Galcanezumab effective and safe in episodic migraine

Article Type
Changed
Wed, 09/07/2022 - 20:59

Key clinical point: A dose of 120 mg galcanezumab monthly was effective and well tolerated in patients with episodic migraine.

 

Major finding: The reduction in mean monthly migraine headache days (MMHD) over 3 months was significantly higher with galcanezumab vs placebo (least squares mean change 3.81 vs 1.99 days; P < .0001), with a higher proportion of patients receiving galcanezumab vs placebo achieving ≥50%, ≥75%, and 100% reductions in MMHD (all P < .0001). The occurrence of serious adverse events was low, with none leading to treatment discontinuation.

 

Study details: Findings are from the phase 3, PERSIST trial including 520 patients with episodic migraine who were randomly assigned to receive monthly 120 mg galcanezumab or placebo.

 

Disclosures: This study was funded by Eli Lilly and Company. J Zhuang reported being a full-time employee, and 8 authors reported receiving clinical research fees from Eli Lilly. S Yu reported serving as an associate editor for the Journal of Headache and Pain.

 

Source: Hu B et al. Galcanezumab in episodic migraine: The phase 3, randomized, double-blind, placebo-controlled PERSIST study. J Headache Pain. 2022;23:90 (Jul 28). Doi: 10.1186/s10194-022-01458-0

Publications
Topics
Sections

Key clinical point: A dose of 120 mg galcanezumab monthly was effective and well tolerated in patients with episodic migraine.

 

Major finding: The reduction in mean monthly migraine headache days (MMHD) over 3 months was significantly higher with galcanezumab vs placebo (least squares mean change 3.81 vs 1.99 days; P < .0001), with a higher proportion of patients receiving galcanezumab vs placebo achieving ≥50%, ≥75%, and 100% reductions in MMHD (all P < .0001). The occurrence of serious adverse events was low, with none leading to treatment discontinuation.

 

Study details: Findings are from the phase 3, PERSIST trial including 520 patients with episodic migraine who were randomly assigned to receive monthly 120 mg galcanezumab or placebo.

 

Disclosures: This study was funded by Eli Lilly and Company. J Zhuang reported being a full-time employee, and 8 authors reported receiving clinical research fees from Eli Lilly. S Yu reported serving as an associate editor for the Journal of Headache and Pain.

 

Source: Hu B et al. Galcanezumab in episodic migraine: The phase 3, randomized, double-blind, placebo-controlled PERSIST study. J Headache Pain. 2022;23:90 (Jul 28). Doi: 10.1186/s10194-022-01458-0

Key clinical point: A dose of 120 mg galcanezumab monthly was effective and well tolerated in patients with episodic migraine.

 

Major finding: The reduction in mean monthly migraine headache days (MMHD) over 3 months was significantly higher with galcanezumab vs placebo (least squares mean change 3.81 vs 1.99 days; P < .0001), with a higher proportion of patients receiving galcanezumab vs placebo achieving ≥50%, ≥75%, and 100% reductions in MMHD (all P < .0001). The occurrence of serious adverse events was low, with none leading to treatment discontinuation.

 

Study details: Findings are from the phase 3, PERSIST trial including 520 patients with episodic migraine who were randomly assigned to receive monthly 120 mg galcanezumab or placebo.

 

Disclosures: This study was funded by Eli Lilly and Company. J Zhuang reported being a full-time employee, and 8 authors reported receiving clinical research fees from Eli Lilly. S Yu reported serving as an associate editor for the Journal of Headache and Pain.

 

Source: Hu B et al. Galcanezumab in episodic migraine: The phase 3, randomized, double-blind, placebo-controlled PERSIST study. J Headache Pain. 2022;23:90 (Jul 28). Doi: 10.1186/s10194-022-01458-0

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan Commentary: Migraine September 2022
Gate On Date
Thu, 02/24/2022 - 17:15
Un-Gate On Date
Thu, 02/24/2022 - 17:15
Use ProPublica
CFC Schedule Remove Status
Thu, 02/24/2022 - 17:15
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Toenail trauma

Article Type
Changed
Mon, 09/19/2022 - 10:37
Display Headline
Toenail trauma

Toenail trauma

The patient’s initial injury was probably a subungual hematoma, which can take 12 to 18 months to resolve (the time it takes for a new toenail to grow). However, the precipitating trauma likely created an opportunity for fungal elements to invade the nail plate, resulting in the current complaint of superficial onychomycosis.

Onychomycosis is a frequently seen condition with increasing prevalence in older patients. It has several clinical presentations: Superficial onychomycosis manifests with chalky white changes on the surface of the nail. Distal subungual onychomycosis develops at the distal aspect of the nail with thickening and subungual debris. Proximal subungual onychomycosis occurs in the proximal aspect of the nail.

Although often asymptomatic, onychomycosis can cause thickening of the nails and development of subsequent deformity or pincer nails (which painfully “pinch” the underlying skin). It is especially concerning in patients with diabetes or peripheral neuropathy, in whom the abnormal thickness and shape of the nails can lead to microtrauma at the proximal and lateral attachments of the nail. These patients have an increased risk of secondary infection, possible complications, and even, for some, amputation.

If the patient is asymptomatic, and does not have diabetes, neuropathy, or other risk factors, treatment is not required. For those who would benefit from treatment, it is usually safe and inexpensive with the current generation of oral antifungal medications.

Some recommend confirmatory testing before treatment intiation,1 but the low adverse effect profile of terbinafine and its current cost below $10/month2 make empiric treatment safe and cost effective in most cases.3 If needed, and with access to microscopy, a potassium hydroxide (KOH) prep can be performed on scrapings from the affected portions of the nail. If that is not available, scrapings or clippings can be sent to the lab for KOH and periodic acid-Schiff staining.

The US Food and Drug Administration previously recommended follow-up liver enzyme tests if terbinafine is used for more than 6 weeks. (Fingernails require only 6 weeks of treatment, but toenails grow more slowly and require 12 weeks of treatment.) However, research has demonstrated that hepatotoxicity risk is extremely low and transaminase elevations are rare.4 In the rare cases that liver dysfunction has occurred, patients developed symptoms of jaundice, malaise, dark urine, or pruritis.4

This patient was counseled regarding the fungal nature of onychomycosis and the general safety of a 90-day course of oral terbinafine 250 mg/d—provided he did not have underlying liver or kidney disease or leukopenia. He reported that he had not had any blood work performed in the past year but was due for his annual wellness evaluation, at which he would discuss his overall health with his primary care provider, obtain baseline blood testing, and determine whether to proceed with treatment. He was advised that if, after starting treatment, he developed any symptoms of jaundice, dark urine, or other difficulties, he should report them to his care team.

Photo courtesy of Daniel Stulberg, MD. Text courtesy of Daniel Stulberg, MD, FAAFP, Department of Family and Community Medicine, University of New Mexico School of Medicine, Albuquerque.

References
  1. Frazier WT, Santiago-Delgado ZM, Stupka KC 2nd. Onychomycosis: rapid evidence review. Am Fam Physician. 2021;104:359-367.
  2. Terbinafine. GoodRx. Accessed August 9, 2022. https://www.goodrx.com/terbinafine
  3. Mikailov A, Cohen J, Joyce C, et al. Cost-effectiveness of confirmatory testing before treatment of onychomycosis. JAMA Dermatol. 2016;152:276-281. doi: 10.1001/jamadermatol.2015.4190
  4. Sun CW, Hsu S. Terbinafine: safety profile and monitoring in treatment of dermatophyte infections. Dermatol Ther. 2019;32:e13111. doi: 10.1111/dth.13111
Issue
The Journal of Family Practice - 71(6)
Publications
Topics
Sections

Toenail trauma

The patient’s initial injury was probably a subungual hematoma, which can take 12 to 18 months to resolve (the time it takes for a new toenail to grow). However, the precipitating trauma likely created an opportunity for fungal elements to invade the nail plate, resulting in the current complaint of superficial onychomycosis.

Onychomycosis is a frequently seen condition with increasing prevalence in older patients. It has several clinical presentations: Superficial onychomycosis manifests with chalky white changes on the surface of the nail. Distal subungual onychomycosis develops at the distal aspect of the nail with thickening and subungual debris. Proximal subungual onychomycosis occurs in the proximal aspect of the nail.

Although often asymptomatic, onychomycosis can cause thickening of the nails and development of subsequent deformity or pincer nails (which painfully “pinch” the underlying skin). It is especially concerning in patients with diabetes or peripheral neuropathy, in whom the abnormal thickness and shape of the nails can lead to microtrauma at the proximal and lateral attachments of the nail. These patients have an increased risk of secondary infection, possible complications, and even, for some, amputation.

If the patient is asymptomatic, and does not have diabetes, neuropathy, or other risk factors, treatment is not required. For those who would benefit from treatment, it is usually safe and inexpensive with the current generation of oral antifungal medications.

Some recommend confirmatory testing before treatment intiation,1 but the low adverse effect profile of terbinafine and its current cost below $10/month2 make empiric treatment safe and cost effective in most cases.3 If needed, and with access to microscopy, a potassium hydroxide (KOH) prep can be performed on scrapings from the affected portions of the nail. If that is not available, scrapings or clippings can be sent to the lab for KOH and periodic acid-Schiff staining.

The US Food and Drug Administration previously recommended follow-up liver enzyme tests if terbinafine is used for more than 6 weeks. (Fingernails require only 6 weeks of treatment, but toenails grow more slowly and require 12 weeks of treatment.) However, research has demonstrated that hepatotoxicity risk is extremely low and transaminase elevations are rare.4 In the rare cases that liver dysfunction has occurred, patients developed symptoms of jaundice, malaise, dark urine, or pruritis.4

This patient was counseled regarding the fungal nature of onychomycosis and the general safety of a 90-day course of oral terbinafine 250 mg/d—provided he did not have underlying liver or kidney disease or leukopenia. He reported that he had not had any blood work performed in the past year but was due for his annual wellness evaluation, at which he would discuss his overall health with his primary care provider, obtain baseline blood testing, and determine whether to proceed with treatment. He was advised that if, after starting treatment, he developed any symptoms of jaundice, dark urine, or other difficulties, he should report them to his care team.

Photo courtesy of Daniel Stulberg, MD. Text courtesy of Daniel Stulberg, MD, FAAFP, Department of Family and Community Medicine, University of New Mexico School of Medicine, Albuquerque.

Toenail trauma

The patient’s initial injury was probably a subungual hematoma, which can take 12 to 18 months to resolve (the time it takes for a new toenail to grow). However, the precipitating trauma likely created an opportunity for fungal elements to invade the nail plate, resulting in the current complaint of superficial onychomycosis.

Onychomycosis is a frequently seen condition with increasing prevalence in older patients. It has several clinical presentations: Superficial onychomycosis manifests with chalky white changes on the surface of the nail. Distal subungual onychomycosis develops at the distal aspect of the nail with thickening and subungual debris. Proximal subungual onychomycosis occurs in the proximal aspect of the nail.

Although often asymptomatic, onychomycosis can cause thickening of the nails and development of subsequent deformity or pincer nails (which painfully “pinch” the underlying skin). It is especially concerning in patients with diabetes or peripheral neuropathy, in whom the abnormal thickness and shape of the nails can lead to microtrauma at the proximal and lateral attachments of the nail. These patients have an increased risk of secondary infection, possible complications, and even, for some, amputation.

If the patient is asymptomatic, and does not have diabetes, neuropathy, or other risk factors, treatment is not required. For those who would benefit from treatment, it is usually safe and inexpensive with the current generation of oral antifungal medications.

Some recommend confirmatory testing before treatment intiation,1 but the low adverse effect profile of terbinafine and its current cost below $10/month2 make empiric treatment safe and cost effective in most cases.3 If needed, and with access to microscopy, a potassium hydroxide (KOH) prep can be performed on scrapings from the affected portions of the nail. If that is not available, scrapings or clippings can be sent to the lab for KOH and periodic acid-Schiff staining.

The US Food and Drug Administration previously recommended follow-up liver enzyme tests if terbinafine is used for more than 6 weeks. (Fingernails require only 6 weeks of treatment, but toenails grow more slowly and require 12 weeks of treatment.) However, research has demonstrated that hepatotoxicity risk is extremely low and transaminase elevations are rare.4 In the rare cases that liver dysfunction has occurred, patients developed symptoms of jaundice, malaise, dark urine, or pruritis.4

This patient was counseled regarding the fungal nature of onychomycosis and the general safety of a 90-day course of oral terbinafine 250 mg/d—provided he did not have underlying liver or kidney disease or leukopenia. He reported that he had not had any blood work performed in the past year but was due for his annual wellness evaluation, at which he would discuss his overall health with his primary care provider, obtain baseline blood testing, and determine whether to proceed with treatment. He was advised that if, after starting treatment, he developed any symptoms of jaundice, dark urine, or other difficulties, he should report them to his care team.

Photo courtesy of Daniel Stulberg, MD. Text courtesy of Daniel Stulberg, MD, FAAFP, Department of Family and Community Medicine, University of New Mexico School of Medicine, Albuquerque.

References
  1. Frazier WT, Santiago-Delgado ZM, Stupka KC 2nd. Onychomycosis: rapid evidence review. Am Fam Physician. 2021;104:359-367.
  2. Terbinafine. GoodRx. Accessed August 9, 2022. https://www.goodrx.com/terbinafine
  3. Mikailov A, Cohen J, Joyce C, et al. Cost-effectiveness of confirmatory testing before treatment of onychomycosis. JAMA Dermatol. 2016;152:276-281. doi: 10.1001/jamadermatol.2015.4190
  4. Sun CW, Hsu S. Terbinafine: safety profile and monitoring in treatment of dermatophyte infections. Dermatol Ther. 2019;32:e13111. doi: 10.1111/dth.13111
References
  1. Frazier WT, Santiago-Delgado ZM, Stupka KC 2nd. Onychomycosis: rapid evidence review. Am Fam Physician. 2021;104:359-367.
  2. Terbinafine. GoodRx. Accessed August 9, 2022. https://www.goodrx.com/terbinafine
  3. Mikailov A, Cohen J, Joyce C, et al. Cost-effectiveness of confirmatory testing before treatment of onychomycosis. JAMA Dermatol. 2016;152:276-281. doi: 10.1001/jamadermatol.2015.4190
  4. Sun CW, Hsu S. Terbinafine: safety profile and monitoring in treatment of dermatophyte infections. Dermatol Ther. 2019;32:e13111. doi: 10.1111/dth.13111
Issue
The Journal of Family Practice - 71(6)
Issue
The Journal of Family Practice - 71(6)
Publications
Publications
Topics
Article Type
Display Headline
Toenail trauma
Display Headline
Toenail trauma
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 08/18/2022 - 08:45
Un-Gate On Date
Thu, 08/18/2022 - 08:45
Use ProPublica
CFC Schedule Remove Status
Thu, 08/18/2022 - 08:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Spotted white fingernails

Article Type
Changed
Wed, 08/31/2022 - 09:00
Display Headline
Spotted white fingernails

Spotted white fingernails

White nail changes are broadly called leukonychia: “leuko” meaning white and “nychia” referring to the nail. Scattered or single asymptomatic cloudy white nail lesions occurring without other associated skin or nail disorders are more specifically called punctate leukonychia.

Punctate leukonychia is theorized to be caused by trauma at the proximal nail matrix, affecting the developing nail.1 The trauma may result from aggressive nail care practices or damage to the cuticle. In many cases, there is no history of known trauma. For this patient with multiple lesions, who performed manual work, multiple small traumas may have induced the punctate leukonychia.

Other causes of leukonychia include superficial onychomycosis (in which discoloration may be whiter than the usual yellow-brown), renal disease, and arsenic toxicity.1 Arsenic toxicity causes transverse leukonychia in a band-like fashion, since it is a systemic insult to the growing nails. Longitudinal leukonychia is due to a more localized insult to the nail matrix, causing the white lines to grow out with the nail along the axis of the digit. Other than avoiding trauma, there is no treatment needed or recommended for punctate leukonychia.

The patient was counseled on the benign nature of his punctate leukonychia and assured that no treatment was necessary.

Photo courtesy of Daniel Stulberg, MD. Text courtesy of Daniel Stulberg, MD, FAAFP, Department of Family and Community Medicine, University of New Mexico School of Medicine, Albuquerque.

References

1. Iorizzo M, Starace M, Pasch MC. Leukonychia: what can white nails tell us? Am J Clin Dermatol. 2022;23:177-193. doi: 10.1007/s40257-022-00671-6

Issue
The Journal of Family Practice - 71(6)
Publications
Topics
Sections

Spotted white fingernails

White nail changes are broadly called leukonychia: “leuko” meaning white and “nychia” referring to the nail. Scattered or single asymptomatic cloudy white nail lesions occurring without other associated skin or nail disorders are more specifically called punctate leukonychia.

Punctate leukonychia is theorized to be caused by trauma at the proximal nail matrix, affecting the developing nail.1 The trauma may result from aggressive nail care practices or damage to the cuticle. In many cases, there is no history of known trauma. For this patient with multiple lesions, who performed manual work, multiple small traumas may have induced the punctate leukonychia.

Other causes of leukonychia include superficial onychomycosis (in which discoloration may be whiter than the usual yellow-brown), renal disease, and arsenic toxicity.1 Arsenic toxicity causes transverse leukonychia in a band-like fashion, since it is a systemic insult to the growing nails. Longitudinal leukonychia is due to a more localized insult to the nail matrix, causing the white lines to grow out with the nail along the axis of the digit. Other than avoiding trauma, there is no treatment needed or recommended for punctate leukonychia.

The patient was counseled on the benign nature of his punctate leukonychia and assured that no treatment was necessary.

Photo courtesy of Daniel Stulberg, MD. Text courtesy of Daniel Stulberg, MD, FAAFP, Department of Family and Community Medicine, University of New Mexico School of Medicine, Albuquerque.

Spotted white fingernails

White nail changes are broadly called leukonychia: “leuko” meaning white and “nychia” referring to the nail. Scattered or single asymptomatic cloudy white nail lesions occurring without other associated skin or nail disorders are more specifically called punctate leukonychia.

Punctate leukonychia is theorized to be caused by trauma at the proximal nail matrix, affecting the developing nail.1 The trauma may result from aggressive nail care practices or damage to the cuticle. In many cases, there is no history of known trauma. For this patient with multiple lesions, who performed manual work, multiple small traumas may have induced the punctate leukonychia.

Other causes of leukonychia include superficial onychomycosis (in which discoloration may be whiter than the usual yellow-brown), renal disease, and arsenic toxicity.1 Arsenic toxicity causes transverse leukonychia in a band-like fashion, since it is a systemic insult to the growing nails. Longitudinal leukonychia is due to a more localized insult to the nail matrix, causing the white lines to grow out with the nail along the axis of the digit. Other than avoiding trauma, there is no treatment needed or recommended for punctate leukonychia.

The patient was counseled on the benign nature of his punctate leukonychia and assured that no treatment was necessary.

Photo courtesy of Daniel Stulberg, MD. Text courtesy of Daniel Stulberg, MD, FAAFP, Department of Family and Community Medicine, University of New Mexico School of Medicine, Albuquerque.

References

1. Iorizzo M, Starace M, Pasch MC. Leukonychia: what can white nails tell us? Am J Clin Dermatol. 2022;23:177-193. doi: 10.1007/s40257-022-00671-6

References

1. Iorizzo M, Starace M, Pasch MC. Leukonychia: what can white nails tell us? Am J Clin Dermatol. 2022;23:177-193. doi: 10.1007/s40257-022-00671-6

Issue
The Journal of Family Practice - 71(6)
Issue
The Journal of Family Practice - 71(6)
Publications
Publications
Topics
Article Type
Display Headline
Spotted white fingernails
Display Headline
Spotted white fingernails
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Thu, 08/18/2022 - 07:45
Un-Gate On Date
Thu, 08/18/2022 - 07:45
Use ProPublica
CFC Schedule Remove Status
Thu, 08/18/2022 - 07:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Sun Protection Factor Testing: A Call for an In Vitro Method

Article Type
Changed
Thu, 08/18/2022 - 10:33
Display Headline
Sun Protection Factor Testing: A Call for an In Vitro Method

The sun protection factor (SPF) value indicates to consumers the level of protection that a given sunscreen formulation provides against erythemally effective UV radiation (UVR). 1 In vivo SPF testing, the gold standard for determining SPF, yields highly variable results and can harm human test participants. 2 In vitro SPF testing methodologies have been under development for years but none have (yet) replaced the in vivo test required by national and international regulatory agencies.

Recent European studies have shown strong data to support a highly standardized in vitro method,1 now under development by the International Organization for Standardization (ISO)—potentially to serve as a new SPF determination standard.1,3 Academia and industry should follow this example and actively take steps to develop and validate a suitable replacement for in vivo SPF testing.

In Vivo SPF Testing

The in vivo SPF test involves comparing doses of UVR necessary to induce erythema in human participants with and without sunscreen applied.2 Although this method has long been the standard for SPF determination, it is associated with the following major disadvantages:

  • Cost: The in vivo test is expensive.
  • Variability: Results of the test are subject to high interlaboratory variability due to the inherent subjectivity of identifying erythema, the variable skin types of human participants, and other laboratory-dependent factors.2 A study found that the average coefficient of variation for SPF values obtained from 3 or 4 laboratories to be 20%—with values exceeding 50% in some cases. With that level of variability, the same sunscreen may be labeled SPF 30, SPF 50, or SPF 50+, thereby posing a health risk to consumers who rely on the accuracy of such claims. In fact, Miksa et al2 concluded that “the largest obstacle to a reliable SPF assessment for consumer health is the in vivo SPF test itself.”
  • Ethical concerns: Human participants are intentionally exposed to harmful UVR until sunburn is achieved. For that reason, there have been calls to abandon the practice of in vivo testing.1

Alternatives to In Vivo SPF Testing

There has been international interest in developing in silico and in vitro alternatives to the in vivo SPF test. These options are attractive because they are relatively inexpensive; avoid exposing human participants to harmful UVR; and have the potential to be more accurate and more reproducible than in vivo tests.

In Vitro Protocols—Many such in vitro tests exist; all generally involve applying a layer of sunscreen to an artificial substrate, exposing it to UVR from a solar simulator, and measuring the UVR transmittance through the product and film by spectrophotometry.1 Prior shortcomings of this method have included suboptimal reproducibility, lack of data on substrate and product properties, and lack of demonstrated equivalency to in vivo SPF testing.4

In Silico Protocols—These tests use data on the UV spectra of sunscreen filters, physical characteristics of sunscreen films on skin, and the unique photoinstability of filters to calculate expected UVR transmittance and SPF of sunscreens based on their ingredients.5 Reports have shown high correlation with in vivo values. Results are not subject to random error; reproducibility is theoretically perfect.5

Regulatory Agencies and In Vitro Testing

In the United States, sunscreens are regulated as over-the-counter drugs. In vivo testing is the only US Food and Drug Administration (FDA)–approved method for determining SPF for labeling purposes.1 In a 2007 Proposed Rule and a 2011 Final Rule, the FDA stated that in vitro SPF tests were an inadequate alternative to in vivo tests because of their shortcomings.4,6

 

 

Acknowledging the potential benefits of in vitro testing, the FDA wrote that it would consider in vitro alternatives if equivalency to the in vivo test could be proved.6 The agency has not published an official stance on in vitro SPF testing since those statements in 2007 and 2011. Of note, the FDA deems in vitro testing sufficient for making claims of broad-spectrum coverage.4

In contrast to the regulatory scenario in the United States, Europe regulates sunscreens as cosmetics, and the European Union (EU) has banned animal testing of cosmetics,7 which poses a problem for the development of new sunscreens. It is not surprising, therefore, that in 2006 the European Commission (the executive arm of the EU) published a mandate that in vitro SPF testing methods be actively developed due to ethical concerns associated with in vivo methods.8 In 2017, the International Organization for Standardization released specific validation criteria for proposed in vitro tests to facilitate the eventual approval of such methods.1

Progress of In Vitro Methods

In recent years, advances in in vitro SPF testing methods have addressed shortcomings noted previously by the FDA, which has led to notably improved reproducibility of results and correlation with in vivo values, in large part due to strict standardization of protocols,1 such as tight temperature control of samples, a multisubstrate approach, robotic product application to ensure even distribution, and pre-irradiation of sunscreen samples.

With these improvements, a 2018 study demonstrated an in vitro SPF testing methodology that exceeded published ISO validation criteria for emulsion-type products.1 This method was found to have low interlaboratory variability and high correlation with in vivo SPF values (Pearson r=0.88). Importantly, the authors noted that the consistency and reliability of in vitro SPF testing requires broad institution of a single unified method.1

The method described in the 2018 study1 has been accepted by the ISO Technical Committee and is undergoing further development3; it is expected to be approved by the European Committee for Standardization. After approval, adoption by member nations of the EU will require individual action, representing the next regulatory hurdle for in vitro SPF testing in Europe.

Final Thoughts and Future Steps

Recent data confirm the potential viability of in vitro testing as a primary method of determining SPF values.1 Although ISO has moved forward with development of this method, the FDA has been quiet on in vitro SPF testing since 2011.4 The agency has, however, acknowledged the disadvantages of in vivo broad-spectrum testing, including exposure of human participants to harmful UVR and poor interlaboratory reproducibility.6

Given the technical developments and substantial potential benefits of in vitro testing, we believe that it is time for the FDA to revisit this matter. We propose that the FDA take 2 steps toward in vitro testing. First, publish specific validation criteria that would be deemed necessary for approval of such a test, similar to what ISO published in 2017. Second, thoroughly assess new data supporting the viability of available in vitro testing to determine if the FDA’s stated position that in vitro testing is inadequate remains true.

 

 

Although these 2 steps will be important to the process, adoption of an in vitro standard will require more than statements from the FDA. Additional funding should be allocated to researchers who are studying in vitro methodologies, and companies that profit from the multibillion-dollar sunscreen industry should be encouraged to invest in the development of more accurate and more ethical alternatives to in vivo SPF testing.

In vitro SPF testing is inexpensive, avoids the moral quandary of intentionally sunburning human participants, and is more reliable than in vivo testing. It is time for the FDA to facilitate the efforts of academia and industry in taking concrete steps toward approval of an in vitro alternative to in vivo SPF testing.

References
  1. Pissavini M, Tricaud C, Wiener G, et al. Validation of an in vitro sun protection factor (SPF) method in blinded ring-testing. Int J Cosmet Sci. 2018;40:263-268. doi:10.1111/ics.12459
  2. Miksa S, Lutz D, Guy C, et al. Sunscreen sun protection factor claim based on in vivo interlaboratory variability. Int J Cosmet Sci. 2016;38:541-549. doi:10.1111/ics.12333
  3. ISO/CD 23675: Cosmetics—sun protection test methods—in vitro determination of sun protection factor. International Organization for Standardization (ISO). July 25, 2020. Accessed May 17, 2022. https://www.iso.org/standard/76616.html
  4. US Food and Drug Administration. Labeling and effectiveness testing; sunscreen drug products for over-the-counter human use. Fed Regist. 2011;76(117):35620-35665. Accessed August 9, 2022. https://www.govinfo.gov/content/pkg/FR-2011-06-17/pdf/2011-14766.pdf
  5. Herzog B, Osterwalder U. Simulation of sunscreen performance. Pure Appl Chem. 2015;87:937-951. doi:10.1515/pac-2015-0401
  6. US Food and Drug Administration. Sunscreen drug products for over-the-counter human use; proposed amendment of final monograph. Fed Regist. 2007;72(165):49070-49122. Published August 27, 2007. Accessed August 9, 2022. https://www.govinfo.gov/content/pkg/FR-2007-08-27/pdf/07-4131.pdf
  7. Regulation (EC) No 1223/2009 of the European Parliament and of the Council of 30 November 2009 on cosmetic products. November 30, 2009. Accessed August 10, 2022. https://eur-lex.europa.eu/legal-content/EN/TXT/?uri=CELEX:02009R1223-20190813
  8. European Commission Recommendation 2006/647/EC. Published September 22, 2006. Accessed August 10, 2022. http://eur-lex.europa.eu/legal-content/EN/TXT/?uri=CELEX%3A32006H0647
Article PDF
Author and Disclosure Information

Dr. Breneman is from the Vagelos College of Physicians & Surgeons, Columbia University, New York, New York. Dr. Belsito is from the Department of Dermatology, Columbia University Irving Medical Center, New York.

Dr. Breneman reports no conflict of interest. Dr. Belsito has received income from Arcutis Biotherapeutics.

Correspondence: Alyssa Breneman, MD, Vagelos College of Physicians & Surgeons, 161 Fort Washington Ave, 12th Floor, New York, NY 10032 ([email protected]).

Issue
Cutis - 110(2)
Publications
Topics
Page Number
E15-E17
Sections
Author and Disclosure Information

Dr. Breneman is from the Vagelos College of Physicians & Surgeons, Columbia University, New York, New York. Dr. Belsito is from the Department of Dermatology, Columbia University Irving Medical Center, New York.

Dr. Breneman reports no conflict of interest. Dr. Belsito has received income from Arcutis Biotherapeutics.

Correspondence: Alyssa Breneman, MD, Vagelos College of Physicians & Surgeons, 161 Fort Washington Ave, 12th Floor, New York, NY 10032 ([email protected]).

Author and Disclosure Information

Dr. Breneman is from the Vagelos College of Physicians & Surgeons, Columbia University, New York, New York. Dr. Belsito is from the Department of Dermatology, Columbia University Irving Medical Center, New York.

Dr. Breneman reports no conflict of interest. Dr. Belsito has received income from Arcutis Biotherapeutics.

Correspondence: Alyssa Breneman, MD, Vagelos College of Physicians & Surgeons, 161 Fort Washington Ave, 12th Floor, New York, NY 10032 ([email protected]).

Article PDF
Article PDF

The sun protection factor (SPF) value indicates to consumers the level of protection that a given sunscreen formulation provides against erythemally effective UV radiation (UVR). 1 In vivo SPF testing, the gold standard for determining SPF, yields highly variable results and can harm human test participants. 2 In vitro SPF testing methodologies have been under development for years but none have (yet) replaced the in vivo test required by national and international regulatory agencies.

Recent European studies have shown strong data to support a highly standardized in vitro method,1 now under development by the International Organization for Standardization (ISO)—potentially to serve as a new SPF determination standard.1,3 Academia and industry should follow this example and actively take steps to develop and validate a suitable replacement for in vivo SPF testing.

In Vivo SPF Testing

The in vivo SPF test involves comparing doses of UVR necessary to induce erythema in human participants with and without sunscreen applied.2 Although this method has long been the standard for SPF determination, it is associated with the following major disadvantages:

  • Cost: The in vivo test is expensive.
  • Variability: Results of the test are subject to high interlaboratory variability due to the inherent subjectivity of identifying erythema, the variable skin types of human participants, and other laboratory-dependent factors.2 A study found that the average coefficient of variation for SPF values obtained from 3 or 4 laboratories to be 20%—with values exceeding 50% in some cases. With that level of variability, the same sunscreen may be labeled SPF 30, SPF 50, or SPF 50+, thereby posing a health risk to consumers who rely on the accuracy of such claims. In fact, Miksa et al2 concluded that “the largest obstacle to a reliable SPF assessment for consumer health is the in vivo SPF test itself.”
  • Ethical concerns: Human participants are intentionally exposed to harmful UVR until sunburn is achieved. For that reason, there have been calls to abandon the practice of in vivo testing.1

Alternatives to In Vivo SPF Testing

There has been international interest in developing in silico and in vitro alternatives to the in vivo SPF test. These options are attractive because they are relatively inexpensive; avoid exposing human participants to harmful UVR; and have the potential to be more accurate and more reproducible than in vivo tests.

In Vitro Protocols—Many such in vitro tests exist; all generally involve applying a layer of sunscreen to an artificial substrate, exposing it to UVR from a solar simulator, and measuring the UVR transmittance through the product and film by spectrophotometry.1 Prior shortcomings of this method have included suboptimal reproducibility, lack of data on substrate and product properties, and lack of demonstrated equivalency to in vivo SPF testing.4

In Silico Protocols—These tests use data on the UV spectra of sunscreen filters, physical characteristics of sunscreen films on skin, and the unique photoinstability of filters to calculate expected UVR transmittance and SPF of sunscreens based on their ingredients.5 Reports have shown high correlation with in vivo values. Results are not subject to random error; reproducibility is theoretically perfect.5

Regulatory Agencies and In Vitro Testing

In the United States, sunscreens are regulated as over-the-counter drugs. In vivo testing is the only US Food and Drug Administration (FDA)–approved method for determining SPF for labeling purposes.1 In a 2007 Proposed Rule and a 2011 Final Rule, the FDA stated that in vitro SPF tests were an inadequate alternative to in vivo tests because of their shortcomings.4,6

 

 

Acknowledging the potential benefits of in vitro testing, the FDA wrote that it would consider in vitro alternatives if equivalency to the in vivo test could be proved.6 The agency has not published an official stance on in vitro SPF testing since those statements in 2007 and 2011. Of note, the FDA deems in vitro testing sufficient for making claims of broad-spectrum coverage.4

In contrast to the regulatory scenario in the United States, Europe regulates sunscreens as cosmetics, and the European Union (EU) has banned animal testing of cosmetics,7 which poses a problem for the development of new sunscreens. It is not surprising, therefore, that in 2006 the European Commission (the executive arm of the EU) published a mandate that in vitro SPF testing methods be actively developed due to ethical concerns associated with in vivo methods.8 In 2017, the International Organization for Standardization released specific validation criteria for proposed in vitro tests to facilitate the eventual approval of such methods.1

Progress of In Vitro Methods

In recent years, advances in in vitro SPF testing methods have addressed shortcomings noted previously by the FDA, which has led to notably improved reproducibility of results and correlation with in vivo values, in large part due to strict standardization of protocols,1 such as tight temperature control of samples, a multisubstrate approach, robotic product application to ensure even distribution, and pre-irradiation of sunscreen samples.

With these improvements, a 2018 study demonstrated an in vitro SPF testing methodology that exceeded published ISO validation criteria for emulsion-type products.1 This method was found to have low interlaboratory variability and high correlation with in vivo SPF values (Pearson r=0.88). Importantly, the authors noted that the consistency and reliability of in vitro SPF testing requires broad institution of a single unified method.1

The method described in the 2018 study1 has been accepted by the ISO Technical Committee and is undergoing further development3; it is expected to be approved by the European Committee for Standardization. After approval, adoption by member nations of the EU will require individual action, representing the next regulatory hurdle for in vitro SPF testing in Europe.

Final Thoughts and Future Steps

Recent data confirm the potential viability of in vitro testing as a primary method of determining SPF values.1 Although ISO has moved forward with development of this method, the FDA has been quiet on in vitro SPF testing since 2011.4 The agency has, however, acknowledged the disadvantages of in vivo broad-spectrum testing, including exposure of human participants to harmful UVR and poor interlaboratory reproducibility.6

Given the technical developments and substantial potential benefits of in vitro testing, we believe that it is time for the FDA to revisit this matter. We propose that the FDA take 2 steps toward in vitro testing. First, publish specific validation criteria that would be deemed necessary for approval of such a test, similar to what ISO published in 2017. Second, thoroughly assess new data supporting the viability of available in vitro testing to determine if the FDA’s stated position that in vitro testing is inadequate remains true.

 

 

Although these 2 steps will be important to the process, adoption of an in vitro standard will require more than statements from the FDA. Additional funding should be allocated to researchers who are studying in vitro methodologies, and companies that profit from the multibillion-dollar sunscreen industry should be encouraged to invest in the development of more accurate and more ethical alternatives to in vivo SPF testing.

In vitro SPF testing is inexpensive, avoids the moral quandary of intentionally sunburning human participants, and is more reliable than in vivo testing. It is time for the FDA to facilitate the efforts of academia and industry in taking concrete steps toward approval of an in vitro alternative to in vivo SPF testing.

The sun protection factor (SPF) value indicates to consumers the level of protection that a given sunscreen formulation provides against erythemally effective UV radiation (UVR). 1 In vivo SPF testing, the gold standard for determining SPF, yields highly variable results and can harm human test participants. 2 In vitro SPF testing methodologies have been under development for years but none have (yet) replaced the in vivo test required by national and international regulatory agencies.

Recent European studies have shown strong data to support a highly standardized in vitro method,1 now under development by the International Organization for Standardization (ISO)—potentially to serve as a new SPF determination standard.1,3 Academia and industry should follow this example and actively take steps to develop and validate a suitable replacement for in vivo SPF testing.

In Vivo SPF Testing

The in vivo SPF test involves comparing doses of UVR necessary to induce erythema in human participants with and without sunscreen applied.2 Although this method has long been the standard for SPF determination, it is associated with the following major disadvantages:

  • Cost: The in vivo test is expensive.
  • Variability: Results of the test are subject to high interlaboratory variability due to the inherent subjectivity of identifying erythema, the variable skin types of human participants, and other laboratory-dependent factors.2 A study found that the average coefficient of variation for SPF values obtained from 3 or 4 laboratories to be 20%—with values exceeding 50% in some cases. With that level of variability, the same sunscreen may be labeled SPF 30, SPF 50, or SPF 50+, thereby posing a health risk to consumers who rely on the accuracy of such claims. In fact, Miksa et al2 concluded that “the largest obstacle to a reliable SPF assessment for consumer health is the in vivo SPF test itself.”
  • Ethical concerns: Human participants are intentionally exposed to harmful UVR until sunburn is achieved. For that reason, there have been calls to abandon the practice of in vivo testing.1

Alternatives to In Vivo SPF Testing

There has been international interest in developing in silico and in vitro alternatives to the in vivo SPF test. These options are attractive because they are relatively inexpensive; avoid exposing human participants to harmful UVR; and have the potential to be more accurate and more reproducible than in vivo tests.

In Vitro Protocols—Many such in vitro tests exist; all generally involve applying a layer of sunscreen to an artificial substrate, exposing it to UVR from a solar simulator, and measuring the UVR transmittance through the product and film by spectrophotometry.1 Prior shortcomings of this method have included suboptimal reproducibility, lack of data on substrate and product properties, and lack of demonstrated equivalency to in vivo SPF testing.4

In Silico Protocols—These tests use data on the UV spectra of sunscreen filters, physical characteristics of sunscreen films on skin, and the unique photoinstability of filters to calculate expected UVR transmittance and SPF of sunscreens based on their ingredients.5 Reports have shown high correlation with in vivo values. Results are not subject to random error; reproducibility is theoretically perfect.5

Regulatory Agencies and In Vitro Testing

In the United States, sunscreens are regulated as over-the-counter drugs. In vivo testing is the only US Food and Drug Administration (FDA)–approved method for determining SPF for labeling purposes.1 In a 2007 Proposed Rule and a 2011 Final Rule, the FDA stated that in vitro SPF tests were an inadequate alternative to in vivo tests because of their shortcomings.4,6

 

 

Acknowledging the potential benefits of in vitro testing, the FDA wrote that it would consider in vitro alternatives if equivalency to the in vivo test could be proved.6 The agency has not published an official stance on in vitro SPF testing since those statements in 2007 and 2011. Of note, the FDA deems in vitro testing sufficient for making claims of broad-spectrum coverage.4

In contrast to the regulatory scenario in the United States, Europe regulates sunscreens as cosmetics, and the European Union (EU) has banned animal testing of cosmetics,7 which poses a problem for the development of new sunscreens. It is not surprising, therefore, that in 2006 the European Commission (the executive arm of the EU) published a mandate that in vitro SPF testing methods be actively developed due to ethical concerns associated with in vivo methods.8 In 2017, the International Organization for Standardization released specific validation criteria for proposed in vitro tests to facilitate the eventual approval of such methods.1

Progress of In Vitro Methods

In recent years, advances in in vitro SPF testing methods have addressed shortcomings noted previously by the FDA, which has led to notably improved reproducibility of results and correlation with in vivo values, in large part due to strict standardization of protocols,1 such as tight temperature control of samples, a multisubstrate approach, robotic product application to ensure even distribution, and pre-irradiation of sunscreen samples.

With these improvements, a 2018 study demonstrated an in vitro SPF testing methodology that exceeded published ISO validation criteria for emulsion-type products.1 This method was found to have low interlaboratory variability and high correlation with in vivo SPF values (Pearson r=0.88). Importantly, the authors noted that the consistency and reliability of in vitro SPF testing requires broad institution of a single unified method.1

The method described in the 2018 study1 has been accepted by the ISO Technical Committee and is undergoing further development3; it is expected to be approved by the European Committee for Standardization. After approval, adoption by member nations of the EU will require individual action, representing the next regulatory hurdle for in vitro SPF testing in Europe.

Final Thoughts and Future Steps

Recent data confirm the potential viability of in vitro testing as a primary method of determining SPF values.1 Although ISO has moved forward with development of this method, the FDA has been quiet on in vitro SPF testing since 2011.4 The agency has, however, acknowledged the disadvantages of in vivo broad-spectrum testing, including exposure of human participants to harmful UVR and poor interlaboratory reproducibility.6

Given the technical developments and substantial potential benefits of in vitro testing, we believe that it is time for the FDA to revisit this matter. We propose that the FDA take 2 steps toward in vitro testing. First, publish specific validation criteria that would be deemed necessary for approval of such a test, similar to what ISO published in 2017. Second, thoroughly assess new data supporting the viability of available in vitro testing to determine if the FDA’s stated position that in vitro testing is inadequate remains true.

 

 

Although these 2 steps will be important to the process, adoption of an in vitro standard will require more than statements from the FDA. Additional funding should be allocated to researchers who are studying in vitro methodologies, and companies that profit from the multibillion-dollar sunscreen industry should be encouraged to invest in the development of more accurate and more ethical alternatives to in vivo SPF testing.

In vitro SPF testing is inexpensive, avoids the moral quandary of intentionally sunburning human participants, and is more reliable than in vivo testing. It is time for the FDA to facilitate the efforts of academia and industry in taking concrete steps toward approval of an in vitro alternative to in vivo SPF testing.

References
  1. Pissavini M, Tricaud C, Wiener G, et al. Validation of an in vitro sun protection factor (SPF) method in blinded ring-testing. Int J Cosmet Sci. 2018;40:263-268. doi:10.1111/ics.12459
  2. Miksa S, Lutz D, Guy C, et al. Sunscreen sun protection factor claim based on in vivo interlaboratory variability. Int J Cosmet Sci. 2016;38:541-549. doi:10.1111/ics.12333
  3. ISO/CD 23675: Cosmetics—sun protection test methods—in vitro determination of sun protection factor. International Organization for Standardization (ISO). July 25, 2020. Accessed May 17, 2022. https://www.iso.org/standard/76616.html
  4. US Food and Drug Administration. Labeling and effectiveness testing; sunscreen drug products for over-the-counter human use. Fed Regist. 2011;76(117):35620-35665. Accessed August 9, 2022. https://www.govinfo.gov/content/pkg/FR-2011-06-17/pdf/2011-14766.pdf
  5. Herzog B, Osterwalder U. Simulation of sunscreen performance. Pure Appl Chem. 2015;87:937-951. doi:10.1515/pac-2015-0401
  6. US Food and Drug Administration. Sunscreen drug products for over-the-counter human use; proposed amendment of final monograph. Fed Regist. 2007;72(165):49070-49122. Published August 27, 2007. Accessed August 9, 2022. https://www.govinfo.gov/content/pkg/FR-2007-08-27/pdf/07-4131.pdf
  7. Regulation (EC) No 1223/2009 of the European Parliament and of the Council of 30 November 2009 on cosmetic products. November 30, 2009. Accessed August 10, 2022. https://eur-lex.europa.eu/legal-content/EN/TXT/?uri=CELEX:02009R1223-20190813
  8. European Commission Recommendation 2006/647/EC. Published September 22, 2006. Accessed August 10, 2022. http://eur-lex.europa.eu/legal-content/EN/TXT/?uri=CELEX%3A32006H0647
References
  1. Pissavini M, Tricaud C, Wiener G, et al. Validation of an in vitro sun protection factor (SPF) method in blinded ring-testing. Int J Cosmet Sci. 2018;40:263-268. doi:10.1111/ics.12459
  2. Miksa S, Lutz D, Guy C, et al. Sunscreen sun protection factor claim based on in vivo interlaboratory variability. Int J Cosmet Sci. 2016;38:541-549. doi:10.1111/ics.12333
  3. ISO/CD 23675: Cosmetics—sun protection test methods—in vitro determination of sun protection factor. International Organization for Standardization (ISO). July 25, 2020. Accessed May 17, 2022. https://www.iso.org/standard/76616.html
  4. US Food and Drug Administration. Labeling and effectiveness testing; sunscreen drug products for over-the-counter human use. Fed Regist. 2011;76(117):35620-35665. Accessed August 9, 2022. https://www.govinfo.gov/content/pkg/FR-2011-06-17/pdf/2011-14766.pdf
  5. Herzog B, Osterwalder U. Simulation of sunscreen performance. Pure Appl Chem. 2015;87:937-951. doi:10.1515/pac-2015-0401
  6. US Food and Drug Administration. Sunscreen drug products for over-the-counter human use; proposed amendment of final monograph. Fed Regist. 2007;72(165):49070-49122. Published August 27, 2007. Accessed August 9, 2022. https://www.govinfo.gov/content/pkg/FR-2007-08-27/pdf/07-4131.pdf
  7. Regulation (EC) No 1223/2009 of the European Parliament and of the Council of 30 November 2009 on cosmetic products. November 30, 2009. Accessed August 10, 2022. https://eur-lex.europa.eu/legal-content/EN/TXT/?uri=CELEX:02009R1223-20190813
  8. European Commission Recommendation 2006/647/EC. Published September 22, 2006. Accessed August 10, 2022. http://eur-lex.europa.eu/legal-content/EN/TXT/?uri=CELEX%3A32006H0647
Issue
Cutis - 110(2)
Issue
Cutis - 110(2)
Page Number
E15-E17
Page Number
E15-E17
Publications
Publications
Topics
Article Type
Display Headline
Sun Protection Factor Testing: A Call for an In Vitro Method
Display Headline
Sun Protection Factor Testing: A Call for an In Vitro Method
Sections
Inside the Article

Practice Points

  • The methodology for determining sun protection factor (SPF) that currently is accepted by the US Food and Drug Administration is an expensive and imprecise in vivo test that exposes human participants to harmful UV radiation.
  • In vitro tests for determining SPF may be viable alternatives to the current in vivo gold standard.
  • Researchers and the sunscreen industry should actively develop these in vitro methodologies to adopt a more accurate and less harmful test for SPF.
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

Intralesional Human Papillomavirus Vaccine Therapy for Recalcitrant Plantar Wart Triggers Gout Flare

Article Type
Changed
Thu, 08/18/2022 - 10:36
Display Headline
Intralesional Human Papillomavirus Vaccine Therapy for Recalcitrant Plantar Wart Triggers Gout Flare

To the Editor:

There is increasing evidence supporting the use of the human papillomavirus (HPV) vaccine in the treatment of recalcitrant common warts.1 We describe a potential complication associated with HPV vaccine treatment of warts that would be of interest to dermatologists.

A 70-year-old woman presented with a plantar wart measuring 6 mm in diameter at the base of the right hallux of 5 years’ duration. Prior failed therapies for wart removal included multiple paring treatments, cryotherapy, and topical salicylic acid 40% to 60%. The patient had no notable comorbidities; no history of gout; and no known risk factors for gout, such as hypertension, renal insufficiency, diuretic use, obesity, family history, or trauma.

Prior reports cited effective treatment of recalcitrant warts with recombinant HPV vaccines, both intralesionally1 and intramuscularly.2,3 With this knowledge in mind, we administered an intralesional injection with 0.1-mL recombinant HPV 9-valent vaccine to the patient’s plantar wart. Gradual erythema and swelling of the right first metatarsophalangeal joint developed over the next 7 days. Synovial fluid analysis demonstrated negatively birefringent crystals. The patient commenced treatment with colchicine and indomethacin and improved over the next 5 days. The wart resolved 3 months later and required no further treatment.

Prophylactic quadrivalent HPV vaccines have shown efficacy in treating HPV-associated precancerous and cancerous lesions.4 Case reports have suggested that HPV vaccines may be an effective treatment option for recalcitrant warts,1-3,5 especially in cases that do not respond to traditional treatment. It is possible that the mechanism of wart treatment involves overlap in the antigenic epitopes of the HPV types targeted by the vaccine vs the HPV types responsible for causing warts.2 Papillomaviruslike particles, based on the L1 capsid protein, can induce a specific CD8+ activation signal, leading to a vaccine-induced cytotoxic T-cell response that targets the wart cells with HPV-like antigens.6 The HPV vaccine contains aluminium, which has been shown to activate NLRP3 inflammasome,5 which may trigger gout by increasing monosodium urate crystal deposition via IL-1β production.7 This may lead to an increased risk for gout flares, an adverse effect of the HPV vaccine. This finding is supported by other studies of aluminium-containing vaccines that show an association with gout.6 It is noted that these vaccines are mostly delivered intramuscularly or subcutaneously in some cases.

We reported a case of gout triggered by intralesional HPV vaccine treatment of warts. It is unclear whether the gout was induced by the vaccine itself or whether it was due to trauma caused by the intralesional injection near the joint space. Based on our findings, we recommend that patients receiving intralesional injections for wart treatment be advised of this potential adverse effect, especially if they have risk factors for gout or have a history of gout.

References
  1. Nofal A, Marei A, Ibrahim AM et al. Intralesional versus intramuscular bivalent human papillomavirus vaccine in the treatment of recalcitrant common warts. J Am Acad Dermatol. 2020;82:94-100.
  2. Venugopal SS, Murrell DF. Recalcitrant cutaneous warts treated with recombinant quadrivalent human papillomavirus vaccine (types 6, 11, 16, and 18) in a developmentally delayed, 31-year-old white man. Arch Dermatol. 2010;146:475-477.
  3. Daniel BS, Murrell DF. Complete resolution of chronic multiple verruca vulgaris treated with quadrivalent human papillomavirus vaccine. JAMA Dermatol. 2013;149:370-372.
  4. Kenter GG, Welters MJ, Valentijn AR, et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med. 2009;361:1838-1847.
  5. Eisenbarth SC, Colegio OR, O’Connor W, et al. Crucial role for the NALP3 inflammasome in the immunostimulatory properties of aluminium adjuvants. Nature. 2008;453:1122-1166.
  6. Bellone S, El-Sahwi K, Cocco E, et al. Human papillomavirus type 16 (HPV-16) virus-like particle L1-specific CD8+ cytotoxic T lymphocytes (CTLs) are equally effective as E7-specific CD8+ CTLs in killing autologous HPV-16-positive tumor cells in cervical cancer patients: implications for L1 dendritic cell-based therapeutic vaccines. J Virol. 2009;83:6779-6789.
  7. Yokose C, McCormick N, Chen C, et al. Risk of gout flares after vaccination: a prospective case cross-over study. Ann Rheum Dis. 2019;78:1601-1604.
Article PDF
Author and Disclosure Information

Dr. Phan is from the St. George Dermatology and Skin Cancer Centre, Kogarah, Sydney, Australia. Dr. Lin is from the Department of Dermatology, Mount Sinai School of Medicine, New York, New York.

The authors report no conflict of interest.

Correspondence: Matthew J. Lin, MD, Mount Sinai School of Medicine, Division of Dermatologic and Cosmetic Surgery, Mount Sinai Skin and Laser Center, 234 E 85th St, New York, NY 10028 ([email protected]).

Issue
Cutis - 110(2)
Publications
Topics
Page Number
E13-E14
Sections
Author and Disclosure Information

Dr. Phan is from the St. George Dermatology and Skin Cancer Centre, Kogarah, Sydney, Australia. Dr. Lin is from the Department of Dermatology, Mount Sinai School of Medicine, New York, New York.

The authors report no conflict of interest.

Correspondence: Matthew J. Lin, MD, Mount Sinai School of Medicine, Division of Dermatologic and Cosmetic Surgery, Mount Sinai Skin and Laser Center, 234 E 85th St, New York, NY 10028 ([email protected]).

Author and Disclosure Information

Dr. Phan is from the St. George Dermatology and Skin Cancer Centre, Kogarah, Sydney, Australia. Dr. Lin is from the Department of Dermatology, Mount Sinai School of Medicine, New York, New York.

The authors report no conflict of interest.

Correspondence: Matthew J. Lin, MD, Mount Sinai School of Medicine, Division of Dermatologic and Cosmetic Surgery, Mount Sinai Skin and Laser Center, 234 E 85th St, New York, NY 10028 ([email protected]).

Article PDF
Article PDF

To the Editor:

There is increasing evidence supporting the use of the human papillomavirus (HPV) vaccine in the treatment of recalcitrant common warts.1 We describe a potential complication associated with HPV vaccine treatment of warts that would be of interest to dermatologists.

A 70-year-old woman presented with a plantar wart measuring 6 mm in diameter at the base of the right hallux of 5 years’ duration. Prior failed therapies for wart removal included multiple paring treatments, cryotherapy, and topical salicylic acid 40% to 60%. The patient had no notable comorbidities; no history of gout; and no known risk factors for gout, such as hypertension, renal insufficiency, diuretic use, obesity, family history, or trauma.

Prior reports cited effective treatment of recalcitrant warts with recombinant HPV vaccines, both intralesionally1 and intramuscularly.2,3 With this knowledge in mind, we administered an intralesional injection with 0.1-mL recombinant HPV 9-valent vaccine to the patient’s plantar wart. Gradual erythema and swelling of the right first metatarsophalangeal joint developed over the next 7 days. Synovial fluid analysis demonstrated negatively birefringent crystals. The patient commenced treatment with colchicine and indomethacin and improved over the next 5 days. The wart resolved 3 months later and required no further treatment.

Prophylactic quadrivalent HPV vaccines have shown efficacy in treating HPV-associated precancerous and cancerous lesions.4 Case reports have suggested that HPV vaccines may be an effective treatment option for recalcitrant warts,1-3,5 especially in cases that do not respond to traditional treatment. It is possible that the mechanism of wart treatment involves overlap in the antigenic epitopes of the HPV types targeted by the vaccine vs the HPV types responsible for causing warts.2 Papillomaviruslike particles, based on the L1 capsid protein, can induce a specific CD8+ activation signal, leading to a vaccine-induced cytotoxic T-cell response that targets the wart cells with HPV-like antigens.6 The HPV vaccine contains aluminium, which has been shown to activate NLRP3 inflammasome,5 which may trigger gout by increasing monosodium urate crystal deposition via IL-1β production.7 This may lead to an increased risk for gout flares, an adverse effect of the HPV vaccine. This finding is supported by other studies of aluminium-containing vaccines that show an association with gout.6 It is noted that these vaccines are mostly delivered intramuscularly or subcutaneously in some cases.

We reported a case of gout triggered by intralesional HPV vaccine treatment of warts. It is unclear whether the gout was induced by the vaccine itself or whether it was due to trauma caused by the intralesional injection near the joint space. Based on our findings, we recommend that patients receiving intralesional injections for wart treatment be advised of this potential adverse effect, especially if they have risk factors for gout or have a history of gout.

To the Editor:

There is increasing evidence supporting the use of the human papillomavirus (HPV) vaccine in the treatment of recalcitrant common warts.1 We describe a potential complication associated with HPV vaccine treatment of warts that would be of interest to dermatologists.

A 70-year-old woman presented with a plantar wart measuring 6 mm in diameter at the base of the right hallux of 5 years’ duration. Prior failed therapies for wart removal included multiple paring treatments, cryotherapy, and topical salicylic acid 40% to 60%. The patient had no notable comorbidities; no history of gout; and no known risk factors for gout, such as hypertension, renal insufficiency, diuretic use, obesity, family history, or trauma.

Prior reports cited effective treatment of recalcitrant warts with recombinant HPV vaccines, both intralesionally1 and intramuscularly.2,3 With this knowledge in mind, we administered an intralesional injection with 0.1-mL recombinant HPV 9-valent vaccine to the patient’s plantar wart. Gradual erythema and swelling of the right first metatarsophalangeal joint developed over the next 7 days. Synovial fluid analysis demonstrated negatively birefringent crystals. The patient commenced treatment with colchicine and indomethacin and improved over the next 5 days. The wart resolved 3 months later and required no further treatment.

Prophylactic quadrivalent HPV vaccines have shown efficacy in treating HPV-associated precancerous and cancerous lesions.4 Case reports have suggested that HPV vaccines may be an effective treatment option for recalcitrant warts,1-3,5 especially in cases that do not respond to traditional treatment. It is possible that the mechanism of wart treatment involves overlap in the antigenic epitopes of the HPV types targeted by the vaccine vs the HPV types responsible for causing warts.2 Papillomaviruslike particles, based on the L1 capsid protein, can induce a specific CD8+ activation signal, leading to a vaccine-induced cytotoxic T-cell response that targets the wart cells with HPV-like antigens.6 The HPV vaccine contains aluminium, which has been shown to activate NLRP3 inflammasome,5 which may trigger gout by increasing monosodium urate crystal deposition via IL-1β production.7 This may lead to an increased risk for gout flares, an adverse effect of the HPV vaccine. This finding is supported by other studies of aluminium-containing vaccines that show an association with gout.6 It is noted that these vaccines are mostly delivered intramuscularly or subcutaneously in some cases.

We reported a case of gout triggered by intralesional HPV vaccine treatment of warts. It is unclear whether the gout was induced by the vaccine itself or whether it was due to trauma caused by the intralesional injection near the joint space. Based on our findings, we recommend that patients receiving intralesional injections for wart treatment be advised of this potential adverse effect, especially if they have risk factors for gout or have a history of gout.

References
  1. Nofal A, Marei A, Ibrahim AM et al. Intralesional versus intramuscular bivalent human papillomavirus vaccine in the treatment of recalcitrant common warts. J Am Acad Dermatol. 2020;82:94-100.
  2. Venugopal SS, Murrell DF. Recalcitrant cutaneous warts treated with recombinant quadrivalent human papillomavirus vaccine (types 6, 11, 16, and 18) in a developmentally delayed, 31-year-old white man. Arch Dermatol. 2010;146:475-477.
  3. Daniel BS, Murrell DF. Complete resolution of chronic multiple verruca vulgaris treated with quadrivalent human papillomavirus vaccine. JAMA Dermatol. 2013;149:370-372.
  4. Kenter GG, Welters MJ, Valentijn AR, et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med. 2009;361:1838-1847.
  5. Eisenbarth SC, Colegio OR, O’Connor W, et al. Crucial role for the NALP3 inflammasome in the immunostimulatory properties of aluminium adjuvants. Nature. 2008;453:1122-1166.
  6. Bellone S, El-Sahwi K, Cocco E, et al. Human papillomavirus type 16 (HPV-16) virus-like particle L1-specific CD8+ cytotoxic T lymphocytes (CTLs) are equally effective as E7-specific CD8+ CTLs in killing autologous HPV-16-positive tumor cells in cervical cancer patients: implications for L1 dendritic cell-based therapeutic vaccines. J Virol. 2009;83:6779-6789.
  7. Yokose C, McCormick N, Chen C, et al. Risk of gout flares after vaccination: a prospective case cross-over study. Ann Rheum Dis. 2019;78:1601-1604.
References
  1. Nofal A, Marei A, Ibrahim AM et al. Intralesional versus intramuscular bivalent human papillomavirus vaccine in the treatment of recalcitrant common warts. J Am Acad Dermatol. 2020;82:94-100.
  2. Venugopal SS, Murrell DF. Recalcitrant cutaneous warts treated with recombinant quadrivalent human papillomavirus vaccine (types 6, 11, 16, and 18) in a developmentally delayed, 31-year-old white man. Arch Dermatol. 2010;146:475-477.
  3. Daniel BS, Murrell DF. Complete resolution of chronic multiple verruca vulgaris treated with quadrivalent human papillomavirus vaccine. JAMA Dermatol. 2013;149:370-372.
  4. Kenter GG, Welters MJ, Valentijn AR, et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med. 2009;361:1838-1847.
  5. Eisenbarth SC, Colegio OR, O’Connor W, et al. Crucial role for the NALP3 inflammasome in the immunostimulatory properties of aluminium adjuvants. Nature. 2008;453:1122-1166.
  6. Bellone S, El-Sahwi K, Cocco E, et al. Human papillomavirus type 16 (HPV-16) virus-like particle L1-specific CD8+ cytotoxic T lymphocytes (CTLs) are equally effective as E7-specific CD8+ CTLs in killing autologous HPV-16-positive tumor cells in cervical cancer patients: implications for L1 dendritic cell-based therapeutic vaccines. J Virol. 2009;83:6779-6789.
  7. Yokose C, McCormick N, Chen C, et al. Risk of gout flares after vaccination: a prospective case cross-over study. Ann Rheum Dis. 2019;78:1601-1604.
Issue
Cutis - 110(2)
Issue
Cutis - 110(2)
Page Number
E13-E14
Page Number
E13-E14
Publications
Publications
Topics
Article Type
Display Headline
Intralesional Human Papillomavirus Vaccine Therapy for Recalcitrant Plantar Wart Triggers Gout Flare
Display Headline
Intralesional Human Papillomavirus Vaccine Therapy for Recalcitrant Plantar Wart Triggers Gout Flare
Sections
Inside the Article

Practice Points

  • Human papillomavirus (HPV) vaccines are increasingly used for recalcitrant warts.
  • We describe an unreported adverse effect of gout flare following HPV vaccine treatment of plantar wart.
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

Hidradenitis Suppurativa Guidelines

Article Type
Changed
Wed, 08/17/2022 - 13:49

Publications
Topics
Sections

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 08/17/2022 - 13:30
Un-Gate On Date
Wed, 08/17/2022 - 13:30
Use ProPublica
CFC Schedule Remove Status
Wed, 08/17/2022 - 13:30
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

Hidradenitis Suppurativa Medications

Article Type
Changed
Wed, 08/17/2022 - 12:43

Publications
Topics
Sections

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 08/17/2022 - 12:45
Un-Gate On Date
Wed, 08/17/2022 - 12:45
Use ProPublica
CFC Schedule Remove Status
Wed, 08/17/2022 - 12:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Exaggerated Facial Lines on the Forehead and Cheeks

Article Type
Changed
Mon, 10/31/2022 - 12:13
Display Headline
Exaggerated Facial Lines on the Forehead and Cheeks

The Diagnosis: Pachydermoperiostosis

Histopathology of the forehead punch biopsy demonstrated sebaceous hyperplasia with an occupation rate of greater than 40%, increased mucin, elastic fiber degeneration, and fibrosis. Pachydermia is graded from 0 to 3 depending on the degree of these changes; our patient met criteria for grade 3 pachydermia (Figure 1). Radiography revealed diffuse cortical thickening of the long bones that was most marked in the left femur (Figure 2); however, no other findings were demonstrative of Paget disease.

A, Histopathology of a forehead biopsy showed increased sebaceous gland occupation (H&E, original magnification ×4). B, Colloidal iron stain demonstrated increased mucin (original magnification ×4). C, Verhoeff-van Gieson stain showed elastic fiber
FIGURE 1. A, Histopathology of a forehead biopsy showed increased sebaceous gland occupation (H&E, original magnification ×4). B, Colloidal iron stain demonstrated increased mucin (original magnification ×4). C, Verhoeff-van Gieson stain showed elastic fiber degeneration (original magnification ×40).

Pachydermoperiostosis (PDP)(also known as Touraine-Solente-Golé syndrome or primary hypertrophic osteoarthropathy) is a rare genetic condition that affects both the dermatologic and skeletal systems. Clinical features of the disease include progressive thickening and furrowing of the skin on the scalp and face (known as pachydermia), digital clubbing, and periostosis. Other potential cutaneous features include seborrhea, acne, hyperhidrosis of the palms and soles, cutis verticis gyrata, eczema, and a burning sensation of the hands and feet. Myelofibrosis and gastrointestinal abnormalities also have been reported.1

Radiography of the left femur demonstrated diffuse cortical thickening
FIGURE 2. Radiography of the left femur demonstrated diffuse cortical thickening.

The disease typically affects males (7:1 ratio); also, men typically display a more severe phenotype of the disease.2 It most commonly begins during puberty and follows a generally progressive course of 5 to 20 years before eventually stabilizing. Both autosomal-dominant with incomplete penetrance and recessive inheritance versions of PDP can occur. Prostaglandin E2 (PGE2) has been implicated in the pathogenesis of PDP; PGE2 is important in the inflammatory response and may evolve from disrupted protein degradation pathways.3 Sasaki et al4 additionally reported that the severity of pachydermia clinically and histologically appeared to correlate with the serum PGE2 levels in affected patients. Prostaglandin E2 causes a vasodilatory effect, perhaps explaining the clubbing observed in PDP, and also modifies the activity of osteoblasts and osteoclasts, causing the bone remodeling observed in the disease.4

In our patient, the initial differential diagnosis included PDP, as well as lepromatous leprosy, acromegaly, Paget disease of the bone, amyloidosis, scleromyxedema, and cutaneous T-cell lymphoma. However, the time course of the disease, lack of numerous symmetric thickened plaques and madarosis, and pathology argued against lepromatous leprosy. Acromegaly was ruled out due to lack of macroglossia as well as laboratory analysis within reference range including IGF-1 levels and thyroid function tests. Biopsy findings ultimately ruled out amyloidosis and cutaneous T-cell lymphoma. The bone scan revealed diffuse cortical thickening consistent with PDP, and there were no other radiologic findings suggestive of Paget disease. Pachydermoperiostosis is diagnosed using the Borochowitz criteria, which entails that 2 of the following 4 fulfillment criteria must be met: familial transmission, pachydermia, digital clubbing, and/or bony involvement with evidence of radiologic alterations or pain. Our patient met all 4 criteria. The clinical manifestations of PDP are variable with respect to skin and bone changes. The various clinical expressions include the complete form (ie, pachydermia, cutis verticis gyrata, periostosis), the incomplete form (ie, absence of cutis verticis gyrata), and forme fruste (ie, pachydermia with minimal or absent periostosis).5

Management for PDP involves surgical correction for cosmesis as well as for functional concerns if present. Symptoms secondary to periostosis should be managed with symptomatic treatment such as nonsteroidal antiinflammatory drugs. Patients managed with etoricoxib, a COX-2–selective nonsteroidal anti-inflammatory drug, have had normalized inflammatory markers that resulted in the lessening of forehead skin folds. Oral aescin has been shown to relieve joint pain due to its antiedematous effect.6 Our patient received treatment with nonsteroidal anti-inflammatory drugs for symptomatic management of the associated joint pain but unfortunately was lost to follow-up.

References
  1. Castori M, Sinibaldi L, Mingarelli R, et al. Pachydermoperiostosis: an update. Clin Genet. 2005;68:477-486.
  2. Reginato AJ, Shipachasse V, Guerrero R. Familial idiopathic hypertrophic osteoarthropathy and cranial suture defects in children. Skel Radiol. 1982;8:105-109.
  3. Coggins KG, Coffman TM, Koller BH. The Hippocratic finger points the blame at PGE2. Nat Genet. 2008;40:691-692.
  4. Sasaki T, Niizeki H, Shimizu A, et al. Identification of mutations in the prostaglandin transporter gene SLCO2A1 and its phenotype-genotype correlation in Japanese patients with pachydermoperiostosis. J Dermatol Sci. 2012;68:36-44.
  5. Bhaskaranand K, Shetty RR, Bhat AK. Pachydermoperiostosis: three case reports. J Orthop Surg (Hong Kong). 2001;9:61-66.
  6. Zhang H, Yang B. Successful treatment of pachydermoperiostosis patients with etoricoxib, aescin, and arthroscopic synovectomy: two case reports. Medicine (Baltimore). 2017;96:E8865.
Article PDF
Author and Disclosure Information

From The University of Texas Health Science Center at San Antonio. Drs. Sitabkhan and Osswald are from the Department of Dermatology, and Dr. Jing is from the Long School of Medicine.

The authors report no conflict of interest.

Correspondence: Amreen Sitabkhan, MD, 12740 Hillcrest Rd #200, Dallas, TX 75230 ([email protected]).

Issue
Cutis - 110(2)
Publications
Topics
Page Number
E18-E20
Sections
Author and Disclosure Information

From The University of Texas Health Science Center at San Antonio. Drs. Sitabkhan and Osswald are from the Department of Dermatology, and Dr. Jing is from the Long School of Medicine.

The authors report no conflict of interest.

Correspondence: Amreen Sitabkhan, MD, 12740 Hillcrest Rd #200, Dallas, TX 75230 ([email protected]).

Author and Disclosure Information

From The University of Texas Health Science Center at San Antonio. Drs. Sitabkhan and Osswald are from the Department of Dermatology, and Dr. Jing is from the Long School of Medicine.

The authors report no conflict of interest.

Correspondence: Amreen Sitabkhan, MD, 12740 Hillcrest Rd #200, Dallas, TX 75230 ([email protected]).

Article PDF
Article PDF
Related Articles

The Diagnosis: Pachydermoperiostosis

Histopathology of the forehead punch biopsy demonstrated sebaceous hyperplasia with an occupation rate of greater than 40%, increased mucin, elastic fiber degeneration, and fibrosis. Pachydermia is graded from 0 to 3 depending on the degree of these changes; our patient met criteria for grade 3 pachydermia (Figure 1). Radiography revealed diffuse cortical thickening of the long bones that was most marked in the left femur (Figure 2); however, no other findings were demonstrative of Paget disease.

A, Histopathology of a forehead biopsy showed increased sebaceous gland occupation (H&E, original magnification ×4). B, Colloidal iron stain demonstrated increased mucin (original magnification ×4). C, Verhoeff-van Gieson stain showed elastic fiber
FIGURE 1. A, Histopathology of a forehead biopsy showed increased sebaceous gland occupation (H&E, original magnification ×4). B, Colloidal iron stain demonstrated increased mucin (original magnification ×4). C, Verhoeff-van Gieson stain showed elastic fiber degeneration (original magnification ×40).

Pachydermoperiostosis (PDP)(also known as Touraine-Solente-Golé syndrome or primary hypertrophic osteoarthropathy) is a rare genetic condition that affects both the dermatologic and skeletal systems. Clinical features of the disease include progressive thickening and furrowing of the skin on the scalp and face (known as pachydermia), digital clubbing, and periostosis. Other potential cutaneous features include seborrhea, acne, hyperhidrosis of the palms and soles, cutis verticis gyrata, eczema, and a burning sensation of the hands and feet. Myelofibrosis and gastrointestinal abnormalities also have been reported.1

Radiography of the left femur demonstrated diffuse cortical thickening
FIGURE 2. Radiography of the left femur demonstrated diffuse cortical thickening.

The disease typically affects males (7:1 ratio); also, men typically display a more severe phenotype of the disease.2 It most commonly begins during puberty and follows a generally progressive course of 5 to 20 years before eventually stabilizing. Both autosomal-dominant with incomplete penetrance and recessive inheritance versions of PDP can occur. Prostaglandin E2 (PGE2) has been implicated in the pathogenesis of PDP; PGE2 is important in the inflammatory response and may evolve from disrupted protein degradation pathways.3 Sasaki et al4 additionally reported that the severity of pachydermia clinically and histologically appeared to correlate with the serum PGE2 levels in affected patients. Prostaglandin E2 causes a vasodilatory effect, perhaps explaining the clubbing observed in PDP, and also modifies the activity of osteoblasts and osteoclasts, causing the bone remodeling observed in the disease.4

In our patient, the initial differential diagnosis included PDP, as well as lepromatous leprosy, acromegaly, Paget disease of the bone, amyloidosis, scleromyxedema, and cutaneous T-cell lymphoma. However, the time course of the disease, lack of numerous symmetric thickened plaques and madarosis, and pathology argued against lepromatous leprosy. Acromegaly was ruled out due to lack of macroglossia as well as laboratory analysis within reference range including IGF-1 levels and thyroid function tests. Biopsy findings ultimately ruled out amyloidosis and cutaneous T-cell lymphoma. The bone scan revealed diffuse cortical thickening consistent with PDP, and there were no other radiologic findings suggestive of Paget disease. Pachydermoperiostosis is diagnosed using the Borochowitz criteria, which entails that 2 of the following 4 fulfillment criteria must be met: familial transmission, pachydermia, digital clubbing, and/or bony involvement with evidence of radiologic alterations or pain. Our patient met all 4 criteria. The clinical manifestations of PDP are variable with respect to skin and bone changes. The various clinical expressions include the complete form (ie, pachydermia, cutis verticis gyrata, periostosis), the incomplete form (ie, absence of cutis verticis gyrata), and forme fruste (ie, pachydermia with minimal or absent periostosis).5

Management for PDP involves surgical correction for cosmesis as well as for functional concerns if present. Symptoms secondary to periostosis should be managed with symptomatic treatment such as nonsteroidal antiinflammatory drugs. Patients managed with etoricoxib, a COX-2–selective nonsteroidal anti-inflammatory drug, have had normalized inflammatory markers that resulted in the lessening of forehead skin folds. Oral aescin has been shown to relieve joint pain due to its antiedematous effect.6 Our patient received treatment with nonsteroidal anti-inflammatory drugs for symptomatic management of the associated joint pain but unfortunately was lost to follow-up.

The Diagnosis: Pachydermoperiostosis

Histopathology of the forehead punch biopsy demonstrated sebaceous hyperplasia with an occupation rate of greater than 40%, increased mucin, elastic fiber degeneration, and fibrosis. Pachydermia is graded from 0 to 3 depending on the degree of these changes; our patient met criteria for grade 3 pachydermia (Figure 1). Radiography revealed diffuse cortical thickening of the long bones that was most marked in the left femur (Figure 2); however, no other findings were demonstrative of Paget disease.

A, Histopathology of a forehead biopsy showed increased sebaceous gland occupation (H&E, original magnification ×4). B, Colloidal iron stain demonstrated increased mucin (original magnification ×4). C, Verhoeff-van Gieson stain showed elastic fiber
FIGURE 1. A, Histopathology of a forehead biopsy showed increased sebaceous gland occupation (H&E, original magnification ×4). B, Colloidal iron stain demonstrated increased mucin (original magnification ×4). C, Verhoeff-van Gieson stain showed elastic fiber degeneration (original magnification ×40).

Pachydermoperiostosis (PDP)(also known as Touraine-Solente-Golé syndrome or primary hypertrophic osteoarthropathy) is a rare genetic condition that affects both the dermatologic and skeletal systems. Clinical features of the disease include progressive thickening and furrowing of the skin on the scalp and face (known as pachydermia), digital clubbing, and periostosis. Other potential cutaneous features include seborrhea, acne, hyperhidrosis of the palms and soles, cutis verticis gyrata, eczema, and a burning sensation of the hands and feet. Myelofibrosis and gastrointestinal abnormalities also have been reported.1

Radiography of the left femur demonstrated diffuse cortical thickening
FIGURE 2. Radiography of the left femur demonstrated diffuse cortical thickening.

The disease typically affects males (7:1 ratio); also, men typically display a more severe phenotype of the disease.2 It most commonly begins during puberty and follows a generally progressive course of 5 to 20 years before eventually stabilizing. Both autosomal-dominant with incomplete penetrance and recessive inheritance versions of PDP can occur. Prostaglandin E2 (PGE2) has been implicated in the pathogenesis of PDP; PGE2 is important in the inflammatory response and may evolve from disrupted protein degradation pathways.3 Sasaki et al4 additionally reported that the severity of pachydermia clinically and histologically appeared to correlate with the serum PGE2 levels in affected patients. Prostaglandin E2 causes a vasodilatory effect, perhaps explaining the clubbing observed in PDP, and also modifies the activity of osteoblasts and osteoclasts, causing the bone remodeling observed in the disease.4

In our patient, the initial differential diagnosis included PDP, as well as lepromatous leprosy, acromegaly, Paget disease of the bone, amyloidosis, scleromyxedema, and cutaneous T-cell lymphoma. However, the time course of the disease, lack of numerous symmetric thickened plaques and madarosis, and pathology argued against lepromatous leprosy. Acromegaly was ruled out due to lack of macroglossia as well as laboratory analysis within reference range including IGF-1 levels and thyroid function tests. Biopsy findings ultimately ruled out amyloidosis and cutaneous T-cell lymphoma. The bone scan revealed diffuse cortical thickening consistent with PDP, and there were no other radiologic findings suggestive of Paget disease. Pachydermoperiostosis is diagnosed using the Borochowitz criteria, which entails that 2 of the following 4 fulfillment criteria must be met: familial transmission, pachydermia, digital clubbing, and/or bony involvement with evidence of radiologic alterations or pain. Our patient met all 4 criteria. The clinical manifestations of PDP are variable with respect to skin and bone changes. The various clinical expressions include the complete form (ie, pachydermia, cutis verticis gyrata, periostosis), the incomplete form (ie, absence of cutis verticis gyrata), and forme fruste (ie, pachydermia with minimal or absent periostosis).5

Management for PDP involves surgical correction for cosmesis as well as for functional concerns if present. Symptoms secondary to periostosis should be managed with symptomatic treatment such as nonsteroidal antiinflammatory drugs. Patients managed with etoricoxib, a COX-2–selective nonsteroidal anti-inflammatory drug, have had normalized inflammatory markers that resulted in the lessening of forehead skin folds. Oral aescin has been shown to relieve joint pain due to its antiedematous effect.6 Our patient received treatment with nonsteroidal anti-inflammatory drugs for symptomatic management of the associated joint pain but unfortunately was lost to follow-up.

References
  1. Castori M, Sinibaldi L, Mingarelli R, et al. Pachydermoperiostosis: an update. Clin Genet. 2005;68:477-486.
  2. Reginato AJ, Shipachasse V, Guerrero R. Familial idiopathic hypertrophic osteoarthropathy and cranial suture defects in children. Skel Radiol. 1982;8:105-109.
  3. Coggins KG, Coffman TM, Koller BH. The Hippocratic finger points the blame at PGE2. Nat Genet. 2008;40:691-692.
  4. Sasaki T, Niizeki H, Shimizu A, et al. Identification of mutations in the prostaglandin transporter gene SLCO2A1 and its phenotype-genotype correlation in Japanese patients with pachydermoperiostosis. J Dermatol Sci. 2012;68:36-44.
  5. Bhaskaranand K, Shetty RR, Bhat AK. Pachydermoperiostosis: three case reports. J Orthop Surg (Hong Kong). 2001;9:61-66.
  6. Zhang H, Yang B. Successful treatment of pachydermoperiostosis patients with etoricoxib, aescin, and arthroscopic synovectomy: two case reports. Medicine (Baltimore). 2017;96:E8865.
References
  1. Castori M, Sinibaldi L, Mingarelli R, et al. Pachydermoperiostosis: an update. Clin Genet. 2005;68:477-486.
  2. Reginato AJ, Shipachasse V, Guerrero R. Familial idiopathic hypertrophic osteoarthropathy and cranial suture defects in children. Skel Radiol. 1982;8:105-109.
  3. Coggins KG, Coffman TM, Koller BH. The Hippocratic finger points the blame at PGE2. Nat Genet. 2008;40:691-692.
  4. Sasaki T, Niizeki H, Shimizu A, et al. Identification of mutations in the prostaglandin transporter gene SLCO2A1 and its phenotype-genotype correlation in Japanese patients with pachydermoperiostosis. J Dermatol Sci. 2012;68:36-44.
  5. Bhaskaranand K, Shetty RR, Bhat AK. Pachydermoperiostosis: three case reports. J Orthop Surg (Hong Kong). 2001;9:61-66.
  6. Zhang H, Yang B. Successful treatment of pachydermoperiostosis patients with etoricoxib, aescin, and arthroscopic synovectomy: two case reports. Medicine (Baltimore). 2017;96:E8865.
Issue
Cutis - 110(2)
Issue
Cutis - 110(2)
Page Number
E18-E20
Page Number
E18-E20
Publications
Publications
Topics
Article Type
Display Headline
Exaggerated Facial Lines on the Forehead and Cheeks
Display Headline
Exaggerated Facial Lines on the Forehead and Cheeks
Sections
Questionnaire Body

A 36-year-old man presented to the emergency department with an olecranon fracture after falling from a tree. The patient had a medical history of type 2 diabetes mellitus and a surgical history of facial cosmetic surgery. He underwent internal fixation with orthopedic surgery for the olecranon fracture, and dermatology subsequently was consulted due to diffuse skin changes on the face. He reported that these dermatologic changes began around 17 years of age and had progressed to the current presentation. He denied itching, burning, pain, or contact with armadillos. A family history revealed the patient’s brother also had a similar appearance. Physical examination revealed exaggerated facial lines on the forehead (top) and cheeks. Digital clubbing and skin thickening were noted on the hands (bottom) and feet; examination of the back revealed multiple hypopigmented patches. Observation of the scalp showed multiple symmetric ridges and grooves with sparse overlying hair consistent with cutis verticis gyrata. A punch biopsy of the forehead was obtained as well as bone radiography taken previously by the primary team.

Exaggerated facial lines on the forehead and cheeks

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 08/17/2022 - 10:15
Un-Gate On Date
Wed, 08/17/2022 - 10:15
Use ProPublica
CFC Schedule Remove Status
Wed, 08/17/2022 - 10:15
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Does PREDICT accurately estimate breast cancer survival?

Article Type
Changed
Thu, 12/15/2022 - 17:19

 

The PREDICT score does not seem to be particularly accurate when it comes to estimating overall survival (OS) in patients with HER2-positive early breast cancer who are treated with modern chemotherapy and anti-HER2 targeted therapies. This is the conclusion of an international study published in the journal npj Breast Cancer. The work was supervised by Matteo Lambertini, MD, PhD, an oncologist at the IRCCS San Martino Polyclinic Hospital in Genoa, Italy.

As the authors explain, “PREDICT is a publicly available online tool that helps to predict the individual prognosis of patients with early breast cancer and to show the impact of adjuvant treatments administered after breast cancer surgery.” The tool uses traditional clinical-pathological factors. The authors also point out that the original version of this tool was validated in several datasets of patients with breast cancer. In 2011, it was updated to include HER2 status.

The investigators noted that, although the use of PREDICT is recommended to aid decision-making in the adjuvant setting, its prognostic role in patients with HER2-positive early breast cancer who are treated with modern chemotherapy and anti-HER2 therapies – even trastuzumab-based ones – remains unclear.

Therefore, they decided to analyze PREDICT’s prognostic performance using data extracted from the ALTTO trial, the largest adjuvant study ever conducted in the field of HER2-positive early breast cancer. That trial “represented a unique opportunity to investigate the reliability and prognostic performance of PREDICT in women with HER2-positive disease,” according to the investigators. They went on to specify that ALTTO evaluated adjuvant lapatinib plus trastuzumab vs. trastuzumab alone in 8,381 patients – 2,794 of whom were included in their own analysis.

What the analysis found was that, overall, PREDICT underestimated 5-year OS by 6.7%. The observed 5-year OS was 94.7%, and the predicted 5-year OS was 88.0%.

“The underestimation was consistent across all subgroups, including those according to the type of anti-HER2 therapy. The highest absolute differences were observed for patients with hormone receptor–negative disease, nodal involvement, and large tumor size (13.0%, 15.8%, and 15.3%, respectively),” they wrote. Furthermore, they reported that “the suboptimal performance of this prognostic tool was observed irrespective of type of anti-HER2 treatment, type of chemotherapy regimen, age of the patients at the time of diagnosis, central hormone receptor status, pathological nodal status, and pathological tumor size.”

To potentially explain the reasons for the underestimation of patients’ OS, the authors questioned whether the population used to validate PREDICT accurately mirrored the real-world population of patients with HER2-positive disease treated in the modern era with effective chemotherapy and anti-HER2 targeted therapies. “Moreover, the current standard of care for early breast cancer is even superior to the treatment received by many patients in the ALTTO study. … As such, the discordance between OS estimated by PREDICT and the current real-world OS is expected to be even higher. Therefore,” the researchers concluded, “our results suggest that the current version of PREDICT should be used with caution for prognostication in HER2-positive early breast cancer patients treated in the modern era with effective chemotherapy and anti-HER2 targeted therapies.”

A version of this article first appeared on Medscape.com. This article was translated from Univadis Italy.

Publications
Topics
Sections

 

The PREDICT score does not seem to be particularly accurate when it comes to estimating overall survival (OS) in patients with HER2-positive early breast cancer who are treated with modern chemotherapy and anti-HER2 targeted therapies. This is the conclusion of an international study published in the journal npj Breast Cancer. The work was supervised by Matteo Lambertini, MD, PhD, an oncologist at the IRCCS San Martino Polyclinic Hospital in Genoa, Italy.

As the authors explain, “PREDICT is a publicly available online tool that helps to predict the individual prognosis of patients with early breast cancer and to show the impact of adjuvant treatments administered after breast cancer surgery.” The tool uses traditional clinical-pathological factors. The authors also point out that the original version of this tool was validated in several datasets of patients with breast cancer. In 2011, it was updated to include HER2 status.

The investigators noted that, although the use of PREDICT is recommended to aid decision-making in the adjuvant setting, its prognostic role in patients with HER2-positive early breast cancer who are treated with modern chemotherapy and anti-HER2 therapies – even trastuzumab-based ones – remains unclear.

Therefore, they decided to analyze PREDICT’s prognostic performance using data extracted from the ALTTO trial, the largest adjuvant study ever conducted in the field of HER2-positive early breast cancer. That trial “represented a unique opportunity to investigate the reliability and prognostic performance of PREDICT in women with HER2-positive disease,” according to the investigators. They went on to specify that ALTTO evaluated adjuvant lapatinib plus trastuzumab vs. trastuzumab alone in 8,381 patients – 2,794 of whom were included in their own analysis.

What the analysis found was that, overall, PREDICT underestimated 5-year OS by 6.7%. The observed 5-year OS was 94.7%, and the predicted 5-year OS was 88.0%.

“The underestimation was consistent across all subgroups, including those according to the type of anti-HER2 therapy. The highest absolute differences were observed for patients with hormone receptor–negative disease, nodal involvement, and large tumor size (13.0%, 15.8%, and 15.3%, respectively),” they wrote. Furthermore, they reported that “the suboptimal performance of this prognostic tool was observed irrespective of type of anti-HER2 treatment, type of chemotherapy regimen, age of the patients at the time of diagnosis, central hormone receptor status, pathological nodal status, and pathological tumor size.”

To potentially explain the reasons for the underestimation of patients’ OS, the authors questioned whether the population used to validate PREDICT accurately mirrored the real-world population of patients with HER2-positive disease treated in the modern era with effective chemotherapy and anti-HER2 targeted therapies. “Moreover, the current standard of care for early breast cancer is even superior to the treatment received by many patients in the ALTTO study. … As such, the discordance between OS estimated by PREDICT and the current real-world OS is expected to be even higher. Therefore,” the researchers concluded, “our results suggest that the current version of PREDICT should be used with caution for prognostication in HER2-positive early breast cancer patients treated in the modern era with effective chemotherapy and anti-HER2 targeted therapies.”

A version of this article first appeared on Medscape.com. This article was translated from Univadis Italy.

 

The PREDICT score does not seem to be particularly accurate when it comes to estimating overall survival (OS) in patients with HER2-positive early breast cancer who are treated with modern chemotherapy and anti-HER2 targeted therapies. This is the conclusion of an international study published in the journal npj Breast Cancer. The work was supervised by Matteo Lambertini, MD, PhD, an oncologist at the IRCCS San Martino Polyclinic Hospital in Genoa, Italy.

As the authors explain, “PREDICT is a publicly available online tool that helps to predict the individual prognosis of patients with early breast cancer and to show the impact of adjuvant treatments administered after breast cancer surgery.” The tool uses traditional clinical-pathological factors. The authors also point out that the original version of this tool was validated in several datasets of patients with breast cancer. In 2011, it was updated to include HER2 status.

The investigators noted that, although the use of PREDICT is recommended to aid decision-making in the adjuvant setting, its prognostic role in patients with HER2-positive early breast cancer who are treated with modern chemotherapy and anti-HER2 therapies – even trastuzumab-based ones – remains unclear.

Therefore, they decided to analyze PREDICT’s prognostic performance using data extracted from the ALTTO trial, the largest adjuvant study ever conducted in the field of HER2-positive early breast cancer. That trial “represented a unique opportunity to investigate the reliability and prognostic performance of PREDICT in women with HER2-positive disease,” according to the investigators. They went on to specify that ALTTO evaluated adjuvant lapatinib plus trastuzumab vs. trastuzumab alone in 8,381 patients – 2,794 of whom were included in their own analysis.

What the analysis found was that, overall, PREDICT underestimated 5-year OS by 6.7%. The observed 5-year OS was 94.7%, and the predicted 5-year OS was 88.0%.

“The underestimation was consistent across all subgroups, including those according to the type of anti-HER2 therapy. The highest absolute differences were observed for patients with hormone receptor–negative disease, nodal involvement, and large tumor size (13.0%, 15.8%, and 15.3%, respectively),” they wrote. Furthermore, they reported that “the suboptimal performance of this prognostic tool was observed irrespective of type of anti-HER2 treatment, type of chemotherapy regimen, age of the patients at the time of diagnosis, central hormone receptor status, pathological nodal status, and pathological tumor size.”

To potentially explain the reasons for the underestimation of patients’ OS, the authors questioned whether the population used to validate PREDICT accurately mirrored the real-world population of patients with HER2-positive disease treated in the modern era with effective chemotherapy and anti-HER2 targeted therapies. “Moreover, the current standard of care for early breast cancer is even superior to the treatment received by many patients in the ALTTO study. … As such, the discordance between OS estimated by PREDICT and the current real-world OS is expected to be even higher. Therefore,” the researchers concluded, “our results suggest that the current version of PREDICT should be used with caution for prognostication in HER2-positive early breast cancer patients treated in the modern era with effective chemotherapy and anti-HER2 targeted therapies.”

A version of this article first appeared on Medscape.com. This article was translated from Univadis Italy.

Publications
Publications
Topics
Article Type
Sections
Article Source

FROM NPJ BREAST CANCER

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

Ulcerative Colitis Medications

Article Type
Changed
Tue, 08/16/2022 - 11:58

Publications
Topics
Sections

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Tue, 08/16/2022 - 12:00
Un-Gate On Date
Tue, 08/16/2022 - 12:00
Use ProPublica
CFC Schedule Remove Status
Tue, 08/16/2022 - 12:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article