junction protein associated with perineuriomas.8 Of note, EMA and GLUT1 usually are positive in both neoplastic and nonneoplastic perineurial cells.9,10 Occasionally, these tumors can be focally positive for SMA and negative for S-100 and glial fibrillary acidic protein. The bipolar, thin, delicate, cytoplasmic processes with long-tapering nuclei may be easier to appreciate on electron microscopy than on conventional light microscopy. In addition, the cells contain pinocytotic vesicles and a discontinuous external lamina, which may be helpful for diagnosis.10

Genetics—Genetic alterations in perineurioma continue to be elucidated. Although many soft tissue perineuriomas possess deletion of chromosome 22q material, this is not a consistent finding and is not pathognomonic. Notably, the NF2 tumor suppressor gene is found on chromosome 22.11 For the sclerosing variant of perineurioma, rearrangements or deletions of chromosome 10q have been described. A study of 14 soft tissue/extraneural perineuriomas using whole-exome sequencing and single nucleotide polymorphism array showed 6 cases of recurrent chromosome 22q deletions containing the NF2 locus and 4 cases with a previously unreported finding of chromosome 17q deletions containing the NF1 locus that were mutually exclusive events in all but 1 case.12 Although perineuriomas can harbor NF1 or NF2 mutations, perineuriomas are not considered to be associated with neurofibromatosis type 1 or 2 (NF1 or NF2, respectively). Patients with NF1 or NF2 and perineurioma are exceedingly rare. One pediatric patient with both soft tissue perineurioma and NF1 has been reported in the literature.13

Differential Diagnosis—Perineuriomas should be distinguished from other benign neural neoplasms of the skin and soft tissue. Commonly considered in the differential diagnosis is schwannoma and neurofibroma. Schwannomas are encapsulated epineurial nerve sheath tumors comprised of a neoplastic proliferation of Schwann cells. Schwannomas morphologically differ from perineuriomas because of the presence of the hypercellular Antoni A with Verocay bodies and the hypocellular myxoid Antoni B patterns of spindle cells with elongated wavy nuclei and tapered ends. Other features include hyalinized vessels, hemosiderin deposition, cystic degeneration, and/or degenerative atypia.3,14 Importantly, the constituent cells of schwannomas are positive for S-100 and SOX10 and negative for EMA.3 Neurofibromas consist of fascicles and whorls of Schwann cells in a background myxoid stroma with scattered mast cells, lymphocytes, fibroblasts, and perineurial cells. Similar to schwannomas, neurofibromas also are positive for S-100 and negative for EMA.3,14 Neurofibromas can have either a somatic or germline mutation of the biallelic NF1 gene on chromosome 17q11.2 with subsequent loss of protein neurofibromin activity.15 Less common but still a consideration are the hybrid peripheral nerve sheath tumors that may present with a biphasic or intermingled morphology. Combinations include neurofibroma-schwannoma, schwannoma-perineurioma, and neurofibroma-perineurioma. The hybrid schwannoma-perineurioma has a mixture of thin and plump spindle cells with tapered nuclei as well as patchy S-100 positivity corresponding to schwannian areas. Similarly, S-100 will highlight the wavy Schwann cells in neurofibroma-perineurioma as well as CD34-highlighting fibroblasts.7,15 In both aforementioned hybrid tumors, EMA will be positive in the perineurial areas. Another potential diagnostic consideration that can occur in both pediatric and adult populations is dermatofibrosarcoma protuberans (DFSP), which is comprised of a dermal proliferation of monomorphic fusiform spindle cells. Although both perineuriomas and DFSP can have a storiform architecture, DFSP is more asymmetric and infiltrative. Dermatofibrosarcoma protuberans is recognized in areas of individual adipocyte trapping, referred to as honeycombing. Dermatofibrosarcoma protuberans typically does not express EMA, though the sclerosing variant of DFSP has been reported to sometimes demonstrate focal EMA reactivity.11,14,16 For morphologically challenging cases, cytogenetic studies will show t(17;22) translocation fusing the COL1A1 and PDGFRB genes.16 Finally, for subcutaneous or deep-seated tumors, one also may consider other mesenchymal neoplasms, including solitary fibrous tumor, low-grade fibromyxoid sarcoma, or low-grade malignant peripheral nerve sheath tumor (MPNST).11

 

 

Management—Perineuriomas are considered benign. The presence of mitotic figures, pleomorphism, and degenerative nuclear atypia akin to ancient change, as seen in ancient schwannoma, does not affect their benign clinical behavior. Treatment of a perineurioma typically is surgical excision with conservative margins and minimal chance of recurrence.1,11 So-called malignant perineuriomas are better classified as MPNSTs with perineural differentiation or perineurial MPNST. They also are positive for EMA and may be distinguished from perineurioma by the presence of major atypia and an infiltrative growth pattern.17,18

Considerations in the Pediatric Population—Few pediatric soft tissue perineuriomas have been reported. A clinicopathologic analysis by Hornick and Fletcher1 of patients with soft tissue perineurioma showed that only 6 of 81 patients were younger than 20 years. The youngest reported case of perineurioma occurred as an extraneural perineurioma on the scalp in an infant.19 Only 1 soft tissue perineural MPNST has been reported in the pediatric population, arising on the face of an 11-year-old boy. In a case series of 11 pediatric perineuriomas, including extraneural and intraneural, there was no evidence of recurrence or metastasis at follow-up.4

Conclusion

Perineuriomas are rare benign peripheral nerve sheath tumors with unique histologic and immunohistochemical features. Soft tissue perineuriomas in the pediatric population are an important diagnostic consideration, especially for the pediatrician or dermatologist when encountering a well-circumscribed nodular soft tissue lesion of the extremity or when encountering a neural-appearing tumor in the subcutaneous tissue.

Acknowledgment—We would like to thank Christopher Fletcher, MD (Boston, Massachusetts), for his expertise in outside consultation for patient 1.

References
  1. Hornick J, Fletcher C. Soft tissue perineurioma. Am J Surg Pathol. 2005;29:845-858.
  2. Tsang WY, Chan JK, Chow LT, et al. Perineurioma: an uncommon soft tissue neoplasm distinct from localized hypertrophic neuropathy and neurofibroma. Am J Surg Pathol. 1992;16:756-763.
  3. Belakhoua SM, Rodriguez FJ. Diagnostic pathology of tumors of peripheral nerve. Neurosurgery. 2021;88:443-456.
  4. Balarezo FS, Muller RC, Weiss RG, et al. Soft tissue perineuriomas in children: report of three cases and review of the literature. Pediatr Dev Pathol. 2003;6:137-141. Published correction appears in Pediatr Dev Pathol. 2003;6:following 364.
  5. Macarenco R, Ellinger F, Oliveira A. Perineurioma: a distinctive and underrecognized peripheral nerve sheath neoplasm. Arch Pathol Lab Med. 2007;131:625-636.
  6. Agaimy A, Buslei R, Coras R, et al. Comparative study of soft tissue perineurioma and meningioma using a five-marker immunohistochemical panel. Histopathology. 2014;65:60-70.
  7. Greenson JK, Hornick JL, Longacre TA, et al. Sternberg’s Diagnostic Surgical Pathology. Wolters Kluwer; 2015.
  8. Folpe A, Billings S, McKenney J, et al. Expression of claudin-1, a recently described tight junction-associated protein, distinguishes soft tissue perineurioma from potential mimics. Am J Surg Pathol. 2002;26:1620-1626.
  9. Hirose T, Tani T, Shimada T, et al. Immunohistochemical demonstration of EMA/Glut1-positive perineurial cells and CD34-positive fibroblastic cells in peripheral nerve sheath tumors. Mod Pathol. 2003;16:293-298.
  10. Fletcher CDM, Bridge JA, Hogendoorn PCW, et al. Perineurioma. WHO Classification of Tumours of Soft Tissue and Bone. IARC Press; 2013:176-178.
  11. Hornick JL. Practical Soft Tissue Pathology: A Diagnostic Approach. Elsevier Saunders; 2013.
  12. Carter JM, Wu Y, Blessing MM, et al. Recurrent genomic alterations in soft tissue perineuriomas. Am J Surg Pathol. 2018;42:1708-1714.
  13. Al-Adnani M. Soft tissue perineurioma in a child with neurofibromatosis type 1: a case report and review of the literature. Pediatr Dev Pathol. 2017;20:444-448.
  14. Reddy VB, David O, Spitz DJ, et al. Gattuso’s Differential Diagnosis in Surgical Pathology. Elsevier Saunders; 2022.
  15. Michal M, Kazakov DV, Michal M. Hybrid peripheral nerve sheath tumors: a review. Cesk Patol. 2017;53:81-88.
  16. Abdaljaleel MY, North JP. Sclerosing dermatofibrosarcoma protuberans shows significant overlap with sclerotic fibroma in both routine and immunohistochemical analysis: a potential diagnostic pitfall. Am J Dermatopathol. 2017;39:83-88.
  17. Rosenberg AS, Langee CL, Stevens GL, et al. Malignant peripheral nerve sheath tumor with perineurial differentiation: “malignant perineurioma.” J Cutan Pathol. 2002;29:362-367.
  18. Mitchell A, Scheithauer BW, Doyon J, et al. Malignant perineurioma (malignant peripheral nerve sheath tumor with perineural differentiation). Clin Neuropathol. 2012;31:424-429.
  19. Duhan A, Rana P, Beniwal K, et al. Perineurioma of scalp in an infant: a case report with short review of literature. Asian J Neurosurg. 2016;11:81-83.
Article PDF
Author and Disclosure Information

Drs. Lyon, Fernandez, and Rampisela are from the Department of Pathology, Baylor Scott & White Health, Temple, Texas. Ms. Burgess is from Texas A&M College of Medicine, Dallas. Dr. Berg is from the Division of Internal Medicine and Health Services Research, University of California, Los Angeles.

The authors report no conflict of interest.

Correspondence: Martin P. Fernandez, MD, 2401 S 31st St, Temple, TX 76508 ([email protected]).

Issue
Cutis - 110(5)
Publications
Topics
Page Number
277-280
Sections
Author and Disclosure Information

Drs. Lyon, Fernandez, and Rampisela are from the Department of Pathology, Baylor Scott & White Health, Temple, Texas. Ms. Burgess is from Texas A&M College of Medicine, Dallas. Dr. Berg is from the Division of Internal Medicine and Health Services Research, University of California, Los Angeles.

The authors report no conflict of interest.

Correspondence: Martin P. Fernandez, MD, 2401 S 31st St, Temple, TX 76508 ([email protected]).

Author and Disclosure Information

Drs. Lyon, Fernandez, and Rampisela are from the Department of Pathology, Baylor Scott & White Health, Temple, Texas. Ms. Burgess is from Texas A&M College of Medicine, Dallas. Dr. Berg is from the Division of Internal Medicine and Health Services Research, University of California, Los Angeles.

The authors report no conflict of interest.

Correspondence: Martin P. Fernandez, MD, 2401 S 31st St, Temple, TX 76508 ([email protected]).

Article PDF
Article PDF

Perineuriomas are benign, slow-growing tumors derived from perineurial cells,1 which form the structurally supportive perineurium that surrounds individual nerve fascicles.2,3 Perineuriomas are classified into 2 main forms: intraneural or extraneural.4 Intraneural perineuriomas are found within the border of the peripheral nerve,5 while extraneural perineuriomas usually are found in soft tissue and skin. Extraneural perineuriomas can be further classified into variants based on their histologic appearance, including reticular, sclerosing, and plexiform subtypes. Extraneural perineuriomas usually present on the extremities or trunk of young to middle-aged adults as a well-circumscribed, painless, subcutaneous masses.1 These tumors are especially unusual in children.4 We present 2 extraneural perineurioma cases in children, and we review the pertinent diagnostic features of perineurioma as well as the presentation in the pediatric population.

Proliferation of spindle cells involving the reticular dermis (H&E, original magnification ×200). Reference bar indicates 500 µm.
FIGURE 1. Proliferation of spindle cells involving the reticular dermis (H&E, original magnification ×200). Reference bar indicates 500 µm.

Case Reports

Patient 1—A 10-year-old boy with a history of cerebral palsy and related comorbidities presented to the clinic for evaluation of a lesion on the thigh with no associated pain, irritation, erythema, or drainage. Physical examination revealed a soft, pedunculated, mobile nodule on the right medial thigh. An elliptical excision was performed. Gross examination demonstrated a 2.0×2.0×1.8-cm polypoid nodule. Histologic examination showed a dermal-based proliferation of bland spindle cells (Figure 1). The cytomorphology was characterized by elongated tapering nuclei and many areas with delicate bipolar cytoplasmic processes. The constituent cells were arranged in a whorled pattern in a variably myxoid to collagenous stroma. The tumor cells were multifocally positive for CD34; focally positive for smooth muscle actin (SMA); and negative for S-100, epithelial membrane antigen (EMA), GLUT1, claudin-1, STAT6, and desmin. Rb protein was intact. The CD34 immunostain highlighted the cytoplasmic processes. Electron microscopy was performed because the immunohistochemical results were nonspecific despite the favorable histologic features for perineurioma and showed pinocytic vesicles with delicate cytoplasmic processes, characteristic of perineurioma (Figure 2). Follow-up visits were related to the management of multiple comorbidities; no known recurrence of the lesion was documented.

Electron microscopy showed long, slender, cytoplasmic processes coated by discontinuous basal lamina and the presence of many pinocytic vesicles (black arrow)(original magnification ×15,000).
FIGURE 2. Electron microscopy showed long, slender, cytoplasmic processes coated by discontinuous basal lamina and the presence of many pinocytic vesicles (black arrow)(original magnification ×15,000).

Patient 2—A 15-year-old adolescent boy with no notable medical history presented to the pediatric clinic for a bump on the right upper arm of 4 to 5 months’ duration. He did not recall an injury to the area and denied change in size, redness, bruising, or pain of the lesion. Ultrasonography demonstrated a 2.6×2.3×1.3-cm hypoechoic and slightly heterogeneous, well-circumscribed, subcutaneous mass with internal vascularity. The patient was then referred to a pediatric surgeon. The clinical differential included a lipoma, lymphadenopathy, or sebaceous cyst. An excision was performed. Gross inspection demonstrated a 7-g, 2.8×2.6×1.8-cm, homogeneous, tan-pink, rubbery nodule with minimal surrounding soft tissue. Histologic examination showed a bland proliferation of spindle cells with storiform and whorled patterns (Figure 3). No notable nuclear atypia or necrosis was identified. The tumor cells were focally positive for EMA (Figure 4), claudin-1, and CD34 and negative for S-100, SOX10, GLUT1, desmin, STAT6, pankeratin AE1/AE3, and SMA. The diagnosis of perineurioma was rendered. No recurrence of the lesion was appreciated clinically on a 6-month follow-up examination.

Spindle cell proliferation with storiform and whorled patterns (H&E, original magnification ×100). Reference bar indicates 100 µm.
FIGURE 3. Spindle cell proliferation with storiform and whorled patterns (H&E, original magnification ×100). Reference bar indicates 100 µm.

Comment

Characteristics of Perineuriomas—On gross evaluation, perineuriomas are firm, gray-white, and well circumscribed but not encapsulated. Histologically, perineuriomas can have a storiform, whorled, or lamellar pattern of spindle cells. Perivascular whorls can be a histologic clue. The spindle cells are bland appearing and typically are elongated and slender but can appear slightly ovoid and plump. The background stroma can be myxoid, collagenous, or mixed. There usually is no atypia, and mitotic figures are rare.2,3,6,7 Intraneural perineuriomas vary architecturally in that they display a unique onion bulb–like appearance in which whorls of cytoplasmic material of variable sizes surround central axons.3

Immunohistochemistry revealed epithelial membrane antigen positivity in spindle cells (original magnification ×100). Reference bar indicates 100 µm.
FIGURE 4. Immunohistochemistry revealed epithelial membrane antigen positivity in spindle cells (original magnification ×100). Reference bar indicates 100 µm.

Diagnosis—The diagnosis of perineuriomas usually requires characteristic immunohistochemical and sometimes ultrastructural features. Perineuriomas are positive for EMA and GLUT1 and variable for CD34.6 Approximately 20% to 91% will be positive for claudin-1, a tight junction protein associated with perineuriomas.8 Of note, EMA and GLUT1 usually are positive in both neoplastic and nonneoplastic perineurial cells.9,10 Occasionally, these tumors can be focally positive for SMA and negative for S-100 and glial fibrillary acidic protein. The bipolar, thin, delicate, cytoplasmic processes with long-tapering nuclei may be easier to appreciate on electron microscopy than on conventional light microscopy. In addition, the cells contain pinocytotic vesicles and a discontinuous external lamina, which may be helpful for diagnosis.10

Genetics—Genetic alterations in perineurioma continue to be elucidated. Although many soft tissue perineuriomas possess deletion of chromosome 22q material, this is not a consistent finding and is not pathognomonic. Notably, the NF2 tumor suppressor gene is found on chromosome 22.11 For the sclerosing variant of perineurioma, rearrangements or deletions of chromosome 10q have been described. A study of 14 soft tissue/extraneural perineuriomas using whole-exome sequencing and single nucleotide polymorphism array showed 6 cases of recurrent chromosome 22q deletions containing the NF2 locus and 4 cases with a previously unreported finding of chromosome 17q deletions containing the NF1 locus that were mutually exclusive events in all but 1 case.12 Although perineuriomas can harbor NF1 or NF2 mutations, perineuriomas are not considered to be associated with neurofibromatosis type 1 or 2 (NF1 or NF2, respectively). Patients with NF1 or NF2 and perineurioma are exceedingly rare. One pediatric patient with both soft tissue perineurioma and NF1 has been reported in the literature.13

Differential Diagnosis—Perineuriomas should be distinguished from other benign neural neoplasms of the skin and soft tissue. Commonly considered in the differential diagnosis is schwannoma and neurofibroma. Schwannomas are encapsulated epineurial nerve sheath tumors comprised of a neoplastic proliferation of Schwann cells. Schwannomas morphologically differ from perineuriomas because of the presence of the hypercellular Antoni A with Verocay bodies and the hypocellular myxoid Antoni B patterns of spindle cells with elongated wavy nuclei and tapered ends. Other features include hyalinized vessels, hemosiderin deposition, cystic degeneration, and/or degenerative atypia.3,14 Importantly, the constituent cells of schwannomas are positive for S-100 and SOX10 and negative for EMA.3 Neurofibromas consist of fascicles and whorls of Schwann cells in a background myxoid stroma with scattered mast cells, lymphocytes, fibroblasts, and perineurial cells. Similar to schwannomas, neurofibromas also are positive for S-100 and negative for EMA.3,14 Neurofibromas can have either a somatic or germline mutation of the biallelic NF1 gene on chromosome 17q11.2 with subsequent loss of protein neurofibromin activity.15 Less common but still a consideration are the hybrid peripheral nerve sheath tumors that may present with a biphasic or intermingled morphology. Combinations include neurofibroma-schwannoma, schwannoma-perineurioma, and neurofibroma-perineurioma. The hybrid schwannoma-perineurioma has a mixture of thin and plump spindle cells with tapered nuclei as well as patchy S-100 positivity corresponding to schwannian areas. Similarly, S-100 will highlight the wavy Schwann cells in neurofibroma-perineurioma as well as CD34-highlighting fibroblasts.7,15 In both aforementioned hybrid tumors, EMA will be positive in the perineurial areas. Another potential diagnostic consideration that can occur in both pediatric and adult populations is dermatofibrosarcoma protuberans (DFSP), which is comprised of a dermal proliferation of monomorphic fusiform spindle cells. Although both perineuriomas and DFSP can have a storiform architecture, DFSP is more asymmetric and infiltrative. Dermatofibrosarcoma protuberans is recognized in areas of individual adipocyte trapping, referred to as honeycombing. Dermatofibrosarcoma protuberans typically does not express EMA, though the sclerosing variant of DFSP has been reported to sometimes demonstrate focal EMA reactivity.11,14,16 For morphologically challenging cases, cytogenetic studies will show t(17;22) translocation fusing the COL1A1 and PDGFRB genes.16 Finally, for subcutaneous or deep-seated tumors, one also may consider other mesenchymal neoplasms, including solitary fibrous tumor, low-grade fibromyxoid sarcoma, or low-grade malignant peripheral nerve sheath tumor (MPNST).11

 

 

Management—Perineuriomas are considered benign. The presence of mitotic figures, pleomorphism, and degenerative nuclear atypia akin to ancient change, as seen in ancient schwannoma, does not affect their benign clinical behavior. Treatment of a perineurioma typically is surgical excision with conservative margins and minimal chance of recurrence.1,11 So-called malignant perineuriomas are better classified as MPNSTs with perineural differentiation or perineurial MPNST. They also are positive for EMA and may be distinguished from perineurioma by the presence of major atypia and an infiltrative growth pattern.17,18

Considerations in the Pediatric Population—Few pediatric soft tissue perineuriomas have been reported. A clinicopathologic analysis by Hornick and Fletcher1 of patients with soft tissue perineurioma showed that only 6 of 81 patients were younger than 20 years. The youngest reported case of perineurioma occurred as an extraneural perineurioma on the scalp in an infant.19 Only 1 soft tissue perineural MPNST has been reported in the pediatric population, arising on the face of an 11-year-old boy. In a case series of 11 pediatric perineuriomas, including extraneural and intraneural, there was no evidence of recurrence or metastasis at follow-up.4

Conclusion

Perineuriomas are rare benign peripheral nerve sheath tumors with unique histologic and immunohistochemical features. Soft tissue perineuriomas in the pediatric population are an important diagnostic consideration, especially for the pediatrician or dermatologist when encountering a well-circumscribed nodular soft tissue lesion of the extremity or when encountering a neural-appearing tumor in the subcutaneous tissue.

Acknowledgment—We would like to thank Christopher Fletcher, MD (Boston, Massachusetts), for his expertise in outside consultation for patient 1.

Perineuriomas are benign, slow-growing tumors derived from perineurial cells,1 which form the structurally supportive perineurium that surrounds individual nerve fascicles.2,3 Perineuriomas are classified into 2 main forms: intraneural or extraneural.4 Intraneural perineuriomas are found within the border of the peripheral nerve,5 while extraneural perineuriomas usually are found in soft tissue and skin. Extraneural perineuriomas can be further classified into variants based on their histologic appearance, including reticular, sclerosing, and plexiform subtypes. Extraneural perineuriomas usually present on the extremities or trunk of young to middle-aged adults as a well-circumscribed, painless, subcutaneous masses.1 These tumors are especially unusual in children.4 We present 2 extraneural perineurioma cases in children, and we review the pertinent diagnostic features of perineurioma as well as the presentation in the pediatric population.

Proliferation of spindle cells involving the reticular dermis (H&E, original magnification ×200). Reference bar indicates 500 µm.
FIGURE 1. Proliferation of spindle cells involving the reticular dermis (H&E, original magnification ×200). Reference bar indicates 500 µm.

Case Reports

Patient 1—A 10-year-old boy with a history of cerebral palsy and related comorbidities presented to the clinic for evaluation of a lesion on the thigh with no associated pain, irritation, erythema, or drainage. Physical examination revealed a soft, pedunculated, mobile nodule on the right medial thigh. An elliptical excision was performed. Gross examination demonstrated a 2.0×2.0×1.8-cm polypoid nodule. Histologic examination showed a dermal-based proliferation of bland spindle cells (Figure 1). The cytomorphology was characterized by elongated tapering nuclei and many areas with delicate bipolar cytoplasmic processes. The constituent cells were arranged in a whorled pattern in a variably myxoid to collagenous stroma. The tumor cells were multifocally positive for CD34; focally positive for smooth muscle actin (SMA); and negative for S-100, epithelial membrane antigen (EMA), GLUT1, claudin-1, STAT6, and desmin. Rb protein was intact. The CD34 immunostain highlighted the cytoplasmic processes. Electron microscopy was performed because the immunohistochemical results were nonspecific despite the favorable histologic features for perineurioma and showed pinocytic vesicles with delicate cytoplasmic processes, characteristic of perineurioma (Figure 2). Follow-up visits were related to the management of multiple comorbidities; no known recurrence of the lesion was documented.

Electron microscopy showed long, slender, cytoplasmic processes coated by discontinuous basal lamina and the presence of many pinocytic vesicles (black arrow)(original magnification ×15,000).
FIGURE 2. Electron microscopy showed long, slender, cytoplasmic processes coated by discontinuous basal lamina and the presence of many pinocytic vesicles (black arrow)(original magnification ×15,000).

Patient 2—A 15-year-old adolescent boy with no notable medical history presented to the pediatric clinic for a bump on the right upper arm of 4 to 5 months’ duration. He did not recall an injury to the area and denied change in size, redness, bruising, or pain of the lesion. Ultrasonography demonstrated a 2.6×2.3×1.3-cm hypoechoic and slightly heterogeneous, well-circumscribed, subcutaneous mass with internal vascularity. The patient was then referred to a pediatric surgeon. The clinical differential included a lipoma, lymphadenopathy, or sebaceous cyst. An excision was performed. Gross inspection demonstrated a 7-g, 2.8×2.6×1.8-cm, homogeneous, tan-pink, rubbery nodule with minimal surrounding soft tissue. Histologic examination showed a bland proliferation of spindle cells with storiform and whorled patterns (Figure 3). No notable nuclear atypia or necrosis was identified. The tumor cells were focally positive for EMA (Figure 4), claudin-1, and CD34 and negative for S-100, SOX10, GLUT1, desmin, STAT6, pankeratin AE1/AE3, and SMA. The diagnosis of perineurioma was rendered. No recurrence of the lesion was appreciated clinically on a 6-month follow-up examination.

Spindle cell proliferation with storiform and whorled patterns (H&E, original magnification ×100). Reference bar indicates 100 µm.
FIGURE 3. Spindle cell proliferation with storiform and whorled patterns (H&E, original magnification ×100). Reference bar indicates 100 µm.

Comment

Characteristics of Perineuriomas—On gross evaluation, perineuriomas are firm, gray-white, and well circumscribed but not encapsulated. Histologically, perineuriomas can have a storiform, whorled, or lamellar pattern of spindle cells. Perivascular whorls can be a histologic clue. The spindle cells are bland appearing and typically are elongated and slender but can appear slightly ovoid and plump. The background stroma can be myxoid, collagenous, or mixed. There usually is no atypia, and mitotic figures are rare.2,3,6,7 Intraneural perineuriomas vary architecturally in that they display a unique onion bulb–like appearance in which whorls of cytoplasmic material of variable sizes surround central axons.3

Immunohistochemistry revealed epithelial membrane antigen positivity in spindle cells (original magnification ×100). Reference bar indicates 100 µm.
FIGURE 4. Immunohistochemistry revealed epithelial membrane antigen positivity in spindle cells (original magnification ×100). Reference bar indicates 100 µm.

Diagnosis—The diagnosis of perineuriomas usually requires characteristic immunohistochemical and sometimes ultrastructural features. Perineuriomas are positive for EMA and GLUT1 and variable for CD34.6 Approximately 20% to 91% will be positive for claudin-1, a tight junction protein associated with perineuriomas.8 Of note, EMA and GLUT1 usually are positive in both neoplastic and nonneoplastic perineurial cells.9,10 Occasionally, these tumors can be focally positive for SMA and negative for S-100 and glial fibrillary acidic protein. The bipolar, thin, delicate, cytoplasmic processes with long-tapering nuclei may be easier to appreciate on electron microscopy than on conventional light microscopy. In addition, the cells contain pinocytotic vesicles and a discontinuous external lamina, which may be helpful for diagnosis.10

Genetics—Genetic alterations in perineurioma continue to be elucidated. Although many soft tissue perineuriomas possess deletion of chromosome 22q material, this is not a consistent finding and is not pathognomonic. Notably, the NF2 tumor suppressor gene is found on chromosome 22.11 For the sclerosing variant of perineurioma, rearrangements or deletions of chromosome 10q have been described. A study of 14 soft tissue/extraneural perineuriomas using whole-exome sequencing and single nucleotide polymorphism array showed 6 cases of recurrent chromosome 22q deletions containing the NF2 locus and 4 cases with a previously unreported finding of chromosome 17q deletions containing the NF1 locus that were mutually exclusive events in all but 1 case.12 Although perineuriomas can harbor NF1 or NF2 mutations, perineuriomas are not considered to be associated with neurofibromatosis type 1 or 2 (NF1 or NF2, respectively). Patients with NF1 or NF2 and perineurioma are exceedingly rare. One pediatric patient with both soft tissue perineurioma and NF1 has been reported in the literature.13

Differential Diagnosis—Perineuriomas should be distinguished from other benign neural neoplasms of the skin and soft tissue. Commonly considered in the differential diagnosis is schwannoma and neurofibroma. Schwannomas are encapsulated epineurial nerve sheath tumors comprised of a neoplastic proliferation of Schwann cells. Schwannomas morphologically differ from perineuriomas because of the presence of the hypercellular Antoni A with Verocay bodies and the hypocellular myxoid Antoni B patterns of spindle cells with elongated wavy nuclei and tapered ends. Other features include hyalinized vessels, hemosiderin deposition, cystic degeneration, and/or degenerative atypia.3,14 Importantly, the constituent cells of schwannomas are positive for S-100 and SOX10 and negative for EMA.3 Neurofibromas consist of fascicles and whorls of Schwann cells in a background myxoid stroma with scattered mast cells, lymphocytes, fibroblasts, and perineurial cells. Similar to schwannomas, neurofibromas also are positive for S-100 and negative for EMA.3,14 Neurofibromas can have either a somatic or germline mutation of the biallelic NF1 gene on chromosome 17q11.2 with subsequent loss of protein neurofibromin activity.15 Less common but still a consideration are the hybrid peripheral nerve sheath tumors that may present with a biphasic or intermingled morphology. Combinations include neurofibroma-schwannoma, schwannoma-perineurioma, and neurofibroma-perineurioma. The hybrid schwannoma-perineurioma has a mixture of thin and plump spindle cells with tapered nuclei as well as patchy S-100 positivity corresponding to schwannian areas. Similarly, S-100 will highlight the wavy Schwann cells in neurofibroma-perineurioma as well as CD34-highlighting fibroblasts.7,15 In both aforementioned hybrid tumors, EMA will be positive in the perineurial areas. Another potential diagnostic consideration that can occur in both pediatric and adult populations is dermatofibrosarcoma protuberans (DFSP), which is comprised of a dermal proliferation of monomorphic fusiform spindle cells. Although both perineuriomas and DFSP can have a storiform architecture, DFSP is more asymmetric and infiltrative. Dermatofibrosarcoma protuberans is recognized in areas of individual adipocyte trapping, referred to as honeycombing. Dermatofibrosarcoma protuberans typically does not express EMA, though the sclerosing variant of DFSP has been reported to sometimes demonstrate focal EMA reactivity.11,14,16 For morphologically challenging cases, cytogenetic studies will show t(17;22) translocation fusing the COL1A1 and PDGFRB genes.16 Finally, for subcutaneous or deep-seated tumors, one also may consider other mesenchymal neoplasms, including solitary fibrous tumor, low-grade fibromyxoid sarcoma, or low-grade malignant peripheral nerve sheath tumor (MPNST).11

 

 

Management—Perineuriomas are considered benign. The presence of mitotic figures, pleomorphism, and degenerative nuclear atypia akin to ancient change, as seen in ancient schwannoma, does not affect their benign clinical behavior. Treatment of a perineurioma typically is surgical excision with conservative margins and minimal chance of recurrence.1,11 So-called malignant perineuriomas are better classified as MPNSTs with perineural differentiation or perineurial MPNST. They also are positive for EMA and may be distinguished from perineurioma by the presence of major atypia and an infiltrative growth pattern.17,18

Considerations in the Pediatric Population—Few pediatric soft tissue perineuriomas have been reported. A clinicopathologic analysis by Hornick and Fletcher1 of patients with soft tissue perineurioma showed that only 6 of 81 patients were younger than 20 years. The youngest reported case of perineurioma occurred as an extraneural perineurioma on the scalp in an infant.19 Only 1 soft tissue perineural MPNST has been reported in the pediatric population, arising on the face of an 11-year-old boy. In a case series of 11 pediatric perineuriomas, including extraneural and intraneural, there was no evidence of recurrence or metastasis at follow-up.4

Conclusion

Perineuriomas are rare benign peripheral nerve sheath tumors with unique histologic and immunohistochemical features. Soft tissue perineuriomas in the pediatric population are an important diagnostic consideration, especially for the pediatrician or dermatologist when encountering a well-circumscribed nodular soft tissue lesion of the extremity or when encountering a neural-appearing tumor in the subcutaneous tissue.

Acknowledgment—We would like to thank Christopher Fletcher, MD (Boston, Massachusetts), for his expertise in outside consultation for patient 1.

References
  1. Hornick J, Fletcher C. Soft tissue perineurioma. Am J Surg Pathol. 2005;29:845-858.
  2. Tsang WY, Chan JK, Chow LT, et al. Perineurioma: an uncommon soft tissue neoplasm distinct from localized hypertrophic neuropathy and neurofibroma. Am J Surg Pathol. 1992;16:756-763.
  3. Belakhoua SM, Rodriguez FJ. Diagnostic pathology of tumors of peripheral nerve. Neurosurgery. 2021;88:443-456.
  4. Balarezo FS, Muller RC, Weiss RG, et al. Soft tissue perineuriomas in children: report of three cases and review of the literature. Pediatr Dev Pathol. 2003;6:137-141. Published correction appears in Pediatr Dev Pathol. 2003;6:following 364.
  5. Macarenco R, Ellinger F, Oliveira A. Perineurioma: a distinctive and underrecognized peripheral nerve sheath neoplasm. Arch Pathol Lab Med. 2007;131:625-636.
  6. Agaimy A, Buslei R, Coras R, et al. Comparative study of soft tissue perineurioma and meningioma using a five-marker immunohistochemical panel. Histopathology. 2014;65:60-70.
  7. Greenson JK, Hornick JL, Longacre TA, et al. Sternberg’s Diagnostic Surgical Pathology. Wolters Kluwer; 2015.
  8. Folpe A, Billings S, McKenney J, et al. Expression of claudin-1, a recently described tight junction-associated protein, distinguishes soft tissue perineurioma from potential mimics. Am J Surg Pathol. 2002;26:1620-1626.
  9. Hirose T, Tani T, Shimada T, et al. Immunohistochemical demonstration of EMA/Glut1-positive perineurial cells and CD34-positive fibroblastic cells in peripheral nerve sheath tumors. Mod Pathol. 2003;16:293-298.
  10. Fletcher CDM, Bridge JA, Hogendoorn PCW, et al. Perineurioma. WHO Classification of Tumours of Soft Tissue and Bone. IARC Press; 2013:176-178.
  11. Hornick JL. Practical Soft Tissue Pathology: A Diagnostic Approach. Elsevier Saunders; 2013.
  12. Carter JM, Wu Y, Blessing MM, et al. Recurrent genomic alterations in soft tissue perineuriomas. Am J Surg Pathol. 2018;42:1708-1714.
  13. Al-Adnani M. Soft tissue perineurioma in a child with neurofibromatosis type 1: a case report and review of the literature. Pediatr Dev Pathol. 2017;20:444-448.
  14. Reddy VB, David O, Spitz DJ, et al. Gattuso’s Differential Diagnosis in Surgical Pathology. Elsevier Saunders; 2022.
  15. Michal M, Kazakov DV, Michal M. Hybrid peripheral nerve sheath tumors: a review. Cesk Patol. 2017;53:81-88.
  16. Abdaljaleel MY, North JP. Sclerosing dermatofibrosarcoma protuberans shows significant overlap with sclerotic fibroma in both routine and immunohistochemical analysis: a potential diagnostic pitfall. Am J Dermatopathol. 2017;39:83-88.
  17. Rosenberg AS, Langee CL, Stevens GL, et al. Malignant peripheral nerve sheath tumor with perineurial differentiation: “malignant perineurioma.” J Cutan Pathol. 2002;29:362-367.
  18. Mitchell A, Scheithauer BW, Doyon J, et al. Malignant perineurioma (malignant peripheral nerve sheath tumor with perineural differentiation). Clin Neuropathol. 2012;31:424-429.
  19. Duhan A, Rana P, Beniwal K, et al. Perineurioma of scalp in an infant: a case report with short review of literature. Asian J Neurosurg. 2016;11:81-83.
References
  1. Hornick J, Fletcher C. Soft tissue perineurioma. Am J Surg Pathol. 2005;29:845-858.
  2. Tsang WY, Chan JK, Chow LT, et al. Perineurioma: an uncommon soft tissue neoplasm distinct from localized hypertrophic neuropathy and neurofibroma. Am J Surg Pathol. 1992;16:756-763.
  3. Belakhoua SM, Rodriguez FJ. Diagnostic pathology of tumors of peripheral nerve. Neurosurgery. 2021;88:443-456.
  4. Balarezo FS, Muller RC, Weiss RG, et al. Soft tissue perineuriomas in children: report of three cases and review of the literature. Pediatr Dev Pathol. 2003;6:137-141. Published correction appears in Pediatr Dev Pathol. 2003;6:following 364.
  5. Macarenco R, Ellinger F, Oliveira A. Perineurioma: a distinctive and underrecognized peripheral nerve sheath neoplasm. Arch Pathol Lab Med. 2007;131:625-636.
  6. Agaimy A, Buslei R, Coras R, et al. Comparative study of soft tissue perineurioma and meningioma using a five-marker immunohistochemical panel. Histopathology. 2014;65:60-70.
  7. Greenson JK, Hornick JL, Longacre TA, et al. Sternberg’s Diagnostic Surgical Pathology. Wolters Kluwer; 2015.
  8. Folpe A, Billings S, McKenney J, et al. Expression of claudin-1, a recently described tight junction-associated protein, distinguishes soft tissue perineurioma from potential mimics. Am J Surg Pathol. 2002;26:1620-1626.
  9. Hirose T, Tani T, Shimada T, et al. Immunohistochemical demonstration of EMA/Glut1-positive perineurial cells and CD34-positive fibroblastic cells in peripheral nerve sheath tumors. Mod Pathol. 2003;16:293-298.
  10. Fletcher CDM, Bridge JA, Hogendoorn PCW, et al. Perineurioma. WHO Classification of Tumours of Soft Tissue and Bone. IARC Press; 2013:176-178.
  11. Hornick JL. Practical Soft Tissue Pathology: A Diagnostic Approach. Elsevier Saunders; 2013.
  12. Carter JM, Wu Y, Blessing MM, et al. Recurrent genomic alterations in soft tissue perineuriomas. Am J Surg Pathol. 2018;42:1708-1714.
  13. Al-Adnani M. Soft tissue perineurioma in a child with neurofibromatosis type 1: a case report and review of the literature. Pediatr Dev Pathol. 2017;20:444-448.
  14. Reddy VB, David O, Spitz DJ, et al. Gattuso’s Differential Diagnosis in Surgical Pathology. Elsevier Saunders; 2022.
  15. Michal M, Kazakov DV, Michal M. Hybrid peripheral nerve sheath tumors: a review. Cesk Patol. 2017;53:81-88.
  16. Abdaljaleel MY, North JP. Sclerosing dermatofibrosarcoma protuberans shows significant overlap with sclerotic fibroma in both routine and immunohistochemical analysis: a potential diagnostic pitfall. Am J Dermatopathol. 2017;39:83-88.
  17. Rosenberg AS, Langee CL, Stevens GL, et al. Malignant peripheral nerve sheath tumor with perineurial differentiation: “malignant perineurioma.” J Cutan Pathol. 2002;29:362-367.
  18. Mitchell A, Scheithauer BW, Doyon J, et al. Malignant perineurioma (malignant peripheral nerve sheath tumor with perineural differentiation). Clin Neuropathol. 2012;31:424-429.
  19. Duhan A, Rana P, Beniwal K, et al. Perineurioma of scalp in an infant: a case report with short review of literature. Asian J Neurosurg. 2016;11:81-83.
Issue
Cutis - 110(5)
Issue
Cutis - 110(5)
Page Number
277-280
Page Number
277-280
Publications
Publications
Topics
Article Type
Display Headline
Cutaneous and Subcutaneous Perineuriomas in 2 Pediatric Patients
Display Headline
Cutaneous and Subcutaneous Perineuriomas in 2 Pediatric Patients
Sections
Inside the Article

Practice Points

  • Perineuriomas are rare benign peripheral nerve sheath tumors that most commonly occur in young to middle-aged adults but rarely can present in children.
  • Immunohistochemically, perineuriomas show positive staining with epithelial membrane antigen, GLUT1, claudin-1, and frequently with CD34; they are negative for S-100 and glial fibrillary acidic protein.
  • Perineuriomas should be considered in the differential diagnosis in children who present with a well-circumscribed nodular lesion in the subcutaneous tissue. 
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

Past, Present, and Future of Pediatric Atopic Dermatitis Management

Article Type
Changed
Tue, 05/02/2023 - 13:40
Display Headline
Past, Present, and Future of Pediatric Atopic Dermatitis Management

Atopic dermatitis (AD), or eczema, is a common inflammatory skin disease notorious for its chronic, relapsing, and often frustrating disease course. Although as many as 25% of children in the United States are affected by this condition and its impact on the quality of life of affected patients and families is profound,1-3 therapeutic advances in the pediatric population have been fairly limited until recently.

Over the last 10 years, there has been robust investigation into pediatric AD therapeutics, with many topical and systemic medications either recently approved or under clinical investigation. These developments are changing the landscape of the management of pediatric AD and raise a set of fascinating questions about how early and aggressive intervention might change the course of this disease. We discuss current limitations in the field that may be addressed with additional research.

New Topical Medications

In the last several years, there has been a rapid increase in efforts to develop new topical agents to manage AD. Until the beginning of the 21st century, the dermatologist’s arsenal was limited to topical corticosteroids (TCs). In the early 2000s, attention shifted to topical calcineurin inhibitors as nonsteroidal alternatives when the US Food and Drug Administration (FDA) approved topical tacrolimus and pimecrolimus for AD. In 2016, crisaborole (a phosphodiesterase-4 [PDE4] inhibitor) was approved by the FDA for use in mild to moderate AD in patients 2 years and older, marking a new age of development for topical AD therapies. In 2021, the FDA approved ruxolitinib (a topical Janus kinase [JAK] 1/2 inhibitor) for use in mild to moderate AD in patients 12 years and older.

Roflumilast (ARQ-151) and difamilast (OPA-15406)(members of the PDE4 inhibitor class) are undergoing investigation for pediatric AD. A phase 3 clinical trial for roflumilast for AD is underway (ClinicalTrial.gov Identifier: NCT04845620); it is already approved for psoriasis in patients 12 years and older. A phase 3 trial of difamilast (NCT03911401) was recently completed, with results supporting the drug’s safety and efficacy in AD management.4 Efforts to synthesize new better-targeted PDE4 inhibitors are ongoing.5

Tapinarof (a novel aryl hydrocarbon receptor-modulating agent) is approved for psoriasis in adults, and a phase 3 trial for management of pediatric AD is underway (NCT05032859) after phase 2 trials revealed promising results.6

Lastly, the microbiome is a target for AD topical therapies. A recently completed phase 1 trial of bacteriotherapy with Staphylococcus hominis A9 transplant lotion showed promising results (NCT03151148).7 Although this bacteriotherapy technique is early in development and has been studied only in adult patients, results are exciting because they represent a gateway to a largely unexplored realm of potential future therapies.

Standard of Care—How will these new topical therapies impact our standard of care for pediatric AD patients? Topical corticosteroids are still a pillar of topical AD therapy, but the potential for nonsteroidal topical agents as alternatives and used in combination therapeutic regimens has expanded exponentially. It is uncertain how we might individualize regimens tailored to patient-specific factors because the standard approach has been to test drugs as monotherapy, with vehicle comparisons or with reference medications in Europe.

 

 

Newer topical nonsteroidal agents may offer several opportunities. First, they may help avoid local and systemic adverse effects that often limit the use of current standard therapy.8 This capability may prove essential in bridging TC treatments and serving as long-term maintenance therapies to decrease the frequency of eczema flares. Second, they can alleviate the need for different medication strengths for different body regions, thereby allowing for simplification of regimens and potentially increased adherence and decreased disease burden—a boon to affected patients and caregivers.

Although the efficacy and long-term safety profile of these new drugs require further study, it does not seem unreasonable to look forward to achieving levels of optimization and individualization with topical regimens for AD in the near future that makes flares in patients with mild to moderate AD a phenomenon of the past.

Advances in Systemic Therapy

Systemic therapeutics in pediatric AD also recently entered an exciting era of development. Traditional systemic agents, including cyclosporine, methotrexate, azathioprine, and mycophenolate mofetil, have existed for decades but have not been widely utilized for moderate to severe AD in the United States, especially in the pediatric population, likely because these drugs lacked FDA approval and they can cause a range of adverse effects, including notable immunosuppression.9

Introduction and approval of dupilumab in 2017 by the FDA was revolutionary in this field. As a monoclonal antibody targeted against IL-4 and IL-13, dupilumab has consistently demonstrated strong long-term efficacy for pediatric AD and has an acceptable safety profile in children and adolescents.10-14 Expansion of the label to include children as young as 6 months with moderate to severe AD seems an important milestone in pediatric AD care.

Since the approval of dupilumab for adolescents and children aged 6 to 12 years, global experience has supported expanded use of systemic agents for patients who have an inadequate response to TCs and previously approved nonsteroidal topical agents. How expansive the use of systemics will be in younger children depends on how their long-term use impacts the disease course, whether therapy is disease modifying, and whether early use can curb the development of comorbidities.

Investigations into targeted systemic therapeutics for eczematous dermatitis are not limited to dupilumab. In a study of adolescents as young as 12 years, tralokinumab (an IL-13 pathway inhibitor) demonstrated an Eczema Area Severity Index-75 of 27.8% to 28.6% and a mean decrease in the SCORing Atopic Dermatitis index of 27.5 to 29.1, with minimal adverse effects.15 Lebrikizumab, another biologic IL-13 inhibitor with strong published safety and efficacy data in adults, has completed short- and longer-term studies in adolescents (NCT04178967 and NCT04146363).16 The drug received FDA Fast Track designation for moderate to severe AD in patients 12 years and older after showing positive data.17

This push to targeted therapy stretches beyond monoclonal antibodies. In the last few years, oral JAK inhibitors have emerged as a new class of systemic therapy for eczematous dermatitis. Upadacitinib, a JAK1 selective inhibitor, was approved by the FDA in 2022 for patients 12 years and older with AD and has data that supports its efficacy in adolescents and adults.18 Other JAK inhibitors including the selective JAK1 inhibitor abrocitinib and the combined JAK1/2 inhibitor baricitinib are being studied for pediatric AD (NCT04564755, NCT03422822, and NCT03952559), with most evidence to date supporting their safety and efficacy, at least over the short-term.19

 

 

The study of these and other advanced systemic therapies for eczematous dermatitis is transforming the toolbox for pediatric AD care. Although long-term data are lacking for some of these medications, it is possible that newer agents may decrease reliance on older immunosuppressants, such as systemic corticosteroids, cyclosporine, and methotrexate. Unanswered questions include: How and which systemic medications may alter the course of the disease? What is the disease modification for AD? What is the impact on comorbidities over time?

What’s Missing?

The field of pediatric AD has experienced exciting new developments with the emergence of targeted therapeutics, but those new agents require more long-term study, though we already have longer-term data on crisaborole and dupilumab.10-14,20 Studies of the long-term use of these new treatments on comorbidities of pediatric AD—mental health outcomes, cardiovascular disease, effects on the family, and other allergic conditions—are needed.21 Furthermore, clinical guidelines that address indications, timing of use, tapering, and discontinuation of new treatments depend on long-term experience and data collection.

Therefore, it is prudent that investigators, companies, payers, patients, and families support phase 4, long-term extension, and registry studies, which will expand our knowledge of AD medications and their impact on the disease over time.

Final Thoughts

Medications to treat AD are reaching a new level of advancement—from topical agents that target novel pathways to revolutionary biologics and systemic medications. Although there are knowledge gaps on these new therapeutics, the standard of care is already rapidly changing as the expectations of clinicians, patients, and families advance with each addition to the provider’s toolbox.

References
  1. Eichenfield LF, Tom WL, Chamlin SL, et al. Guidelines of care for the management of atopic dermatitis: part 1. diagnosis and assessment of atopic dermatitis. J Am Acad Dermatol. 2014;70:338-351. doi:10.1016/j.jaad.2013.10.010
  2. Kiebert G, Sorensen SV, Revicki D, et al. Atopic dermatitis is associated with a decrement in health-related quality of life. Int J Dermatol. 2002;41:151-158. doi:10.1046/j.1365-4362.2002.01436.x
  3. Al Shobaili HA. The impact of childhood atopic dermatitis on the patients’ family. Pediatr Dermatol. 2010;27:618-623. doi:10.1111/j.1525-1470.2010.01215.x
  4. Saeki H, Baba N, Ito K, et al. Difamilast, a selective phosphodiesterase 4 inhibitor, ointment in paediatric patients with atopic dermatitis: a phase III randomized double-blind, vehicle-controlled trial [published online November 1, 2021]. Br J Dermatol. 2022;186:40-49. doi:10.1111/bjd.20655
  5. Chu Z, Xu Q, Zhu Q, et al. Design, synthesis and biological evaluation of novel benzoxaborole derivatives as potent PDE4 inhibitors for topical treatment of atopic dermatitis. Eur J Med Chem. 2021;213:113171. doi:10.1016/j.ejmech.2021.113171
  6. Paller AS, Stein Gold L, Soung J, et al. Efficacy and patient-reported outcomes from a phase 2b, randomized clinical trial of tapinarof cream for the treatment of adolescents and adults with atopic dermatitis. J Am Acad Dermatol. 2021;84:632-638. doi:10.1016/j.jaad.2020.05.135
  7. Nakatsuji T, Hata TR, Tong Y, et al. Development of a human skin commensal microbe for bacteriotherapy of atopic dermatitis and use in a phase 1 randomized clinical trial. Nat Med. 2021;27:700-709. doi:10.1038/s41591-021-01256-2
  8. Eichenfield LF, Tom WL, Berger TG, et al. Guidelines of care for the management of atopic dermatitis: part 2. management and treatment of atopic dermatitis with topical therapies. J Am Acad Dermatol. 2014;71:116-132. doi:10.1016/j.jaad.2014.03.023
  9. Sidbury R, Davis DM, Cohen DE, et al. Guidelines of care for the management of atopic dermatitis: part 3. management and treatment with phototherapy and systemic agents. J Am Acad Dermatol. 2014;71:327-349. doi:10.1016/j.jaad.2014.03.030
  10. Gooderham MJ, Hong HC-H, Eshtiaghi P, et al. Dupilumab: a review of its use in the treatment of atopic dermatitis. J Am Acad Dermatol. 2018;78(3 suppl 1):S28-S36. doi:10.1016/j.jaad.2017.12.022
  11. Simpson EL, Paller AS, Siegfried EC, et al. Efficacy and safety of dupilumab in adolescents with uncontrolled moderate to severe atopic dermatitis: a phase 3 randomized clinical trial. JAMA Dermatol. 2020;156:44-56. doi:10.1001/jamadermatol.2019.3336
  12. Blauvelt A, Guttman-Yassky E, Paller AS, et al. Long-term efficacy and safety of dupilumab in adolescents with moderate-to-severe atopicdermatitis: results through week 52 from a phase III open-label extension trial (LIBERTY AD PED-OLE). Am J Clin Dermatol. 2022;23:365-383. doi:10.1007/s40257-022-00683-2
  13. Cork MJ, Thaçi D, Eichenfield LF, et al. Dupilumab provides favourable long-term safety and efficacy in children aged ≥ 6 to < 12 years with uncontrolled severe atopic dermatitis: results from an open-label phase IIa study and subsequent phase III open-label extension study. Br J Dermatol. 2021;184:857-870. doi:10.1111/bjd.19460
  14. Simpson EL, Paller AS, Siegfried EC, et al. Dupilumab demonstrates rapid and consistent improvement in extent and signs of atopic dermatitis across all anatomical regions in pediatric patients 6 years of age and older. Dermatol Ther (Heidelb). 2021;11:1643-1656. doi:10.1007/s13555-021-00568-y
  15. Paller A, Blauvelt A, Soong W, et al. Efficacy and safety of tralokinumab in adolescents with moderate-to-severe atopic dermatitis: results of the phase 3 ECZTRA 6 trial. SKIN. 2022;6:S29. doi:10.25251/skin.6.supp.s29
  16. Guttman-Yassky E, Blauvelt A, Eichenfield LF, et al. Efficacy and safety of lebrikizumab, a high-affinity interleukin 13 inhibitor, in adults with moderate to severe atopic dermatitis: a phase 2b randomized clinical trial. JAMA Dermatol. 2020;156:411-420. doi:10.1001/jamadermatol.2020.0079
  17. Lebrikizumab dosed every four weeks maintained durable skin clearance in Lilly’s phase 3 monotherapy atopic dermatitis trials [news release]. Eli Lilly and Company; September 8, 2022. Accessed October 19, 2022. https://investor.lilly.com/news-releases/news-release-details/lebrikizumab-dosed-every-four-weeks-maintained-durable-skin
  18. Guttman-Yassky E, Teixeira HD, Simpson EL, et al. Once-daily upadacitinib versus placebo in adolescents and adults with moderate-to-severe atopic dermatitis (Measure Up 1 and Measure Up 2): results from two replicate double-blind, randomised controlled phase 3 trials. Lancet. 2021;397:2151-2168. doi:10.1016/S0140-6736(21)00588-2
  19. Chovatiya R, Paller AS. JAK inhibitors in the treatment of atopic dermatitis. J Allergy Clin Immunol. 2021;148:927-940. doi:10.1016/j.jaci.2021.08.009
  20. Geng B, Hebert AA, Takiya L, et al. Efficacy and safety trends with continuous, long-term crisaborole use in patients aged ≥ 2 years with mild-to-moderate atopic dermatitis. Dermatol Ther (Heidelb). 2021;11:1667-1678. doi:10.1007/s13555-021-00584-y
  21. Appiah MM, Haft MA, Kleinman E, et al. Atopic dermatitis: review of comorbidities and therapeutics. Ann Allergy Asthma Immunol. 2022;129:142-149. doi:10.1016/j.anai.2022.05.015
Article PDF
Author and Disclosure Information

Drs. Haft and Eichenfield and Ms. Sui are from the Division of Pediatric and Adolescent Dermatology, Rady Children’s Hospital–San Diego, and the Departments of Dermatology and Pediatrics, UC San Diego School of Medicine. Dr. Choi is from the Department of Dermatology, Inje University Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea.

Drs. Haft and Choi and Ms. Sui report no conflict of interest. Dr. Eichenfield has served as an advisory board member and/or speaker, consultant, or clinical trial investigator for AbbVie, Almirall, Amgen, Arcutis, Arena Pharmaceuticals, Aslan Pharmaceuticals, Castle Biosciences, Dermavant Sciences, Eli Lilly and Company, Forte Biosciences, Galderma, Incyte, LEO Pharma, Novartis, Ortho, Otsuka, Pfizer, Regeneron, Sanofi Genzyme, and UCB.

Correspondence: Lawrence F. Eichenfield, MD, 3020 Children’s Way, Mail Code 5092, San Diego, CA 92123 ([email protected]).

Issue
Cutis - 110(5)
Publications
Topics
Page Number
259-261
Sections
Author and Disclosure Information

Drs. Haft and Eichenfield and Ms. Sui are from the Division of Pediatric and Adolescent Dermatology, Rady Children’s Hospital–San Diego, and the Departments of Dermatology and Pediatrics, UC San Diego School of Medicine. Dr. Choi is from the Department of Dermatology, Inje University Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea.

Drs. Haft and Choi and Ms. Sui report no conflict of interest. Dr. Eichenfield has served as an advisory board member and/or speaker, consultant, or clinical trial investigator for AbbVie, Almirall, Amgen, Arcutis, Arena Pharmaceuticals, Aslan Pharmaceuticals, Castle Biosciences, Dermavant Sciences, Eli Lilly and Company, Forte Biosciences, Galderma, Incyte, LEO Pharma, Novartis, Ortho, Otsuka, Pfizer, Regeneron, Sanofi Genzyme, and UCB.

Correspondence: Lawrence F. Eichenfield, MD, 3020 Children’s Way, Mail Code 5092, San Diego, CA 92123 ([email protected]).

Author and Disclosure Information

Drs. Haft and Eichenfield and Ms. Sui are from the Division of Pediatric and Adolescent Dermatology, Rady Children’s Hospital–San Diego, and the Departments of Dermatology and Pediatrics, UC San Diego School of Medicine. Dr. Choi is from the Department of Dermatology, Inje University Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea.

Drs. Haft and Choi and Ms. Sui report no conflict of interest. Dr. Eichenfield has served as an advisory board member and/or speaker, consultant, or clinical trial investigator for AbbVie, Almirall, Amgen, Arcutis, Arena Pharmaceuticals, Aslan Pharmaceuticals, Castle Biosciences, Dermavant Sciences, Eli Lilly and Company, Forte Biosciences, Galderma, Incyte, LEO Pharma, Novartis, Ortho, Otsuka, Pfizer, Regeneron, Sanofi Genzyme, and UCB.

Correspondence: Lawrence F. Eichenfield, MD, 3020 Children’s Way, Mail Code 5092, San Diego, CA 92123 ([email protected]).

Article PDF
Article PDF

Atopic dermatitis (AD), or eczema, is a common inflammatory skin disease notorious for its chronic, relapsing, and often frustrating disease course. Although as many as 25% of children in the United States are affected by this condition and its impact on the quality of life of affected patients and families is profound,1-3 therapeutic advances in the pediatric population have been fairly limited until recently.

Over the last 10 years, there has been robust investigation into pediatric AD therapeutics, with many topical and systemic medications either recently approved or under clinical investigation. These developments are changing the landscape of the management of pediatric AD and raise a set of fascinating questions about how early and aggressive intervention might change the course of this disease. We discuss current limitations in the field that may be addressed with additional research.

New Topical Medications

In the last several years, there has been a rapid increase in efforts to develop new topical agents to manage AD. Until the beginning of the 21st century, the dermatologist’s arsenal was limited to topical corticosteroids (TCs). In the early 2000s, attention shifted to topical calcineurin inhibitors as nonsteroidal alternatives when the US Food and Drug Administration (FDA) approved topical tacrolimus and pimecrolimus for AD. In 2016, crisaborole (a phosphodiesterase-4 [PDE4] inhibitor) was approved by the FDA for use in mild to moderate AD in patients 2 years and older, marking a new age of development for topical AD therapies. In 2021, the FDA approved ruxolitinib (a topical Janus kinase [JAK] 1/2 inhibitor) for use in mild to moderate AD in patients 12 years and older.

Roflumilast (ARQ-151) and difamilast (OPA-15406)(members of the PDE4 inhibitor class) are undergoing investigation for pediatric AD. A phase 3 clinical trial for roflumilast for AD is underway (ClinicalTrial.gov Identifier: NCT04845620); it is already approved for psoriasis in patients 12 years and older. A phase 3 trial of difamilast (NCT03911401) was recently completed, with results supporting the drug’s safety and efficacy in AD management.4 Efforts to synthesize new better-targeted PDE4 inhibitors are ongoing.5

Tapinarof (a novel aryl hydrocarbon receptor-modulating agent) is approved for psoriasis in adults, and a phase 3 trial for management of pediatric AD is underway (NCT05032859) after phase 2 trials revealed promising results.6

Lastly, the microbiome is a target for AD topical therapies. A recently completed phase 1 trial of bacteriotherapy with Staphylococcus hominis A9 transplant lotion showed promising results (NCT03151148).7 Although this bacteriotherapy technique is early in development and has been studied only in adult patients, results are exciting because they represent a gateway to a largely unexplored realm of potential future therapies.

Standard of Care—How will these new topical therapies impact our standard of care for pediatric AD patients? Topical corticosteroids are still a pillar of topical AD therapy, but the potential for nonsteroidal topical agents as alternatives and used in combination therapeutic regimens has expanded exponentially. It is uncertain how we might individualize regimens tailored to patient-specific factors because the standard approach has been to test drugs as monotherapy, with vehicle comparisons or with reference medications in Europe.

 

 

Newer topical nonsteroidal agents may offer several opportunities. First, they may help avoid local and systemic adverse effects that often limit the use of current standard therapy.8 This capability may prove essential in bridging TC treatments and serving as long-term maintenance therapies to decrease the frequency of eczema flares. Second, they can alleviate the need for different medication strengths for different body regions, thereby allowing for simplification of regimens and potentially increased adherence and decreased disease burden—a boon to affected patients and caregivers.

Although the efficacy and long-term safety profile of these new drugs require further study, it does not seem unreasonable to look forward to achieving levels of optimization and individualization with topical regimens for AD in the near future that makes flares in patients with mild to moderate AD a phenomenon of the past.

Advances in Systemic Therapy

Systemic therapeutics in pediatric AD also recently entered an exciting era of development. Traditional systemic agents, including cyclosporine, methotrexate, azathioprine, and mycophenolate mofetil, have existed for decades but have not been widely utilized for moderate to severe AD in the United States, especially in the pediatric population, likely because these drugs lacked FDA approval and they can cause a range of adverse effects, including notable immunosuppression.9

Introduction and approval of dupilumab in 2017 by the FDA was revolutionary in this field. As a monoclonal antibody targeted against IL-4 and IL-13, dupilumab has consistently demonstrated strong long-term efficacy for pediatric AD and has an acceptable safety profile in children and adolescents.10-14 Expansion of the label to include children as young as 6 months with moderate to severe AD seems an important milestone in pediatric AD care.

Since the approval of dupilumab for adolescents and children aged 6 to 12 years, global experience has supported expanded use of systemic agents for patients who have an inadequate response to TCs and previously approved nonsteroidal topical agents. How expansive the use of systemics will be in younger children depends on how their long-term use impacts the disease course, whether therapy is disease modifying, and whether early use can curb the development of comorbidities.

Investigations into targeted systemic therapeutics for eczematous dermatitis are not limited to dupilumab. In a study of adolescents as young as 12 years, tralokinumab (an IL-13 pathway inhibitor) demonstrated an Eczema Area Severity Index-75 of 27.8% to 28.6% and a mean decrease in the SCORing Atopic Dermatitis index of 27.5 to 29.1, with minimal adverse effects.15 Lebrikizumab, another biologic IL-13 inhibitor with strong published safety and efficacy data in adults, has completed short- and longer-term studies in adolescents (NCT04178967 and NCT04146363).16 The drug received FDA Fast Track designation for moderate to severe AD in patients 12 years and older after showing positive data.17

This push to targeted therapy stretches beyond monoclonal antibodies. In the last few years, oral JAK inhibitors have emerged as a new class of systemic therapy for eczematous dermatitis. Upadacitinib, a JAK1 selective inhibitor, was approved by the FDA in 2022 for patients 12 years and older with AD and has data that supports its efficacy in adolescents and adults.18 Other JAK inhibitors including the selective JAK1 inhibitor abrocitinib and the combined JAK1/2 inhibitor baricitinib are being studied for pediatric AD (NCT04564755, NCT03422822, and NCT03952559), with most evidence to date supporting their safety and efficacy, at least over the short-term.19

 

 

The study of these and other advanced systemic therapies for eczematous dermatitis is transforming the toolbox for pediatric AD care. Although long-term data are lacking for some of these medications, it is possible that newer agents may decrease reliance on older immunosuppressants, such as systemic corticosteroids, cyclosporine, and methotrexate. Unanswered questions include: How and which systemic medications may alter the course of the disease? What is the disease modification for AD? What is the impact on comorbidities over time?

What’s Missing?

The field of pediatric AD has experienced exciting new developments with the emergence of targeted therapeutics, but those new agents require more long-term study, though we already have longer-term data on crisaborole and dupilumab.10-14,20 Studies of the long-term use of these new treatments on comorbidities of pediatric AD—mental health outcomes, cardiovascular disease, effects on the family, and other allergic conditions—are needed.21 Furthermore, clinical guidelines that address indications, timing of use, tapering, and discontinuation of new treatments depend on long-term experience and data collection.

Therefore, it is prudent that investigators, companies, payers, patients, and families support phase 4, long-term extension, and registry studies, which will expand our knowledge of AD medications and their impact on the disease over time.

Final Thoughts

Medications to treat AD are reaching a new level of advancement—from topical agents that target novel pathways to revolutionary biologics and systemic medications. Although there are knowledge gaps on these new therapeutics, the standard of care is already rapidly changing as the expectations of clinicians, patients, and families advance with each addition to the provider’s toolbox.

Atopic dermatitis (AD), or eczema, is a common inflammatory skin disease notorious for its chronic, relapsing, and often frustrating disease course. Although as many as 25% of children in the United States are affected by this condition and its impact on the quality of life of affected patients and families is profound,1-3 therapeutic advances in the pediatric population have been fairly limited until recently.

Over the last 10 years, there has been robust investigation into pediatric AD therapeutics, with many topical and systemic medications either recently approved or under clinical investigation. These developments are changing the landscape of the management of pediatric AD and raise a set of fascinating questions about how early and aggressive intervention might change the course of this disease. We discuss current limitations in the field that may be addressed with additional research.

New Topical Medications

In the last several years, there has been a rapid increase in efforts to develop new topical agents to manage AD. Until the beginning of the 21st century, the dermatologist’s arsenal was limited to topical corticosteroids (TCs). In the early 2000s, attention shifted to topical calcineurin inhibitors as nonsteroidal alternatives when the US Food and Drug Administration (FDA) approved topical tacrolimus and pimecrolimus for AD. In 2016, crisaborole (a phosphodiesterase-4 [PDE4] inhibitor) was approved by the FDA for use in mild to moderate AD in patients 2 years and older, marking a new age of development for topical AD therapies. In 2021, the FDA approved ruxolitinib (a topical Janus kinase [JAK] 1/2 inhibitor) for use in mild to moderate AD in patients 12 years and older.

Roflumilast (ARQ-151) and difamilast (OPA-15406)(members of the PDE4 inhibitor class) are undergoing investigation for pediatric AD. A phase 3 clinical trial for roflumilast for AD is underway (ClinicalTrial.gov Identifier: NCT04845620); it is already approved for psoriasis in patients 12 years and older. A phase 3 trial of difamilast (NCT03911401) was recently completed, with results supporting the drug’s safety and efficacy in AD management.4 Efforts to synthesize new better-targeted PDE4 inhibitors are ongoing.5

Tapinarof (a novel aryl hydrocarbon receptor-modulating agent) is approved for psoriasis in adults, and a phase 3 trial for management of pediatric AD is underway (NCT05032859) after phase 2 trials revealed promising results.6

Lastly, the microbiome is a target for AD topical therapies. A recently completed phase 1 trial of bacteriotherapy with Staphylococcus hominis A9 transplant lotion showed promising results (NCT03151148).7 Although this bacteriotherapy technique is early in development and has been studied only in adult patients, results are exciting because they represent a gateway to a largely unexplored realm of potential future therapies.

Standard of Care—How will these new topical therapies impact our standard of care for pediatric AD patients? Topical corticosteroids are still a pillar of topical AD therapy, but the potential for nonsteroidal topical agents as alternatives and used in combination therapeutic regimens has expanded exponentially. It is uncertain how we might individualize regimens tailored to patient-specific factors because the standard approach has been to test drugs as monotherapy, with vehicle comparisons or with reference medications in Europe.

 

 

Newer topical nonsteroidal agents may offer several opportunities. First, they may help avoid local and systemic adverse effects that often limit the use of current standard therapy.8 This capability may prove essential in bridging TC treatments and serving as long-term maintenance therapies to decrease the frequency of eczema flares. Second, they can alleviate the need for different medication strengths for different body regions, thereby allowing for simplification of regimens and potentially increased adherence and decreased disease burden—a boon to affected patients and caregivers.

Although the efficacy and long-term safety profile of these new drugs require further study, it does not seem unreasonable to look forward to achieving levels of optimization and individualization with topical regimens for AD in the near future that makes flares in patients with mild to moderate AD a phenomenon of the past.

Advances in Systemic Therapy

Systemic therapeutics in pediatric AD also recently entered an exciting era of development. Traditional systemic agents, including cyclosporine, methotrexate, azathioprine, and mycophenolate mofetil, have existed for decades but have not been widely utilized for moderate to severe AD in the United States, especially in the pediatric population, likely because these drugs lacked FDA approval and they can cause a range of adverse effects, including notable immunosuppression.9

Introduction and approval of dupilumab in 2017 by the FDA was revolutionary in this field. As a monoclonal antibody targeted against IL-4 and IL-13, dupilumab has consistently demonstrated strong long-term efficacy for pediatric AD and has an acceptable safety profile in children and adolescents.10-14 Expansion of the label to include children as young as 6 months with moderate to severe AD seems an important milestone in pediatric AD care.

Since the approval of dupilumab for adolescents and children aged 6 to 12 years, global experience has supported expanded use of systemic agents for patients who have an inadequate response to TCs and previously approved nonsteroidal topical agents. How expansive the use of systemics will be in younger children depends on how their long-term use impacts the disease course, whether therapy is disease modifying, and whether early use can curb the development of comorbidities.

Investigations into targeted systemic therapeutics for eczematous dermatitis are not limited to dupilumab. In a study of adolescents as young as 12 years, tralokinumab (an IL-13 pathway inhibitor) demonstrated an Eczema Area Severity Index-75 of 27.8% to 28.6% and a mean decrease in the SCORing Atopic Dermatitis index of 27.5 to 29.1, with minimal adverse effects.15 Lebrikizumab, another biologic IL-13 inhibitor with strong published safety and efficacy data in adults, has completed short- and longer-term studies in adolescents (NCT04178967 and NCT04146363).16 The drug received FDA Fast Track designation for moderate to severe AD in patients 12 years and older after showing positive data.17

This push to targeted therapy stretches beyond monoclonal antibodies. In the last few years, oral JAK inhibitors have emerged as a new class of systemic therapy for eczematous dermatitis. Upadacitinib, a JAK1 selective inhibitor, was approved by the FDA in 2022 for patients 12 years and older with AD and has data that supports its efficacy in adolescents and adults.18 Other JAK inhibitors including the selective JAK1 inhibitor abrocitinib and the combined JAK1/2 inhibitor baricitinib are being studied for pediatric AD (NCT04564755, NCT03422822, and NCT03952559), with most evidence to date supporting their safety and efficacy, at least over the short-term.19

 

 

The study of these and other advanced systemic therapies for eczematous dermatitis is transforming the toolbox for pediatric AD care. Although long-term data are lacking for some of these medications, it is possible that newer agents may decrease reliance on older immunosuppressants, such as systemic corticosteroids, cyclosporine, and methotrexate. Unanswered questions include: How and which systemic medications may alter the course of the disease? What is the disease modification for AD? What is the impact on comorbidities over time?

What’s Missing?

The field of pediatric AD has experienced exciting new developments with the emergence of targeted therapeutics, but those new agents require more long-term study, though we already have longer-term data on crisaborole and dupilumab.10-14,20 Studies of the long-term use of these new treatments on comorbidities of pediatric AD—mental health outcomes, cardiovascular disease, effects on the family, and other allergic conditions—are needed.21 Furthermore, clinical guidelines that address indications, timing of use, tapering, and discontinuation of new treatments depend on long-term experience and data collection.

Therefore, it is prudent that investigators, companies, payers, patients, and families support phase 4, long-term extension, and registry studies, which will expand our knowledge of AD medications and their impact on the disease over time.

Final Thoughts

Medications to treat AD are reaching a new level of advancement—from topical agents that target novel pathways to revolutionary biologics and systemic medications. Although there are knowledge gaps on these new therapeutics, the standard of care is already rapidly changing as the expectations of clinicians, patients, and families advance with each addition to the provider’s toolbox.

References
  1. Eichenfield LF, Tom WL, Chamlin SL, et al. Guidelines of care for the management of atopic dermatitis: part 1. diagnosis and assessment of atopic dermatitis. J Am Acad Dermatol. 2014;70:338-351. doi:10.1016/j.jaad.2013.10.010
  2. Kiebert G, Sorensen SV, Revicki D, et al. Atopic dermatitis is associated with a decrement in health-related quality of life. Int J Dermatol. 2002;41:151-158. doi:10.1046/j.1365-4362.2002.01436.x
  3. Al Shobaili HA. The impact of childhood atopic dermatitis on the patients’ family. Pediatr Dermatol. 2010;27:618-623. doi:10.1111/j.1525-1470.2010.01215.x
  4. Saeki H, Baba N, Ito K, et al. Difamilast, a selective phosphodiesterase 4 inhibitor, ointment in paediatric patients with atopic dermatitis: a phase III randomized double-blind, vehicle-controlled trial [published online November 1, 2021]. Br J Dermatol. 2022;186:40-49. doi:10.1111/bjd.20655
  5. Chu Z, Xu Q, Zhu Q, et al. Design, synthesis and biological evaluation of novel benzoxaborole derivatives as potent PDE4 inhibitors for topical treatment of atopic dermatitis. Eur J Med Chem. 2021;213:113171. doi:10.1016/j.ejmech.2021.113171
  6. Paller AS, Stein Gold L, Soung J, et al. Efficacy and patient-reported outcomes from a phase 2b, randomized clinical trial of tapinarof cream for the treatment of adolescents and adults with atopic dermatitis. J Am Acad Dermatol. 2021;84:632-638. doi:10.1016/j.jaad.2020.05.135
  7. Nakatsuji T, Hata TR, Tong Y, et al. Development of a human skin commensal microbe for bacteriotherapy of atopic dermatitis and use in a phase 1 randomized clinical trial. Nat Med. 2021;27:700-709. doi:10.1038/s41591-021-01256-2
  8. Eichenfield LF, Tom WL, Berger TG, et al. Guidelines of care for the management of atopic dermatitis: part 2. management and treatment of atopic dermatitis with topical therapies. J Am Acad Dermatol. 2014;71:116-132. doi:10.1016/j.jaad.2014.03.023
  9. Sidbury R, Davis DM, Cohen DE, et al. Guidelines of care for the management of atopic dermatitis: part 3. management and treatment with phototherapy and systemic agents. J Am Acad Dermatol. 2014;71:327-349. doi:10.1016/j.jaad.2014.03.030
  10. Gooderham MJ, Hong HC-H, Eshtiaghi P, et al. Dupilumab: a review of its use in the treatment of atopic dermatitis. J Am Acad Dermatol. 2018;78(3 suppl 1):S28-S36. doi:10.1016/j.jaad.2017.12.022
  11. Simpson EL, Paller AS, Siegfried EC, et al. Efficacy and safety of dupilumab in adolescents with uncontrolled moderate to severe atopic dermatitis: a phase 3 randomized clinical trial. JAMA Dermatol. 2020;156:44-56. doi:10.1001/jamadermatol.2019.3336
  12. Blauvelt A, Guttman-Yassky E, Paller AS, et al. Long-term efficacy and safety of dupilumab in adolescents with moderate-to-severe atopicdermatitis: results through week 52 from a phase III open-label extension trial (LIBERTY AD PED-OLE). Am J Clin Dermatol. 2022;23:365-383. doi:10.1007/s40257-022-00683-2
  13. Cork MJ, Thaçi D, Eichenfield LF, et al. Dupilumab provides favourable long-term safety and efficacy in children aged ≥ 6 to < 12 years with uncontrolled severe atopic dermatitis: results from an open-label phase IIa study and subsequent phase III open-label extension study. Br J Dermatol. 2021;184:857-870. doi:10.1111/bjd.19460
  14. Simpson EL, Paller AS, Siegfried EC, et al. Dupilumab demonstrates rapid and consistent improvement in extent and signs of atopic dermatitis across all anatomical regions in pediatric patients 6 years of age and older. Dermatol Ther (Heidelb). 2021;11:1643-1656. doi:10.1007/s13555-021-00568-y
  15. Paller A, Blauvelt A, Soong W, et al. Efficacy and safety of tralokinumab in adolescents with moderate-to-severe atopic dermatitis: results of the phase 3 ECZTRA 6 trial. SKIN. 2022;6:S29. doi:10.25251/skin.6.supp.s29
  16. Guttman-Yassky E, Blauvelt A, Eichenfield LF, et al. Efficacy and safety of lebrikizumab, a high-affinity interleukin 13 inhibitor, in adults with moderate to severe atopic dermatitis: a phase 2b randomized clinical trial. JAMA Dermatol. 2020;156:411-420. doi:10.1001/jamadermatol.2020.0079
  17. Lebrikizumab dosed every four weeks maintained durable skin clearance in Lilly’s phase 3 monotherapy atopic dermatitis trials [news release]. Eli Lilly and Company; September 8, 2022. Accessed October 19, 2022. https://investor.lilly.com/news-releases/news-release-details/lebrikizumab-dosed-every-four-weeks-maintained-durable-skin
  18. Guttman-Yassky E, Teixeira HD, Simpson EL, et al. Once-daily upadacitinib versus placebo in adolescents and adults with moderate-to-severe atopic dermatitis (Measure Up 1 and Measure Up 2): results from two replicate double-blind, randomised controlled phase 3 trials. Lancet. 2021;397:2151-2168. doi:10.1016/S0140-6736(21)00588-2
  19. Chovatiya R, Paller AS. JAK inhibitors in the treatment of atopic dermatitis. J Allergy Clin Immunol. 2021;148:927-940. doi:10.1016/j.jaci.2021.08.009
  20. Geng B, Hebert AA, Takiya L, et al. Efficacy and safety trends with continuous, long-term crisaborole use in patients aged ≥ 2 years with mild-to-moderate atopic dermatitis. Dermatol Ther (Heidelb). 2021;11:1667-1678. doi:10.1007/s13555-021-00584-y
  21. Appiah MM, Haft MA, Kleinman E, et al. Atopic dermatitis: review of comorbidities and therapeutics. Ann Allergy Asthma Immunol. 2022;129:142-149. doi:10.1016/j.anai.2022.05.015
References
  1. Eichenfield LF, Tom WL, Chamlin SL, et al. Guidelines of care for the management of atopic dermatitis: part 1. diagnosis and assessment of atopic dermatitis. J Am Acad Dermatol. 2014;70:338-351. doi:10.1016/j.jaad.2013.10.010
  2. Kiebert G, Sorensen SV, Revicki D, et al. Atopic dermatitis is associated with a decrement in health-related quality of life. Int J Dermatol. 2002;41:151-158. doi:10.1046/j.1365-4362.2002.01436.x
  3. Al Shobaili HA. The impact of childhood atopic dermatitis on the patients’ family. Pediatr Dermatol. 2010;27:618-623. doi:10.1111/j.1525-1470.2010.01215.x
  4. Saeki H, Baba N, Ito K, et al. Difamilast, a selective phosphodiesterase 4 inhibitor, ointment in paediatric patients with atopic dermatitis: a phase III randomized double-blind, vehicle-controlled trial [published online November 1, 2021]. Br J Dermatol. 2022;186:40-49. doi:10.1111/bjd.20655
  5. Chu Z, Xu Q, Zhu Q, et al. Design, synthesis and biological evaluation of novel benzoxaborole derivatives as potent PDE4 inhibitors for topical treatment of atopic dermatitis. Eur J Med Chem. 2021;213:113171. doi:10.1016/j.ejmech.2021.113171
  6. Paller AS, Stein Gold L, Soung J, et al. Efficacy and patient-reported outcomes from a phase 2b, randomized clinical trial of tapinarof cream for the treatment of adolescents and adults with atopic dermatitis. J Am Acad Dermatol. 2021;84:632-638. doi:10.1016/j.jaad.2020.05.135
  7. Nakatsuji T, Hata TR, Tong Y, et al. Development of a human skin commensal microbe for bacteriotherapy of atopic dermatitis and use in a phase 1 randomized clinical trial. Nat Med. 2021;27:700-709. doi:10.1038/s41591-021-01256-2
  8. Eichenfield LF, Tom WL, Berger TG, et al. Guidelines of care for the management of atopic dermatitis: part 2. management and treatment of atopic dermatitis with topical therapies. J Am Acad Dermatol. 2014;71:116-132. doi:10.1016/j.jaad.2014.03.023
  9. Sidbury R, Davis DM, Cohen DE, et al. Guidelines of care for the management of atopic dermatitis: part 3. management and treatment with phototherapy and systemic agents. J Am Acad Dermatol. 2014;71:327-349. doi:10.1016/j.jaad.2014.03.030
  10. Gooderham MJ, Hong HC-H, Eshtiaghi P, et al. Dupilumab: a review of its use in the treatment of atopic dermatitis. J Am Acad Dermatol. 2018;78(3 suppl 1):S28-S36. doi:10.1016/j.jaad.2017.12.022
  11. Simpson EL, Paller AS, Siegfried EC, et al. Efficacy and safety of dupilumab in adolescents with uncontrolled moderate to severe atopic dermatitis: a phase 3 randomized clinical trial. JAMA Dermatol. 2020;156:44-56. doi:10.1001/jamadermatol.2019.3336
  12. Blauvelt A, Guttman-Yassky E, Paller AS, et al. Long-term efficacy and safety of dupilumab in adolescents with moderate-to-severe atopicdermatitis: results through week 52 from a phase III open-label extension trial (LIBERTY AD PED-OLE). Am J Clin Dermatol. 2022;23:365-383. doi:10.1007/s40257-022-00683-2
  13. Cork MJ, Thaçi D, Eichenfield LF, et al. Dupilumab provides favourable long-term safety and efficacy in children aged ≥ 6 to < 12 years with uncontrolled severe atopic dermatitis: results from an open-label phase IIa study and subsequent phase III open-label extension study. Br J Dermatol. 2021;184:857-870. doi:10.1111/bjd.19460
  14. Simpson EL, Paller AS, Siegfried EC, et al. Dupilumab demonstrates rapid and consistent improvement in extent and signs of atopic dermatitis across all anatomical regions in pediatric patients 6 years of age and older. Dermatol Ther (Heidelb). 2021;11:1643-1656. doi:10.1007/s13555-021-00568-y
  15. Paller A, Blauvelt A, Soong W, et al. Efficacy and safety of tralokinumab in adolescents with moderate-to-severe atopic dermatitis: results of the phase 3 ECZTRA 6 trial. SKIN. 2022;6:S29. doi:10.25251/skin.6.supp.s29
  16. Guttman-Yassky E, Blauvelt A, Eichenfield LF, et al. Efficacy and safety of lebrikizumab, a high-affinity interleukin 13 inhibitor, in adults with moderate to severe atopic dermatitis: a phase 2b randomized clinical trial. JAMA Dermatol. 2020;156:411-420. doi:10.1001/jamadermatol.2020.0079
  17. Lebrikizumab dosed every four weeks maintained durable skin clearance in Lilly’s phase 3 monotherapy atopic dermatitis trials [news release]. Eli Lilly and Company; September 8, 2022. Accessed October 19, 2022. https://investor.lilly.com/news-releases/news-release-details/lebrikizumab-dosed-every-four-weeks-maintained-durable-skin
  18. Guttman-Yassky E, Teixeira HD, Simpson EL, et al. Once-daily upadacitinib versus placebo in adolescents and adults with moderate-to-severe atopic dermatitis (Measure Up 1 and Measure Up 2): results from two replicate double-blind, randomised controlled phase 3 trials. Lancet. 2021;397:2151-2168. doi:10.1016/S0140-6736(21)00588-2
  19. Chovatiya R, Paller AS. JAK inhibitors in the treatment of atopic dermatitis. J Allergy Clin Immunol. 2021;148:927-940. doi:10.1016/j.jaci.2021.08.009
  20. Geng B, Hebert AA, Takiya L, et al. Efficacy and safety trends with continuous, long-term crisaborole use in patients aged ≥ 2 years with mild-to-moderate atopic dermatitis. Dermatol Ther (Heidelb). 2021;11:1667-1678. doi:10.1007/s13555-021-00584-y
  21. Appiah MM, Haft MA, Kleinman E, et al. Atopic dermatitis: review of comorbidities and therapeutics. Ann Allergy Asthma Immunol. 2022;129:142-149. doi:10.1016/j.anai.2022.05.015
Issue
Cutis - 110(5)
Issue
Cutis - 110(5)
Page Number
259-261
Page Number
259-261
Publications
Publications
Topics
Article Type
Display Headline
Past, Present, and Future of Pediatric Atopic Dermatitis Management
Display Headline
Past, Present, and Future of Pediatric Atopic Dermatitis Management
Sections
Inside the Article

PRACTICE POINTS

  • Pediatric atopic dermatitis (AD) therapeutics have rapidly evolved over the last decade and dermatologists should be aware of new tools in their treatment arsenal.
  • New topical nonsteroidal agents serve as useful alternatives to topical corticosteroids through mitigating adverse effects from current standard therapy and potentially simplifying topical regimens.
  • Monoclonal antibodies and Janus kinase inhibitors are part of an important set of new systemic therapeutics for pediatric AD.
  • Long-term data on these new therapeutics is required to better understand their impact on pediatric AD comorbidities and impact on the longitudinal disease course.
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
Gating Strategy
No Gating
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Acquired Acrodermatitis Enteropathica in an Infant

Article Type
Changed
Tue, 05/02/2023 - 13:45
Display Headline
Acquired Acrodermatitis Enteropathica in an Infant

Acrodermatitis enteropathica (AE) is a rare disorder of zinc metabolism that typically presents in infancy.1 Although it is clinically characterized by acral and periorificial dermatitis, alopecia, and diarrhea, only 20% of cases present with this triad.2 Zinc deficiency in AE can either be acquired or inborn (congenital). Acquired forms can occur from dietary inadequacy or malabsorption, whereas genetic causes are related to an autosomal-recessive disorder affecting zinc transporters.1 We report a case of a 3-month-old female infant with acquired AE who was successfully treated with zinc supplementation over the course of 3 weeks.

Case Report

A 3-month-old female infant presented to the emergency department with a rash of 2 weeks’ duration. She was born full term with no birth complications. The patient’s mother reported that the rash started on the cheeks, then enlarged and spread to the neck, back, and perineum. The patient also had been having diarrhea during this time. She previously had received mupirocin and cephalexin with no response to treatment. Maternal history was negative for lupus, and the mother’s diet consisted of a variety of foods but not many vegetables. The patient was exclusively breastfed, and there was no pertinent history of similar rashes occurring in other family members.

Physical examination revealed the patient had annular and polycyclic, hyperkeratotic, crusted papules and plaques on the cheeks, neck, back, and axillae, as well as the perineum/groin and perianal regions (Figure 1). The differential diagnosis at the time included neonatal lupus, zinc deficiency, and syphilis. Relevant laboratory testing and a shave biopsy of the left axilla were obtained.

A, Annular and polycyclic, hyperkeratotic, crusted papules and plaques on the cheeks. B, Similar lesions were present in the perineum/groin and perianal regions.
FIGURE 1. A, Annular and polycyclic, hyperkeratotic, crusted papules and plaques on the cheeks. B, Similar lesions were present in the perineum/groin and perianal regions.


Pertinent laboratory findings included a low zinc level (23 μg/dL [reference range, 26–141 μg/dL]), low alkaline phosphatase level (74 U/L [reference range, 94–486 U/L]), and thrombocytosis (826×109/L [reference range, 150–400×109/L). Results for antinuclear antibody and anti–Sjögren syndrome–related antigen A and B antibody testing were negative. A rapid plasma reagin test was nonreactive. Histologic examination revealed psoriasiform hyperplasia with overlying confluent parakeratosis, focal spongiosis, multiple dyskeratotic keratinocytes, and mitotic figures (Figure 2). Ballooning was evident in focal cells in the subcorneal region in addition to an accompanying lymphocytic infiltrate and occasional neutrophils.

Biopsy of the left axilla showed psoriasiform hyperplasia with overlying confluent parakeratosis, focal spongiosis, multiple dyskeratotic keratinocytes, and mitotic figures
FIGURE 2. Biopsy of the left axilla showed psoriasiform hyperplasia with overlying confluent parakeratosis, focal spongiosis, multiple dyskeratotic keratinocytes, and mitotic figures (H&E, original magnification ×10). Focal cells in the subcorneal region showed ballooning with a lymphocytic infiltrate and neutrophils (inset: H&E, original magnification ×40).


The patient was given a 10-mg/mL suspension of elemental zinc and was advised to take 1 mL (10 mg) by mouth twice daily with food. This dosage equated to 3 mg/kg/d. On follow-up 3 weeks later, the skin began to clear (Figure 3). Follow-up laboratory testing showed an increase in zinc (114 μg/dL) and alkaline phosphatase levels (313 U/L). The patient was able to discontinue the zinc supplementation, and follow-up during the next year revealed no recurrence.

A, Three weeks after treatment with zinc supplementation, the annular crusted papules and plaques were no longer evident on the cheeks. B, The perineum/groin and perianal regions showed similar clearance.
FIGURE 3. A, Three weeks after treatment with zinc supplementation, the annular crusted papules and plaques were no longer evident on the cheeks. B, The perineum/groin and perianal regions showed similar clearance.

Comment

Etiology of AE—Acrodermatitis enteropathica was first identified in 1942 as an acral rash associated with diarrhea3; in 1973, Barnes and Moynahan4 discovered zinc deficiency as a causal agent for these findings. The causes of AE are further subclassified as either an acquired or inborn etiology. Congenital causes commonly are seen in infants within the first few months of life, whereas acquired forms are seen at any age. Acquired forms in infants can occur from failure of the mother to secrete zinc in breast milk, low maternal serum zinc levels, or other reasons causing low nutritional intake. A single mutation in the SLC30A2 gene has been found to markedly reduce zinc concentrations in breast milk, thus causing zinc deficiency in breastfed infants.5 Other acquired forms can be caused by malabsorption, sometimes after surgery such as intestinal bypass or from intravenous nutrition without sufficient zinc.1 The congenital form of AE is an autosomal-recessive disorder occurring from mutations in the SLC39A4 gene located on band 8q24.3. Affected individuals have a decreased ability to absorb zinc in the small intestine because of defects in zinc transporters ZIP and ZnT.6 Based on our patient’s laboratory findings and history, it is believed that the zinc deficiency was acquired, as the condition normalized with repletion and has not required any supplementation in the year of follow-up. In addition, the absence of a pertinent family history supported an acquired diagnosis, which has various etiologies, whereas the congenital form primarily is a genetic disease.

Diagnosis of AE—The characteristic clinical features of AE include erythematous, dry, scaly papules and plaques that may evolve into crusted, erosive, pustular lesions. These lesions typically are distributed in a periorificial and acral pattern.1,2 Although AE includes the clinical triad of acral and periorificial dermatitis, alopecia, and diarrhea, most cases present with only partial features of this syndrome, as seen in our patient, who presented with only 2 symptoms—dermatitis and diarrhea. The diagnosis of AE is based on clinical and laboratory abnormalities, especially a low serum zinc level. Low levels of zinc-dependent enzymes, such as alkaline phosphatase, may support the diagnosis, as seen in our patient. Histologic evaluation is characteristic but is not diagnostic, as the same findings can be seen in other nutritional disorders. Such findings include confluent parakeratosis associated with a reduced granular layer in early lesions and subsequent ballooning of subcorneal keratinocytes, upper epidermal pallor, and intraepidermal clefts. Late lesions exhibit psoriasiform hyperplasia of the epidermis with less epidermal pallor.7

 

 

Management—Treatment of AE includes supplementation with oral elemental zinc; however, there are scant evidence-based recommendations on the exact dose of zinc to be given. Generally, the recommended amount is 3 mg/kg/d.8 For individuals with the congenital form of AE, lifelong zinc supplementation is additionally recommended.9 It is important to recognize this presentation because the patient can develop worsening irritability, severe diarrhea, nail dystrophy, hair loss, immune dysfunction, and numerous ophthalmic disorders if left untreated. Acute zinc toxicity due to excess administration is rare, with symptoms of nausea and vomiting occurring with dosages of 50 to 100 mg/d. Additionally, dosages of up to 70 mg twice weekly have been provided without any toxic effect.10 In our case, 3 mg/kg/d of oral zinc supplementation proved to be effective in resolving the patient’s symptoms of acquired zinc deficiency.

Differential Diagnosis—It is important to note that deficiencies of other nutrients may present as an AE-like eruption called acrodermatitis dysmetabolica (AD). Both diseases may present with the triad of dermatitis, alopecia, and diarrhea; however, AD is associated with inborn errors of metabolism. There have been cases that describe AD in patients with a zinc deficiency in conjunction with a deficiency of branched-chain amino acids.11,12 It is important to consider AD in the differential diagnosis of an AE eruption, especially in the context of a metabolic disorder, as it may affect the treatment plan. One case described the dermatitis of AD as not responding to zinc supplementation alone, while another described improvement after increasing an isoleucine supplementation dose.11,12

Other considerations in the differential diagnoses include AE-like conditions such as biotinidase deficiency, multiple carboxylase deficiency, and essential fatty acid deficiency. An AE-like condition may present with the triad of dermatitis, alopecia, and diarrhea. However, unlike in true AE, zinc and alkaline phosphatase levels tend to be normal in these conditions. Other features seen in AE-like conditions depend on the underlying cause but often include failure to thrive, neurologic defects, ophthalmic abnormalities, and metabolic abnormalities.13
References
  1. Acrodermatitis enteropathica. National Organization for Rare Disorders. Accessed October 16, 2022. https://rarediseases.org/rare-diseases/acrodermatitis-enteropathica/
  2. Perafán-Riveros C, França LFS, Alves ACF, et al. Acrodermatitis enteropathica: case report and review of the literature. Pediatr Dermatol. 2002;19:426-431.
  3. Danbolt N. Acrodermatitis enteropathica. Br J Dermatol. 1979;100:37-40.
  4. Barnes PM, Moynahan EJ. Zinc deficiency in acrodermatitis enteropathica: multiple dietary intolerance treated with synthetic diet. Proc R Soc Med. 1973;66:327-329.
  5. Lee S, Zhou Y, Gill DL, et al. A genetic variant in SLC30A2 causes breast dysfunction during lactation by inducing ER stress, oxidative stress and epithelial barrier defects. Sci Rep. 2018;8:3542.
  6. Kaur S, Sangwan A, Sahu P, et al. Clinical variants of acrodermatitis enteropathica and its co-relation with genetics. Indian J Paediatr Dermatol. 2016;17:35-37.
  7. Dela Rosa KM, James WD. Acrodermatitis enteropathica workup. Medscape. Updated June 4, 2021. Accessed October 16, 2022. https://emedicine.medscape.com/article/1102575-workup#showall
  8. Ngan V, Gangakhedkar A, Oakley A. Acrodermatitis enteropathica. DermNet. Accessed October 16, 2022. https://dermnetnz.org/topics/acrodermatitis-enteropathica/
  9. Ranugha P, Sethi P, Veeranna S. Acrodermatitis enteropathica: the need for sustained high dose zinc supplementation. Dermatol Online J. 2018;24:13030/qt1w9002sr.
  10. Larson CP, Roy SK, Khan AI, et al. Zinc treatment to under-five children: applications to improve child survival and reduce burden of disease. J Health Popul Nutr. 2008;26:356-365.
  11. Samady JA, Schwartz RA, Shih LY, et al. Acrodermatitis enteropathica-like eruption in an infant with nonketotic hyperglycinemia. J Dermatol. 2000;27:604-608.
  12. Flores K, Chikowski R, Morrell DS. Acrodermatitis dysmetabolica in an infant with maple syrup urine disease. Clin Exp Dermatol. 2016;41:651-654.
  13. Jones L, Oakley A. Acrodermatitis enteropathica-like conditions. DermNet. Accessed August 30, 2022. https://dermnetnz.org/topics/acrodermatitis-enteropathica-like-conditions
Article PDF
Author and Disclosure Information

Ms. Vu and Drs. Becker and Osswald are from the University of Texas Health Science Center at San Antonio. Ms. Vu is from the Long School of Medicine, and Drs. Becker and Osswald are from the Department of Dermatology. Dr. Gillooly is from Wright-Patterson Medical Center, Wright-Patterson Air Force Base, Ohio.

The authors report no conflict of interest.

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

Correspondence: Marie Vu, BSA, University of Texas Health Science Center at San Antonio, 7979 Wurzbach Rd, Grossman, 3rd Floor, San Antonio, TX 78229 ([email protected]).

Issue
Cutis - 110(5)
Publications
Topics
Page Number
281-283
Sections
Author and Disclosure Information

Ms. Vu and Drs. Becker and Osswald are from the University of Texas Health Science Center at San Antonio. Ms. Vu is from the Long School of Medicine, and Drs. Becker and Osswald are from the Department of Dermatology. Dr. Gillooly is from Wright-Patterson Medical Center, Wright-Patterson Air Force Base, Ohio.

The authors report no conflict of interest.

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

Correspondence: Marie Vu, BSA, University of Texas Health Science Center at San Antonio, 7979 Wurzbach Rd, Grossman, 3rd Floor, San Antonio, TX 78229 ([email protected]).

Author and Disclosure Information

Ms. Vu and Drs. Becker and Osswald are from the University of Texas Health Science Center at San Antonio. Ms. Vu is from the Long School of Medicine, and Drs. Becker and Osswald are from the Department of Dermatology. Dr. Gillooly is from Wright-Patterson Medical Center, Wright-Patterson Air Force Base, Ohio.

The authors report no conflict of interest.

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

Correspondence: Marie Vu, BSA, University of Texas Health Science Center at San Antonio, 7979 Wurzbach Rd, Grossman, 3rd Floor, San Antonio, TX 78229 ([email protected]).

Article PDF
Article PDF

Acrodermatitis enteropathica (AE) is a rare disorder of zinc metabolism that typically presents in infancy.1 Although it is clinically characterized by acral and periorificial dermatitis, alopecia, and diarrhea, only 20% of cases present with this triad.2 Zinc deficiency in AE can either be acquired or inborn (congenital). Acquired forms can occur from dietary inadequacy or malabsorption, whereas genetic causes are related to an autosomal-recessive disorder affecting zinc transporters.1 We report a case of a 3-month-old female infant with acquired AE who was successfully treated with zinc supplementation over the course of 3 weeks.

Case Report

A 3-month-old female infant presented to the emergency department with a rash of 2 weeks’ duration. She was born full term with no birth complications. The patient’s mother reported that the rash started on the cheeks, then enlarged and spread to the neck, back, and perineum. The patient also had been having diarrhea during this time. She previously had received mupirocin and cephalexin with no response to treatment. Maternal history was negative for lupus, and the mother’s diet consisted of a variety of foods but not many vegetables. The patient was exclusively breastfed, and there was no pertinent history of similar rashes occurring in other family members.

Physical examination revealed the patient had annular and polycyclic, hyperkeratotic, crusted papules and plaques on the cheeks, neck, back, and axillae, as well as the perineum/groin and perianal regions (Figure 1). The differential diagnosis at the time included neonatal lupus, zinc deficiency, and syphilis. Relevant laboratory testing and a shave biopsy of the left axilla were obtained.

A, Annular and polycyclic, hyperkeratotic, crusted papules and plaques on the cheeks. B, Similar lesions were present in the perineum/groin and perianal regions.
FIGURE 1. A, Annular and polycyclic, hyperkeratotic, crusted papules and plaques on the cheeks. B, Similar lesions were present in the perineum/groin and perianal regions.


Pertinent laboratory findings included a low zinc level (23 μg/dL [reference range, 26–141 μg/dL]), low alkaline phosphatase level (74 U/L [reference range, 94–486 U/L]), and thrombocytosis (826×109/L [reference range, 150–400×109/L). Results for antinuclear antibody and anti–Sjögren syndrome–related antigen A and B antibody testing were negative. A rapid plasma reagin test was nonreactive. Histologic examination revealed psoriasiform hyperplasia with overlying confluent parakeratosis, focal spongiosis, multiple dyskeratotic keratinocytes, and mitotic figures (Figure 2). Ballooning was evident in focal cells in the subcorneal region in addition to an accompanying lymphocytic infiltrate and occasional neutrophils.

Biopsy of the left axilla showed psoriasiform hyperplasia with overlying confluent parakeratosis, focal spongiosis, multiple dyskeratotic keratinocytes, and mitotic figures
FIGURE 2. Biopsy of the left axilla showed psoriasiform hyperplasia with overlying confluent parakeratosis, focal spongiosis, multiple dyskeratotic keratinocytes, and mitotic figures (H&E, original magnification ×10). Focal cells in the subcorneal region showed ballooning with a lymphocytic infiltrate and neutrophils (inset: H&E, original magnification ×40).


The patient was given a 10-mg/mL suspension of elemental zinc and was advised to take 1 mL (10 mg) by mouth twice daily with food. This dosage equated to 3 mg/kg/d. On follow-up 3 weeks later, the skin began to clear (Figure 3). Follow-up laboratory testing showed an increase in zinc (114 μg/dL) and alkaline phosphatase levels (313 U/L). The patient was able to discontinue the zinc supplementation, and follow-up during the next year revealed no recurrence.

A, Three weeks after treatment with zinc supplementation, the annular crusted papules and plaques were no longer evident on the cheeks. B, The perineum/groin and perianal regions showed similar clearance.
FIGURE 3. A, Three weeks after treatment with zinc supplementation, the annular crusted papules and plaques were no longer evident on the cheeks. B, The perineum/groin and perianal regions showed similar clearance.

Comment

Etiology of AE—Acrodermatitis enteropathica was first identified in 1942 as an acral rash associated with diarrhea3; in 1973, Barnes and Moynahan4 discovered zinc deficiency as a causal agent for these findings. The causes of AE are further subclassified as either an acquired or inborn etiology. Congenital causes commonly are seen in infants within the first few months of life, whereas acquired forms are seen at any age. Acquired forms in infants can occur from failure of the mother to secrete zinc in breast milk, low maternal serum zinc levels, or other reasons causing low nutritional intake. A single mutation in the SLC30A2 gene has been found to markedly reduce zinc concentrations in breast milk, thus causing zinc deficiency in breastfed infants.5 Other acquired forms can be caused by malabsorption, sometimes after surgery such as intestinal bypass or from intravenous nutrition without sufficient zinc.1 The congenital form of AE is an autosomal-recessive disorder occurring from mutations in the SLC39A4 gene located on band 8q24.3. Affected individuals have a decreased ability to absorb zinc in the small intestine because of defects in zinc transporters ZIP and ZnT.6 Based on our patient’s laboratory findings and history, it is believed that the zinc deficiency was acquired, as the condition normalized with repletion and has not required any supplementation in the year of follow-up. In addition, the absence of a pertinent family history supported an acquired diagnosis, which has various etiologies, whereas the congenital form primarily is a genetic disease.

Diagnosis of AE—The characteristic clinical features of AE include erythematous, dry, scaly papules and plaques that may evolve into crusted, erosive, pustular lesions. These lesions typically are distributed in a periorificial and acral pattern.1,2 Although AE includes the clinical triad of acral and periorificial dermatitis, alopecia, and diarrhea, most cases present with only partial features of this syndrome, as seen in our patient, who presented with only 2 symptoms—dermatitis and diarrhea. The diagnosis of AE is based on clinical and laboratory abnormalities, especially a low serum zinc level. Low levels of zinc-dependent enzymes, such as alkaline phosphatase, may support the diagnosis, as seen in our patient. Histologic evaluation is characteristic but is not diagnostic, as the same findings can be seen in other nutritional disorders. Such findings include confluent parakeratosis associated with a reduced granular layer in early lesions and subsequent ballooning of subcorneal keratinocytes, upper epidermal pallor, and intraepidermal clefts. Late lesions exhibit psoriasiform hyperplasia of the epidermis with less epidermal pallor.7

 

 

Management—Treatment of AE includes supplementation with oral elemental zinc; however, there are scant evidence-based recommendations on the exact dose of zinc to be given. Generally, the recommended amount is 3 mg/kg/d.8 For individuals with the congenital form of AE, lifelong zinc supplementation is additionally recommended.9 It is important to recognize this presentation because the patient can develop worsening irritability, severe diarrhea, nail dystrophy, hair loss, immune dysfunction, and numerous ophthalmic disorders if left untreated. Acute zinc toxicity due to excess administration is rare, with symptoms of nausea and vomiting occurring with dosages of 50 to 100 mg/d. Additionally, dosages of up to 70 mg twice weekly have been provided without any toxic effect.10 In our case, 3 mg/kg/d of oral zinc supplementation proved to be effective in resolving the patient’s symptoms of acquired zinc deficiency.

Differential Diagnosis—It is important to note that deficiencies of other nutrients may present as an AE-like eruption called acrodermatitis dysmetabolica (AD). Both diseases may present with the triad of dermatitis, alopecia, and diarrhea; however, AD is associated with inborn errors of metabolism. There have been cases that describe AD in patients with a zinc deficiency in conjunction with a deficiency of branched-chain amino acids.11,12 It is important to consider AD in the differential diagnosis of an AE eruption, especially in the context of a metabolic disorder, as it may affect the treatment plan. One case described the dermatitis of AD as not responding to zinc supplementation alone, while another described improvement after increasing an isoleucine supplementation dose.11,12

Other considerations in the differential diagnoses include AE-like conditions such as biotinidase deficiency, multiple carboxylase deficiency, and essential fatty acid deficiency. An AE-like condition may present with the triad of dermatitis, alopecia, and diarrhea. However, unlike in true AE, zinc and alkaline phosphatase levels tend to be normal in these conditions. Other features seen in AE-like conditions depend on the underlying cause but often include failure to thrive, neurologic defects, ophthalmic abnormalities, and metabolic abnormalities.13

Acrodermatitis enteropathica (AE) is a rare disorder of zinc metabolism that typically presents in infancy.1 Although it is clinically characterized by acral and periorificial dermatitis, alopecia, and diarrhea, only 20% of cases present with this triad.2 Zinc deficiency in AE can either be acquired or inborn (congenital). Acquired forms can occur from dietary inadequacy or malabsorption, whereas genetic causes are related to an autosomal-recessive disorder affecting zinc transporters.1 We report a case of a 3-month-old female infant with acquired AE who was successfully treated with zinc supplementation over the course of 3 weeks.

Case Report

A 3-month-old female infant presented to the emergency department with a rash of 2 weeks’ duration. She was born full term with no birth complications. The patient’s mother reported that the rash started on the cheeks, then enlarged and spread to the neck, back, and perineum. The patient also had been having diarrhea during this time. She previously had received mupirocin and cephalexin with no response to treatment. Maternal history was negative for lupus, and the mother’s diet consisted of a variety of foods but not many vegetables. The patient was exclusively breastfed, and there was no pertinent history of similar rashes occurring in other family members.

Physical examination revealed the patient had annular and polycyclic, hyperkeratotic, crusted papules and plaques on the cheeks, neck, back, and axillae, as well as the perineum/groin and perianal regions (Figure 1). The differential diagnosis at the time included neonatal lupus, zinc deficiency, and syphilis. Relevant laboratory testing and a shave biopsy of the left axilla were obtained.

A, Annular and polycyclic, hyperkeratotic, crusted papules and plaques on the cheeks. B, Similar lesions were present in the perineum/groin and perianal regions.
FIGURE 1. A, Annular and polycyclic, hyperkeratotic, crusted papules and plaques on the cheeks. B, Similar lesions were present in the perineum/groin and perianal regions.


Pertinent laboratory findings included a low zinc level (23 μg/dL [reference range, 26–141 μg/dL]), low alkaline phosphatase level (74 U/L [reference range, 94–486 U/L]), and thrombocytosis (826×109/L [reference range, 150–400×109/L). Results for antinuclear antibody and anti–Sjögren syndrome–related antigen A and B antibody testing were negative. A rapid plasma reagin test was nonreactive. Histologic examination revealed psoriasiform hyperplasia with overlying confluent parakeratosis, focal spongiosis, multiple dyskeratotic keratinocytes, and mitotic figures (Figure 2). Ballooning was evident in focal cells in the subcorneal region in addition to an accompanying lymphocytic infiltrate and occasional neutrophils.

Biopsy of the left axilla showed psoriasiform hyperplasia with overlying confluent parakeratosis, focal spongiosis, multiple dyskeratotic keratinocytes, and mitotic figures
FIGURE 2. Biopsy of the left axilla showed psoriasiform hyperplasia with overlying confluent parakeratosis, focal spongiosis, multiple dyskeratotic keratinocytes, and mitotic figures (H&E, original magnification ×10). Focal cells in the subcorneal region showed ballooning with a lymphocytic infiltrate and neutrophils (inset: H&E, original magnification ×40).


The patient was given a 10-mg/mL suspension of elemental zinc and was advised to take 1 mL (10 mg) by mouth twice daily with food. This dosage equated to 3 mg/kg/d. On follow-up 3 weeks later, the skin began to clear (Figure 3). Follow-up laboratory testing showed an increase in zinc (114 μg/dL) and alkaline phosphatase levels (313 U/L). The patient was able to discontinue the zinc supplementation, and follow-up during the next year revealed no recurrence.

A, Three weeks after treatment with zinc supplementation, the annular crusted papules and plaques were no longer evident on the cheeks. B, The perineum/groin and perianal regions showed similar clearance.
FIGURE 3. A, Three weeks after treatment with zinc supplementation, the annular crusted papules and plaques were no longer evident on the cheeks. B, The perineum/groin and perianal regions showed similar clearance.

Comment

Etiology of AE—Acrodermatitis enteropathica was first identified in 1942 as an acral rash associated with diarrhea3; in 1973, Barnes and Moynahan4 discovered zinc deficiency as a causal agent for these findings. The causes of AE are further subclassified as either an acquired or inborn etiology. Congenital causes commonly are seen in infants within the first few months of life, whereas acquired forms are seen at any age. Acquired forms in infants can occur from failure of the mother to secrete zinc in breast milk, low maternal serum zinc levels, or other reasons causing low nutritional intake. A single mutation in the SLC30A2 gene has been found to markedly reduce zinc concentrations in breast milk, thus causing zinc deficiency in breastfed infants.5 Other acquired forms can be caused by malabsorption, sometimes after surgery such as intestinal bypass or from intravenous nutrition without sufficient zinc.1 The congenital form of AE is an autosomal-recessive disorder occurring from mutations in the SLC39A4 gene located on band 8q24.3. Affected individuals have a decreased ability to absorb zinc in the small intestine because of defects in zinc transporters ZIP and ZnT.6 Based on our patient’s laboratory findings and history, it is believed that the zinc deficiency was acquired, as the condition normalized with repletion and has not required any supplementation in the year of follow-up. In addition, the absence of a pertinent family history supported an acquired diagnosis, which has various etiologies, whereas the congenital form primarily is a genetic disease.

Diagnosis of AE—The characteristic clinical features of AE include erythematous, dry, scaly papules and plaques that may evolve into crusted, erosive, pustular lesions. These lesions typically are distributed in a periorificial and acral pattern.1,2 Although AE includes the clinical triad of acral and periorificial dermatitis, alopecia, and diarrhea, most cases present with only partial features of this syndrome, as seen in our patient, who presented with only 2 symptoms—dermatitis and diarrhea. The diagnosis of AE is based on clinical and laboratory abnormalities, especially a low serum zinc level. Low levels of zinc-dependent enzymes, such as alkaline phosphatase, may support the diagnosis, as seen in our patient. Histologic evaluation is characteristic but is not diagnostic, as the same findings can be seen in other nutritional disorders. Such findings include confluent parakeratosis associated with a reduced granular layer in early lesions and subsequent ballooning of subcorneal keratinocytes, upper epidermal pallor, and intraepidermal clefts. Late lesions exhibit psoriasiform hyperplasia of the epidermis with less epidermal pallor.7

 

 

Management—Treatment of AE includes supplementation with oral elemental zinc; however, there are scant evidence-based recommendations on the exact dose of zinc to be given. Generally, the recommended amount is 3 mg/kg/d.8 For individuals with the congenital form of AE, lifelong zinc supplementation is additionally recommended.9 It is important to recognize this presentation because the patient can develop worsening irritability, severe diarrhea, nail dystrophy, hair loss, immune dysfunction, and numerous ophthalmic disorders if left untreated. Acute zinc toxicity due to excess administration is rare, with symptoms of nausea and vomiting occurring with dosages of 50 to 100 mg/d. Additionally, dosages of up to 70 mg twice weekly have been provided without any toxic effect.10 In our case, 3 mg/kg/d of oral zinc supplementation proved to be effective in resolving the patient’s symptoms of acquired zinc deficiency.

Differential Diagnosis—It is important to note that deficiencies of other nutrients may present as an AE-like eruption called acrodermatitis dysmetabolica (AD). Both diseases may present with the triad of dermatitis, alopecia, and diarrhea; however, AD is associated with inborn errors of metabolism. There have been cases that describe AD in patients with a zinc deficiency in conjunction with a deficiency of branched-chain amino acids.11,12 It is important to consider AD in the differential diagnosis of an AE eruption, especially in the context of a metabolic disorder, as it may affect the treatment plan. One case described the dermatitis of AD as not responding to zinc supplementation alone, while another described improvement after increasing an isoleucine supplementation dose.11,12

Other considerations in the differential diagnoses include AE-like conditions such as biotinidase deficiency, multiple carboxylase deficiency, and essential fatty acid deficiency. An AE-like condition may present with the triad of dermatitis, alopecia, and diarrhea. However, unlike in true AE, zinc and alkaline phosphatase levels tend to be normal in these conditions. Other features seen in AE-like conditions depend on the underlying cause but often include failure to thrive, neurologic defects, ophthalmic abnormalities, and metabolic abnormalities.13
References
  1. Acrodermatitis enteropathica. National Organization for Rare Disorders. Accessed October 16, 2022. https://rarediseases.org/rare-diseases/acrodermatitis-enteropathica/
  2. Perafán-Riveros C, França LFS, Alves ACF, et al. Acrodermatitis enteropathica: case report and review of the literature. Pediatr Dermatol. 2002;19:426-431.
  3. Danbolt N. Acrodermatitis enteropathica. Br J Dermatol. 1979;100:37-40.
  4. Barnes PM, Moynahan EJ. Zinc deficiency in acrodermatitis enteropathica: multiple dietary intolerance treated with synthetic diet. Proc R Soc Med. 1973;66:327-329.
  5. Lee S, Zhou Y, Gill DL, et al. A genetic variant in SLC30A2 causes breast dysfunction during lactation by inducing ER stress, oxidative stress and epithelial barrier defects. Sci Rep. 2018;8:3542.
  6. Kaur S, Sangwan A, Sahu P, et al. Clinical variants of acrodermatitis enteropathica and its co-relation with genetics. Indian J Paediatr Dermatol. 2016;17:35-37.
  7. Dela Rosa KM, James WD. Acrodermatitis enteropathica workup. Medscape. Updated June 4, 2021. Accessed October 16, 2022. https://emedicine.medscape.com/article/1102575-workup#showall
  8. Ngan V, Gangakhedkar A, Oakley A. Acrodermatitis enteropathica. DermNet. Accessed October 16, 2022. https://dermnetnz.org/topics/acrodermatitis-enteropathica/
  9. Ranugha P, Sethi P, Veeranna S. Acrodermatitis enteropathica: the need for sustained high dose zinc supplementation. Dermatol Online J. 2018;24:13030/qt1w9002sr.
  10. Larson CP, Roy SK, Khan AI, et al. Zinc treatment to under-five children: applications to improve child survival and reduce burden of disease. J Health Popul Nutr. 2008;26:356-365.
  11. Samady JA, Schwartz RA, Shih LY, et al. Acrodermatitis enteropathica-like eruption in an infant with nonketotic hyperglycinemia. J Dermatol. 2000;27:604-608.
  12. Flores K, Chikowski R, Morrell DS. Acrodermatitis dysmetabolica in an infant with maple syrup urine disease. Clin Exp Dermatol. 2016;41:651-654.
  13. Jones L, Oakley A. Acrodermatitis enteropathica-like conditions. DermNet. Accessed August 30, 2022. https://dermnetnz.org/topics/acrodermatitis-enteropathica-like-conditions
References
  1. Acrodermatitis enteropathica. National Organization for Rare Disorders. Accessed October 16, 2022. https://rarediseases.org/rare-diseases/acrodermatitis-enteropathica/
  2. Perafán-Riveros C, França LFS, Alves ACF, et al. Acrodermatitis enteropathica: case report and review of the literature. Pediatr Dermatol. 2002;19:426-431.
  3. Danbolt N. Acrodermatitis enteropathica. Br J Dermatol. 1979;100:37-40.
  4. Barnes PM, Moynahan EJ. Zinc deficiency in acrodermatitis enteropathica: multiple dietary intolerance treated with synthetic diet. Proc R Soc Med. 1973;66:327-329.
  5. Lee S, Zhou Y, Gill DL, et al. A genetic variant in SLC30A2 causes breast dysfunction during lactation by inducing ER stress, oxidative stress and epithelial barrier defects. Sci Rep. 2018;8:3542.
  6. Kaur S, Sangwan A, Sahu P, et al. Clinical variants of acrodermatitis enteropathica and its co-relation with genetics. Indian J Paediatr Dermatol. 2016;17:35-37.
  7. Dela Rosa KM, James WD. Acrodermatitis enteropathica workup. Medscape. Updated June 4, 2021. Accessed October 16, 2022. https://emedicine.medscape.com/article/1102575-workup#showall
  8. Ngan V, Gangakhedkar A, Oakley A. Acrodermatitis enteropathica. DermNet. Accessed October 16, 2022. https://dermnetnz.org/topics/acrodermatitis-enteropathica/
  9. Ranugha P, Sethi P, Veeranna S. Acrodermatitis enteropathica: the need for sustained high dose zinc supplementation. Dermatol Online J. 2018;24:13030/qt1w9002sr.
  10. Larson CP, Roy SK, Khan AI, et al. Zinc treatment to under-five children: applications to improve child survival and reduce burden of disease. J Health Popul Nutr. 2008;26:356-365.
  11. Samady JA, Schwartz RA, Shih LY, et al. Acrodermatitis enteropathica-like eruption in an infant with nonketotic hyperglycinemia. J Dermatol. 2000;27:604-608.
  12. Flores K, Chikowski R, Morrell DS. Acrodermatitis dysmetabolica in an infant with maple syrup urine disease. Clin Exp Dermatol. 2016;41:651-654.
  13. Jones L, Oakley A. Acrodermatitis enteropathica-like conditions. DermNet. Accessed August 30, 2022. https://dermnetnz.org/topics/acrodermatitis-enteropathica-like-conditions
Issue
Cutis - 110(5)
Issue
Cutis - 110(5)
Page Number
281-283
Page Number
281-283
Publications
Publications
Topics
Article Type
Display Headline
Acquired Acrodermatitis Enteropathica in an Infant
Display Headline
Acquired Acrodermatitis Enteropathica in an Infant
Sections
Inside the Article

Practice Points

  • Although clinically characterized by the triad of acral and periorificial dermatitis, alopecia, and diarrhea, most cases of acrodermatitis enteropathica (AE) present with only partial features of this syndrome.
  • Low levels of zinc-dependent enzymes such as alkaline phosphatase may support the diagnosis of AE.
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

Photoallergic Contact Dermatitis: No Fun in the Sun

Article Type
Changed
Wed, 11/09/2022 - 09:56
Display Headline
Photoallergic Contact Dermatitis: No Fun in the Sun

Photoallergic contact dermatitis (PACD), a subtype of allergic contact dermatitis that occurs because of the specific combination of exposure to an exogenous chemical applied topically to the skin and UV radiation, may be more common than was once thought.1 Although the incidence in the general population is unknown, current research points to approximately 20% to 40% of patients with suspected photosensitivity having a PACD diagnosis.2 Recently, the North American Contact Dermatitis Group (NACDG) reported that 21% of 373 patients undergoing photopatch testing (PPT) were diagnosed with PACD2; however, PPT is not routinely performed, which may contribute to underdiagnosis.

Mechanism of Disease

Similar to allergic contact dermatitis, PACD is a delayed type IV hypersensitivity reaction; however, it only occurs when an exogenous chemical is applied topically to the skin with concomitant exposure to UV radiation, usually in the UVA range (315–400 nm).3,4 When exposed to UV radiation, it is thought that the exogenous chemical combines with a protein in the skin and transforms into a photoantigen. In the sensitization phase, the photoantigen is taken up by antigen-presenting cells in the epidermis and transported to local lymph nodes where antigen-specific T cells are generated.5 In the elicitation phase, the inflammatory reaction of PACD occurs upon subsequent exposure to the same chemical plus UV radiation.4 Development of PACD does not necessarily depend on the dose of the chemical or the amount of UV radiation.6 Why certain individuals may be more susceptible is unknown, though major histocompatibility complex haplotypes could be influential.7,8

Clinical Manifestations

Photoallergic contact dermatitis primarily presents in sun-exposed areas of the skin (eg, face, neck, V area of the chest, dorsal upper extremities) with sparing of naturally photoprotected sites, such as the upper eyelids and nasolabial and retroauricular folds. Other than its characteristic photodistribution, PACD often is clinically indistinguishable from routine allergic contact dermatitis. It manifests as a pruritic, poorly demarcated, eczematous or sometimes vesiculobullous eruption that develops in a delayed fashion—24 to 72 hours after sun exposure. The dermatitis may extend to other parts of the body either through spread of the chemical agent by the hands or clothing or due to the systemic nature of the immune response. The severity of the presentation can vary depending on multiple factors, such as concentration and absorption of the agent, length of exposure, intensity and duration of UV radiation exposure, and individual susceptibility.4 Chronic PACD may become lichenified. Generally, rashes resolve after discontinuation of the causative agent; however, long-term exposure may lead to development of chronic actinic dermatitis, with persistent photodistributed eczema regardless of contact with the initial inciting agent.9

Differential Diagnosis

The differential diagnosis for patients presenting with photodistributed dermatitis is broad; therefore, taking a thorough history is important. Considerations include age of onset, timing and persistence of reactions, use of topical and systemic medications (both prescription and over-the-counter [OTC]), personal care products, occupation, and hobbies, as well as a thorough review of systems.

It is important to distinguish PACD from phototoxic contact dermatitis (PTCD)(also known as photoirritant contact dermatitis)(Table). Asking about the onset and timing of the eruption may be critical for distinction, as PTCD can occur within minutes to hours of the first exposure to a chemical and UV radiation, while there is a sensitization delay in PACD.6 Phytophotodermatitis is a well-known type of PTCD caused by exposure to furocoumarin-containing plants, most commonly limes.10 Other causes of PTCD include tar products and certain medications.11 Importantly, PPT to a known phototoxic chemical should never be performed because it will cause a strong reaction in anyone tested, regardless of exposure history.

Comparison of Phototoxic and Photoallergic Contact Dermatitis


Other diagnoses to consider include photoaggravated dermatoses (eg, atopic dermatitis, lupus erythematosus, dermatomyositis) and idiopathic photodermatoses (eg, chronic actinic dermatitis, actinic prurigo, polymorphous light eruption). Although atopic dermatitis usually improves with UV light exposure, photoaggravated atopic dermatitis is suggested in eczema patients who flare with sun exposure, in a seasonal pattern, or after phototherapy; this condition is challenging to differentiate from PACD if PPT is not performed.12 The diagnosis of idiopathic photodermatoses is nuanced; however, asking about the timeline of the reaction including onset, duration, and persistence, as well as characterization of unique clinical features, can help in differentiation.13 In certain scenarios, a biopsy may be helpful. A thorough review of systems will help to assess for autoimmune connective tissue disorders, and relevant serologies should be checked as indicated.

Diagnosis

Histologically, PACD presents similarly to allergic contact dermatitis with spongiotic dermatitis; therefore, biopsy cannot be relied upon to make the diagnosis.6 Photopatch testing is required for definitive diagnosis. It is reasonable to perform PPT in any patient with chronic dermatitis primarily affecting sun-exposed areas without a clear alternative diagnosis.14,15 Of note, at present there are no North American consensus guidelines for PPT, but typically duplicate sets of photoallergens are applied to both sides of the patient’s back and one side is exposed to UVA radiation. The reactions are compared after 48 to 96 hours.15 A positive reaction only at the irradiated site is consistent with photoallergy, while a reaction of equal strength at both the irradiated and nonirradiated sites indicates regular contact allergy. The case of a reaction occurring at both sites with a stronger response at the irradiated site is known as photoaggravated contact allergy, which can be thought of as allergic contact dermatitis that worsens but does not solely occur with exposure to sunlight.

 

 

Although PPT is necessary for the accurate diagnosis of PACD, it is infrequently used. Two surveys of 112 and 117 American Contact Dermatitis Society members, respectively, have revealed that only around half performed PPT, most of them testing fewer than 20 times per year.16,17 Additionally, there was variability in the test methodology and allergens employed. Nevertheless, most respondents tested sunscreens, nonsteroidal anti-inflammatory drugs (NSAIDs), fragrances, and their patients’ own products.16,17 The most common reasons for not performing PPT were lack of equipment, insufficient skills, rare clinical suspicion, and cost. Dermatologists at academic centers performed more PPT than those in other practice settings, including multispecialty group practices and private offices.16 These findings highlight multiple factors that may contribute to reduced patient access to PPT and thus potential underdiagnosis of PACD.

Common Photoallergens

The most common photoallergens change over time in response to market trends; for example, fragrance was once a top photoallergen in the United States in the 1970s and 1980s but declined in prominence after musk ambrette—the primary allergen associated with PACD at the time—was removed as an ingredient in fragrances.18

In the largest and most recent PPT series from North America (1999-2009),2 sunscreens comprised 7 of the top 10 most common photoallergens, which is consistent with other studies showing sunscreens to be the most common North American photoallergens.19-22 The frequency of PACD due to sunscreens likely relates to their increasing use worldwide as awareness of photocarcinogenesis and photoaging grows, as well as the common use of UV filters in nonsunscreen personal care products, ranging from lip balms to perfumes and bodywashes. Chemical (organic) UV filters—in particular oxybenzone (benzophenone-3) and avobenzone (butyl methoxydibenzoylmethane)—are the most common sunscreen photoallergens.2,23 Para-aminobenzoic acid was once a common photoallergen, but it is no longer used in US sunscreens due to safety concerns.19,20 The physical (inorganic) UV filters zinc oxide and titanium dioxide are not known photosensitizers.

Methylisothiazolinone (MI) is a highly allergenic preservative commonly used in a wide array of personal care products, including sunscreens.24 In the most recent NACDG patch test data, MI was the second most common contact allergen.25 Allergic contact dermatitis caused by MI in sunscreen can mimic PACD.26 In addition, MI can cause photoaggravated contact dermatitis, with some affected patients experiencing ongoing photosensitivity even after avoiding this allergen.26-30 The European Union and Canada have introduced restrictions on the use of MI in personal care products, but no such regulatory measures have been taken in the United States to date.25,31,32

After sunscreens, another common cause of PACD are topical NSAIDs, which are frequently used for musculoskeletal pain relief. These are of particular concern in Europe, where a variety of formulations are widely available OTC.33 Ketoprofen and etofenamate are responsible for the largest number of PACD reactions in Europe.2,34,35 Meanwhile, the only OTC topical NSAID available in the United States is diclofenac gel, which was approved in 2020. Cases of PACD due to use of diclofenac gel have been reported in the literature, but testing in larger populations is needed.36-39

Notably, ketoprofen may co- or cross-react with certain UV filters—oxybenzone and octocrylene—and the lipid-lowering agent fenofibrate due to chemical similarities.40-43 Despite the relatively high number of photoallergic reactions to ketoprofen in the NACDG photopatch series, only 25% (5/20) were considered clinically relevant (ie, the allergen could not be verified as present in the known skin contactants of the patient, and the patient was not exposed to circumstances in which contact with materials known to contain the allergen would likely occur), which suggests that they likely represented cross-reactions in patients sensitized to sunscreens.2

Other agents that may cause PACD include antimicrobials, plants and plant derivatives, and pesticides.2,4,18 The antimicrobial fentichlor is a common cause of positive PPT reactions, but it rarely is clinically relevant.44

Treatment

The primary management of PACD centers on identification of the causative photoallergen to avoid future exposure. Patients should be educated on the various names by which the causative allergen can be identified on product labels and should be given a list of safe products that are free from relevant allergens and cross-reacting chemicals.45 Additionally, sun protection education should be provided. Exposure to UVA radiation can occur through windows, making the use of broad-spectrum sunscreens and protective clothing crucial. In cases of sunscreen-induced PACD, the responsible chemical UV filter(s) should be avoided, or alternatively, patients may use physical sunscreens containing only zinc oxide and/or titanium dioxide as active ingredients, as these are not known to cause PACD.4

When avoidance alone is insufficient, topical corticosteroids are the usual first-line treatment for localized PACD. When steroid-sparing treatments are preferred, topical calcineurin inhibitors such as tacrolimus and pimecrolimus may be used. If PACD is more widespread and severe, systemic therapy using steroids or steroid-sparing agents may be necessary to provide symptomatic relief.4

Final Interpretation

Photoallergic contact dermatitis is not uncommon, particularly among photosensitive patients. Most cases are due to sunscreens or topical NSAIDs. Consideration of PPT should be given in any patient with a chronic photodistributed dermatitis to evaluate for the possibility of PACD.

References
  1. Darvay A, White IR, Rycroft RJ, et al. Photoallergic contact dermatitis is uncommon. Br J Dermatol. 2001;145:597-601.
  2. DeLeo VA, Adler BL, Warshaw EM, et al. Photopatch test results of the North American contact dermatitis group, 1999-2009. Photodermatol Photoimmunol Photomed. 2022;38:288-291.
  3. Kerr A, Ferguson J. Photoallergic contact dermatitis. Photodermatol Photoimmunol Photomed. 2010;26:56-65.
  4. As¸kın Ö, Cesur SK, Engin B, et al. Photoallergic contact dermatitis. Curr Derm Rep. 2019;8:157-163.
  5. Wilm A, Berneburg M. Photoallergy. J Dtsch Dermatol Ges. 2015;13:7-13.
  6. DeLeo VA. Photocontact dermatitis. Dermatol Ther. 2004;17:279-288. 
  7. Imai S, Atarashi K, Ikesue K, et al. Establishment of murine model of allergic photocontact dermatitis to ketoprofen and characterization of pathogenic T cells. J Dermatol Sci. 2006;41:127-136.
  8. Tokura Y, Yagi H, Satoh T, et al. Inhibitory effect of melanin pigment on sensitization and elicitation of murine contact photosensitivity: mechanism of low responsiveness in C57BL/10 background mice. J Invest Dermatol. 1993;101:673-678.
  9. Stein KR, Scheinfeld NS. Drug-induced photoallergic and phototoxic reactions. Expert Opin Drug Saf. 2007;6:431-443.
  10. Janusz SC, Schwartz RA. Botanical briefs: phytophotodermatitis is an occupational and recreational dermatosis in the limelight. Cutis. 2021;107:187-189.
  11. Atwal SK, Chen A, Adler BL. Phototoxic contact dermatitis from over-the-counter 8-methoxypsoralen. Cutis. 2022;109:E2-E3.
  12. Rutter KJ, Farrar MD, Marjanovic EJ, et al. Clinicophotobiological characterization of photoaggravated atopic dermatitis [published online July 27, 2022]. JAMA Dermatol. doi:10.1001/jamadermatol.2022.2823
  13. Lecha M. Idiopathic photodermatoses: clinical, diagnostic and therapeutic aspects. J Eur Acad Dermatol Venereol. 2001;15:499-505.
  14. Marks JG Jr, Anderson BE, DeLeo VA. Contact & Occupational Dermatology. 4th ed. Jaypee Brothers; 2016.
  15. Bruynzeel DP, Ferguson J, Andersen K, et al. Photopatch testing: a consensus methodology for Europe. J Eur Acad Dermatol Venereol. 2004;18:679-682. 
  16. Kim T, Taylor JS, Maibach HI, et al. Photopatch testing among members of the American Contact Dermatitis Society. Dermatitis. 2020;31:59-67.
  17. Asemota E, Crawford G, Kovarik C, et al. A survey examining photopatch test and phototest methodologies of contact dermatologists in the United States: platform for developing a consensus. Dermatitis. 2017;28:265-269.
  18. Scalf LA, Davis MD, Rohlinger AL, et al. Photopatch testing of 182 patients: a 6-year experience at the Mayo Clinic. Dermatitis. 2009;20:44-52.
  19. Greenspoon J, Ahluwalia R, Juma N, et al. Allergic and photoallergic contact dermatitis: a 10-year experience. Dermatitis. 2013;24:29-32.
  20. Victor FC, Cohen DE, Soter NA. A 20-year analysis of previous and emerging allergens that elicit photoallergic contact dermatitis. J Am Acad Dermatol. 2010;62:605-610.
  21. Schauder S, Ippen H. Contact and photocontact sensitivity to sunscreens. review of a 15-year experience and of the literature. Contact Dermatitis. 1997;37:221-232. 
  22. Collaris EJ, Frank J. Photoallergic contact dermatitis caused by ultraviolet filters in different sunscreens. Int J Dermatol. 2008;47(suppl 1):35-37.
  23. Heurung AR, Raju SI, Warshaw EM. Adverse reactions to sunscreen agents: epidemiology, responsible irritants and allergens, clinical characteristics, and management. Dermatitis. 2014;25:289-326.
  24. Reeder M, Atwater AR. Methylisothiazolinone and isothiazolinone allergy. Cutis. 2019;104:94-96.
  25. DeKoven JG, Silverberg JI, Warshaw EM, et al. North American Contact Dermatitis Group Patch Test Results: 2017-2018. Dermatitis. 2021;32:111-123.
  26. Kullberg SA, Voller LM, Warshaw EM. Methylisothiazolinone in “dermatology-recommended” sunscreens: an important mimicker of photoallergic contact dermatitis. Photodermatol Photoimmunol Photomed. 2021;37:366-370. 
  27. Herman A, Aerts O, de Montjoye L, et al. Isothiazolinone derivatives and allergic contact dermatitis: a review and update. J Eur Acad Dermatol Venereol. 2019;33:267-276.
  28. Adler BL, Houle MC, Pratt M. Photoaggravated contact dermatitis to methylisothiazolinone and associated photosensitivity: a case series [published online January 25, 2022]. Dermatitis. doi:10.1097/DER.0000000000000833
  29. Aerts O, Goossens A, Marguery MC, et al. Photoaggravated allergic contact dermatitis and transient photosensitivity caused by methylisothiazolinone. Contact Dermatitis. 2018;78:241-245.
  30. Pirmez R, Fernandes AL, Melo MG. Photoaggravated contact dermatitis to Kathon CG (methylchloroisothiazolinone/methylisothiazolinone): a novel pattern of involvement in a growing epidemic?. Br J Dermatol. 2015;173:1343-1344.
  31. Uter W, Aalto-Korte K, Agner T, et al. The epidemic of methylisothiazolinone contact allergy in Europe: follow-up on changing exposures.J Eur Acad Dermatol Venereol. 2020;34:333-339.
  32. Government of Canada. Changes to the cosmetic ingredient hotlist. December 3, 2019. Updated August 26, 2022. Accessed October 20, 2022. https://www.canada.ca/en/health-canada/services/consumer-product-safety/cosmetics/cosmetic-ingredient-hotlist-prohibited-restricted-ingredients/changes.html
  33. Barkin RL. Topical nonsteroidal anti-inflammatory drugs: the importance of drug, delivery, and therapeutic outcome. Am J Ther. 2015;22:388-407.
  34. European Multicentre Photopatch Test Study (EMCPPTS) Taskforce. A European multicentre photopatch test study. Br J Dermatol. 2012;166:1002-1009.
  35. Ophaswongse S, Maibach H. Topical nonsteroidal antiinflammatory drugs: allergic and photoallergic contact dermatitis and phototoxicity. Contact Dermatitis. 1993;29:57-64. 
  36. Kowalzick L, Ziegler H. Photoallergic contact dermatitis from topical diclofenac in Solaraze gel. Contact Dermatitis. 2006;54:348-349.
  37. Montoro J, Rodríguez M, Díaz M, et al. Photoallergic contact dermatitis due to diclofenac. Contact Dermatitis. 2003;48:115.
  38. Fernández-Jorge B, Goday-Buján JJ, Murga M, et al. Photoallergic contact dermatitis due to diclofenac with cross-reaction to aceclofenac: two case reports. Contact Dermatitis. 2009;61:236-237.
  39. Akat PB. Severe photosensitivity reaction induced by topical diclofenac. Indian J Pharmacol. 2013;45:408-409.
  40. Leroy D, Dompmartin A, Szczurko C, et al. Photodermatitis from ketoprofen with cross-reactivity to fenofibrate and benzophenones. Photodermatol Photoimmunol Photomed. 1997;13:93-97.
  41. Devleeschouwer V, Roelandts R, Garmyn M, et al. Allergic and photoallergic contact dermatitis from ketoprofen: results of (photo) patch testing and follow-up of 42 patients. Contact Dermatitis. 2008;58:159-166.
  42. Matsushita T, Kamide R. Five cases of photocontact dermatitisdue to topical ketoprofen: photopatch testing and cross-reaction study. Photodermatol Photoimmunol Photomed. 2001;17:26-31.
  43. de Groot AC, Roberts DW. Contact and photocontact allergy to octocrylene: a review. Contact Dermatitis. 2014;70:193-204.
  44. Wolverton JE, Soter NA, Cohen DE. Fentichlor photocontact dermatitis: a persistent enigma. Dermatitis. 2013;24:77-81.
  45. Mowad CM, Anderson B, Scheinman P, et al. Allergic contact dermatitis: patient management and education. J Am Acad Dermatol. 2016;74:1043-1054.
Article PDF
Author and Disclosure Information

Ms. Guenther and Dr. Adler are from the Keck School of Medicine, University of Southern California, Los Angeles. Dr. Adler is from the Department of Dermatology. Ms. Johnson is from the University of Minnesota Medical School, Minneapolis. Dr. Yu is from the Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston.

Ms. Guenther and Ms. Johnson report no conflict of interest. Dr. Yu has served as a speaker for the National Eczema Foundation, has received research grants from the Dermatology Foundation and the Pediatric Dermatology Foundation, and has received income from Dynamed. Dr. Adler has served as a research investigator and/or consultant to AbbVie and Skin Research Institute, LLC.

Correspondence: Brandon L. Adler, MD, 1441 Eastlake Ave, Ezralow Tower, Ste 5301, Los Angeles, CA 90033 ([email protected]).

Issue
Cutis - 110(5)
Publications
Topics
Page Number
241-243,267
Sections
Author and Disclosure Information

Ms. Guenther and Dr. Adler are from the Keck School of Medicine, University of Southern California, Los Angeles. Dr. Adler is from the Department of Dermatology. Ms. Johnson is from the University of Minnesota Medical School, Minneapolis. Dr. Yu is from the Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston.

Ms. Guenther and Ms. Johnson report no conflict of interest. Dr. Yu has served as a speaker for the National Eczema Foundation, has received research grants from the Dermatology Foundation and the Pediatric Dermatology Foundation, and has received income from Dynamed. Dr. Adler has served as a research investigator and/or consultant to AbbVie and Skin Research Institute, LLC.

Correspondence: Brandon L. Adler, MD, 1441 Eastlake Ave, Ezralow Tower, Ste 5301, Los Angeles, CA 90033 ([email protected]).

Author and Disclosure Information

Ms. Guenther and Dr. Adler are from the Keck School of Medicine, University of Southern California, Los Angeles. Dr. Adler is from the Department of Dermatology. Ms. Johnson is from the University of Minnesota Medical School, Minneapolis. Dr. Yu is from the Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston.

Ms. Guenther and Ms. Johnson report no conflict of interest. Dr. Yu has served as a speaker for the National Eczema Foundation, has received research grants from the Dermatology Foundation and the Pediatric Dermatology Foundation, and has received income from Dynamed. Dr. Adler has served as a research investigator and/or consultant to AbbVie and Skin Research Institute, LLC.

Correspondence: Brandon L. Adler, MD, 1441 Eastlake Ave, Ezralow Tower, Ste 5301, Los Angeles, CA 90033 ([email protected]).

Article PDF
Article PDF

Photoallergic contact dermatitis (PACD), a subtype of allergic contact dermatitis that occurs because of the specific combination of exposure to an exogenous chemical applied topically to the skin and UV radiation, may be more common than was once thought.1 Although the incidence in the general population is unknown, current research points to approximately 20% to 40% of patients with suspected photosensitivity having a PACD diagnosis.2 Recently, the North American Contact Dermatitis Group (NACDG) reported that 21% of 373 patients undergoing photopatch testing (PPT) were diagnosed with PACD2; however, PPT is not routinely performed, which may contribute to underdiagnosis.

Mechanism of Disease

Similar to allergic contact dermatitis, PACD is a delayed type IV hypersensitivity reaction; however, it only occurs when an exogenous chemical is applied topically to the skin with concomitant exposure to UV radiation, usually in the UVA range (315–400 nm).3,4 When exposed to UV radiation, it is thought that the exogenous chemical combines with a protein in the skin and transforms into a photoantigen. In the sensitization phase, the photoantigen is taken up by antigen-presenting cells in the epidermis and transported to local lymph nodes where antigen-specific T cells are generated.5 In the elicitation phase, the inflammatory reaction of PACD occurs upon subsequent exposure to the same chemical plus UV radiation.4 Development of PACD does not necessarily depend on the dose of the chemical or the amount of UV radiation.6 Why certain individuals may be more susceptible is unknown, though major histocompatibility complex haplotypes could be influential.7,8

Clinical Manifestations

Photoallergic contact dermatitis primarily presents in sun-exposed areas of the skin (eg, face, neck, V area of the chest, dorsal upper extremities) with sparing of naturally photoprotected sites, such as the upper eyelids and nasolabial and retroauricular folds. Other than its characteristic photodistribution, PACD often is clinically indistinguishable from routine allergic contact dermatitis. It manifests as a pruritic, poorly demarcated, eczematous or sometimes vesiculobullous eruption that develops in a delayed fashion—24 to 72 hours after sun exposure. The dermatitis may extend to other parts of the body either through spread of the chemical agent by the hands or clothing or due to the systemic nature of the immune response. The severity of the presentation can vary depending on multiple factors, such as concentration and absorption of the agent, length of exposure, intensity and duration of UV radiation exposure, and individual susceptibility.4 Chronic PACD may become lichenified. Generally, rashes resolve after discontinuation of the causative agent; however, long-term exposure may lead to development of chronic actinic dermatitis, with persistent photodistributed eczema regardless of contact with the initial inciting agent.9

Differential Diagnosis

The differential diagnosis for patients presenting with photodistributed dermatitis is broad; therefore, taking a thorough history is important. Considerations include age of onset, timing and persistence of reactions, use of topical and systemic medications (both prescription and over-the-counter [OTC]), personal care products, occupation, and hobbies, as well as a thorough review of systems.

It is important to distinguish PACD from phototoxic contact dermatitis (PTCD)(also known as photoirritant contact dermatitis)(Table). Asking about the onset and timing of the eruption may be critical for distinction, as PTCD can occur within minutes to hours of the first exposure to a chemical and UV radiation, while there is a sensitization delay in PACD.6 Phytophotodermatitis is a well-known type of PTCD caused by exposure to furocoumarin-containing plants, most commonly limes.10 Other causes of PTCD include tar products and certain medications.11 Importantly, PPT to a known phototoxic chemical should never be performed because it will cause a strong reaction in anyone tested, regardless of exposure history.

Comparison of Phototoxic and Photoallergic Contact Dermatitis


Other diagnoses to consider include photoaggravated dermatoses (eg, atopic dermatitis, lupus erythematosus, dermatomyositis) and idiopathic photodermatoses (eg, chronic actinic dermatitis, actinic prurigo, polymorphous light eruption). Although atopic dermatitis usually improves with UV light exposure, photoaggravated atopic dermatitis is suggested in eczema patients who flare with sun exposure, in a seasonal pattern, or after phototherapy; this condition is challenging to differentiate from PACD if PPT is not performed.12 The diagnosis of idiopathic photodermatoses is nuanced; however, asking about the timeline of the reaction including onset, duration, and persistence, as well as characterization of unique clinical features, can help in differentiation.13 In certain scenarios, a biopsy may be helpful. A thorough review of systems will help to assess for autoimmune connective tissue disorders, and relevant serologies should be checked as indicated.

Diagnosis

Histologically, PACD presents similarly to allergic contact dermatitis with spongiotic dermatitis; therefore, biopsy cannot be relied upon to make the diagnosis.6 Photopatch testing is required for definitive diagnosis. It is reasonable to perform PPT in any patient with chronic dermatitis primarily affecting sun-exposed areas without a clear alternative diagnosis.14,15 Of note, at present there are no North American consensus guidelines for PPT, but typically duplicate sets of photoallergens are applied to both sides of the patient’s back and one side is exposed to UVA radiation. The reactions are compared after 48 to 96 hours.15 A positive reaction only at the irradiated site is consistent with photoallergy, while a reaction of equal strength at both the irradiated and nonirradiated sites indicates regular contact allergy. The case of a reaction occurring at both sites with a stronger response at the irradiated site is known as photoaggravated contact allergy, which can be thought of as allergic contact dermatitis that worsens but does not solely occur with exposure to sunlight.

 

 

Although PPT is necessary for the accurate diagnosis of PACD, it is infrequently used. Two surveys of 112 and 117 American Contact Dermatitis Society members, respectively, have revealed that only around half performed PPT, most of them testing fewer than 20 times per year.16,17 Additionally, there was variability in the test methodology and allergens employed. Nevertheless, most respondents tested sunscreens, nonsteroidal anti-inflammatory drugs (NSAIDs), fragrances, and their patients’ own products.16,17 The most common reasons for not performing PPT were lack of equipment, insufficient skills, rare clinical suspicion, and cost. Dermatologists at academic centers performed more PPT than those in other practice settings, including multispecialty group practices and private offices.16 These findings highlight multiple factors that may contribute to reduced patient access to PPT and thus potential underdiagnosis of PACD.

Common Photoallergens

The most common photoallergens change over time in response to market trends; for example, fragrance was once a top photoallergen in the United States in the 1970s and 1980s but declined in prominence after musk ambrette—the primary allergen associated with PACD at the time—was removed as an ingredient in fragrances.18

In the largest and most recent PPT series from North America (1999-2009),2 sunscreens comprised 7 of the top 10 most common photoallergens, which is consistent with other studies showing sunscreens to be the most common North American photoallergens.19-22 The frequency of PACD due to sunscreens likely relates to their increasing use worldwide as awareness of photocarcinogenesis and photoaging grows, as well as the common use of UV filters in nonsunscreen personal care products, ranging from lip balms to perfumes and bodywashes. Chemical (organic) UV filters—in particular oxybenzone (benzophenone-3) and avobenzone (butyl methoxydibenzoylmethane)—are the most common sunscreen photoallergens.2,23 Para-aminobenzoic acid was once a common photoallergen, but it is no longer used in US sunscreens due to safety concerns.19,20 The physical (inorganic) UV filters zinc oxide and titanium dioxide are not known photosensitizers.

Methylisothiazolinone (MI) is a highly allergenic preservative commonly used in a wide array of personal care products, including sunscreens.24 In the most recent NACDG patch test data, MI was the second most common contact allergen.25 Allergic contact dermatitis caused by MI in sunscreen can mimic PACD.26 In addition, MI can cause photoaggravated contact dermatitis, with some affected patients experiencing ongoing photosensitivity even after avoiding this allergen.26-30 The European Union and Canada have introduced restrictions on the use of MI in personal care products, but no such regulatory measures have been taken in the United States to date.25,31,32

After sunscreens, another common cause of PACD are topical NSAIDs, which are frequently used for musculoskeletal pain relief. These are of particular concern in Europe, where a variety of formulations are widely available OTC.33 Ketoprofen and etofenamate are responsible for the largest number of PACD reactions in Europe.2,34,35 Meanwhile, the only OTC topical NSAID available in the United States is diclofenac gel, which was approved in 2020. Cases of PACD due to use of diclofenac gel have been reported in the literature, but testing in larger populations is needed.36-39

Notably, ketoprofen may co- or cross-react with certain UV filters—oxybenzone and octocrylene—and the lipid-lowering agent fenofibrate due to chemical similarities.40-43 Despite the relatively high number of photoallergic reactions to ketoprofen in the NACDG photopatch series, only 25% (5/20) were considered clinically relevant (ie, the allergen could not be verified as present in the known skin contactants of the patient, and the patient was not exposed to circumstances in which contact with materials known to contain the allergen would likely occur), which suggests that they likely represented cross-reactions in patients sensitized to sunscreens.2

Other agents that may cause PACD include antimicrobials, plants and plant derivatives, and pesticides.2,4,18 The antimicrobial fentichlor is a common cause of positive PPT reactions, but it rarely is clinically relevant.44

Treatment

The primary management of PACD centers on identification of the causative photoallergen to avoid future exposure. Patients should be educated on the various names by which the causative allergen can be identified on product labels and should be given a list of safe products that are free from relevant allergens and cross-reacting chemicals.45 Additionally, sun protection education should be provided. Exposure to UVA radiation can occur through windows, making the use of broad-spectrum sunscreens and protective clothing crucial. In cases of sunscreen-induced PACD, the responsible chemical UV filter(s) should be avoided, or alternatively, patients may use physical sunscreens containing only zinc oxide and/or titanium dioxide as active ingredients, as these are not known to cause PACD.4

When avoidance alone is insufficient, topical corticosteroids are the usual first-line treatment for localized PACD. When steroid-sparing treatments are preferred, topical calcineurin inhibitors such as tacrolimus and pimecrolimus may be used. If PACD is more widespread and severe, systemic therapy using steroids or steroid-sparing agents may be necessary to provide symptomatic relief.4

Final Interpretation

Photoallergic contact dermatitis is not uncommon, particularly among photosensitive patients. Most cases are due to sunscreens or topical NSAIDs. Consideration of PPT should be given in any patient with a chronic photodistributed dermatitis to evaluate for the possibility of PACD.

Photoallergic contact dermatitis (PACD), a subtype of allergic contact dermatitis that occurs because of the specific combination of exposure to an exogenous chemical applied topically to the skin and UV radiation, may be more common than was once thought.1 Although the incidence in the general population is unknown, current research points to approximately 20% to 40% of patients with suspected photosensitivity having a PACD diagnosis.2 Recently, the North American Contact Dermatitis Group (NACDG) reported that 21% of 373 patients undergoing photopatch testing (PPT) were diagnosed with PACD2; however, PPT is not routinely performed, which may contribute to underdiagnosis.

Mechanism of Disease

Similar to allergic contact dermatitis, PACD is a delayed type IV hypersensitivity reaction; however, it only occurs when an exogenous chemical is applied topically to the skin with concomitant exposure to UV radiation, usually in the UVA range (315–400 nm).3,4 When exposed to UV radiation, it is thought that the exogenous chemical combines with a protein in the skin and transforms into a photoantigen. In the sensitization phase, the photoantigen is taken up by antigen-presenting cells in the epidermis and transported to local lymph nodes where antigen-specific T cells are generated.5 In the elicitation phase, the inflammatory reaction of PACD occurs upon subsequent exposure to the same chemical plus UV radiation.4 Development of PACD does not necessarily depend on the dose of the chemical or the amount of UV radiation.6 Why certain individuals may be more susceptible is unknown, though major histocompatibility complex haplotypes could be influential.7,8

Clinical Manifestations

Photoallergic contact dermatitis primarily presents in sun-exposed areas of the skin (eg, face, neck, V area of the chest, dorsal upper extremities) with sparing of naturally photoprotected sites, such as the upper eyelids and nasolabial and retroauricular folds. Other than its characteristic photodistribution, PACD often is clinically indistinguishable from routine allergic contact dermatitis. It manifests as a pruritic, poorly demarcated, eczematous or sometimes vesiculobullous eruption that develops in a delayed fashion—24 to 72 hours after sun exposure. The dermatitis may extend to other parts of the body either through spread of the chemical agent by the hands or clothing or due to the systemic nature of the immune response. The severity of the presentation can vary depending on multiple factors, such as concentration and absorption of the agent, length of exposure, intensity and duration of UV radiation exposure, and individual susceptibility.4 Chronic PACD may become lichenified. Generally, rashes resolve after discontinuation of the causative agent; however, long-term exposure may lead to development of chronic actinic dermatitis, with persistent photodistributed eczema regardless of contact with the initial inciting agent.9

Differential Diagnosis

The differential diagnosis for patients presenting with photodistributed dermatitis is broad; therefore, taking a thorough history is important. Considerations include age of onset, timing and persistence of reactions, use of topical and systemic medications (both prescription and over-the-counter [OTC]), personal care products, occupation, and hobbies, as well as a thorough review of systems.

It is important to distinguish PACD from phototoxic contact dermatitis (PTCD)(also known as photoirritant contact dermatitis)(Table). Asking about the onset and timing of the eruption may be critical for distinction, as PTCD can occur within minutes to hours of the first exposure to a chemical and UV radiation, while there is a sensitization delay in PACD.6 Phytophotodermatitis is a well-known type of PTCD caused by exposure to furocoumarin-containing plants, most commonly limes.10 Other causes of PTCD include tar products and certain medications.11 Importantly, PPT to a known phototoxic chemical should never be performed because it will cause a strong reaction in anyone tested, regardless of exposure history.

Comparison of Phototoxic and Photoallergic Contact Dermatitis


Other diagnoses to consider include photoaggravated dermatoses (eg, atopic dermatitis, lupus erythematosus, dermatomyositis) and idiopathic photodermatoses (eg, chronic actinic dermatitis, actinic prurigo, polymorphous light eruption). Although atopic dermatitis usually improves with UV light exposure, photoaggravated atopic dermatitis is suggested in eczema patients who flare with sun exposure, in a seasonal pattern, or after phototherapy; this condition is challenging to differentiate from PACD if PPT is not performed.12 The diagnosis of idiopathic photodermatoses is nuanced; however, asking about the timeline of the reaction including onset, duration, and persistence, as well as characterization of unique clinical features, can help in differentiation.13 In certain scenarios, a biopsy may be helpful. A thorough review of systems will help to assess for autoimmune connective tissue disorders, and relevant serologies should be checked as indicated.

Diagnosis

Histologically, PACD presents similarly to allergic contact dermatitis with spongiotic dermatitis; therefore, biopsy cannot be relied upon to make the diagnosis.6 Photopatch testing is required for definitive diagnosis. It is reasonable to perform PPT in any patient with chronic dermatitis primarily affecting sun-exposed areas without a clear alternative diagnosis.14,15 Of note, at present there are no North American consensus guidelines for PPT, but typically duplicate sets of photoallergens are applied to both sides of the patient’s back and one side is exposed to UVA radiation. The reactions are compared after 48 to 96 hours.15 A positive reaction only at the irradiated site is consistent with photoallergy, while a reaction of equal strength at both the irradiated and nonirradiated sites indicates regular contact allergy. The case of a reaction occurring at both sites with a stronger response at the irradiated site is known as photoaggravated contact allergy, which can be thought of as allergic contact dermatitis that worsens but does not solely occur with exposure to sunlight.

 

 

Although PPT is necessary for the accurate diagnosis of PACD, it is infrequently used. Two surveys of 112 and 117 American Contact Dermatitis Society members, respectively, have revealed that only around half performed PPT, most of them testing fewer than 20 times per year.16,17 Additionally, there was variability in the test methodology and allergens employed. Nevertheless, most respondents tested sunscreens, nonsteroidal anti-inflammatory drugs (NSAIDs), fragrances, and their patients’ own products.16,17 The most common reasons for not performing PPT were lack of equipment, insufficient skills, rare clinical suspicion, and cost. Dermatologists at academic centers performed more PPT than those in other practice settings, including multispecialty group practices and private offices.16 These findings highlight multiple factors that may contribute to reduced patient access to PPT and thus potential underdiagnosis of PACD.

Common Photoallergens

The most common photoallergens change over time in response to market trends; for example, fragrance was once a top photoallergen in the United States in the 1970s and 1980s but declined in prominence after musk ambrette—the primary allergen associated with PACD at the time—was removed as an ingredient in fragrances.18

In the largest and most recent PPT series from North America (1999-2009),2 sunscreens comprised 7 of the top 10 most common photoallergens, which is consistent with other studies showing sunscreens to be the most common North American photoallergens.19-22 The frequency of PACD due to sunscreens likely relates to their increasing use worldwide as awareness of photocarcinogenesis and photoaging grows, as well as the common use of UV filters in nonsunscreen personal care products, ranging from lip balms to perfumes and bodywashes. Chemical (organic) UV filters—in particular oxybenzone (benzophenone-3) and avobenzone (butyl methoxydibenzoylmethane)—are the most common sunscreen photoallergens.2,23 Para-aminobenzoic acid was once a common photoallergen, but it is no longer used in US sunscreens due to safety concerns.19,20 The physical (inorganic) UV filters zinc oxide and titanium dioxide are not known photosensitizers.

Methylisothiazolinone (MI) is a highly allergenic preservative commonly used in a wide array of personal care products, including sunscreens.24 In the most recent NACDG patch test data, MI was the second most common contact allergen.25 Allergic contact dermatitis caused by MI in sunscreen can mimic PACD.26 In addition, MI can cause photoaggravated contact dermatitis, with some affected patients experiencing ongoing photosensitivity even after avoiding this allergen.26-30 The European Union and Canada have introduced restrictions on the use of MI in personal care products, but no such regulatory measures have been taken in the United States to date.25,31,32

After sunscreens, another common cause of PACD are topical NSAIDs, which are frequently used for musculoskeletal pain relief. These are of particular concern in Europe, where a variety of formulations are widely available OTC.33 Ketoprofen and etofenamate are responsible for the largest number of PACD reactions in Europe.2,34,35 Meanwhile, the only OTC topical NSAID available in the United States is diclofenac gel, which was approved in 2020. Cases of PACD due to use of diclofenac gel have been reported in the literature, but testing in larger populations is needed.36-39

Notably, ketoprofen may co- or cross-react with certain UV filters—oxybenzone and octocrylene—and the lipid-lowering agent fenofibrate due to chemical similarities.40-43 Despite the relatively high number of photoallergic reactions to ketoprofen in the NACDG photopatch series, only 25% (5/20) were considered clinically relevant (ie, the allergen could not be verified as present in the known skin contactants of the patient, and the patient was not exposed to circumstances in which contact with materials known to contain the allergen would likely occur), which suggests that they likely represented cross-reactions in patients sensitized to sunscreens.2

Other agents that may cause PACD include antimicrobials, plants and plant derivatives, and pesticides.2,4,18 The antimicrobial fentichlor is a common cause of positive PPT reactions, but it rarely is clinically relevant.44

Treatment

The primary management of PACD centers on identification of the causative photoallergen to avoid future exposure. Patients should be educated on the various names by which the causative allergen can be identified on product labels and should be given a list of safe products that are free from relevant allergens and cross-reacting chemicals.45 Additionally, sun protection education should be provided. Exposure to UVA radiation can occur through windows, making the use of broad-spectrum sunscreens and protective clothing crucial. In cases of sunscreen-induced PACD, the responsible chemical UV filter(s) should be avoided, or alternatively, patients may use physical sunscreens containing only zinc oxide and/or titanium dioxide as active ingredients, as these are not known to cause PACD.4

When avoidance alone is insufficient, topical corticosteroids are the usual first-line treatment for localized PACD. When steroid-sparing treatments are preferred, topical calcineurin inhibitors such as tacrolimus and pimecrolimus may be used. If PACD is more widespread and severe, systemic therapy using steroids or steroid-sparing agents may be necessary to provide symptomatic relief.4

Final Interpretation

Photoallergic contact dermatitis is not uncommon, particularly among photosensitive patients. Most cases are due to sunscreens or topical NSAIDs. Consideration of PPT should be given in any patient with a chronic photodistributed dermatitis to evaluate for the possibility of PACD.

References
  1. Darvay A, White IR, Rycroft RJ, et al. Photoallergic contact dermatitis is uncommon. Br J Dermatol. 2001;145:597-601.
  2. DeLeo VA, Adler BL, Warshaw EM, et al. Photopatch test results of the North American contact dermatitis group, 1999-2009. Photodermatol Photoimmunol Photomed. 2022;38:288-291.
  3. Kerr A, Ferguson J. Photoallergic contact dermatitis. Photodermatol Photoimmunol Photomed. 2010;26:56-65.
  4. As¸kın Ö, Cesur SK, Engin B, et al. Photoallergic contact dermatitis. Curr Derm Rep. 2019;8:157-163.
  5. Wilm A, Berneburg M. Photoallergy. J Dtsch Dermatol Ges. 2015;13:7-13.
  6. DeLeo VA. Photocontact dermatitis. Dermatol Ther. 2004;17:279-288. 
  7. Imai S, Atarashi K, Ikesue K, et al. Establishment of murine model of allergic photocontact dermatitis to ketoprofen and characterization of pathogenic T cells. J Dermatol Sci. 2006;41:127-136.
  8. Tokura Y, Yagi H, Satoh T, et al. Inhibitory effect of melanin pigment on sensitization and elicitation of murine contact photosensitivity: mechanism of low responsiveness in C57BL/10 background mice. J Invest Dermatol. 1993;101:673-678.
  9. Stein KR, Scheinfeld NS. Drug-induced photoallergic and phototoxic reactions. Expert Opin Drug Saf. 2007;6:431-443.
  10. Janusz SC, Schwartz RA. Botanical briefs: phytophotodermatitis is an occupational and recreational dermatosis in the limelight. Cutis. 2021;107:187-189.
  11. Atwal SK, Chen A, Adler BL. Phototoxic contact dermatitis from over-the-counter 8-methoxypsoralen. Cutis. 2022;109:E2-E3.
  12. Rutter KJ, Farrar MD, Marjanovic EJ, et al. Clinicophotobiological characterization of photoaggravated atopic dermatitis [published online July 27, 2022]. JAMA Dermatol. doi:10.1001/jamadermatol.2022.2823
  13. Lecha M. Idiopathic photodermatoses: clinical, diagnostic and therapeutic aspects. J Eur Acad Dermatol Venereol. 2001;15:499-505.
  14. Marks JG Jr, Anderson BE, DeLeo VA. Contact & Occupational Dermatology. 4th ed. Jaypee Brothers; 2016.
  15. Bruynzeel DP, Ferguson J, Andersen K, et al. Photopatch testing: a consensus methodology for Europe. J Eur Acad Dermatol Venereol. 2004;18:679-682. 
  16. Kim T, Taylor JS, Maibach HI, et al. Photopatch testing among members of the American Contact Dermatitis Society. Dermatitis. 2020;31:59-67.
  17. Asemota E, Crawford G, Kovarik C, et al. A survey examining photopatch test and phototest methodologies of contact dermatologists in the United States: platform for developing a consensus. Dermatitis. 2017;28:265-269.
  18. Scalf LA, Davis MD, Rohlinger AL, et al. Photopatch testing of 182 patients: a 6-year experience at the Mayo Clinic. Dermatitis. 2009;20:44-52.
  19. Greenspoon J, Ahluwalia R, Juma N, et al. Allergic and photoallergic contact dermatitis: a 10-year experience. Dermatitis. 2013;24:29-32.
  20. Victor FC, Cohen DE, Soter NA. A 20-year analysis of previous and emerging allergens that elicit photoallergic contact dermatitis. J Am Acad Dermatol. 2010;62:605-610.
  21. Schauder S, Ippen H. Contact and photocontact sensitivity to sunscreens. review of a 15-year experience and of the literature. Contact Dermatitis. 1997;37:221-232. 
  22. Collaris EJ, Frank J. Photoallergic contact dermatitis caused by ultraviolet filters in different sunscreens. Int J Dermatol. 2008;47(suppl 1):35-37.
  23. Heurung AR, Raju SI, Warshaw EM. Adverse reactions to sunscreen agents: epidemiology, responsible irritants and allergens, clinical characteristics, and management. Dermatitis. 2014;25:289-326.
  24. Reeder M, Atwater AR. Methylisothiazolinone and isothiazolinone allergy. Cutis. 2019;104:94-96.
  25. DeKoven JG, Silverberg JI, Warshaw EM, et al. North American Contact Dermatitis Group Patch Test Results: 2017-2018. Dermatitis. 2021;32:111-123.
  26. Kullberg SA, Voller LM, Warshaw EM. Methylisothiazolinone in “dermatology-recommended” sunscreens: an important mimicker of photoallergic contact dermatitis. Photodermatol Photoimmunol Photomed. 2021;37:366-370. 
  27. Herman A, Aerts O, de Montjoye L, et al. Isothiazolinone derivatives and allergic contact dermatitis: a review and update. J Eur Acad Dermatol Venereol. 2019;33:267-276.
  28. Adler BL, Houle MC, Pratt M. Photoaggravated contact dermatitis to methylisothiazolinone and associated photosensitivity: a case series [published online January 25, 2022]. Dermatitis. doi:10.1097/DER.0000000000000833
  29. Aerts O, Goossens A, Marguery MC, et al. Photoaggravated allergic contact dermatitis and transient photosensitivity caused by methylisothiazolinone. Contact Dermatitis. 2018;78:241-245.
  30. Pirmez R, Fernandes AL, Melo MG. Photoaggravated contact dermatitis to Kathon CG (methylchloroisothiazolinone/methylisothiazolinone): a novel pattern of involvement in a growing epidemic?. Br J Dermatol. 2015;173:1343-1344.
  31. Uter W, Aalto-Korte K, Agner T, et al. The epidemic of methylisothiazolinone contact allergy in Europe: follow-up on changing exposures.J Eur Acad Dermatol Venereol. 2020;34:333-339.
  32. Government of Canada. Changes to the cosmetic ingredient hotlist. December 3, 2019. Updated August 26, 2022. Accessed October 20, 2022. https://www.canada.ca/en/health-canada/services/consumer-product-safety/cosmetics/cosmetic-ingredient-hotlist-prohibited-restricted-ingredients/changes.html
  33. Barkin RL. Topical nonsteroidal anti-inflammatory drugs: the importance of drug, delivery, and therapeutic outcome. Am J Ther. 2015;22:388-407.
  34. European Multicentre Photopatch Test Study (EMCPPTS) Taskforce. A European multicentre photopatch test study. Br J Dermatol. 2012;166:1002-1009.
  35. Ophaswongse S, Maibach H. Topical nonsteroidal antiinflammatory drugs: allergic and photoallergic contact dermatitis and phototoxicity. Contact Dermatitis. 1993;29:57-64. 
  36. Kowalzick L, Ziegler H. Photoallergic contact dermatitis from topical diclofenac in Solaraze gel. Contact Dermatitis. 2006;54:348-349.
  37. Montoro J, Rodríguez M, Díaz M, et al. Photoallergic contact dermatitis due to diclofenac. Contact Dermatitis. 2003;48:115.
  38. Fernández-Jorge B, Goday-Buján JJ, Murga M, et al. Photoallergic contact dermatitis due to diclofenac with cross-reaction to aceclofenac: two case reports. Contact Dermatitis. 2009;61:236-237.
  39. Akat PB. Severe photosensitivity reaction induced by topical diclofenac. Indian J Pharmacol. 2013;45:408-409.
  40. Leroy D, Dompmartin A, Szczurko C, et al. Photodermatitis from ketoprofen with cross-reactivity to fenofibrate and benzophenones. Photodermatol Photoimmunol Photomed. 1997;13:93-97.
  41. Devleeschouwer V, Roelandts R, Garmyn M, et al. Allergic and photoallergic contact dermatitis from ketoprofen: results of (photo) patch testing and follow-up of 42 patients. Contact Dermatitis. 2008;58:159-166.
  42. Matsushita T, Kamide R. Five cases of photocontact dermatitisdue to topical ketoprofen: photopatch testing and cross-reaction study. Photodermatol Photoimmunol Photomed. 2001;17:26-31.
  43. de Groot AC, Roberts DW. Contact and photocontact allergy to octocrylene: a review. Contact Dermatitis. 2014;70:193-204.
  44. Wolverton JE, Soter NA, Cohen DE. Fentichlor photocontact dermatitis: a persistent enigma. Dermatitis. 2013;24:77-81.
  45. Mowad CM, Anderson B, Scheinman P, et al. Allergic contact dermatitis: patient management and education. J Am Acad Dermatol. 2016;74:1043-1054.
References
  1. Darvay A, White IR, Rycroft RJ, et al. Photoallergic contact dermatitis is uncommon. Br J Dermatol. 2001;145:597-601.
  2. DeLeo VA, Adler BL, Warshaw EM, et al. Photopatch test results of the North American contact dermatitis group, 1999-2009. Photodermatol Photoimmunol Photomed. 2022;38:288-291.
  3. Kerr A, Ferguson J. Photoallergic contact dermatitis. Photodermatol Photoimmunol Photomed. 2010;26:56-65.
  4. As¸kın Ö, Cesur SK, Engin B, et al. Photoallergic contact dermatitis. Curr Derm Rep. 2019;8:157-163.
  5. Wilm A, Berneburg M. Photoallergy. J Dtsch Dermatol Ges. 2015;13:7-13.
  6. DeLeo VA. Photocontact dermatitis. Dermatol Ther. 2004;17:279-288. 
  7. Imai S, Atarashi K, Ikesue K, et al. Establishment of murine model of allergic photocontact dermatitis to ketoprofen and characterization of pathogenic T cells. J Dermatol Sci. 2006;41:127-136.
  8. Tokura Y, Yagi H, Satoh T, et al. Inhibitory effect of melanin pigment on sensitization and elicitation of murine contact photosensitivity: mechanism of low responsiveness in C57BL/10 background mice. J Invest Dermatol. 1993;101:673-678.
  9. Stein KR, Scheinfeld NS. Drug-induced photoallergic and phototoxic reactions. Expert Opin Drug Saf. 2007;6:431-443.
  10. Janusz SC, Schwartz RA. Botanical briefs: phytophotodermatitis is an occupational and recreational dermatosis in the limelight. Cutis. 2021;107:187-189.
  11. Atwal SK, Chen A, Adler BL. Phototoxic contact dermatitis from over-the-counter 8-methoxypsoralen. Cutis. 2022;109:E2-E3.
  12. Rutter KJ, Farrar MD, Marjanovic EJ, et al. Clinicophotobiological characterization of photoaggravated atopic dermatitis [published online July 27, 2022]. JAMA Dermatol. doi:10.1001/jamadermatol.2022.2823
  13. Lecha M. Idiopathic photodermatoses: clinical, diagnostic and therapeutic aspects. J Eur Acad Dermatol Venereol. 2001;15:499-505.
  14. Marks JG Jr, Anderson BE, DeLeo VA. Contact & Occupational Dermatology. 4th ed. Jaypee Brothers; 2016.
  15. Bruynzeel DP, Ferguson J, Andersen K, et al. Photopatch testing: a consensus methodology for Europe. J Eur Acad Dermatol Venereol. 2004;18:679-682. 
  16. Kim T, Taylor JS, Maibach HI, et al. Photopatch testing among members of the American Contact Dermatitis Society. Dermatitis. 2020;31:59-67.
  17. Asemota E, Crawford G, Kovarik C, et al. A survey examining photopatch test and phototest methodologies of contact dermatologists in the United States: platform for developing a consensus. Dermatitis. 2017;28:265-269.
  18. Scalf LA, Davis MD, Rohlinger AL, et al. Photopatch testing of 182 patients: a 6-year experience at the Mayo Clinic. Dermatitis. 2009;20:44-52.
  19. Greenspoon J, Ahluwalia R, Juma N, et al. Allergic and photoallergic contact dermatitis: a 10-year experience. Dermatitis. 2013;24:29-32.
  20. Victor FC, Cohen DE, Soter NA. A 20-year analysis of previous and emerging allergens that elicit photoallergic contact dermatitis. J Am Acad Dermatol. 2010;62:605-610.
  21. Schauder S, Ippen H. Contact and photocontact sensitivity to sunscreens. review of a 15-year experience and of the literature. Contact Dermatitis. 1997;37:221-232. 
  22. Collaris EJ, Frank J. Photoallergic contact dermatitis caused by ultraviolet filters in different sunscreens. Int J Dermatol. 2008;47(suppl 1):35-37.
  23. Heurung AR, Raju SI, Warshaw EM. Adverse reactions to sunscreen agents: epidemiology, responsible irritants and allergens, clinical characteristics, and management. Dermatitis. 2014;25:289-326.
  24. Reeder M, Atwater AR. Methylisothiazolinone and isothiazolinone allergy. Cutis. 2019;104:94-96.
  25. DeKoven JG, Silverberg JI, Warshaw EM, et al. North American Contact Dermatitis Group Patch Test Results: 2017-2018. Dermatitis. 2021;32:111-123.
  26. Kullberg SA, Voller LM, Warshaw EM. Methylisothiazolinone in “dermatology-recommended” sunscreens: an important mimicker of photoallergic contact dermatitis. Photodermatol Photoimmunol Photomed. 2021;37:366-370. 
  27. Herman A, Aerts O, de Montjoye L, et al. Isothiazolinone derivatives and allergic contact dermatitis: a review and update. J Eur Acad Dermatol Venereol. 2019;33:267-276.
  28. Adler BL, Houle MC, Pratt M. Photoaggravated contact dermatitis to methylisothiazolinone and associated photosensitivity: a case series [published online January 25, 2022]. Dermatitis. doi:10.1097/DER.0000000000000833
  29. Aerts O, Goossens A, Marguery MC, et al. Photoaggravated allergic contact dermatitis and transient photosensitivity caused by methylisothiazolinone. Contact Dermatitis. 2018;78:241-245.
  30. Pirmez R, Fernandes AL, Melo MG. Photoaggravated contact dermatitis to Kathon CG (methylchloroisothiazolinone/methylisothiazolinone): a novel pattern of involvement in a growing epidemic?. Br J Dermatol. 2015;173:1343-1344.
  31. Uter W, Aalto-Korte K, Agner T, et al. The epidemic of methylisothiazolinone contact allergy in Europe: follow-up on changing exposures.J Eur Acad Dermatol Venereol. 2020;34:333-339.
  32. Government of Canada. Changes to the cosmetic ingredient hotlist. December 3, 2019. Updated August 26, 2022. Accessed October 20, 2022. https://www.canada.ca/en/health-canada/services/consumer-product-safety/cosmetics/cosmetic-ingredient-hotlist-prohibited-restricted-ingredients/changes.html
  33. Barkin RL. Topical nonsteroidal anti-inflammatory drugs: the importance of drug, delivery, and therapeutic outcome. Am J Ther. 2015;22:388-407.
  34. European Multicentre Photopatch Test Study (EMCPPTS) Taskforce. A European multicentre photopatch test study. Br J Dermatol. 2012;166:1002-1009.
  35. Ophaswongse S, Maibach H. Topical nonsteroidal antiinflammatory drugs: allergic and photoallergic contact dermatitis and phototoxicity. Contact Dermatitis. 1993;29:57-64. 
  36. Kowalzick L, Ziegler H. Photoallergic contact dermatitis from topical diclofenac in Solaraze gel. Contact Dermatitis. 2006;54:348-349.
  37. Montoro J, Rodríguez M, Díaz M, et al. Photoallergic contact dermatitis due to diclofenac. Contact Dermatitis. 2003;48:115.
  38. Fernández-Jorge B, Goday-Buján JJ, Murga M, et al. Photoallergic contact dermatitis due to diclofenac with cross-reaction to aceclofenac: two case reports. Contact Dermatitis. 2009;61:236-237.
  39. Akat PB. Severe photosensitivity reaction induced by topical diclofenac. Indian J Pharmacol. 2013;45:408-409.
  40. Leroy D, Dompmartin A, Szczurko C, et al. Photodermatitis from ketoprofen with cross-reactivity to fenofibrate and benzophenones. Photodermatol Photoimmunol Photomed. 1997;13:93-97.
  41. Devleeschouwer V, Roelandts R, Garmyn M, et al. Allergic and photoallergic contact dermatitis from ketoprofen: results of (photo) patch testing and follow-up of 42 patients. Contact Dermatitis. 2008;58:159-166.
  42. Matsushita T, Kamide R. Five cases of photocontact dermatitisdue to topical ketoprofen: photopatch testing and cross-reaction study. Photodermatol Photoimmunol Photomed. 2001;17:26-31.
  43. de Groot AC, Roberts DW. Contact and photocontact allergy to octocrylene: a review. Contact Dermatitis. 2014;70:193-204.
  44. Wolverton JE, Soter NA, Cohen DE. Fentichlor photocontact dermatitis: a persistent enigma. Dermatitis. 2013;24:77-81.
  45. Mowad CM, Anderson B, Scheinman P, et al. Allergic contact dermatitis: patient management and education. J Am Acad Dermatol. 2016;74:1043-1054.
Issue
Cutis - 110(5)
Issue
Cutis - 110(5)
Page Number
241-243,267
Page Number
241-243,267
Publications
Publications
Topics
Article Type
Display Headline
Photoallergic Contact Dermatitis: No Fun in the Sun
Display Headline
Photoallergic Contact Dermatitis: No Fun in the Sun
Sections
Inside the Article

Practice Points

  • Photoallergic contact dermatitis (PACD) presents clinically and histologically similar to allergic contact dermatitis but is concentrated in sun-exposed body sites.
  • Sunscreens currently are the most common photoallergens in North America, whereas topical nonsteroidal anti-inflammatory drugs are more common culprits in Europe.
  • Photopatch testing is required to diagnose PACD; however, it is infrequently performed, and there currently are no North American consensus guidelines.
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

Update on Tinea Capitis Diagnosis and Treatment

Article Type
Changed
Fri, 10/06/2023 - 13:43
Display Headline
Update on Tinea Capitis Diagnosis and Treatment

Tinea capitis (TC) most often is caused by Trichophyton tonsurans and Microsporum canis. The peak incidence is between 3 and 7 years of age. Noninflammatory TC typically presents as fine scaling with single or multiple scaly patches of circular alopecia (grey patches); diffuse or patchy, fine, white, adherent scaling of the scalp resembling generalized dandruff with subtle hair loss; or single or multiple patches of well-demarcated areas of alopecia with fine scale studded with broken-off hairs at the scalp surface, resulting in a black dot appearance. Inflammatory variants of TC include kerion and favus.1 Herein, updates on diagnosis, treatment, and monitoring of TC are provided, as well as a discussion of changes in the fungal microbiome associated with TC. Lastly, insights to some queries that practitioners may encounter when treating children with TC are provided.

Genetic Susceptibility

Molecular techniques have identified a number of macrophage regulator, leukocyte activation and migration, and cutaneous permeability genes associated with susceptibility to TC. These findings indicate that genetically determined deficiency in adaptive immune responses may affect the predisposition to dermatophyte infections.2

Clinical Varieties of Infection

Dermatophytes causing ringworm are capable of invading the hair shafts and can simultaneously invade smooth or glabrous skin (eg, T tonsurans, Trichophyton schoenleinii, Trichophyton violaceum). Some causative dermatophytes can even penetrate the nails (eg, Trichophyton soudanense). The clinical presentation is dependent on 3 main patterns of hair invasion3:

• Ectothrix: A mid-follicular pattern of invasion with hyphae growing down to the hair bulb that commonly is caused by Microsporum species. It clinically presents with scaling and inflammation with hair shafts breaking 2 to 3 mm above the scalp level.

• Endothrix: This pattern is nonfluorescent on Wood lamp examination, and hairs often break at the scalp level (black dot type). Trichophyton tonsurans, T soudanense, Trichophyton rubrum, and T violaceum are common causes.

• Favus: In this pattern, T schoenleinii is a common cause, and hairs grow to considerable lengths above the scalp with less damage than the other patterns. The hair shafts present with characteristic air spaces, and hyphae form clusters at the level of the epidermis.

Diagnosis

Optimal treatment of TC relies on proper identification of the causative agent. Fungal culture remains the gold standard of mycologic diagnosis regardless of its delayed results, which may take up to 4 weeks for proper identification of the fungal colonies and require ample expertise to interpret the morphologic features of the grown colonies.4

 

 

Other tests such as the potassium hydroxide preparation are nonspecific and do not identify the dermatophyte species. Although this method has been reported to have 5% to 15% false-negative results in routine practice depending on the skill of the observer and the quality of sampling, microscopic examination is essential, as it may allow the clinician to start treatment sooner pending culture results. The use of a Wood lamp is not suitable for definitive species identification, as this technique primarily is useful for observing fluorescence in ectothrix infection caused by Microsporum species, with the exception of T schoenleinii; otherwise, Trichophyton species, which cause endothrix infections, do not fluoresce.5Polymerase chain reaction is a sensitive technique that can help identify both the genus and species of common dermatophytes. Common target sequences include the ribosomal internal transcribed spacer and translation elongation factor 1α. The use of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry also has become popular for dermatophyte identification.6Trichoscopic diagnosis of TC, which is simple and noninvasive, is becoming increasingly popular. Features such as short, broken, black dot, comma, corkscrew, and/or zigzag hairs, as well as perifollicular scaling, are helpful for diagnosing TC (Figure). Moreover, trichoscopy can be useful for differentiating other common causes of hair loss, such as trichotillomania and alopecia areata. It had been reported that the trichoscopic features of TC can be seen as early as 2 weeks after starting treatment and therefore this can be a reliable period in which to follow-up with the patient to evaluate progress. The disappearance of black dots and comma hairs can be appreciated from 2 weeks onwards by trichoscopic evaluation.4

A, A 10-year-old boy with the black dot type of tinea capitis. B, Trichoscopy showed black dot hairs (pink circle), comma hairs (red arrows), corkscrew hairs (green arrows), short broken hairs (blue arrow), and perifollicular scaling (yellow arrow).
A, A 10-year-old boy with the black dot type of tinea capitis. B, Trichoscopy showed black dot hairs (pink circle), comma hairs (red arrows), corkscrew hairs (green arrows), short broken hairs (blue arrow), and perifollicular scaling (yellow arrow). C, Clinical resolution was achieved after 8 weeks of griseofulvin therapy. D, Posttreatment trichoscopy showed disappearance of dystrophic hairs.

Treatment

The common recommendation for first-line treatment of TC is the use of systemic antifungals with the use of a topical agent as an adjuvant to prevent the spread of fungal spores. For almost 6 decades, griseofulvin had been the gold-standard fungistatic used for treating TC in patients older than 2 years until the 2007 US Food and Drug Administration (FDA) approval of terbinafine fungicidal oral granules for treatment of TC in patients older than 4 years.7

Meta-analyses have demonstrated comparable efficacy for a 4-week course of terbinafine compared to 6 weeks of griseofulvin for TC based on the infectious organism. Terbinafine demonstrated superiority in treating T tonsurans and a similar efficacy in treating T violaceum, while griseofulvin was superior in treating M canis and other Microsporum species.8,9

The off-label use of fluconazole and itraconazole to treat TC is gaining popularity, with limited trials showing increased evidence of their effectiveness. There is not much clinical evidence to support the use of other oral antifungals, including the newer azoles such as voriconazole or posaconazole.9

Newer limited evidence has shown the off-label use of photodynamic therapy to be a promising alternative to systemic antifungal therapy in treating TC, pending validation by larger sample trials.10In my practice, I have found that severe cases of TC demonstrating inflammation or possible widespread id reactions are better treated with oral steroids. Ketoconazole shampoo or selenium sulfide used 2 to 3 times weekly to prevent spread in the early phases of therapy is a good adjunct to systemic treatment. Cases with kerions should be assessed for the possibility of a coexisting bacterial infection under the crusts, and if confirmed, antibiotics should be started.9The commonly used systemic antifungals generally are safe with a low side-effect profile, but there is a risk for hepatotoxicity. The FDA recommends that baseline alanine transaminase and aspartate transaminase levels should be obtained prior to beginning a terbinafine-based treatment regimen.11 The American Academy of Pediatrics has specifically stated that laboratory testing of serum hepatic enzymes is not a requirement if a griseofulvin-based regimen does not exceed 8 weeks; however, transaminase levels (alanine transaminase and aspartate transaminase) should be considered in patients using terbinafine at baseline or if treatment is prolonged beyond 4 to 6 weeks.12 In agreement with the FDA guidelines, the Canadian Pediatric Society has suggested that liver enzymes should be periodically monitored in patients being treated with terbinafine beyond 4 to 6 weeks.13

Changes in the Fungal Microbiome

Research has shown that changes in the fungal microbiome were associated with an altered bacterial community in patients with TC. During fungal infection, the relative abundances of Cutibacterium and Corynebacterium increased, and the relative abundance of Streptococcus decreased. In addition, some uncommon bacterial genera such as Herbaspirillum and Methylorubrum were detected on the scalp in TC.14

Carrier State

Carrier state is determined for those siblings and contacts of cases with a clinically normal scalp that are positive on culture. Those individuals could represent a potential reservoir responsible for contamination (or recontamination) of the patient as well as treatment failure. Opinions remain divided as to whether to use oral antifungal therapy in these carriers or maintain therapy on antifungal shampoos containing ketoconazole or povidone-iodine. Due to the paucity of available data, my experience has shown that it is sufficient to use antifungal shampoos for such carriers. In zoophilic infections, it is important to identify and treat the animal source.6-9

Final Thoughts

Successful treatment of TC requires accurate identification of the pathogen, which commonly is achieved via fungal culture. Despite its practical value, the conventional identification of dermatophytes based on morphologic features can be highly challenging due to the low positive rate and delayed results. Trichoscopy is a quick, handy, and noninvasive tool that can better indicate the diagnosis and also is helpful for follow-up on treatment progress. Due to better understanding of the immunology and genetic susceptibility associated with TC spread, the current treatment pipeline holds more insight into better control of this condition. Increased surveillance, prompt diagnosis, and early onset of systemic treatment are the key to proper prevention of spread of TC.

References
  1. Leung AKC, Hon KL, Leong KF, et al. Tinea capitis: an updated review. Recent Pat Inflamm Allergy Drug Discov. 2020;14:58-68.
  2. Abdel-Rahman SM, Preuett BL. Genetic predictors of susceptibility to cutaneous fungal infections: a pilot genome wide association study to refine a candidate gene search. J Dermatol Sci. 2012;67:147-152.
  3. Hay RJ. Tinea capitis: current status. Mycopathologia. 2017;182:87-93.
  4. Wahbah HR, Atallah RB, Eldahshan RM, et al. A prospective clinical and trichoscopic study of tinea capitis in children during treatment [published online May 23, 2022]. Dermatol Ther. 2022;35:E15582. doi:10.1111/dth.15582
  5. Salehi Z, Shams-Ghahfarokhi M, Razzaghi-Abyaneh M. Molecular epidemiology, genetic diversity, and antifungal susceptibility of major pathogenic dermatophytes isolated from human dermatophytosis. Front Microbiol. 2021;12:643509.
  6. Lamisil. Package insert. Novartis; 2011. Accessed October 17, 2022. https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/020539s021lbl.pdf
  7. Gupta AK, Drummond-Main C. Meta-analysis of randomized, controlled trials comparing particular doses of griseofulvin and terbinafine for the treatment of tinea capitis. Pediatr Dermatol. 2013;30:1-6.
  8. Tey HL, Tan AS, Chan YC. Meta-analysis of randomized, controlled trials comparing griseofulvin and terbinafine in the treatment of tinea capitis. J Am Acad Dermatol. 2011;64:663-670.
  9. Gupta AK, Friedlander SF, Simkovich AJ. Tinea capitis: an update. Pediatr Dermatol. 2022;39:167-172.
  10. Aspiroz C, Melcon B, Cerro PA, et al. Tinea capitis caused by Microsporum canis treated with methyl-aminolevulinate daylight photodynamic therapy and ketoconazole shampooing. Photodermatol Photoimmunol Photomed. 2021;37:567-568.
  11. Aleohin N, Bar J, Bar-Ilan E, et al. Laboratory monitoring during antifungal treatment of paediatric tinea capitis. Mycoses. 2021;64:157-161.
  12. Kimberlin DW, Brady MT, Jackson MA, et al, eds. Tinea capitis. In: Red Book 2018-2021: Report of the Committee of Infectious Diseases. American Academy of Pediatrics; 2018:798-801.
  13. Bortolussi R, Martin S, Audcent T, et al. Antifungal agents for common outpatient paediatric infections. Canadian Paediatric Society website. Published June 20, 2019. Accessed October 4, 2022. https://www.cps.ca/en/documents/position/antifungal-agents-common-infections
  14. Tao R, Zhu P, Zhou Y, et al. Altered skin fungal and bacterial community compositions in tinea capitis. Mycoses. 2022;65:834-840.
Article PDF
Author and Disclosure Information

From the Department of Dermatology, Medical and Clinical Research Institute, National Research Centre, Egypt, and the Miller School of Medicine, University of Miami, Florida.

The author reports no conflict of interest.

Correspondence: Mohamed L. Elsaie, MD ([email protected]).

Issue
Cutis - 110(5)
Publications
Topics
Page Number
238-240
Sections
Author and Disclosure Information

From the Department of Dermatology, Medical and Clinical Research Institute, National Research Centre, Egypt, and the Miller School of Medicine, University of Miami, Florida.

The author reports no conflict of interest.

Correspondence: Mohamed L. Elsaie, MD ([email protected]).

Author and Disclosure Information

From the Department of Dermatology, Medical and Clinical Research Institute, National Research Centre, Egypt, and the Miller School of Medicine, University of Miami, Florida.

The author reports no conflict of interest.

Correspondence: Mohamed L. Elsaie, MD ([email protected]).

Article PDF
Article PDF

Tinea capitis (TC) most often is caused by Trichophyton tonsurans and Microsporum canis. The peak incidence is between 3 and 7 years of age. Noninflammatory TC typically presents as fine scaling with single or multiple scaly patches of circular alopecia (grey patches); diffuse or patchy, fine, white, adherent scaling of the scalp resembling generalized dandruff with subtle hair loss; or single or multiple patches of well-demarcated areas of alopecia with fine scale studded with broken-off hairs at the scalp surface, resulting in a black dot appearance. Inflammatory variants of TC include kerion and favus.1 Herein, updates on diagnosis, treatment, and monitoring of TC are provided, as well as a discussion of changes in the fungal microbiome associated with TC. Lastly, insights to some queries that practitioners may encounter when treating children with TC are provided.

Genetic Susceptibility

Molecular techniques have identified a number of macrophage regulator, leukocyte activation and migration, and cutaneous permeability genes associated with susceptibility to TC. These findings indicate that genetically determined deficiency in adaptive immune responses may affect the predisposition to dermatophyte infections.2

Clinical Varieties of Infection

Dermatophytes causing ringworm are capable of invading the hair shafts and can simultaneously invade smooth or glabrous skin (eg, T tonsurans, Trichophyton schoenleinii, Trichophyton violaceum). Some causative dermatophytes can even penetrate the nails (eg, Trichophyton soudanense). The clinical presentation is dependent on 3 main patterns of hair invasion3:

• Ectothrix: A mid-follicular pattern of invasion with hyphae growing down to the hair bulb that commonly is caused by Microsporum species. It clinically presents with scaling and inflammation with hair shafts breaking 2 to 3 mm above the scalp level.

• Endothrix: This pattern is nonfluorescent on Wood lamp examination, and hairs often break at the scalp level (black dot type). Trichophyton tonsurans, T soudanense, Trichophyton rubrum, and T violaceum are common causes.

• Favus: In this pattern, T schoenleinii is a common cause, and hairs grow to considerable lengths above the scalp with less damage than the other patterns. The hair shafts present with characteristic air spaces, and hyphae form clusters at the level of the epidermis.

Diagnosis

Optimal treatment of TC relies on proper identification of the causative agent. Fungal culture remains the gold standard of mycologic diagnosis regardless of its delayed results, which may take up to 4 weeks for proper identification of the fungal colonies and require ample expertise to interpret the morphologic features of the grown colonies.4

 

 

Other tests such as the potassium hydroxide preparation are nonspecific and do not identify the dermatophyte species. Although this method has been reported to have 5% to 15% false-negative results in routine practice depending on the skill of the observer and the quality of sampling, microscopic examination is essential, as it may allow the clinician to start treatment sooner pending culture results. The use of a Wood lamp is not suitable for definitive species identification, as this technique primarily is useful for observing fluorescence in ectothrix infection caused by Microsporum species, with the exception of T schoenleinii; otherwise, Trichophyton species, which cause endothrix infections, do not fluoresce.5Polymerase chain reaction is a sensitive technique that can help identify both the genus and species of common dermatophytes. Common target sequences include the ribosomal internal transcribed spacer and translation elongation factor 1α. The use of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry also has become popular for dermatophyte identification.6Trichoscopic diagnosis of TC, which is simple and noninvasive, is becoming increasingly popular. Features such as short, broken, black dot, comma, corkscrew, and/or zigzag hairs, as well as perifollicular scaling, are helpful for diagnosing TC (Figure). Moreover, trichoscopy can be useful for differentiating other common causes of hair loss, such as trichotillomania and alopecia areata. It had been reported that the trichoscopic features of TC can be seen as early as 2 weeks after starting treatment and therefore this can be a reliable period in which to follow-up with the patient to evaluate progress. The disappearance of black dots and comma hairs can be appreciated from 2 weeks onwards by trichoscopic evaluation.4

A, A 10-year-old boy with the black dot type of tinea capitis. B, Trichoscopy showed black dot hairs (pink circle), comma hairs (red arrows), corkscrew hairs (green arrows), short broken hairs (blue arrow), and perifollicular scaling (yellow arrow).
A, A 10-year-old boy with the black dot type of tinea capitis. B, Trichoscopy showed black dot hairs (pink circle), comma hairs (red arrows), corkscrew hairs (green arrows), short broken hairs (blue arrow), and perifollicular scaling (yellow arrow). C, Clinical resolution was achieved after 8 weeks of griseofulvin therapy. D, Posttreatment trichoscopy showed disappearance of dystrophic hairs.

Treatment

The common recommendation for first-line treatment of TC is the use of systemic antifungals with the use of a topical agent as an adjuvant to prevent the spread of fungal spores. For almost 6 decades, griseofulvin had been the gold-standard fungistatic used for treating TC in patients older than 2 years until the 2007 US Food and Drug Administration (FDA) approval of terbinafine fungicidal oral granules for treatment of TC in patients older than 4 years.7

Meta-analyses have demonstrated comparable efficacy for a 4-week course of terbinafine compared to 6 weeks of griseofulvin for TC based on the infectious organism. Terbinafine demonstrated superiority in treating T tonsurans and a similar efficacy in treating T violaceum, while griseofulvin was superior in treating M canis and other Microsporum species.8,9

The off-label use of fluconazole and itraconazole to treat TC is gaining popularity, with limited trials showing increased evidence of their effectiveness. There is not much clinical evidence to support the use of other oral antifungals, including the newer azoles such as voriconazole or posaconazole.9

Newer limited evidence has shown the off-label use of photodynamic therapy to be a promising alternative to systemic antifungal therapy in treating TC, pending validation by larger sample trials.10In my practice, I have found that severe cases of TC demonstrating inflammation or possible widespread id reactions are better treated with oral steroids. Ketoconazole shampoo or selenium sulfide used 2 to 3 times weekly to prevent spread in the early phases of therapy is a good adjunct to systemic treatment. Cases with kerions should be assessed for the possibility of a coexisting bacterial infection under the crusts, and if confirmed, antibiotics should be started.9The commonly used systemic antifungals generally are safe with a low side-effect profile, but there is a risk for hepatotoxicity. The FDA recommends that baseline alanine transaminase and aspartate transaminase levels should be obtained prior to beginning a terbinafine-based treatment regimen.11 The American Academy of Pediatrics has specifically stated that laboratory testing of serum hepatic enzymes is not a requirement if a griseofulvin-based regimen does not exceed 8 weeks; however, transaminase levels (alanine transaminase and aspartate transaminase) should be considered in patients using terbinafine at baseline or if treatment is prolonged beyond 4 to 6 weeks.12 In agreement with the FDA guidelines, the Canadian Pediatric Society has suggested that liver enzymes should be periodically monitored in patients being treated with terbinafine beyond 4 to 6 weeks.13

Changes in the Fungal Microbiome

Research has shown that changes in the fungal microbiome were associated with an altered bacterial community in patients with TC. During fungal infection, the relative abundances of Cutibacterium and Corynebacterium increased, and the relative abundance of Streptococcus decreased. In addition, some uncommon bacterial genera such as Herbaspirillum and Methylorubrum were detected on the scalp in TC.14

Carrier State

Carrier state is determined for those siblings and contacts of cases with a clinically normal scalp that are positive on culture. Those individuals could represent a potential reservoir responsible for contamination (or recontamination) of the patient as well as treatment failure. Opinions remain divided as to whether to use oral antifungal therapy in these carriers or maintain therapy on antifungal shampoos containing ketoconazole or povidone-iodine. Due to the paucity of available data, my experience has shown that it is sufficient to use antifungal shampoos for such carriers. In zoophilic infections, it is important to identify and treat the animal source.6-9

Final Thoughts

Successful treatment of TC requires accurate identification of the pathogen, which commonly is achieved via fungal culture. Despite its practical value, the conventional identification of dermatophytes based on morphologic features can be highly challenging due to the low positive rate and delayed results. Trichoscopy is a quick, handy, and noninvasive tool that can better indicate the diagnosis and also is helpful for follow-up on treatment progress. Due to better understanding of the immunology and genetic susceptibility associated with TC spread, the current treatment pipeline holds more insight into better control of this condition. Increased surveillance, prompt diagnosis, and early onset of systemic treatment are the key to proper prevention of spread of TC.

Tinea capitis (TC) most often is caused by Trichophyton tonsurans and Microsporum canis. The peak incidence is between 3 and 7 years of age. Noninflammatory TC typically presents as fine scaling with single or multiple scaly patches of circular alopecia (grey patches); diffuse or patchy, fine, white, adherent scaling of the scalp resembling generalized dandruff with subtle hair loss; or single or multiple patches of well-demarcated areas of alopecia with fine scale studded with broken-off hairs at the scalp surface, resulting in a black dot appearance. Inflammatory variants of TC include kerion and favus.1 Herein, updates on diagnosis, treatment, and monitoring of TC are provided, as well as a discussion of changes in the fungal microbiome associated with TC. Lastly, insights to some queries that practitioners may encounter when treating children with TC are provided.

Genetic Susceptibility

Molecular techniques have identified a number of macrophage regulator, leukocyte activation and migration, and cutaneous permeability genes associated with susceptibility to TC. These findings indicate that genetically determined deficiency in adaptive immune responses may affect the predisposition to dermatophyte infections.2

Clinical Varieties of Infection

Dermatophytes causing ringworm are capable of invading the hair shafts and can simultaneously invade smooth or glabrous skin (eg, T tonsurans, Trichophyton schoenleinii, Trichophyton violaceum). Some causative dermatophytes can even penetrate the nails (eg, Trichophyton soudanense). The clinical presentation is dependent on 3 main patterns of hair invasion3:

• Ectothrix: A mid-follicular pattern of invasion with hyphae growing down to the hair bulb that commonly is caused by Microsporum species. It clinically presents with scaling and inflammation with hair shafts breaking 2 to 3 mm above the scalp level.

• Endothrix: This pattern is nonfluorescent on Wood lamp examination, and hairs often break at the scalp level (black dot type). Trichophyton tonsurans, T soudanense, Trichophyton rubrum, and T violaceum are common causes.

• Favus: In this pattern, T schoenleinii is a common cause, and hairs grow to considerable lengths above the scalp with less damage than the other patterns. The hair shafts present with characteristic air spaces, and hyphae form clusters at the level of the epidermis.

Diagnosis

Optimal treatment of TC relies on proper identification of the causative agent. Fungal culture remains the gold standard of mycologic diagnosis regardless of its delayed results, which may take up to 4 weeks for proper identification of the fungal colonies and require ample expertise to interpret the morphologic features of the grown colonies.4

 

 

Other tests such as the potassium hydroxide preparation are nonspecific and do not identify the dermatophyte species. Although this method has been reported to have 5% to 15% false-negative results in routine practice depending on the skill of the observer and the quality of sampling, microscopic examination is essential, as it may allow the clinician to start treatment sooner pending culture results. The use of a Wood lamp is not suitable for definitive species identification, as this technique primarily is useful for observing fluorescence in ectothrix infection caused by Microsporum species, with the exception of T schoenleinii; otherwise, Trichophyton species, which cause endothrix infections, do not fluoresce.5Polymerase chain reaction is a sensitive technique that can help identify both the genus and species of common dermatophytes. Common target sequences include the ribosomal internal transcribed spacer and translation elongation factor 1α. The use of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry also has become popular for dermatophyte identification.6Trichoscopic diagnosis of TC, which is simple and noninvasive, is becoming increasingly popular. Features such as short, broken, black dot, comma, corkscrew, and/or zigzag hairs, as well as perifollicular scaling, are helpful for diagnosing TC (Figure). Moreover, trichoscopy can be useful for differentiating other common causes of hair loss, such as trichotillomania and alopecia areata. It had been reported that the trichoscopic features of TC can be seen as early as 2 weeks after starting treatment and therefore this can be a reliable period in which to follow-up with the patient to evaluate progress. The disappearance of black dots and comma hairs can be appreciated from 2 weeks onwards by trichoscopic evaluation.4

A, A 10-year-old boy with the black dot type of tinea capitis. B, Trichoscopy showed black dot hairs (pink circle), comma hairs (red arrows), corkscrew hairs (green arrows), short broken hairs (blue arrow), and perifollicular scaling (yellow arrow).
A, A 10-year-old boy with the black dot type of tinea capitis. B, Trichoscopy showed black dot hairs (pink circle), comma hairs (red arrows), corkscrew hairs (green arrows), short broken hairs (blue arrow), and perifollicular scaling (yellow arrow). C, Clinical resolution was achieved after 8 weeks of griseofulvin therapy. D, Posttreatment trichoscopy showed disappearance of dystrophic hairs.

Treatment

The common recommendation for first-line treatment of TC is the use of systemic antifungals with the use of a topical agent as an adjuvant to prevent the spread of fungal spores. For almost 6 decades, griseofulvin had been the gold-standard fungistatic used for treating TC in patients older than 2 years until the 2007 US Food and Drug Administration (FDA) approval of terbinafine fungicidal oral granules for treatment of TC in patients older than 4 years.7

Meta-analyses have demonstrated comparable efficacy for a 4-week course of terbinafine compared to 6 weeks of griseofulvin for TC based on the infectious organism. Terbinafine demonstrated superiority in treating T tonsurans and a similar efficacy in treating T violaceum, while griseofulvin was superior in treating M canis and other Microsporum species.8,9

The off-label use of fluconazole and itraconazole to treat TC is gaining popularity, with limited trials showing increased evidence of their effectiveness. There is not much clinical evidence to support the use of other oral antifungals, including the newer azoles such as voriconazole or posaconazole.9

Newer limited evidence has shown the off-label use of photodynamic therapy to be a promising alternative to systemic antifungal therapy in treating TC, pending validation by larger sample trials.10In my practice, I have found that severe cases of TC demonstrating inflammation or possible widespread id reactions are better treated with oral steroids. Ketoconazole shampoo or selenium sulfide used 2 to 3 times weekly to prevent spread in the early phases of therapy is a good adjunct to systemic treatment. Cases with kerions should be assessed for the possibility of a coexisting bacterial infection under the crusts, and if confirmed, antibiotics should be started.9The commonly used systemic antifungals generally are safe with a low side-effect profile, but there is a risk for hepatotoxicity. The FDA recommends that baseline alanine transaminase and aspartate transaminase levels should be obtained prior to beginning a terbinafine-based treatment regimen.11 The American Academy of Pediatrics has specifically stated that laboratory testing of serum hepatic enzymes is not a requirement if a griseofulvin-based regimen does not exceed 8 weeks; however, transaminase levels (alanine transaminase and aspartate transaminase) should be considered in patients using terbinafine at baseline or if treatment is prolonged beyond 4 to 6 weeks.12 In agreement with the FDA guidelines, the Canadian Pediatric Society has suggested that liver enzymes should be periodically monitored in patients being treated with terbinafine beyond 4 to 6 weeks.13

Changes in the Fungal Microbiome

Research has shown that changes in the fungal microbiome were associated with an altered bacterial community in patients with TC. During fungal infection, the relative abundances of Cutibacterium and Corynebacterium increased, and the relative abundance of Streptococcus decreased. In addition, some uncommon bacterial genera such as Herbaspirillum and Methylorubrum were detected on the scalp in TC.14

Carrier State

Carrier state is determined for those siblings and contacts of cases with a clinically normal scalp that are positive on culture. Those individuals could represent a potential reservoir responsible for contamination (or recontamination) of the patient as well as treatment failure. Opinions remain divided as to whether to use oral antifungal therapy in these carriers or maintain therapy on antifungal shampoos containing ketoconazole or povidone-iodine. Due to the paucity of available data, my experience has shown that it is sufficient to use antifungal shampoos for such carriers. In zoophilic infections, it is important to identify and treat the animal source.6-9

Final Thoughts

Successful treatment of TC requires accurate identification of the pathogen, which commonly is achieved via fungal culture. Despite its practical value, the conventional identification of dermatophytes based on morphologic features can be highly challenging due to the low positive rate and delayed results. Trichoscopy is a quick, handy, and noninvasive tool that can better indicate the diagnosis and also is helpful for follow-up on treatment progress. Due to better understanding of the immunology and genetic susceptibility associated with TC spread, the current treatment pipeline holds more insight into better control of this condition. Increased surveillance, prompt diagnosis, and early onset of systemic treatment are the key to proper prevention of spread of TC.

References
  1. Leung AKC, Hon KL, Leong KF, et al. Tinea capitis: an updated review. Recent Pat Inflamm Allergy Drug Discov. 2020;14:58-68.
  2. Abdel-Rahman SM, Preuett BL. Genetic predictors of susceptibility to cutaneous fungal infections: a pilot genome wide association study to refine a candidate gene search. J Dermatol Sci. 2012;67:147-152.
  3. Hay RJ. Tinea capitis: current status. Mycopathologia. 2017;182:87-93.
  4. Wahbah HR, Atallah RB, Eldahshan RM, et al. A prospective clinical and trichoscopic study of tinea capitis in children during treatment [published online May 23, 2022]. Dermatol Ther. 2022;35:E15582. doi:10.1111/dth.15582
  5. Salehi Z, Shams-Ghahfarokhi M, Razzaghi-Abyaneh M. Molecular epidemiology, genetic diversity, and antifungal susceptibility of major pathogenic dermatophytes isolated from human dermatophytosis. Front Microbiol. 2021;12:643509.
  6. Lamisil. Package insert. Novartis; 2011. Accessed October 17, 2022. https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/020539s021lbl.pdf
  7. Gupta AK, Drummond-Main C. Meta-analysis of randomized, controlled trials comparing particular doses of griseofulvin and terbinafine for the treatment of tinea capitis. Pediatr Dermatol. 2013;30:1-6.
  8. Tey HL, Tan AS, Chan YC. Meta-analysis of randomized, controlled trials comparing griseofulvin and terbinafine in the treatment of tinea capitis. J Am Acad Dermatol. 2011;64:663-670.
  9. Gupta AK, Friedlander SF, Simkovich AJ. Tinea capitis: an update. Pediatr Dermatol. 2022;39:167-172.
  10. Aspiroz C, Melcon B, Cerro PA, et al. Tinea capitis caused by Microsporum canis treated with methyl-aminolevulinate daylight photodynamic therapy and ketoconazole shampooing. Photodermatol Photoimmunol Photomed. 2021;37:567-568.
  11. Aleohin N, Bar J, Bar-Ilan E, et al. Laboratory monitoring during antifungal treatment of paediatric tinea capitis. Mycoses. 2021;64:157-161.
  12. Kimberlin DW, Brady MT, Jackson MA, et al, eds. Tinea capitis. In: Red Book 2018-2021: Report of the Committee of Infectious Diseases. American Academy of Pediatrics; 2018:798-801.
  13. Bortolussi R, Martin S, Audcent T, et al. Antifungal agents for common outpatient paediatric infections. Canadian Paediatric Society website. Published June 20, 2019. Accessed October 4, 2022. https://www.cps.ca/en/documents/position/antifungal-agents-common-infections
  14. Tao R, Zhu P, Zhou Y, et al. Altered skin fungal and bacterial community compositions in tinea capitis. Mycoses. 2022;65:834-840.
References
  1. Leung AKC, Hon KL, Leong KF, et al. Tinea capitis: an updated review. Recent Pat Inflamm Allergy Drug Discov. 2020;14:58-68.
  2. Abdel-Rahman SM, Preuett BL. Genetic predictors of susceptibility to cutaneous fungal infections: a pilot genome wide association study to refine a candidate gene search. J Dermatol Sci. 2012;67:147-152.
  3. Hay RJ. Tinea capitis: current status. Mycopathologia. 2017;182:87-93.
  4. Wahbah HR, Atallah RB, Eldahshan RM, et al. A prospective clinical and trichoscopic study of tinea capitis in children during treatment [published online May 23, 2022]. Dermatol Ther. 2022;35:E15582. doi:10.1111/dth.15582
  5. Salehi Z, Shams-Ghahfarokhi M, Razzaghi-Abyaneh M. Molecular epidemiology, genetic diversity, and antifungal susceptibility of major pathogenic dermatophytes isolated from human dermatophytosis. Front Microbiol. 2021;12:643509.
  6. Lamisil. Package insert. Novartis; 2011. Accessed October 17, 2022. https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/020539s021lbl.pdf
  7. Gupta AK, Drummond-Main C. Meta-analysis of randomized, controlled trials comparing particular doses of griseofulvin and terbinafine for the treatment of tinea capitis. Pediatr Dermatol. 2013;30:1-6.
  8. Tey HL, Tan AS, Chan YC. Meta-analysis of randomized, controlled trials comparing griseofulvin and terbinafine in the treatment of tinea capitis. J Am Acad Dermatol. 2011;64:663-670.
  9. Gupta AK, Friedlander SF, Simkovich AJ. Tinea capitis: an update. Pediatr Dermatol. 2022;39:167-172.
  10. Aspiroz C, Melcon B, Cerro PA, et al. Tinea capitis caused by Microsporum canis treated with methyl-aminolevulinate daylight photodynamic therapy and ketoconazole shampooing. Photodermatol Photoimmunol Photomed. 2021;37:567-568.
  11. Aleohin N, Bar J, Bar-Ilan E, et al. Laboratory monitoring during antifungal treatment of paediatric tinea capitis. Mycoses. 2021;64:157-161.
  12. Kimberlin DW, Brady MT, Jackson MA, et al, eds. Tinea capitis. In: Red Book 2018-2021: Report of the Committee of Infectious Diseases. American Academy of Pediatrics; 2018:798-801.
  13. Bortolussi R, Martin S, Audcent T, et al. Antifungal agents for common outpatient paediatric infections. Canadian Paediatric Society website. Published June 20, 2019. Accessed October 4, 2022. https://www.cps.ca/en/documents/position/antifungal-agents-common-infections
  14. Tao R, Zhu P, Zhou Y, et al. Altered skin fungal and bacterial community compositions in tinea capitis. Mycoses. 2022;65:834-840.
Issue
Cutis - 110(5)
Issue
Cutis - 110(5)
Page Number
238-240
Page Number
238-240
Publications
Publications
Topics
Article Type
Display Headline
Update on Tinea Capitis Diagnosis and Treatment
Display Headline
Update on Tinea Capitis Diagnosis and Treatment
Sections
Disallow All Ads
Content Gating
Open Access (article Unlocked/Open Access)
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

ACC/AHA issues updated guidance on aortic disease

Article Type
Changed
Fri, 11/04/2022 - 13:50

The American College of Cardiology and the American Heart Association have published an updated guideline on the diagnosis and management of aortic disease, focusing on surgical intervention considerations, consistent imaging practices, genetic and familial screenings, and the importance of multidisciplinary care.

“There has been a host of new evidence-based research available for clinicians in the past decade when it comes to aortic disease. It was time to reevaluate and update the previous, existing guidelines,” Eric M. Isselbacher, MD, MSc, chair of the writing committee, said in a statement.

“We hope this new guideline can inform clinical practices with up-to-date and synthesized recommendations, targeted toward a full multidisciplinary aortic team working to provide the best possible care for this vulnerable patient population,” added Dr. Isselbacher, codirector of the Thoracic Aortic Center at Massachusetts General Hospital, Boston.

The 2022 ACC/AHA Guideline for the Diagnosis and Management of Aortic Disease was simultaneously published online in the Journal of the American College of Cardiology and Circulation.

The new guideline replaces the 2010 ACCF/AHA Guidelines for the Diagnosis and Management of Patients With Thoracic Aortic Disease and the 2015 Surgery for Aortic Dilation in Patients With Bicuspid Aortic Valves: A Statement of Clarification From the ACC/AHA Task Force on Clinical Practice Guidelines.

The new guideline is intended to be used with the 2020 ACC/AHA Guideline for the Management of Patients With Valvular Heart Disease.

It brings together guidelines for both the thoracic and abdominal aorta and is targeted to cardiovascular clinicians involved in the care of people with aortic disease, including general cardiovascular care clinicians and emergency medicine clinicians, the writing group says.

Among the key recommendations in the new guideline are the following:

  • Screen first-degree relatives of individuals diagnosed with aneurysms of the aortic root or ascending thoracic aorta, or those with aortic dissection to identify individuals most at risk for aortic disease. Screening would include genetic testing and imaging.
  • Be consistent in the way CT or MRI are obtained and reported; in the measurement of aortic size and features; and in how often images are used for monitoring before and after repair surgery or other intervention. Ideally, all surveillance imaging for an individual should be done using the same modality and in the same lab, the guideline notes.
  • For individuals who require aortic intervention, know that outcomes are optimized when surgery is performed by an experienced surgeon working in a multidisciplinary aortic team. The new guideline recommends “a specialized hospital team with expertise in the evaluation and management of aortic disease, in which care is delivered in a comprehensive, multidisciplinary manner.”
  • At centers with multidisciplinary aortic teams and experienced surgeons, the threshold for surgical intervention for sporadic aortic root and ascending aortic aneurysms has been lowered from 5.5 cm to 5.0 cm in select individuals, and even lower in specific scenarios among patients with heritable thoracic aortic aneurysms.
  • In patients who are significantly smaller or taller than average, surgical thresholds may incorporate indexing of the aortic root or ascending aortic diameter to either patient body surface area or height, or aortic cross-sectional area to patient height.
  • Rapid aortic growth is a risk factor for rupture and the definition for rapid aneurysm growth rate has been updated. Surgery is now recommended for patients with aneurysms of aortic root and ascending thoracic aorta with a confirmed growth rate of ≥ 0.3 cm per year across 2 consecutive years or ≥ 0.5 cm in 1 year.
  • In patients undergoing aortic root replacement surgery, valve-sparing aortic root replacement is reasonable if the valve is suitable for repair and when performed by experienced surgeons in a multidisciplinary aortic team.
  • Patients with acute type A aortic dissection, if clinically stable, should be considered for transfer to a high-volume aortic center to improve survival. The operative repair of type A aortic dissection should entail at least an open distal anastomosis rather than just a simple supracoronary interposition graft.
  • For management of uncomplicated type B aortic dissection, there is an increasing role for . Clinical trials of repair of thoracoabdominal aortic aneurysms with endografts are reporting results that suggest endovascular repair is an option for patients with suitable anatomy.
  • Shared decision-making between the patient and multidisciplinary aortic team is highly encouraged, especially when the patient is on the borderline of thresholds for repair or eligible for different types of surgical repair.
  • Shared decision-making should also be used with individuals who are pregnant or may become pregnant to consider the risks of pregnancy in individuals with aortic disease.

The guideline was developed in collaboration with and endorsed by the American Association for Thoracic Surgery, the American College of Radiology, the Society of Cardiovascular Anesthesiologists, the Society for Cardiovascular Angiography and Interventions, the Society of Thoracic Surgeons, and the Society for Vascular Medicine.

It has been endorsed by the Society of Interventional Radiology and the Society for Vascular Surgery.

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

Publications
Topics
Sections

The American College of Cardiology and the American Heart Association have published an updated guideline on the diagnosis and management of aortic disease, focusing on surgical intervention considerations, consistent imaging practices, genetic and familial screenings, and the importance of multidisciplinary care.

“There has been a host of new evidence-based research available for clinicians in the past decade when it comes to aortic disease. It was time to reevaluate and update the previous, existing guidelines,” Eric M. Isselbacher, MD, MSc, chair of the writing committee, said in a statement.

“We hope this new guideline can inform clinical practices with up-to-date and synthesized recommendations, targeted toward a full multidisciplinary aortic team working to provide the best possible care for this vulnerable patient population,” added Dr. Isselbacher, codirector of the Thoracic Aortic Center at Massachusetts General Hospital, Boston.

The 2022 ACC/AHA Guideline for the Diagnosis and Management of Aortic Disease was simultaneously published online in the Journal of the American College of Cardiology and Circulation.

The new guideline replaces the 2010 ACCF/AHA Guidelines for the Diagnosis and Management of Patients With Thoracic Aortic Disease and the 2015 Surgery for Aortic Dilation in Patients With Bicuspid Aortic Valves: A Statement of Clarification From the ACC/AHA Task Force on Clinical Practice Guidelines.

The new guideline is intended to be used with the 2020 ACC/AHA Guideline for the Management of Patients With Valvular Heart Disease.

It brings together guidelines for both the thoracic and abdominal aorta and is targeted to cardiovascular clinicians involved in the care of people with aortic disease, including general cardiovascular care clinicians and emergency medicine clinicians, the writing group says.

Among the key recommendations in the new guideline are the following:

  • Screen first-degree relatives of individuals diagnosed with aneurysms of the aortic root or ascending thoracic aorta, or those with aortic dissection to identify individuals most at risk for aortic disease. Screening would include genetic testing and imaging.
  • Be consistent in the way CT or MRI are obtained and reported; in the measurement of aortic size and features; and in how often images are used for monitoring before and after repair surgery or other intervention. Ideally, all surveillance imaging for an individual should be done using the same modality and in the same lab, the guideline notes.
  • For individuals who require aortic intervention, know that outcomes are optimized when surgery is performed by an experienced surgeon working in a multidisciplinary aortic team. The new guideline recommends “a specialized hospital team with expertise in the evaluation and management of aortic disease, in which care is delivered in a comprehensive, multidisciplinary manner.”
  • At centers with multidisciplinary aortic teams and experienced surgeons, the threshold for surgical intervention for sporadic aortic root and ascending aortic aneurysms has been lowered from 5.5 cm to 5.0 cm in select individuals, and even lower in specific scenarios among patients with heritable thoracic aortic aneurysms.
  • In patients who are significantly smaller or taller than average, surgical thresholds may incorporate indexing of the aortic root or ascending aortic diameter to either patient body surface area or height, or aortic cross-sectional area to patient height.
  • Rapid aortic growth is a risk factor for rupture and the definition for rapid aneurysm growth rate has been updated. Surgery is now recommended for patients with aneurysms of aortic root and ascending thoracic aorta with a confirmed growth rate of ≥ 0.3 cm per year across 2 consecutive years or ≥ 0.5 cm in 1 year.
  • In patients undergoing aortic root replacement surgery, valve-sparing aortic root replacement is reasonable if the valve is suitable for repair and when performed by experienced surgeons in a multidisciplinary aortic team.
  • Patients with acute type A aortic dissection, if clinically stable, should be considered for transfer to a high-volume aortic center to improve survival. The operative repair of type A aortic dissection should entail at least an open distal anastomosis rather than just a simple supracoronary interposition graft.
  • For management of uncomplicated type B aortic dissection, there is an increasing role for . Clinical trials of repair of thoracoabdominal aortic aneurysms with endografts are reporting results that suggest endovascular repair is an option for patients with suitable anatomy.
  • Shared decision-making between the patient and multidisciplinary aortic team is highly encouraged, especially when the patient is on the borderline of thresholds for repair or eligible for different types of surgical repair.
  • Shared decision-making should also be used with individuals who are pregnant or may become pregnant to consider the risks of pregnancy in individuals with aortic disease.

The guideline was developed in collaboration with and endorsed by the American Association for Thoracic Surgery, the American College of Radiology, the Society of Cardiovascular Anesthesiologists, the Society for Cardiovascular Angiography and Interventions, the Society of Thoracic Surgeons, and the Society for Vascular Medicine.

It has been endorsed by the Society of Interventional Radiology and the Society for Vascular Surgery.

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

The American College of Cardiology and the American Heart Association have published an updated guideline on the diagnosis and management of aortic disease, focusing on surgical intervention considerations, consistent imaging practices, genetic and familial screenings, and the importance of multidisciplinary care.

“There has been a host of new evidence-based research available for clinicians in the past decade when it comes to aortic disease. It was time to reevaluate and update the previous, existing guidelines,” Eric M. Isselbacher, MD, MSc, chair of the writing committee, said in a statement.

“We hope this new guideline can inform clinical practices with up-to-date and synthesized recommendations, targeted toward a full multidisciplinary aortic team working to provide the best possible care for this vulnerable patient population,” added Dr. Isselbacher, codirector of the Thoracic Aortic Center at Massachusetts General Hospital, Boston.

The 2022 ACC/AHA Guideline for the Diagnosis and Management of Aortic Disease was simultaneously published online in the Journal of the American College of Cardiology and Circulation.

The new guideline replaces the 2010 ACCF/AHA Guidelines for the Diagnosis and Management of Patients With Thoracic Aortic Disease and the 2015 Surgery for Aortic Dilation in Patients With Bicuspid Aortic Valves: A Statement of Clarification From the ACC/AHA Task Force on Clinical Practice Guidelines.

The new guideline is intended to be used with the 2020 ACC/AHA Guideline for the Management of Patients With Valvular Heart Disease.

It brings together guidelines for both the thoracic and abdominal aorta and is targeted to cardiovascular clinicians involved in the care of people with aortic disease, including general cardiovascular care clinicians and emergency medicine clinicians, the writing group says.

Among the key recommendations in the new guideline are the following:

  • Screen first-degree relatives of individuals diagnosed with aneurysms of the aortic root or ascending thoracic aorta, or those with aortic dissection to identify individuals most at risk for aortic disease. Screening would include genetic testing and imaging.
  • Be consistent in the way CT or MRI are obtained and reported; in the measurement of aortic size and features; and in how often images are used for monitoring before and after repair surgery or other intervention. Ideally, all surveillance imaging for an individual should be done using the same modality and in the same lab, the guideline notes.
  • For individuals who require aortic intervention, know that outcomes are optimized when surgery is performed by an experienced surgeon working in a multidisciplinary aortic team. The new guideline recommends “a specialized hospital team with expertise in the evaluation and management of aortic disease, in which care is delivered in a comprehensive, multidisciplinary manner.”
  • At centers with multidisciplinary aortic teams and experienced surgeons, the threshold for surgical intervention for sporadic aortic root and ascending aortic aneurysms has been lowered from 5.5 cm to 5.0 cm in select individuals, and even lower in specific scenarios among patients with heritable thoracic aortic aneurysms.
  • In patients who are significantly smaller or taller than average, surgical thresholds may incorporate indexing of the aortic root or ascending aortic diameter to either patient body surface area or height, or aortic cross-sectional area to patient height.
  • Rapid aortic growth is a risk factor for rupture and the definition for rapid aneurysm growth rate has been updated. Surgery is now recommended for patients with aneurysms of aortic root and ascending thoracic aorta with a confirmed growth rate of ≥ 0.3 cm per year across 2 consecutive years or ≥ 0.5 cm in 1 year.
  • In patients undergoing aortic root replacement surgery, valve-sparing aortic root replacement is reasonable if the valve is suitable for repair and when performed by experienced surgeons in a multidisciplinary aortic team.
  • Patients with acute type A aortic dissection, if clinically stable, should be considered for transfer to a high-volume aortic center to improve survival. The operative repair of type A aortic dissection should entail at least an open distal anastomosis rather than just a simple supracoronary interposition graft.
  • For management of uncomplicated type B aortic dissection, there is an increasing role for . Clinical trials of repair of thoracoabdominal aortic aneurysms with endografts are reporting results that suggest endovascular repair is an option for patients with suitable anatomy.
  • Shared decision-making between the patient and multidisciplinary aortic team is highly encouraged, especially when the patient is on the borderline of thresholds for repair or eligible for different types of surgical repair.
  • Shared decision-making should also be used with individuals who are pregnant or may become pregnant to consider the risks of pregnancy in individuals with aortic disease.

The guideline was developed in collaboration with and endorsed by the American Association for Thoracic Surgery, the American College of Radiology, the Society of Cardiovascular Anesthesiologists, the Society for Cardiovascular Angiography and Interventions, the Society of Thoracic Surgeons, and the Society for Vascular Medicine.

It has been endorsed by the Society of Interventional Radiology and the Society for Vascular Surgery.

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

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CIRCULATION

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

Pulmonologist consult at COPD admission reduces risk of return

Article Type
Changed
Mon, 11/14/2022 - 09:48

. – If a pulmonologist becomes involved early in the care of patients admitted to the hospital for an acute exacerbation of chronic obstructive pulmonary disease (AECOPD), the rate of readmission is reduced substantially relative to no pulmonologist involvement, according to a retrospective cohort review presented at the annual meeting of the American College of Chest Physicians (CHEST).

“When stratified by severity of COPD at the time of admission, the difference in the readmission rate was even greater,” reported Nakisa Hekmat-Joo, MD, a third-year resident at Staten Island University Hospital, New York.

Just as protocols have been developed for prompt initiation of antibiotics in patients with septicemia or prompt revascularization in patients with ST-segment elevated myocardial infarction (STEMI), Dr. Hekmat-Joo said the data from this study warrant a larger trial to evaluate whether an AECOPD admission protocol is warranted to improve outcomes and lower costs.

In this study, all AECOPD admissions were included from a recent 2-year period at two Staten Island hospitals. Of these, 198 patients received a pulmonologist consult within 24 hours. The remaining 92 patients were not evaluated by pulmonologists but were admitted and then managed by residents, internists, or others.

The primary outcome was length of stay (LOS). Although the slightly lower LOS in pulmonologist-treated group did not approach significance (4.16 vs. 4.21 days; P = .88), the readmission rate at 90 days, which was a secondary outcome, was reduced by almost half (30.1% vs. 57.6%; P < .0001).

At admission, there was no significant difference between those receiving a pulmonologist consult and those who did not. The average O2 saturation was lower in the group seen by a pulmonologist (93% vs. 95.4%; P < .0001), but the most striking difference was the low relative readmission rate, which remained significant after controlling for severity and pulmonary function.

“When we stratified patients for baseline severity, the advantage of a pulmonologist consult was even greater for those with the most severe disease,” Dr. Hekmat-Joo said. Among those with the greatest severity, the 90-day readmission rate was nearly three times greater in the absence of a pulmonologist consult (72% vs. 28%).

Although the comparison of outcomes for those receiving a pulmonologist consult vs. those who did not was adjusted for COPD severity, the potential for pulmonologist consults to be ordered for those patients who looked the sickest would have likely worked against the study result.

“We speculate that pulmonologists were more likely than internists to treat beyond standard guidelines, particularly in the event of greater severity,” Dr. Hekmat-Joo explained. These steps might include earlier use of noninvasive positive pressure ventilation or earlier initiation of rehabilitation strategies.

There were several signals that a pulmonologist consult led to more rigorous care.

“The average time to follow-up after hospitalization was 23 days for the pulmonologist group and 66 days for the nonpulmonologist group,” said Dr. Hekmat-Joo, noting this difference was highly significant (P = .0052).

Based on these results, Dr. Hekmat-Joo and her co-investigators are now working on a protocol for COPD admissions that involves a pulmonologist consult within 24 hours of admission. She hopes to test this protocol in a prospective trial.

“COPD remains a major cause of death and consumes enormous health care resources. About 30% of the cost of COPD care is due to readmissions,” she said, noting that readmissions adversely impact quality of life.

Asked if there was sufficient staff at her institution to allow for a pulmonologist consult with every COPD admission, Dr. Hekmat-Joo acknowledged that this has to be demonstrated, but compelling evidence of a benefit might prompt a redistribution of resources.

“If we can show that readmissions are substantially reduced, adding staff to perform these consults would be a good investment,” said Dr. Hekmat-Joo, indicating that improved outcomes could also attract the attention of third-party payers and those tracking quality-of-care metrics.

There is a strong rationale for a randomized prospective trial to confirm the value of a pulmonologist consultation following admission for an acute exacerbation of COPD, according to Nicola A. Hanania, MD, director, Airways Clinical Research Center, Baylor College of Medicine, Houston.

The potential for benefit as seen in this retrospective study is a rational expectation and might be related to more appropriate therapy upon discharge as well as to earlier and more rigorous follow-up, according to Dr. Hanania. Although he cautioned that there is a meaningful risk of selection bias in a retrospective study, he thinks this study “is certainly probing an important issue.”

“Mortality from a hospitalized COPD exacerbation exceeds that of a myocardial infarction,” Dr. Hanania pointed out. Noting that all patients with an MI are evaluated by a cardiologist, he sees the logic of a pulmonologist consult – although he acknowledged that evidence is needed.

“I strongly believe that a prospective study is feasible and will answer the question in an unbiased manner if done properly,” he said in an interview. If a multicenter, well-controlled study was positive, it could change practice.

In the event of a study showing major clinical benefits, particularly a reduction in mortality, “I believe it is feasible to have a pulmonary consult to see every COPD exacerbation patient admitted to the hospital,” Dr. Hanania said.

Dr. Hekmat-Joo reports no relevant financial relationships. Dr. Hanania has financial relationships with AstraZeneca, Boehringer Ingelheim, GlaxoSmithKline, Mylan, Novartis, Regeneron, Sanofi, and Sunovion.

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

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

. – If a pulmonologist becomes involved early in the care of patients admitted to the hospital for an acute exacerbation of chronic obstructive pulmonary disease (AECOPD), the rate of readmission is reduced substantially relative to no pulmonologist involvement, according to a retrospective cohort review presented at the annual meeting of the American College of Chest Physicians (CHEST).

“When stratified by severity of COPD at the time of admission, the difference in the readmission rate was even greater,” reported Nakisa Hekmat-Joo, MD, a third-year resident at Staten Island University Hospital, New York.

Just as protocols have been developed for prompt initiation of antibiotics in patients with septicemia or prompt revascularization in patients with ST-segment elevated myocardial infarction (STEMI), Dr. Hekmat-Joo said the data from this study warrant a larger trial to evaluate whether an AECOPD admission protocol is warranted to improve outcomes and lower costs.

In this study, all AECOPD admissions were included from a recent 2-year period at two Staten Island hospitals. Of these, 198 patients received a pulmonologist consult within 24 hours. The remaining 92 patients were not evaluated by pulmonologists but were admitted and then managed by residents, internists, or others.

The primary outcome was length of stay (LOS). Although the slightly lower LOS in pulmonologist-treated group did not approach significance (4.16 vs. 4.21 days; P = .88), the readmission rate at 90 days, which was a secondary outcome, was reduced by almost half (30.1% vs. 57.6%; P < .0001).

At admission, there was no significant difference between those receiving a pulmonologist consult and those who did not. The average O2 saturation was lower in the group seen by a pulmonologist (93% vs. 95.4%; P < .0001), but the most striking difference was the low relative readmission rate, which remained significant after controlling for severity and pulmonary function.

“When we stratified patients for baseline severity, the advantage of a pulmonologist consult was even greater for those with the most severe disease,” Dr. Hekmat-Joo said. Among those with the greatest severity, the 90-day readmission rate was nearly three times greater in the absence of a pulmonologist consult (72% vs. 28%).

Although the comparison of outcomes for those receiving a pulmonologist consult vs. those who did not was adjusted for COPD severity, the potential for pulmonologist consults to be ordered for those patients who looked the sickest would have likely worked against the study result.

“We speculate that pulmonologists were more likely than internists to treat beyond standard guidelines, particularly in the event of greater severity,” Dr. Hekmat-Joo explained. These steps might include earlier use of noninvasive positive pressure ventilation or earlier initiation of rehabilitation strategies.

There were several signals that a pulmonologist consult led to more rigorous care.

“The average time to follow-up after hospitalization was 23 days for the pulmonologist group and 66 days for the nonpulmonologist group,” said Dr. Hekmat-Joo, noting this difference was highly significant (P = .0052).

Based on these results, Dr. Hekmat-Joo and her co-investigators are now working on a protocol for COPD admissions that involves a pulmonologist consult within 24 hours of admission. She hopes to test this protocol in a prospective trial.

“COPD remains a major cause of death and consumes enormous health care resources. About 30% of the cost of COPD care is due to readmissions,” she said, noting that readmissions adversely impact quality of life.

Asked if there was sufficient staff at her institution to allow for a pulmonologist consult with every COPD admission, Dr. Hekmat-Joo acknowledged that this has to be demonstrated, but compelling evidence of a benefit might prompt a redistribution of resources.

“If we can show that readmissions are substantially reduced, adding staff to perform these consults would be a good investment,” said Dr. Hekmat-Joo, indicating that improved outcomes could also attract the attention of third-party payers and those tracking quality-of-care metrics.

There is a strong rationale for a randomized prospective trial to confirm the value of a pulmonologist consultation following admission for an acute exacerbation of COPD, according to Nicola A. Hanania, MD, director, Airways Clinical Research Center, Baylor College of Medicine, Houston.

The potential for benefit as seen in this retrospective study is a rational expectation and might be related to more appropriate therapy upon discharge as well as to earlier and more rigorous follow-up, according to Dr. Hanania. Although he cautioned that there is a meaningful risk of selection bias in a retrospective study, he thinks this study “is certainly probing an important issue.”

“Mortality from a hospitalized COPD exacerbation exceeds that of a myocardial infarction,” Dr. Hanania pointed out. Noting that all patients with an MI are evaluated by a cardiologist, he sees the logic of a pulmonologist consult – although he acknowledged that evidence is needed.

“I strongly believe that a prospective study is feasible and will answer the question in an unbiased manner if done properly,” he said in an interview. If a multicenter, well-controlled study was positive, it could change practice.

In the event of a study showing major clinical benefits, particularly a reduction in mortality, “I believe it is feasible to have a pulmonary consult to see every COPD exacerbation patient admitted to the hospital,” Dr. Hanania said.

Dr. Hekmat-Joo reports no relevant financial relationships. Dr. Hanania has financial relationships with AstraZeneca, Boehringer Ingelheim, GlaxoSmithKline, Mylan, Novartis, Regeneron, Sanofi, and Sunovion.

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

. – If a pulmonologist becomes involved early in the care of patients admitted to the hospital for an acute exacerbation of chronic obstructive pulmonary disease (AECOPD), the rate of readmission is reduced substantially relative to no pulmonologist involvement, according to a retrospective cohort review presented at the annual meeting of the American College of Chest Physicians (CHEST).

“When stratified by severity of COPD at the time of admission, the difference in the readmission rate was even greater,” reported Nakisa Hekmat-Joo, MD, a third-year resident at Staten Island University Hospital, New York.

Just as protocols have been developed for prompt initiation of antibiotics in patients with septicemia or prompt revascularization in patients with ST-segment elevated myocardial infarction (STEMI), Dr. Hekmat-Joo said the data from this study warrant a larger trial to evaluate whether an AECOPD admission protocol is warranted to improve outcomes and lower costs.

In this study, all AECOPD admissions were included from a recent 2-year period at two Staten Island hospitals. Of these, 198 patients received a pulmonologist consult within 24 hours. The remaining 92 patients were not evaluated by pulmonologists but were admitted and then managed by residents, internists, or others.

The primary outcome was length of stay (LOS). Although the slightly lower LOS in pulmonologist-treated group did not approach significance (4.16 vs. 4.21 days; P = .88), the readmission rate at 90 days, which was a secondary outcome, was reduced by almost half (30.1% vs. 57.6%; P < .0001).

At admission, there was no significant difference between those receiving a pulmonologist consult and those who did not. The average O2 saturation was lower in the group seen by a pulmonologist (93% vs. 95.4%; P < .0001), but the most striking difference was the low relative readmission rate, which remained significant after controlling for severity and pulmonary function.

“When we stratified patients for baseline severity, the advantage of a pulmonologist consult was even greater for those with the most severe disease,” Dr. Hekmat-Joo said. Among those with the greatest severity, the 90-day readmission rate was nearly three times greater in the absence of a pulmonologist consult (72% vs. 28%).

Although the comparison of outcomes for those receiving a pulmonologist consult vs. those who did not was adjusted for COPD severity, the potential for pulmonologist consults to be ordered for those patients who looked the sickest would have likely worked against the study result.

“We speculate that pulmonologists were more likely than internists to treat beyond standard guidelines, particularly in the event of greater severity,” Dr. Hekmat-Joo explained. These steps might include earlier use of noninvasive positive pressure ventilation or earlier initiation of rehabilitation strategies.

There were several signals that a pulmonologist consult led to more rigorous care.

“The average time to follow-up after hospitalization was 23 days for the pulmonologist group and 66 days for the nonpulmonologist group,” said Dr. Hekmat-Joo, noting this difference was highly significant (P = .0052).

Based on these results, Dr. Hekmat-Joo and her co-investigators are now working on a protocol for COPD admissions that involves a pulmonologist consult within 24 hours of admission. She hopes to test this protocol in a prospective trial.

“COPD remains a major cause of death and consumes enormous health care resources. About 30% of the cost of COPD care is due to readmissions,” she said, noting that readmissions adversely impact quality of life.

Asked if there was sufficient staff at her institution to allow for a pulmonologist consult with every COPD admission, Dr. Hekmat-Joo acknowledged that this has to be demonstrated, but compelling evidence of a benefit might prompt a redistribution of resources.

“If we can show that readmissions are substantially reduced, adding staff to perform these consults would be a good investment,” said Dr. Hekmat-Joo, indicating that improved outcomes could also attract the attention of third-party payers and those tracking quality-of-care metrics.

There is a strong rationale for a randomized prospective trial to confirm the value of a pulmonologist consultation following admission for an acute exacerbation of COPD, according to Nicola A. Hanania, MD, director, Airways Clinical Research Center, Baylor College of Medicine, Houston.

The potential for benefit as seen in this retrospective study is a rational expectation and might be related to more appropriate therapy upon discharge as well as to earlier and more rigorous follow-up, according to Dr. Hanania. Although he cautioned that there is a meaningful risk of selection bias in a retrospective study, he thinks this study “is certainly probing an important issue.”

“Mortality from a hospitalized COPD exacerbation exceeds that of a myocardial infarction,” Dr. Hanania pointed out. Noting that all patients with an MI are evaluated by a cardiologist, he sees the logic of a pulmonologist consult – although he acknowledged that evidence is needed.

“I strongly believe that a prospective study is feasible and will answer the question in an unbiased manner if done properly,” he said in an interview. If a multicenter, well-controlled study was positive, it could change practice.

In the event of a study showing major clinical benefits, particularly a reduction in mortality, “I believe it is feasible to have a pulmonary consult to see every COPD exacerbation patient admitted to the hospital,” Dr. Hanania said.

Dr. Hekmat-Joo reports no relevant financial relationships. Dr. Hanania has financial relationships with AstraZeneca, Boehringer Ingelheim, GlaxoSmithKline, Mylan, Novartis, Regeneron, Sanofi, and Sunovion.

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

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM CHEST 2022

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

Working while sick: Why doctors don’t stay home when ill

Article Type
Changed
Fri, 11/04/2022 - 13:19

Before the pandemic, physicians came to work sick, as people do in many other professions. The reasons are likely as varied as, “you weren’t feeling bad enough to miss work,” “you couldn’t afford to miss pay,” “you had too many patients to see,” or “too much work to do.”

In Medscape’s Employed Physicians Report: Loving the Focus, Hating the Bureaucracy, 61% of physicians reported that they sometimes or often come to work sick. Only 2% of respondents said they never come to work unwell.

Medscape wanted to know more about how often you call in sick, how often you come to work feeling unwell, what symptoms you have, and the dogma of your workplace culture regarding sick days. Not to mention the brutal ethos that starts in medical school, in which calling in sick shows weakness or is unacceptable.

So, we polled 2,347 physicians in the United States and abroad and asked them about their sniffling, sneezing, cold, flu, and fever symptoms, and, of course, COVID. Results were split about 50-50 among male and female physicians. The poll ran from Sept. 28 through Oct. 11.
 

Coming to work sick

It’s no surprise that the majority of physicians who were polled (85%) have come to work sick in 2022. In the last prepandemic year (2019), about 70% came to work feeling sick one to five times, and 13% worked while sick six to ten times.

When asked about the symptoms that they’ve previously come to work with, 48% of U.S. physicians said multiple symptoms. They gave high marks for runny nose, cough, congestion, and sore throat. Only 27% have worked with a fever, 22% have worked with other symptoms, and 7% have worked with both strep throat and COVID.

“My workplace, especially in the COVID years, accommodates persons who honestly do not feel well enough to report. Sooner or later, everyone covers for someone else who has to be out,” says Kenneth Abbott, MD, an oncologist in Maryland.
 

The culture of working while sick

Why doctors come to work when they’re sick is complicated. The overwhelming majority of U.S. respondents cited professional obligations; 73% noted that they feel a professional obligation to their patients, and 72% feel a professional obligation to their co-workers. Half of the polled U.S. physicians said they didn’t feel bad enough to stay home, while 48% said they had too much work to do to stay home.

Some 45% said the expectation at their workplace is to come to work unless seriously ill; 43% had too many patients to see; and 18% didn’t think they were contagious when they headed to work sick. Unfortunately, 15% chose to work while sick because otherwise they would lose pay.

In light of these responses, it’s not surprising that 93% reported they’d seen other medical professionals working when sick.

“My schedule is almost always booked weeks in advance. If someone misses or has to cancel their appointment, they typically have 2-4 weeks to wait to get back in. If I was sick and a full day of patients (or God forbid more than a day) had to be canceled because I called in, it’s so much more work when I return,” says Caitlin Briggs, MD, a psychiatrist in Lexington, Ky.
 

 

 

Doctors’ workplace sick day policy

Most employees’ benefits allow at least a few sick days, but doctors who treat society’s ill patients don’t seem to stay home from work when they’re suffering. So, we asked physicians, official policy aside, whether they thought going to work sick was expected in their workplace. The majority (76%) said yes, while 24% said no.

“Unless I’m dying or extremely contagious, I usually work. At least now, I have the telehealth option. Not saying any of this is right, but it’s the reality we deal with and the choice we must make,” says Dr. Briggs.

Additionally, 58% of polled physicians said their workplace did not have a clearly defined policy against coming to work sick, while 20% said theirs did, and 22% weren’t sure.

“The first thing I heard on the subject as a medical student was that sick people come to the hospital, so if you’re sick, then you come to the hospital too ... to work. If you can’t work, then you will be admitted. Another aphorism was from Churchill, that ‘most of the world’s work is done by people who don’t feel very well,’ ” says Paul Andreason, MD, a psychiatrist in Bethesda, Md.
 

Working in the time of COVID

Working while ill during ordinary times is one thing, but what about working in the time of COVID? Has the pandemic changed the culture of coming to work sick because medical facilities, such as doctor’s offices and hospitals, don’t want their staff coming in when they have COVID?

Surprisingly, when we asked physicians whether the pandemic has made it more or less acceptable to come to work sick, only 61% thought COVID has made it less acceptable to work while sick, while 16% thought it made it more acceptable, and 23% said there’s no change.

“I draw the line at fevers/chills, feeling like you’ve just been run over, or significant enteritis,” says Dr. Abbott. “Also, if I have to take palliative meds that interfere with alertness, I’m not doing my patients any favors.”

While a minority of physicians may call in sick, most still suffer through their sneezing, coughing, chills, and fever while seeing patients as usual.

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

Publications
Topics
Sections

Before the pandemic, physicians came to work sick, as people do in many other professions. The reasons are likely as varied as, “you weren’t feeling bad enough to miss work,” “you couldn’t afford to miss pay,” “you had too many patients to see,” or “too much work to do.”

In Medscape’s Employed Physicians Report: Loving the Focus, Hating the Bureaucracy, 61% of physicians reported that they sometimes or often come to work sick. Only 2% of respondents said they never come to work unwell.

Medscape wanted to know more about how often you call in sick, how often you come to work feeling unwell, what symptoms you have, and the dogma of your workplace culture regarding sick days. Not to mention the brutal ethos that starts in medical school, in which calling in sick shows weakness or is unacceptable.

So, we polled 2,347 physicians in the United States and abroad and asked them about their sniffling, sneezing, cold, flu, and fever symptoms, and, of course, COVID. Results were split about 50-50 among male and female physicians. The poll ran from Sept. 28 through Oct. 11.
 

Coming to work sick

It’s no surprise that the majority of physicians who were polled (85%) have come to work sick in 2022. In the last prepandemic year (2019), about 70% came to work feeling sick one to five times, and 13% worked while sick six to ten times.

When asked about the symptoms that they’ve previously come to work with, 48% of U.S. physicians said multiple symptoms. They gave high marks for runny nose, cough, congestion, and sore throat. Only 27% have worked with a fever, 22% have worked with other symptoms, and 7% have worked with both strep throat and COVID.

“My workplace, especially in the COVID years, accommodates persons who honestly do not feel well enough to report. Sooner or later, everyone covers for someone else who has to be out,” says Kenneth Abbott, MD, an oncologist in Maryland.
 

The culture of working while sick

Why doctors come to work when they’re sick is complicated. The overwhelming majority of U.S. respondents cited professional obligations; 73% noted that they feel a professional obligation to their patients, and 72% feel a professional obligation to their co-workers. Half of the polled U.S. physicians said they didn’t feel bad enough to stay home, while 48% said they had too much work to do to stay home.

Some 45% said the expectation at their workplace is to come to work unless seriously ill; 43% had too many patients to see; and 18% didn’t think they were contagious when they headed to work sick. Unfortunately, 15% chose to work while sick because otherwise they would lose pay.

In light of these responses, it’s not surprising that 93% reported they’d seen other medical professionals working when sick.

“My schedule is almost always booked weeks in advance. If someone misses or has to cancel their appointment, they typically have 2-4 weeks to wait to get back in. If I was sick and a full day of patients (or God forbid more than a day) had to be canceled because I called in, it’s so much more work when I return,” says Caitlin Briggs, MD, a psychiatrist in Lexington, Ky.
 

 

 

Doctors’ workplace sick day policy

Most employees’ benefits allow at least a few sick days, but doctors who treat society’s ill patients don’t seem to stay home from work when they’re suffering. So, we asked physicians, official policy aside, whether they thought going to work sick was expected in their workplace. The majority (76%) said yes, while 24% said no.

“Unless I’m dying or extremely contagious, I usually work. At least now, I have the telehealth option. Not saying any of this is right, but it’s the reality we deal with and the choice we must make,” says Dr. Briggs.

Additionally, 58% of polled physicians said their workplace did not have a clearly defined policy against coming to work sick, while 20% said theirs did, and 22% weren’t sure.

“The first thing I heard on the subject as a medical student was that sick people come to the hospital, so if you’re sick, then you come to the hospital too ... to work. If you can’t work, then you will be admitted. Another aphorism was from Churchill, that ‘most of the world’s work is done by people who don’t feel very well,’ ” says Paul Andreason, MD, a psychiatrist in Bethesda, Md.
 

Working in the time of COVID

Working while ill during ordinary times is one thing, but what about working in the time of COVID? Has the pandemic changed the culture of coming to work sick because medical facilities, such as doctor’s offices and hospitals, don’t want their staff coming in when they have COVID?

Surprisingly, when we asked physicians whether the pandemic has made it more or less acceptable to come to work sick, only 61% thought COVID has made it less acceptable to work while sick, while 16% thought it made it more acceptable, and 23% said there’s no change.

“I draw the line at fevers/chills, feeling like you’ve just been run over, or significant enteritis,” says Dr. Abbott. “Also, if I have to take palliative meds that interfere with alertness, I’m not doing my patients any favors.”

While a minority of physicians may call in sick, most still suffer through their sneezing, coughing, chills, and fever while seeing patients as usual.

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

Before the pandemic, physicians came to work sick, as people do in many other professions. The reasons are likely as varied as, “you weren’t feeling bad enough to miss work,” “you couldn’t afford to miss pay,” “you had too many patients to see,” or “too much work to do.”

In Medscape’s Employed Physicians Report: Loving the Focus, Hating the Bureaucracy, 61% of physicians reported that they sometimes or often come to work sick. Only 2% of respondents said they never come to work unwell.

Medscape wanted to know more about how often you call in sick, how often you come to work feeling unwell, what symptoms you have, and the dogma of your workplace culture regarding sick days. Not to mention the brutal ethos that starts in medical school, in which calling in sick shows weakness or is unacceptable.

So, we polled 2,347 physicians in the United States and abroad and asked them about their sniffling, sneezing, cold, flu, and fever symptoms, and, of course, COVID. Results were split about 50-50 among male and female physicians. The poll ran from Sept. 28 through Oct. 11.
 

Coming to work sick

It’s no surprise that the majority of physicians who were polled (85%) have come to work sick in 2022. In the last prepandemic year (2019), about 70% came to work feeling sick one to five times, and 13% worked while sick six to ten times.

When asked about the symptoms that they’ve previously come to work with, 48% of U.S. physicians said multiple symptoms. They gave high marks for runny nose, cough, congestion, and sore throat. Only 27% have worked with a fever, 22% have worked with other symptoms, and 7% have worked with both strep throat and COVID.

“My workplace, especially in the COVID years, accommodates persons who honestly do not feel well enough to report. Sooner or later, everyone covers for someone else who has to be out,” says Kenneth Abbott, MD, an oncologist in Maryland.
 

The culture of working while sick

Why doctors come to work when they’re sick is complicated. The overwhelming majority of U.S. respondents cited professional obligations; 73% noted that they feel a professional obligation to their patients, and 72% feel a professional obligation to their co-workers. Half of the polled U.S. physicians said they didn’t feel bad enough to stay home, while 48% said they had too much work to do to stay home.

Some 45% said the expectation at their workplace is to come to work unless seriously ill; 43% had too many patients to see; and 18% didn’t think they were contagious when they headed to work sick. Unfortunately, 15% chose to work while sick because otherwise they would lose pay.

In light of these responses, it’s not surprising that 93% reported they’d seen other medical professionals working when sick.

“My schedule is almost always booked weeks in advance. If someone misses or has to cancel their appointment, they typically have 2-4 weeks to wait to get back in. If I was sick and a full day of patients (or God forbid more than a day) had to be canceled because I called in, it’s so much more work when I return,” says Caitlin Briggs, MD, a psychiatrist in Lexington, Ky.
 

 

 

Doctors’ workplace sick day policy

Most employees’ benefits allow at least a few sick days, but doctors who treat society’s ill patients don’t seem to stay home from work when they’re suffering. So, we asked physicians, official policy aside, whether they thought going to work sick was expected in their workplace. The majority (76%) said yes, while 24% said no.

“Unless I’m dying or extremely contagious, I usually work. At least now, I have the telehealth option. Not saying any of this is right, but it’s the reality we deal with and the choice we must make,” says Dr. Briggs.

Additionally, 58% of polled physicians said their workplace did not have a clearly defined policy against coming to work sick, while 20% said theirs did, and 22% weren’t sure.

“The first thing I heard on the subject as a medical student was that sick people come to the hospital, so if you’re sick, then you come to the hospital too ... to work. If you can’t work, then you will be admitted. Another aphorism was from Churchill, that ‘most of the world’s work is done by people who don’t feel very well,’ ” says Paul Andreason, MD, a psychiatrist in Bethesda, Md.
 

Working in the time of COVID

Working while ill during ordinary times is one thing, but what about working in the time of COVID? Has the pandemic changed the culture of coming to work sick because medical facilities, such as doctor’s offices and hospitals, don’t want their staff coming in when they have COVID?

Surprisingly, when we asked physicians whether the pandemic has made it more or less acceptable to come to work sick, only 61% thought COVID has made it less acceptable to work while sick, while 16% thought it made it more acceptable, and 23% said there’s no change.

“I draw the line at fevers/chills, feeling like you’ve just been run over, or significant enteritis,” says Dr. Abbott. “Also, if I have to take palliative meds that interfere with alertness, I’m not doing my patients any favors.”

While a minority of physicians may call in sick, most still suffer through their sneezing, coughing, chills, and fever while seeing patients as usual.

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

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

Stroke risk rises with years of drinking in young adults

Article Type
Changed
Tue, 11/22/2022 - 11:12

Young adults who are moderate to heavy drinkers are at increased risk of suffering a stroke – and the risk goes up with more years of imbibing, a new study suggests.

“The rate of stroke among young adults has been increasing over the last few decades, and stroke in young adults causes death and serious disability,” study coauthor Eue-Keun Choi, MD, PhD, of Seoul National University, Republic of Korea, said in a statement.

“If we could prevent stroke in young adults by reducing alcohol consumption, that could potentially have a substantial impact on the health of individuals and the overall burden of stroke on society,” Dr. Choi added.

The study was published online in Neurology.

Compounding effects

Using data from a Korean national health database, the researchers identified roughly 1.5 million adults aged 20-39 years (mean age 29.5 years, 72% male) who had four consecutive annual health examinations during which they were asked about their alcohol use.

During a median follow-up of roughly 6 years, 3,153 individuals suffered a stroke.

After multivariate adjustment accounting for other factors that could affect the risk for stroke, such as hypertension, smoking, and body mass index, the risk of stroke increased steadily with the number of years of moderate to heavy drinking, defined as 105 grams or more of alcohol per week.

Compared with light drinkers or teetotalers, stroke risk increased 19% with 2 years of moderate to heavy drinking and 22% and 23%, respectively, for 3 and 4 years of moderate or heaving drinking.

The positive dose-response relationship was chiefly driven by increased risk of hemorrhagic stroke; with 2, 3 and 4 years of moderate to heavy drinking, hemorrhagic stroke risk increased 30%, 42% and 36%, respectively, relative to light/no drinking.

“Since more than 90% of the burden of stroke overall can be attributed to potentially modifiable risk factors, including alcohol consumption, and since stroke in young adults severely impacts both the individual and society by limiting their activities during their most productive years, reducing alcohol consumption should be emphasized in young adults with heavy drinking habits as part of any strategy to prevent stroke,” Dr. Choi said.

A limitation of the study is that only Korean people were included, so the results may not apply to people of other races and ethnicities, they noted. In addition, people filled out questionnaires about their alcohol consumption, which may introduce recall bias.
 

Consistent evidence

“For decades, the evidence was suggestive that a moderate amount of alcohol daily is actually beneficial – one to two drinks in men and one drink in women – at reducing major vascular outcomes,” Pierre Fayad, MD, professor, department of neurological sciences, and director of the Nebraska Stroke Center, University of Nebraska Medical Center, Omaha, said in commenting on the research.

Yet, over the past few years, some research has found no evidence of benefit with moderate alcohol intake. There is, however, “consistent evidence” that shows a detrimental effect of excessive alcohol, particularly binge drinking, especially in young adults, Dr. Fayad said.

This study, he said, “adds to the known detrimental effects of excessive alcohol intake, in increasing the risk of stroke, particularly in young adults.

“The bottom line: Young adults who usually have a low risk of stroke increase their risk significantly by heavy alcohol drinking, and Koreans are equally at risk as other populations,” Dr. Fayad said.

The study was supported by the Korea Medical Device Development Fund and the Korea National Research Foundation. Dr. Choi and Dr. Fayad report no relevant financial relationships.

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

Issue
Neurology Reviews - 30(12)
Publications
Topics
Sections

Young adults who are moderate to heavy drinkers are at increased risk of suffering a stroke – and the risk goes up with more years of imbibing, a new study suggests.

“The rate of stroke among young adults has been increasing over the last few decades, and stroke in young adults causes death and serious disability,” study coauthor Eue-Keun Choi, MD, PhD, of Seoul National University, Republic of Korea, said in a statement.

“If we could prevent stroke in young adults by reducing alcohol consumption, that could potentially have a substantial impact on the health of individuals and the overall burden of stroke on society,” Dr. Choi added.

The study was published online in Neurology.

Compounding effects

Using data from a Korean national health database, the researchers identified roughly 1.5 million adults aged 20-39 years (mean age 29.5 years, 72% male) who had four consecutive annual health examinations during which they were asked about their alcohol use.

During a median follow-up of roughly 6 years, 3,153 individuals suffered a stroke.

After multivariate adjustment accounting for other factors that could affect the risk for stroke, such as hypertension, smoking, and body mass index, the risk of stroke increased steadily with the number of years of moderate to heavy drinking, defined as 105 grams or more of alcohol per week.

Compared with light drinkers or teetotalers, stroke risk increased 19% with 2 years of moderate to heavy drinking and 22% and 23%, respectively, for 3 and 4 years of moderate or heaving drinking.

The positive dose-response relationship was chiefly driven by increased risk of hemorrhagic stroke; with 2, 3 and 4 years of moderate to heavy drinking, hemorrhagic stroke risk increased 30%, 42% and 36%, respectively, relative to light/no drinking.

“Since more than 90% of the burden of stroke overall can be attributed to potentially modifiable risk factors, including alcohol consumption, and since stroke in young adults severely impacts both the individual and society by limiting their activities during their most productive years, reducing alcohol consumption should be emphasized in young adults with heavy drinking habits as part of any strategy to prevent stroke,” Dr. Choi said.

A limitation of the study is that only Korean people were included, so the results may not apply to people of other races and ethnicities, they noted. In addition, people filled out questionnaires about their alcohol consumption, which may introduce recall bias.
 

Consistent evidence

“For decades, the evidence was suggestive that a moderate amount of alcohol daily is actually beneficial – one to two drinks in men and one drink in women – at reducing major vascular outcomes,” Pierre Fayad, MD, professor, department of neurological sciences, and director of the Nebraska Stroke Center, University of Nebraska Medical Center, Omaha, said in commenting on the research.

Yet, over the past few years, some research has found no evidence of benefit with moderate alcohol intake. There is, however, “consistent evidence” that shows a detrimental effect of excessive alcohol, particularly binge drinking, especially in young adults, Dr. Fayad said.

This study, he said, “adds to the known detrimental effects of excessive alcohol intake, in increasing the risk of stroke, particularly in young adults.

“The bottom line: Young adults who usually have a low risk of stroke increase their risk significantly by heavy alcohol drinking, and Koreans are equally at risk as other populations,” Dr. Fayad said.

The study was supported by the Korea Medical Device Development Fund and the Korea National Research Foundation. Dr. Choi and Dr. Fayad report no relevant financial relationships.

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

Young adults who are moderate to heavy drinkers are at increased risk of suffering a stroke – and the risk goes up with more years of imbibing, a new study suggests.

“The rate of stroke among young adults has been increasing over the last few decades, and stroke in young adults causes death and serious disability,” study coauthor Eue-Keun Choi, MD, PhD, of Seoul National University, Republic of Korea, said in a statement.

“If we could prevent stroke in young adults by reducing alcohol consumption, that could potentially have a substantial impact on the health of individuals and the overall burden of stroke on society,” Dr. Choi added.

The study was published online in Neurology.

Compounding effects

Using data from a Korean national health database, the researchers identified roughly 1.5 million adults aged 20-39 years (mean age 29.5 years, 72% male) who had four consecutive annual health examinations during which they were asked about their alcohol use.

During a median follow-up of roughly 6 years, 3,153 individuals suffered a stroke.

After multivariate adjustment accounting for other factors that could affect the risk for stroke, such as hypertension, smoking, and body mass index, the risk of stroke increased steadily with the number of years of moderate to heavy drinking, defined as 105 grams or more of alcohol per week.

Compared with light drinkers or teetotalers, stroke risk increased 19% with 2 years of moderate to heavy drinking and 22% and 23%, respectively, for 3 and 4 years of moderate or heaving drinking.

The positive dose-response relationship was chiefly driven by increased risk of hemorrhagic stroke; with 2, 3 and 4 years of moderate to heavy drinking, hemorrhagic stroke risk increased 30%, 42% and 36%, respectively, relative to light/no drinking.

“Since more than 90% of the burden of stroke overall can be attributed to potentially modifiable risk factors, including alcohol consumption, and since stroke in young adults severely impacts both the individual and society by limiting their activities during their most productive years, reducing alcohol consumption should be emphasized in young adults with heavy drinking habits as part of any strategy to prevent stroke,” Dr. Choi said.

A limitation of the study is that only Korean people were included, so the results may not apply to people of other races and ethnicities, they noted. In addition, people filled out questionnaires about their alcohol consumption, which may introduce recall bias.
 

Consistent evidence

“For decades, the evidence was suggestive that a moderate amount of alcohol daily is actually beneficial – one to two drinks in men and one drink in women – at reducing major vascular outcomes,” Pierre Fayad, MD, professor, department of neurological sciences, and director of the Nebraska Stroke Center, University of Nebraska Medical Center, Omaha, said in commenting on the research.

Yet, over the past few years, some research has found no evidence of benefit with moderate alcohol intake. There is, however, “consistent evidence” that shows a detrimental effect of excessive alcohol, particularly binge drinking, especially in young adults, Dr. Fayad said.

This study, he said, “adds to the known detrimental effects of excessive alcohol intake, in increasing the risk of stroke, particularly in young adults.

“The bottom line: Young adults who usually have a low risk of stroke increase their risk significantly by heavy alcohol drinking, and Koreans are equally at risk as other populations,” Dr. Fayad said.

The study was supported by the Korea Medical Device Development Fund and the Korea National Research Foundation. Dr. Choi and Dr. Fayad report no relevant financial relationships.

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

Issue
Neurology Reviews - 30(12)
Issue
Neurology Reviews - 30(12)
Publications
Publications
Topics
Article Type
Sections
Article Source

FROM NEUROLOGY

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