COVID-19/heart connection: What hospitalists need to know

Article Type
Changed
Thu, 08/26/2021 - 16:02

The heart-related manifestations of COVID-19 are a serious matter, but no one should make the mistake of thinking of COVID-19 as primarily a cardiac disease, according to Jeffrey C. Trost, MD, a cardiologist at Johns Hopkins University, Baltimore.

“One of my take-home messages is this is not a heart illness. This is still an infectious pulmonary illness that most likely causes stress on the heart in both healthy people and those with preexisting heart disease,” he said in offering a preview of his upcoming clinical update at HM20 Virtual, hosted by the Society of Hospital Medicine.

For this reason, in his clinical update talk, titled “COVID-19 and the Heart: What Every Hospitalist Should Know,” he’ll urge hospitalists to be conservative in ordering cardiac biomarker tests such troponin and natriuretic peptide levels. The focus should appropriately be on the subset of COVID-19 patients having the same symptoms suggestive of acute coronary syndrome, heart failure, or new-onset cardiomyopathy that would trigger laboratory testing in non–COVID-19 patients.

“Be more selective. Definitely do not routinely monitor troponin or [N-terminal of the prohormone brain natriuretic peptide] in patients just because they have COVID-19. A lot of patients with COVID-19 have these labs drawn, especially in the emergency department. We see a high signal-to-noise ratio: not infrequently the values are abnormal, and yet we don’t really know what that means,” said Dr. Trost, who is also director of the cardiac catheterization laboratory at Johns Hopkins Bayview Medical Center.

COVID-19 patients with preexisting heart disease are clearly at increased risk of severe forms of the infectious illness. In his talk, Dr. Trost will review the epidemiology of this association. He’ll also discuss the varied cardiac manifestations of COVID-19, consisting of myocarditis or other forms of new-onset cardiomyopathy, acute coronary syndrome, heart failure, and arrhythmias.

Many questions regarding COVID-19 and the heart remain unanswered for now, such as the mechanism and long-term implications of the phenomenon of ST-elevation acute coronary syndrome with chest pain in the presence of unobstructed coronary arteries, which Dr. Trost and others have encountered. Or the extent to which COVID-19–associated myocarditis is directly virus mediated as opposed to an autoimmune process.

“We’re relying completely on case reports at this point,” according to the cardiologist.

But one major issue has, thankfully, been put to rest on the basis of persuasive evidence which Dr. Trost plans to highlight: Millions of patients on ACE inhibitors or angiotensin receptor blockers can now rest assured that taking those medications doesn’t place them at increased risk of becoming infected with the novel coronavirus or, if infected, developing severe complications of COVID-19. Earlier in the pandemic that had been a legitimate theoretic concern based upon a plausible mechanism.

“I think we as physicians can now confidently say that we don’t need to stop these medicines in folks,” Dr. Trost said.

COVID-19 and the Heart: What Every Hospitalist Should Know

Live Q&A: Wednesday, Aug. 19, 3:30 p.m. to 4:30 p.m. ET

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

The heart-related manifestations of COVID-19 are a serious matter, but no one should make the mistake of thinking of COVID-19 as primarily a cardiac disease, according to Jeffrey C. Trost, MD, a cardiologist at Johns Hopkins University, Baltimore.

“One of my take-home messages is this is not a heart illness. This is still an infectious pulmonary illness that most likely causes stress on the heart in both healthy people and those with preexisting heart disease,” he said in offering a preview of his upcoming clinical update at HM20 Virtual, hosted by the Society of Hospital Medicine.

For this reason, in his clinical update talk, titled “COVID-19 and the Heart: What Every Hospitalist Should Know,” he’ll urge hospitalists to be conservative in ordering cardiac biomarker tests such troponin and natriuretic peptide levels. The focus should appropriately be on the subset of COVID-19 patients having the same symptoms suggestive of acute coronary syndrome, heart failure, or new-onset cardiomyopathy that would trigger laboratory testing in non–COVID-19 patients.

“Be more selective. Definitely do not routinely monitor troponin or [N-terminal of the prohormone brain natriuretic peptide] in patients just because they have COVID-19. A lot of patients with COVID-19 have these labs drawn, especially in the emergency department. We see a high signal-to-noise ratio: not infrequently the values are abnormal, and yet we don’t really know what that means,” said Dr. Trost, who is also director of the cardiac catheterization laboratory at Johns Hopkins Bayview Medical Center.

COVID-19 patients with preexisting heart disease are clearly at increased risk of severe forms of the infectious illness. In his talk, Dr. Trost will review the epidemiology of this association. He’ll also discuss the varied cardiac manifestations of COVID-19, consisting of myocarditis or other forms of new-onset cardiomyopathy, acute coronary syndrome, heart failure, and arrhythmias.

Many questions regarding COVID-19 and the heart remain unanswered for now, such as the mechanism and long-term implications of the phenomenon of ST-elevation acute coronary syndrome with chest pain in the presence of unobstructed coronary arteries, which Dr. Trost and others have encountered. Or the extent to which COVID-19–associated myocarditis is directly virus mediated as opposed to an autoimmune process.

“We’re relying completely on case reports at this point,” according to the cardiologist.

But one major issue has, thankfully, been put to rest on the basis of persuasive evidence which Dr. Trost plans to highlight: Millions of patients on ACE inhibitors or angiotensin receptor blockers can now rest assured that taking those medications doesn’t place them at increased risk of becoming infected with the novel coronavirus or, if infected, developing severe complications of COVID-19. Earlier in the pandemic that had been a legitimate theoretic concern based upon a plausible mechanism.

“I think we as physicians can now confidently say that we don’t need to stop these medicines in folks,” Dr. Trost said.

COVID-19 and the Heart: What Every Hospitalist Should Know

Live Q&A: Wednesday, Aug. 19, 3:30 p.m. to 4:30 p.m. ET

The heart-related manifestations of COVID-19 are a serious matter, but no one should make the mistake of thinking of COVID-19 as primarily a cardiac disease, according to Jeffrey C. Trost, MD, a cardiologist at Johns Hopkins University, Baltimore.

“One of my take-home messages is this is not a heart illness. This is still an infectious pulmonary illness that most likely causes stress on the heart in both healthy people and those with preexisting heart disease,” he said in offering a preview of his upcoming clinical update at HM20 Virtual, hosted by the Society of Hospital Medicine.

For this reason, in his clinical update talk, titled “COVID-19 and the Heart: What Every Hospitalist Should Know,” he’ll urge hospitalists to be conservative in ordering cardiac biomarker tests such troponin and natriuretic peptide levels. The focus should appropriately be on the subset of COVID-19 patients having the same symptoms suggestive of acute coronary syndrome, heart failure, or new-onset cardiomyopathy that would trigger laboratory testing in non–COVID-19 patients.

“Be more selective. Definitely do not routinely monitor troponin or [N-terminal of the prohormone brain natriuretic peptide] in patients just because they have COVID-19. A lot of patients with COVID-19 have these labs drawn, especially in the emergency department. We see a high signal-to-noise ratio: not infrequently the values are abnormal, and yet we don’t really know what that means,” said Dr. Trost, who is also director of the cardiac catheterization laboratory at Johns Hopkins Bayview Medical Center.

COVID-19 patients with preexisting heart disease are clearly at increased risk of severe forms of the infectious illness. In his talk, Dr. Trost will review the epidemiology of this association. He’ll also discuss the varied cardiac manifestations of COVID-19, consisting of myocarditis or other forms of new-onset cardiomyopathy, acute coronary syndrome, heart failure, and arrhythmias.

Many questions regarding COVID-19 and the heart remain unanswered for now, such as the mechanism and long-term implications of the phenomenon of ST-elevation acute coronary syndrome with chest pain in the presence of unobstructed coronary arteries, which Dr. Trost and others have encountered. Or the extent to which COVID-19–associated myocarditis is directly virus mediated as opposed to an autoimmune process.

“We’re relying completely on case reports at this point,” according to the cardiologist.

But one major issue has, thankfully, been put to rest on the basis of persuasive evidence which Dr. Trost plans to highlight: Millions of patients on ACE inhibitors or angiotensin receptor blockers can now rest assured that taking those medications doesn’t place them at increased risk of becoming infected with the novel coronavirus or, if infected, developing severe complications of COVID-19. Earlier in the pandemic that had been a legitimate theoretic concern based upon a plausible mechanism.

“I think we as physicians can now confidently say that we don’t need to stop these medicines in folks,” Dr. Trost said.

COVID-19 and the Heart: What Every Hospitalist Should Know

Live Q&A: Wednesday, Aug. 19, 3:30 p.m. to 4:30 p.m. ET

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

Scaly hand papule

Article Type
Changed
Thu, 08/13/2020 - 13:55
Display Headline
Scaly hand papule

Scaly hand papule

This pink raised nodule underlying a scaly surface was suspicious for squamous cell carcinoma (SCC). Since this was a virtual visit, and the lesion required pathology due to the likelihood of cancer, the patient was brought into the clinic for additional evaluation. A broad-based deep shave biopsy was performed to remove the visible lesion. Pathology showed SCC in situ, with borders uninvolved.

Patients who have had AKs are extremely likely to develop additional AKs. A notable percentage of AKs will, over time, develop into SCC in situ and then invasive SCC if not treated. While cryosurgery of an SK should not result in SCC, it’s most likely that in this case, an AK adjacent to the SK progressed to the SCC in situ.

There are multiple treatments available for SCC in situ. Topical imiquimod has been shown to be somewhat effective in stimulating the immune system, thus leading to resolution of SCC in situ. But there is a significant risk of recurrence. Topical 5-FU can be utilized on a daily or twice daily basis for 2 weeks (or up to several months). The risk of recurrence ranges from 7% to 33%. Electrodesiccation and curettage is often used for SCC in situ, with recurrence rates of 2% to 19%. Cryosurgery for SCC in situ requires an aggressive freeze, with freeze times of up to 30 seconds. Photodynamic therapy also is an option; however, it requires multiple sessions and is more costly than other treatment options.

This patient’s borders were uninvolved on pathology, but it was possible that there was some residual SCC in situ due to the standard “bread loaf slicing” used for routine pathology. To treat possible residual SCC in situ at the wound site and surrounding tissue, the patient was given a prescription for topical 5-FU to apply twice daily for 6 weeks. The patient was instructed to return for follow-up in 6 months, or sooner, if any problems arose.

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

References

Shimizu I, Cruz A, Chang KH, et al. Treatment of squamous cell carcinoma in situ: a review. Dermatol Surg. 2011;37:1394-1411.

Issue
The Journal of Family Practice - 69(6)
Publications
Topics
Sections

Scaly hand papule

This pink raised nodule underlying a scaly surface was suspicious for squamous cell carcinoma (SCC). Since this was a virtual visit, and the lesion required pathology due to the likelihood of cancer, the patient was brought into the clinic for additional evaluation. A broad-based deep shave biopsy was performed to remove the visible lesion. Pathology showed SCC in situ, with borders uninvolved.

Patients who have had AKs are extremely likely to develop additional AKs. A notable percentage of AKs will, over time, develop into SCC in situ and then invasive SCC if not treated. While cryosurgery of an SK should not result in SCC, it’s most likely that in this case, an AK adjacent to the SK progressed to the SCC in situ.

There are multiple treatments available for SCC in situ. Topical imiquimod has been shown to be somewhat effective in stimulating the immune system, thus leading to resolution of SCC in situ. But there is a significant risk of recurrence. Topical 5-FU can be utilized on a daily or twice daily basis for 2 weeks (or up to several months). The risk of recurrence ranges from 7% to 33%. Electrodesiccation and curettage is often used for SCC in situ, with recurrence rates of 2% to 19%. Cryosurgery for SCC in situ requires an aggressive freeze, with freeze times of up to 30 seconds. Photodynamic therapy also is an option; however, it requires multiple sessions and is more costly than other treatment options.

This patient’s borders were uninvolved on pathology, but it was possible that there was some residual SCC in situ due to the standard “bread loaf slicing” used for routine pathology. To treat possible residual SCC in situ at the wound site and surrounding tissue, the patient was given a prescription for topical 5-FU to apply twice daily for 6 weeks. The patient was instructed to return for follow-up in 6 months, or sooner, if any problems arose.

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

Scaly hand papule

This pink raised nodule underlying a scaly surface was suspicious for squamous cell carcinoma (SCC). Since this was a virtual visit, and the lesion required pathology due to the likelihood of cancer, the patient was brought into the clinic for additional evaluation. A broad-based deep shave biopsy was performed to remove the visible lesion. Pathology showed SCC in situ, with borders uninvolved.

Patients who have had AKs are extremely likely to develop additional AKs. A notable percentage of AKs will, over time, develop into SCC in situ and then invasive SCC if not treated. While cryosurgery of an SK should not result in SCC, it’s most likely that in this case, an AK adjacent to the SK progressed to the SCC in situ.

There are multiple treatments available for SCC in situ. Topical imiquimod has been shown to be somewhat effective in stimulating the immune system, thus leading to resolution of SCC in situ. But there is a significant risk of recurrence. Topical 5-FU can be utilized on a daily or twice daily basis for 2 weeks (or up to several months). The risk of recurrence ranges from 7% to 33%. Electrodesiccation and curettage is often used for SCC in situ, with recurrence rates of 2% to 19%. Cryosurgery for SCC in situ requires an aggressive freeze, with freeze times of up to 30 seconds. Photodynamic therapy also is an option; however, it requires multiple sessions and is more costly than other treatment options.

This patient’s borders were uninvolved on pathology, but it was possible that there was some residual SCC in situ due to the standard “bread loaf slicing” used for routine pathology. To treat possible residual SCC in situ at the wound site and surrounding tissue, the patient was given a prescription for topical 5-FU to apply twice daily for 6 weeks. The patient was instructed to return for follow-up in 6 months, or sooner, if any problems arose.

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

References

Shimizu I, Cruz A, Chang KH, et al. Treatment of squamous cell carcinoma in situ: a review. Dermatol Surg. 2011;37:1394-1411.

References

Shimizu I, Cruz A, Chang KH, et al. Treatment of squamous cell carcinoma in situ: a review. Dermatol Surg. 2011;37:1394-1411.

Issue
The Journal of Family Practice - 69(6)
Issue
The Journal of Family Practice - 69(6)
Publications
Publications
Topics
Article Type
Display Headline
Scaly hand papule
Display Headline
Scaly hand papule
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 08/12/2020 - 15:00
Un-Gate On Date
Wed, 08/12/2020 - 15:00
Use ProPublica
CFC Schedule Remove Status
Wed, 08/12/2020 - 15:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article

Psoriasis in Patients of Color: Differences in Morphology, Clinical Presentation, and Treatment

Article Type
Changed
Thu, 10/29/2020 - 15:06

Psoriasis is a chronic inflammatory skin disease that affects 2% to 3% of individuals worldwide.1 Despite extensive research, the majority of clinical data are in white patients with limited data in patients of color, yet a number of differences are known. The prevalence of psoriasis differs among racial and ethnic groups, with lower prevalence in racial minorities.2 A cross-sectional American study using data from 2009 through 2010 showed the prevalence for psoriasis was 3.6% in white patients, 1.9% in black patients, 1.6% in Hispanic patients, and 1.4% in other racial groups.3 Psoriasis presents differently in patients of color, both in morphology and severity. Cultural differences and stigma may contribute to the differences seen in severity but also to the psychological impact and treatment choices in patients of color compared to white patients.4 It has even been theorized that treatment efficacy could differ because of potential genetic differences.5 Psoriasis in patients of color is an emerging clinical issue that requires further attention so that dermatologists can learn about, diagnose, and treat them.

We report 3 cases of patients of color with psoriasis who presented to an urban and racially diverse dermatology clinic affiliated with Scarborough General Hospital in Toronto, Ontario, Canada. A retrospective chart review was performed on these high-yield representative cases to demonstrate differences in color and morphology, disease severity, and treatment in patients of various races seen at our clinic. After informed consent was obtained, photographs were taken of patient cutaneous findings to illustrate these differences. Discussion with these selected patients yielded supplementary qualitative data, highlighting individual perspectives of their disease.

Case Series

Patient 1
A 53-year-old black man from Grenada presented to our clinic with a history of psoriasis for a number of years that presented as violaceous plaques throughout large portions of the body (Figure 1). He previously had achieved inadequate results while using topical therapies, methotrexate, acitretin, apremilast, ustekinumab, ixekizumab, and guselkumab at adequate or even maximum doses. His disease affected 30% of the body surface area, with a psoriasis area and severity index score of 27 and a dermatology life quality index score of 23. The patient’s life was quite affected by psoriasis, with emphasis on choice of clothing worn and effect on body image. He also discussed the stigma psoriasis may have in black patients, stating that he has been told multiple times that “black people do not get psoriasis.”

Figure 1. A and B, Violaceous plaques on the back and arm in a black patient with psoriasis.

Patient 2
A 27-year-old man from India presented with guttate psoriasis (Figure 2). He was treated with methotrexate 2 years prior and currently is on maintenance therapy with topical treatments alone. His main concerns pertained to the persistent dyschromia that occurred secondary to the psoriatic lesions. Through discussion, the patient stated that he “would do anything to get rid of it.”

Figure 2. A and B, Postinflammatory hyperpigmentation from guttate psoriasis in a patient from India


Patient 3
A 49-year-old man from the Philippines presented to our clinic with plaque psoriasis that predominantly affected the trunk and scalp (Figure 3). He had been treated with methotrexate and phototherapy with suboptimal efficacy and was planning for biologic therapy. Although he had active plaques on the trunk, the patient stated, “I am most bothered by my scalp,” particularly referring to the itch and scale and their effects on hair and hairstyling.

Figure 3. A and B, Plaque psoriasis on the scalp and back, respectively, in a patient from the Philippines.

Comment

Clinical differences in patients of color with psoriasis affect the management of the disease. Special consideration should be given to variances in morphology, presentation, treatment, and psychosocial factors in the management of psoriasis for these patient populations, as summarized in the eTable.

Morphology
At our clinic, patients of color have been found to have differences in morphology, including lesions that are more violaceous in color, as seen in patient 1; less noticeable inflammation; and more postinflammatory hypopigmentation and hyperpigmentation changes, as seen in patient 2. These changes are supported by the literature and differ from typical psoriasis plaques, which are pink-red and have more overlying scale. The varied morphology also may affect the differential, and other mimickers may be considered, such as lichen planus, cutaneous lupus erythematosus, and sarcoidosis.2



Presentation
There are differences in presentation among patients of color, particularly in distribution, type of psoriasis, and severity. As seen in patient 3, Asian and black patients are more likely to present with scalp psoriasis.2,5 Hairstyling and hair care practices can differ considerably between racial groups. Given the differences in hairstyling, scalp psoriasis also may have a greater impact on patient quality of life (QOL).

 

 



Racial differences affect the type of psoriasis seen. Asian patients are more likely to present with pustular and erythrodermic psoriasis and less likely to present with inverse psoriasis compared to white patients. Hispanic patients are more likely to present with pustular psoriasis.11 Black patients have been reported to have lower frequencies of psoriatic arthritis compared to white patients.12 Recognition of these differences may help guide initial choice for therapeutics.



Notably, patients of color may present with much more severe psoriasis, particularly Asian and Hispanic patients.7 One retrospective study looking at patients with psoriasis treated with etanercept found that Asian patients were more likely to have greater baseline body surface area involvement.6 An American cross-sectional study reported higher psoriasis area and severity index scores in black patients compared to white patients,12 possibly because patients of color do not normalize the experience of having psoriasis and feel stigmatized, which can cause delays in seeking medical attention and worsen disease burden. For patient 1, the stigma of black patients having psoriasis affected his body image and may have led to a delay in seeking medical attention due to him not believing it was possible for people of his skin color to have psoriasis. Increased disease severity may contribute to treatment resistance or numerous trials of topicals or biologics before the disease improves. Patient education in the community as well as patient support groups are paramount, and increased awareness of psoriasis can help improve disease management.

Treatment
Topical therapies are the first-line treatment of psoriasis. Although there is no evidence showing differences in topical treatment efficacy, patient preference for different topical treatments may vary based on race. For example, patients with Afro-textured hair may prefer foams and lotions and would avoid shampoo therapies, as frequent hair washing may not be feasible with certain hairstyles and may cause hair breakage or dryness.2

UV therapy can be an effective treatment modality for patients with psoriasis. The strength of therapy tends to be dictated by the Fitzpatrick skin phototype rather than race. Darker-skinned individuals may have an increased risk for hyperpigmentation, so caution should be taken to prevent burning during therapy. Suberythemogenic dosing—70% of minimal erythema dose—of narrowband UVB treatments has shown the same efficacy as using minimal erythema dose in patients with darker skin types in addition to fair-skinned patients.8

Although we found poor efficacy of systemic treatments in patient 1, to our knowledge, studies examining the efficacy of systemic therapeutic options have not shown differences in patients of color.6,13 Studies show similar efficacy in treatments among races, particularly biologic therapies.5 However, patients with skin of color historically have been underrepresented in clinical trials,9 which may contribute to these patients, particularly black patients, being less familiar with biologics as a treatment option for psoriasis, as reported by Takeshita et al.10 Therefore, patient-centered discussions regarding treatment choices are important to ensure patients understand all options available to manage their disease.

Psychosocial Impact
Because of its chronic remitting course, psoriasis has a notable psychosocial impact on the lives of all patients, though the literature suggests there may be more of an impact on QOL in patients of color. Higher baseline dermatology life quality index scores have been reported in patients of color compared to white patients.6 Kerr et al12 reported significantly greater psoriasis area and severity index scores (P=.06) and greater psychological impact in black patients compared to white patients. Stress also was more likely to be reported as a trigger for psoriasis in patients of Hispanic background compared to white patients.14 Many patients report body image issues with large physical lesions; however, the difference may lie in personal and cultural views about psoriasis, as one of our patients stated, “black people do not get psoriasis.” In addition to the cosmetic challenges that patients face with active lesions, postinflammatory pigmentary changes can be equally as burdensome to patients, as one of our patients stated he “would do anything to get rid of it.” Increased rates of depression and anxiety in patients of color can worsen their outlook on the condition.15,16 The increased stigma and burden of psoriasis in patients of color calls for clinicians to counsel and address psoriasis in a holistic way and refer patients to psoriasis support groups when appropriate. Although the burden of psoriasis is clear, more studies can be carried out to investigate the impact on QOL in different ethnic populations.

Dermatology Education
Although differences have been found in patients of color with psoriasis, dissemination of this knowledge continues to be a challenge. In dermatology residency programs, the majority of teaching is provided with examples of skin diseases in white patients, which can complicate pattern recognition and diagnostic ability for trainees. Although dermatologists recognize that ethnic skin has unique dermatologic considerations, there is a persistent need for increasing skin of color education within dermatology residency programs.17,18 Implementing more educational programs on skin of color has been proposed, and these programs will continue to be in demand as our population increasingly diversifies.19

Conclusion

Psoriasis in patients of color carries unique challenges when compared to psoriasis in white patients. Differences in morphology and presentation can make the disease difficult to accurately diagnose. These differences in addition to cultural differences may contribute to a greater impact on QOL and psychological health. Although treatment preferences and recognition may differ, treatment efficacy has so far been similar, albeit with a low proportion of patients with skin of color included in clinical trials.

Further focus should now lie within knowledge translation of these differences, which would normalize the condition for patients, support them seeking medical attention sooner, and inform them of all treatment options possible. For clinicians, more attention on the differences would help make earlier diagnoses, personalize physician-patient conversations, and advocate for further education on this issue in residency training programs.

References
  1. National Psoriasis Foundation. Statistics. https://www.psoriasis.org/content/statistics. Accessed July 14, 2020.
  2.  Alexis AF, Blackcloud P. Psoriasis in skin of color: epidemiology, genetics, clinical presentation, and treatment nuances. J Clin Aesthet Dermatol. 2014;7:16-24.
  3. Rachakonda TD, Schupp CW, Armstrong AW. Psoriasis prevalence among adults in the United States. J Am Acad Dermatol. 2014;70:512-516.
  4. Goff KL, Karimkhani C, Boyers LN, et al. The global burden of psoriatic skin disease. Br J Dermatol. 2015;172:1665-1668.
  5. Kaufman BP, Alexis AF. Psoriasis in skin of color: insights into the epidemiology, clinical presentation, genetics, quality-of-life impact, and treatment of psoriasis in non-white racial/ethnic groups. Am J Clin Dermatol. 2018;19:405-423.
  6. Shah SK, Arthur A, Yang YC, et al. A retrospective study to investigate racial and ethnic variations in the treatment of psoriasis with etanercept. J Drugs Dermatol. 2011;10:866-872.
  7. Abrouk M, Lee K, Brodsky M, et al. Ethnicity affects the presenting severity of psoriasis. J Am Acad Dermatol. 2017;77:180-182.
  8. Youssef RM, Mahgoub D, Mashaly HM, et al. Different narrowband UVB dosage regimens in dark skinned psoriatics: a preliminary study. Photodermatol Photoimmunol Photomed. 2008;24:256-259.
  9. Charrow A, Xia F Di, Joyce C, et al. Diversity in dermatology clinical trials: a systematic review. JAMA Dermatol. 2017;153:193-198.
  10. Takeshita J, Eriksen WT, Raziano VT, et al. Racial differences in perceptions of psoriasis therapies: implications for racial disparities in psoriasis treatment. J Invest Dermatol. 2019;139:1672-1679.
  11. Yan D, Afifi L, Jeon C, et al. A cross-sectional study of the distribution of psoriasis subtypes in different ethno-racial groups. Dermatol Online J. 2018;24. pii:13030/qt5z21q4k2.
  12. Kerr GS, Qaiyumi S, Richards J, et al. Psoriasis and psoriatic arthritis in African-American patients—the need to measure disease burden. Clin Rheumatol. 2015;34:1753-1759.
  13. Edson-Heredia E, Sterling KL, Alatorre CI, et al. Heterogeneity of response to biologic treatment: perspective for psoriasis. J Invest Dermatol. 2014;134:18-23.
  14. Yan D, Afifi L, Jeon C, et al. A cross-sectional study of psoriasis triggers among different ethno-racial groups. J Am Acad Dermatol. 2017;77:756-758.
  15.  Bailey RK, Mokonogho J, Kumar A. Racial and ethnic differences in depression: current perspectives. Neuropsychiatr Dis Treat. 2019;15:603-609.
  16.  Jackson C, Maibach H. Ethnic and socioeconomic disparities in dermatology. J Dermatolog Treat. 2016;27:290-291.
  17. Salam A, Dadzie OE. Dermatology training in the U.K.: does it reflect the changing demographics of our population? Br J Dermatol. 2013;169:1360-1362.
  18. Nijhawan RI, Jacob SE, Woolery-Lloyd H. Skin of color education in dermatology residency programs: does residency training reflect the changing demographics of the United States? J Am Acad Dermatol. 2008;59:615-618.
  19. Ogunyemi B, Miller-Monthrope Y. The state of ethnic dermatology in Canada. J Cutan Med Surg. 2017;21:464-466.
Article PDF
Author and Disclosure Information

From the University of Toronto, Ontario, Canada. Drs. Nicholas and Hessami-Booshehri are from the Division of Dermatology, Department of Medicine, and Ms. Chan is from the Faculty of Medicine. Dr. Nicholas also is from the Division of Dermatology, Women’s College Hospital, Toronto. Dr. Hessami-Booshehri also is from Scarborough General Hospital, Ontario, Canada.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Mathew N. Nicholas, MD, Division of Dermatology, Women’s College Hospital, 3rd Floor, 76 Grenville St, Toronto, ON M5S 1B2 Canada ([email protected]).

Issue
Cutis - 106(2S)
Publications
Topics
Page Number
7-10, E10
Sections
Author and Disclosure Information

From the University of Toronto, Ontario, Canada. Drs. Nicholas and Hessami-Booshehri are from the Division of Dermatology, Department of Medicine, and Ms. Chan is from the Faculty of Medicine. Dr. Nicholas also is from the Division of Dermatology, Women’s College Hospital, Toronto. Dr. Hessami-Booshehri also is from Scarborough General Hospital, Ontario, Canada.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Mathew N. Nicholas, MD, Division of Dermatology, Women’s College Hospital, 3rd Floor, 76 Grenville St, Toronto, ON M5S 1B2 Canada ([email protected]).

Author and Disclosure Information

From the University of Toronto, Ontario, Canada. Drs. Nicholas and Hessami-Booshehri are from the Division of Dermatology, Department of Medicine, and Ms. Chan is from the Faculty of Medicine. Dr. Nicholas also is from the Division of Dermatology, Women’s College Hospital, Toronto. Dr. Hessami-Booshehri also is from Scarborough General Hospital, Ontario, Canada.

The authors report no conflict of interest.

The eTable is available in the Appendix online at www.mdedge.com/dermatology.

Correspondence: Mathew N. Nicholas, MD, Division of Dermatology, Women’s College Hospital, 3rd Floor, 76 Grenville St, Toronto, ON M5S 1B2 Canada ([email protected]).

Article PDF
Article PDF

Psoriasis is a chronic inflammatory skin disease that affects 2% to 3% of individuals worldwide.1 Despite extensive research, the majority of clinical data are in white patients with limited data in patients of color, yet a number of differences are known. The prevalence of psoriasis differs among racial and ethnic groups, with lower prevalence in racial minorities.2 A cross-sectional American study using data from 2009 through 2010 showed the prevalence for psoriasis was 3.6% in white patients, 1.9% in black patients, 1.6% in Hispanic patients, and 1.4% in other racial groups.3 Psoriasis presents differently in patients of color, both in morphology and severity. Cultural differences and stigma may contribute to the differences seen in severity but also to the psychological impact and treatment choices in patients of color compared to white patients.4 It has even been theorized that treatment efficacy could differ because of potential genetic differences.5 Psoriasis in patients of color is an emerging clinical issue that requires further attention so that dermatologists can learn about, diagnose, and treat them.

We report 3 cases of patients of color with psoriasis who presented to an urban and racially diverse dermatology clinic affiliated with Scarborough General Hospital in Toronto, Ontario, Canada. A retrospective chart review was performed on these high-yield representative cases to demonstrate differences in color and morphology, disease severity, and treatment in patients of various races seen at our clinic. After informed consent was obtained, photographs were taken of patient cutaneous findings to illustrate these differences. Discussion with these selected patients yielded supplementary qualitative data, highlighting individual perspectives of their disease.

Case Series

Patient 1
A 53-year-old black man from Grenada presented to our clinic with a history of psoriasis for a number of years that presented as violaceous plaques throughout large portions of the body (Figure 1). He previously had achieved inadequate results while using topical therapies, methotrexate, acitretin, apremilast, ustekinumab, ixekizumab, and guselkumab at adequate or even maximum doses. His disease affected 30% of the body surface area, with a psoriasis area and severity index score of 27 and a dermatology life quality index score of 23. The patient’s life was quite affected by psoriasis, with emphasis on choice of clothing worn and effect on body image. He also discussed the stigma psoriasis may have in black patients, stating that he has been told multiple times that “black people do not get psoriasis.”

Figure 1. A and B, Violaceous plaques on the back and arm in a black patient with psoriasis.

Patient 2
A 27-year-old man from India presented with guttate psoriasis (Figure 2). He was treated with methotrexate 2 years prior and currently is on maintenance therapy with topical treatments alone. His main concerns pertained to the persistent dyschromia that occurred secondary to the psoriatic lesions. Through discussion, the patient stated that he “would do anything to get rid of it.”

Figure 2. A and B, Postinflammatory hyperpigmentation from guttate psoriasis in a patient from India


Patient 3
A 49-year-old man from the Philippines presented to our clinic with plaque psoriasis that predominantly affected the trunk and scalp (Figure 3). He had been treated with methotrexate and phototherapy with suboptimal efficacy and was planning for biologic therapy. Although he had active plaques on the trunk, the patient stated, “I am most bothered by my scalp,” particularly referring to the itch and scale and their effects on hair and hairstyling.

Figure 3. A and B, Plaque psoriasis on the scalp and back, respectively, in a patient from the Philippines.

Comment

Clinical differences in patients of color with psoriasis affect the management of the disease. Special consideration should be given to variances in morphology, presentation, treatment, and psychosocial factors in the management of psoriasis for these patient populations, as summarized in the eTable.

Morphology
At our clinic, patients of color have been found to have differences in morphology, including lesions that are more violaceous in color, as seen in patient 1; less noticeable inflammation; and more postinflammatory hypopigmentation and hyperpigmentation changes, as seen in patient 2. These changes are supported by the literature and differ from typical psoriasis plaques, which are pink-red and have more overlying scale. The varied morphology also may affect the differential, and other mimickers may be considered, such as lichen planus, cutaneous lupus erythematosus, and sarcoidosis.2



Presentation
There are differences in presentation among patients of color, particularly in distribution, type of psoriasis, and severity. As seen in patient 3, Asian and black patients are more likely to present with scalp psoriasis.2,5 Hairstyling and hair care practices can differ considerably between racial groups. Given the differences in hairstyling, scalp psoriasis also may have a greater impact on patient quality of life (QOL).

 

 



Racial differences affect the type of psoriasis seen. Asian patients are more likely to present with pustular and erythrodermic psoriasis and less likely to present with inverse psoriasis compared to white patients. Hispanic patients are more likely to present with pustular psoriasis.11 Black patients have been reported to have lower frequencies of psoriatic arthritis compared to white patients.12 Recognition of these differences may help guide initial choice for therapeutics.



Notably, patients of color may present with much more severe psoriasis, particularly Asian and Hispanic patients.7 One retrospective study looking at patients with psoriasis treated with etanercept found that Asian patients were more likely to have greater baseline body surface area involvement.6 An American cross-sectional study reported higher psoriasis area and severity index scores in black patients compared to white patients,12 possibly because patients of color do not normalize the experience of having psoriasis and feel stigmatized, which can cause delays in seeking medical attention and worsen disease burden. For patient 1, the stigma of black patients having psoriasis affected his body image and may have led to a delay in seeking medical attention due to him not believing it was possible for people of his skin color to have psoriasis. Increased disease severity may contribute to treatment resistance or numerous trials of topicals or biologics before the disease improves. Patient education in the community as well as patient support groups are paramount, and increased awareness of psoriasis can help improve disease management.

Treatment
Topical therapies are the first-line treatment of psoriasis. Although there is no evidence showing differences in topical treatment efficacy, patient preference for different topical treatments may vary based on race. For example, patients with Afro-textured hair may prefer foams and lotions and would avoid shampoo therapies, as frequent hair washing may not be feasible with certain hairstyles and may cause hair breakage or dryness.2

UV therapy can be an effective treatment modality for patients with psoriasis. The strength of therapy tends to be dictated by the Fitzpatrick skin phototype rather than race. Darker-skinned individuals may have an increased risk for hyperpigmentation, so caution should be taken to prevent burning during therapy. Suberythemogenic dosing—70% of minimal erythema dose—of narrowband UVB treatments has shown the same efficacy as using minimal erythema dose in patients with darker skin types in addition to fair-skinned patients.8

Although we found poor efficacy of systemic treatments in patient 1, to our knowledge, studies examining the efficacy of systemic therapeutic options have not shown differences in patients of color.6,13 Studies show similar efficacy in treatments among races, particularly biologic therapies.5 However, patients with skin of color historically have been underrepresented in clinical trials,9 which may contribute to these patients, particularly black patients, being less familiar with biologics as a treatment option for psoriasis, as reported by Takeshita et al.10 Therefore, patient-centered discussions regarding treatment choices are important to ensure patients understand all options available to manage their disease.

Psychosocial Impact
Because of its chronic remitting course, psoriasis has a notable psychosocial impact on the lives of all patients, though the literature suggests there may be more of an impact on QOL in patients of color. Higher baseline dermatology life quality index scores have been reported in patients of color compared to white patients.6 Kerr et al12 reported significantly greater psoriasis area and severity index scores (P=.06) and greater psychological impact in black patients compared to white patients. Stress also was more likely to be reported as a trigger for psoriasis in patients of Hispanic background compared to white patients.14 Many patients report body image issues with large physical lesions; however, the difference may lie in personal and cultural views about psoriasis, as one of our patients stated, “black people do not get psoriasis.” In addition to the cosmetic challenges that patients face with active lesions, postinflammatory pigmentary changes can be equally as burdensome to patients, as one of our patients stated he “would do anything to get rid of it.” Increased rates of depression and anxiety in patients of color can worsen their outlook on the condition.15,16 The increased stigma and burden of psoriasis in patients of color calls for clinicians to counsel and address psoriasis in a holistic way and refer patients to psoriasis support groups when appropriate. Although the burden of psoriasis is clear, more studies can be carried out to investigate the impact on QOL in different ethnic populations.

Dermatology Education
Although differences have been found in patients of color with psoriasis, dissemination of this knowledge continues to be a challenge. In dermatology residency programs, the majority of teaching is provided with examples of skin diseases in white patients, which can complicate pattern recognition and diagnostic ability for trainees. Although dermatologists recognize that ethnic skin has unique dermatologic considerations, there is a persistent need for increasing skin of color education within dermatology residency programs.17,18 Implementing more educational programs on skin of color has been proposed, and these programs will continue to be in demand as our population increasingly diversifies.19

Conclusion

Psoriasis in patients of color carries unique challenges when compared to psoriasis in white patients. Differences in morphology and presentation can make the disease difficult to accurately diagnose. These differences in addition to cultural differences may contribute to a greater impact on QOL and psychological health. Although treatment preferences and recognition may differ, treatment efficacy has so far been similar, albeit with a low proportion of patients with skin of color included in clinical trials.

Further focus should now lie within knowledge translation of these differences, which would normalize the condition for patients, support them seeking medical attention sooner, and inform them of all treatment options possible. For clinicians, more attention on the differences would help make earlier diagnoses, personalize physician-patient conversations, and advocate for further education on this issue in residency training programs.

Psoriasis is a chronic inflammatory skin disease that affects 2% to 3% of individuals worldwide.1 Despite extensive research, the majority of clinical data are in white patients with limited data in patients of color, yet a number of differences are known. The prevalence of psoriasis differs among racial and ethnic groups, with lower prevalence in racial minorities.2 A cross-sectional American study using data from 2009 through 2010 showed the prevalence for psoriasis was 3.6% in white patients, 1.9% in black patients, 1.6% in Hispanic patients, and 1.4% in other racial groups.3 Psoriasis presents differently in patients of color, both in morphology and severity. Cultural differences and stigma may contribute to the differences seen in severity but also to the psychological impact and treatment choices in patients of color compared to white patients.4 It has even been theorized that treatment efficacy could differ because of potential genetic differences.5 Psoriasis in patients of color is an emerging clinical issue that requires further attention so that dermatologists can learn about, diagnose, and treat them.

We report 3 cases of patients of color with psoriasis who presented to an urban and racially diverse dermatology clinic affiliated with Scarborough General Hospital in Toronto, Ontario, Canada. A retrospective chart review was performed on these high-yield representative cases to demonstrate differences in color and morphology, disease severity, and treatment in patients of various races seen at our clinic. After informed consent was obtained, photographs were taken of patient cutaneous findings to illustrate these differences. Discussion with these selected patients yielded supplementary qualitative data, highlighting individual perspectives of their disease.

Case Series

Patient 1
A 53-year-old black man from Grenada presented to our clinic with a history of psoriasis for a number of years that presented as violaceous plaques throughout large portions of the body (Figure 1). He previously had achieved inadequate results while using topical therapies, methotrexate, acitretin, apremilast, ustekinumab, ixekizumab, and guselkumab at adequate or even maximum doses. His disease affected 30% of the body surface area, with a psoriasis area and severity index score of 27 and a dermatology life quality index score of 23. The patient’s life was quite affected by psoriasis, with emphasis on choice of clothing worn and effect on body image. He also discussed the stigma psoriasis may have in black patients, stating that he has been told multiple times that “black people do not get psoriasis.”

Figure 1. A and B, Violaceous plaques on the back and arm in a black patient with psoriasis.

Patient 2
A 27-year-old man from India presented with guttate psoriasis (Figure 2). He was treated with methotrexate 2 years prior and currently is on maintenance therapy with topical treatments alone. His main concerns pertained to the persistent dyschromia that occurred secondary to the psoriatic lesions. Through discussion, the patient stated that he “would do anything to get rid of it.”

Figure 2. A and B, Postinflammatory hyperpigmentation from guttate psoriasis in a patient from India


Patient 3
A 49-year-old man from the Philippines presented to our clinic with plaque psoriasis that predominantly affected the trunk and scalp (Figure 3). He had been treated with methotrexate and phototherapy with suboptimal efficacy and was planning for biologic therapy. Although he had active plaques on the trunk, the patient stated, “I am most bothered by my scalp,” particularly referring to the itch and scale and their effects on hair and hairstyling.

Figure 3. A and B, Plaque psoriasis on the scalp and back, respectively, in a patient from the Philippines.

Comment

Clinical differences in patients of color with psoriasis affect the management of the disease. Special consideration should be given to variances in morphology, presentation, treatment, and psychosocial factors in the management of psoriasis for these patient populations, as summarized in the eTable.

Morphology
At our clinic, patients of color have been found to have differences in morphology, including lesions that are more violaceous in color, as seen in patient 1; less noticeable inflammation; and more postinflammatory hypopigmentation and hyperpigmentation changes, as seen in patient 2. These changes are supported by the literature and differ from typical psoriasis plaques, which are pink-red and have more overlying scale. The varied morphology also may affect the differential, and other mimickers may be considered, such as lichen planus, cutaneous lupus erythematosus, and sarcoidosis.2



Presentation
There are differences in presentation among patients of color, particularly in distribution, type of psoriasis, and severity. As seen in patient 3, Asian and black patients are more likely to present with scalp psoriasis.2,5 Hairstyling and hair care practices can differ considerably between racial groups. Given the differences in hairstyling, scalp psoriasis also may have a greater impact on patient quality of life (QOL).

 

 



Racial differences affect the type of psoriasis seen. Asian patients are more likely to present with pustular and erythrodermic psoriasis and less likely to present with inverse psoriasis compared to white patients. Hispanic patients are more likely to present with pustular psoriasis.11 Black patients have been reported to have lower frequencies of psoriatic arthritis compared to white patients.12 Recognition of these differences may help guide initial choice for therapeutics.



Notably, patients of color may present with much more severe psoriasis, particularly Asian and Hispanic patients.7 One retrospective study looking at patients with psoriasis treated with etanercept found that Asian patients were more likely to have greater baseline body surface area involvement.6 An American cross-sectional study reported higher psoriasis area and severity index scores in black patients compared to white patients,12 possibly because patients of color do not normalize the experience of having psoriasis and feel stigmatized, which can cause delays in seeking medical attention and worsen disease burden. For patient 1, the stigma of black patients having psoriasis affected his body image and may have led to a delay in seeking medical attention due to him not believing it was possible for people of his skin color to have psoriasis. Increased disease severity may contribute to treatment resistance or numerous trials of topicals or biologics before the disease improves. Patient education in the community as well as patient support groups are paramount, and increased awareness of psoriasis can help improve disease management.

Treatment
Topical therapies are the first-line treatment of psoriasis. Although there is no evidence showing differences in topical treatment efficacy, patient preference for different topical treatments may vary based on race. For example, patients with Afro-textured hair may prefer foams and lotions and would avoid shampoo therapies, as frequent hair washing may not be feasible with certain hairstyles and may cause hair breakage or dryness.2

UV therapy can be an effective treatment modality for patients with psoriasis. The strength of therapy tends to be dictated by the Fitzpatrick skin phototype rather than race. Darker-skinned individuals may have an increased risk for hyperpigmentation, so caution should be taken to prevent burning during therapy. Suberythemogenic dosing—70% of minimal erythema dose—of narrowband UVB treatments has shown the same efficacy as using minimal erythema dose in patients with darker skin types in addition to fair-skinned patients.8

Although we found poor efficacy of systemic treatments in patient 1, to our knowledge, studies examining the efficacy of systemic therapeutic options have not shown differences in patients of color.6,13 Studies show similar efficacy in treatments among races, particularly biologic therapies.5 However, patients with skin of color historically have been underrepresented in clinical trials,9 which may contribute to these patients, particularly black patients, being less familiar with biologics as a treatment option for psoriasis, as reported by Takeshita et al.10 Therefore, patient-centered discussions regarding treatment choices are important to ensure patients understand all options available to manage their disease.

Psychosocial Impact
Because of its chronic remitting course, psoriasis has a notable psychosocial impact on the lives of all patients, though the literature suggests there may be more of an impact on QOL in patients of color. Higher baseline dermatology life quality index scores have been reported in patients of color compared to white patients.6 Kerr et al12 reported significantly greater psoriasis area and severity index scores (P=.06) and greater psychological impact in black patients compared to white patients. Stress also was more likely to be reported as a trigger for psoriasis in patients of Hispanic background compared to white patients.14 Many patients report body image issues with large physical lesions; however, the difference may lie in personal and cultural views about psoriasis, as one of our patients stated, “black people do not get psoriasis.” In addition to the cosmetic challenges that patients face with active lesions, postinflammatory pigmentary changes can be equally as burdensome to patients, as one of our patients stated he “would do anything to get rid of it.” Increased rates of depression and anxiety in patients of color can worsen their outlook on the condition.15,16 The increased stigma and burden of psoriasis in patients of color calls for clinicians to counsel and address psoriasis in a holistic way and refer patients to psoriasis support groups when appropriate. Although the burden of psoriasis is clear, more studies can be carried out to investigate the impact on QOL in different ethnic populations.

Dermatology Education
Although differences have been found in patients of color with psoriasis, dissemination of this knowledge continues to be a challenge. In dermatology residency programs, the majority of teaching is provided with examples of skin diseases in white patients, which can complicate pattern recognition and diagnostic ability for trainees. Although dermatologists recognize that ethnic skin has unique dermatologic considerations, there is a persistent need for increasing skin of color education within dermatology residency programs.17,18 Implementing more educational programs on skin of color has been proposed, and these programs will continue to be in demand as our population increasingly diversifies.19

Conclusion

Psoriasis in patients of color carries unique challenges when compared to psoriasis in white patients. Differences in morphology and presentation can make the disease difficult to accurately diagnose. These differences in addition to cultural differences may contribute to a greater impact on QOL and psychological health. Although treatment preferences and recognition may differ, treatment efficacy has so far been similar, albeit with a low proportion of patients with skin of color included in clinical trials.

Further focus should now lie within knowledge translation of these differences, which would normalize the condition for patients, support them seeking medical attention sooner, and inform them of all treatment options possible. For clinicians, more attention on the differences would help make earlier diagnoses, personalize physician-patient conversations, and advocate for further education on this issue in residency training programs.

References
  1. National Psoriasis Foundation. Statistics. https://www.psoriasis.org/content/statistics. Accessed July 14, 2020.
  2.  Alexis AF, Blackcloud P. Psoriasis in skin of color: epidemiology, genetics, clinical presentation, and treatment nuances. J Clin Aesthet Dermatol. 2014;7:16-24.
  3. Rachakonda TD, Schupp CW, Armstrong AW. Psoriasis prevalence among adults in the United States. J Am Acad Dermatol. 2014;70:512-516.
  4. Goff KL, Karimkhani C, Boyers LN, et al. The global burden of psoriatic skin disease. Br J Dermatol. 2015;172:1665-1668.
  5. Kaufman BP, Alexis AF. Psoriasis in skin of color: insights into the epidemiology, clinical presentation, genetics, quality-of-life impact, and treatment of psoriasis in non-white racial/ethnic groups. Am J Clin Dermatol. 2018;19:405-423.
  6. Shah SK, Arthur A, Yang YC, et al. A retrospective study to investigate racial and ethnic variations in the treatment of psoriasis with etanercept. J Drugs Dermatol. 2011;10:866-872.
  7. Abrouk M, Lee K, Brodsky M, et al. Ethnicity affects the presenting severity of psoriasis. J Am Acad Dermatol. 2017;77:180-182.
  8. Youssef RM, Mahgoub D, Mashaly HM, et al. Different narrowband UVB dosage regimens in dark skinned psoriatics: a preliminary study. Photodermatol Photoimmunol Photomed. 2008;24:256-259.
  9. Charrow A, Xia F Di, Joyce C, et al. Diversity in dermatology clinical trials: a systematic review. JAMA Dermatol. 2017;153:193-198.
  10. Takeshita J, Eriksen WT, Raziano VT, et al. Racial differences in perceptions of psoriasis therapies: implications for racial disparities in psoriasis treatment. J Invest Dermatol. 2019;139:1672-1679.
  11. Yan D, Afifi L, Jeon C, et al. A cross-sectional study of the distribution of psoriasis subtypes in different ethno-racial groups. Dermatol Online J. 2018;24. pii:13030/qt5z21q4k2.
  12. Kerr GS, Qaiyumi S, Richards J, et al. Psoriasis and psoriatic arthritis in African-American patients—the need to measure disease burden. Clin Rheumatol. 2015;34:1753-1759.
  13. Edson-Heredia E, Sterling KL, Alatorre CI, et al. Heterogeneity of response to biologic treatment: perspective for psoriasis. J Invest Dermatol. 2014;134:18-23.
  14. Yan D, Afifi L, Jeon C, et al. A cross-sectional study of psoriasis triggers among different ethno-racial groups. J Am Acad Dermatol. 2017;77:756-758.
  15.  Bailey RK, Mokonogho J, Kumar A. Racial and ethnic differences in depression: current perspectives. Neuropsychiatr Dis Treat. 2019;15:603-609.
  16.  Jackson C, Maibach H. Ethnic and socioeconomic disparities in dermatology. J Dermatolog Treat. 2016;27:290-291.
  17. Salam A, Dadzie OE. Dermatology training in the U.K.: does it reflect the changing demographics of our population? Br J Dermatol. 2013;169:1360-1362.
  18. Nijhawan RI, Jacob SE, Woolery-Lloyd H. Skin of color education in dermatology residency programs: does residency training reflect the changing demographics of the United States? J Am Acad Dermatol. 2008;59:615-618.
  19. Ogunyemi B, Miller-Monthrope Y. The state of ethnic dermatology in Canada. J Cutan Med Surg. 2017;21:464-466.
References
  1. National Psoriasis Foundation. Statistics. https://www.psoriasis.org/content/statistics. Accessed July 14, 2020.
  2.  Alexis AF, Blackcloud P. Psoriasis in skin of color: epidemiology, genetics, clinical presentation, and treatment nuances. J Clin Aesthet Dermatol. 2014;7:16-24.
  3. Rachakonda TD, Schupp CW, Armstrong AW. Psoriasis prevalence among adults in the United States. J Am Acad Dermatol. 2014;70:512-516.
  4. Goff KL, Karimkhani C, Boyers LN, et al. The global burden of psoriatic skin disease. Br J Dermatol. 2015;172:1665-1668.
  5. Kaufman BP, Alexis AF. Psoriasis in skin of color: insights into the epidemiology, clinical presentation, genetics, quality-of-life impact, and treatment of psoriasis in non-white racial/ethnic groups. Am J Clin Dermatol. 2018;19:405-423.
  6. Shah SK, Arthur A, Yang YC, et al. A retrospective study to investigate racial and ethnic variations in the treatment of psoriasis with etanercept. J Drugs Dermatol. 2011;10:866-872.
  7. Abrouk M, Lee K, Brodsky M, et al. Ethnicity affects the presenting severity of psoriasis. J Am Acad Dermatol. 2017;77:180-182.
  8. Youssef RM, Mahgoub D, Mashaly HM, et al. Different narrowband UVB dosage regimens in dark skinned psoriatics: a preliminary study. Photodermatol Photoimmunol Photomed. 2008;24:256-259.
  9. Charrow A, Xia F Di, Joyce C, et al. Diversity in dermatology clinical trials: a systematic review. JAMA Dermatol. 2017;153:193-198.
  10. Takeshita J, Eriksen WT, Raziano VT, et al. Racial differences in perceptions of psoriasis therapies: implications for racial disparities in psoriasis treatment. J Invest Dermatol. 2019;139:1672-1679.
  11. Yan D, Afifi L, Jeon C, et al. A cross-sectional study of the distribution of psoriasis subtypes in different ethno-racial groups. Dermatol Online J. 2018;24. pii:13030/qt5z21q4k2.
  12. Kerr GS, Qaiyumi S, Richards J, et al. Psoriasis and psoriatic arthritis in African-American patients—the need to measure disease burden. Clin Rheumatol. 2015;34:1753-1759.
  13. Edson-Heredia E, Sterling KL, Alatorre CI, et al. Heterogeneity of response to biologic treatment: perspective for psoriasis. J Invest Dermatol. 2014;134:18-23.
  14. Yan D, Afifi L, Jeon C, et al. A cross-sectional study of psoriasis triggers among different ethno-racial groups. J Am Acad Dermatol. 2017;77:756-758.
  15.  Bailey RK, Mokonogho J, Kumar A. Racial and ethnic differences in depression: current perspectives. Neuropsychiatr Dis Treat. 2019;15:603-609.
  16.  Jackson C, Maibach H. Ethnic and socioeconomic disparities in dermatology. J Dermatolog Treat. 2016;27:290-291.
  17. Salam A, Dadzie OE. Dermatology training in the U.K.: does it reflect the changing demographics of our population? Br J Dermatol. 2013;169:1360-1362.
  18. Nijhawan RI, Jacob SE, Woolery-Lloyd H. Skin of color education in dermatology residency programs: does residency training reflect the changing demographics of the United States? J Am Acad Dermatol. 2008;59:615-618.
  19. Ogunyemi B, Miller-Monthrope Y. The state of ethnic dermatology in Canada. J Cutan Med Surg. 2017;21:464-466.
Issue
Cutis - 106(2S)
Issue
Cutis - 106(2S)
Page Number
7-10, E10
Page Number
7-10, E10
Publications
Publications
Topics
Article Type
Sections
Citation Override
Cutis. 2020 August;106(2S):7-10, E10
Inside the Article

Practice Points

  • There are key differences in psoriasis in patients with skin of color, including the morphology, clinical presentation, treatment, and psychosocial impact.
  • Recognition and awareness of these differences may normalize the condition for patients, support them seeking medical attention sooner, and better inform them of all possible treatment options.
  • Advocating further education on these differences in residency training and continuing medical education programs may help physicians make earlier diagnoses and personalize physician-patient conversations.
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
Article PDF Media

Tales of the Pandemic

Article Type
Changed
Thu, 08/20/2020 - 00:03

After learning about coronavirus disease 2019 (COVID-19) on the news, we were all aware that it would eventually affect our lives and our dermatology practices. However, once the COVID-19 pandemic arrived in the United States, we were under a shelter-in-place order, schools were shut, and most businesses were closed within a few weeks.

As dermatologists, we were considered essential workers, and our offices could remain open. However, as the numbers of cases accelerated in New York City—the global epicenter of the pandemic—and we approached our peak, I closed down my practice, except for emergencies.

One of the first medical challenges dermatologists faced in the early days of the COVID-19 pandemic was the proper management of our psoriasis patients. The major concern was that patients on biologics and other immunomodulatory therapies might be at an increased risk for COVID-19 infection and increased morbidity if affected. I received a multitude of telephone calls from patients taking these therapies who expressed high levels of concern and anxiety and were looking for direction as to whether they should continue their medications.

Early on, several of our professional societies provided guidelines regarding the use of systemic immunosuppressive agents during the COVID-19 pandemic. On April 15, 2020, the American Academy of Dermatology (AAD) advised, “Dermatologists must delicately balance the risk of immunosuppression with the risk of disease flare requiring urgent intervention with patient-specific risks.”1 The AAD strongly recommended that patients should not stop their ongoing systemic immunosuppressive therapy without consulting their physicians. The AAD’s guidance provided specific recommendations for the following groups: (1) patients on systemic immunosuppressive agents who have not tested positive or exhibited signs/symptoms of COVID-19, (2) patients on systemic immunosuppressive agents who have tested positive for COVID-19 or exhibit signs/symptoms of COVID-19, (3) patients who have halted systemic immunosuppressive therapy after testing positive for COVID-19 (in whom it recommended physicians could reinitiate treatment), and (4) patients being considered for systemic immunosuppressive agents.1

The National Psoriasis Foundation (NPF) also recognized the need for additional guidelines for health care providers and patients on managing psoriatic disease during the COVID-19 pandemic. In June 2020, the NPF formed a COVID-19 Task Force, which released its own recommendations for adult and pediatric patients with psoriatic disease.2 Similar to the AAD, the NPF COVID-19 Task Force recommended that patients do not stop biologic or oral therapies for psoriasis during the current health crisis, stating the following: “While some uncertainties remain, initial data suggest that the benefit of continuing treatments for psoriatic diseases outweighs the hypothetical risks associated with immune modulating treatment of poor COVID-19–related outcomes for most patients.” Individuals in high-risk groups were advised to consult their health care providers regarding whether they should continue or alter therapy during the pandemic, and the clinical decision would be guided by the specific treatment regimen; the patient’s age, disease characteristics, and underlying medical conditions; or any particular concerns. Additionally, the task force emphasized that patients with psoriatic disease should continue to follow common sense measures to lower the risk of becoming infected with COVID-19, including practicing physical distancing, wearing face coverings in public settings, and washing their hands regularly.2



We remain in the midst of the COVID-19 pandemic with no true guidance as to the future course and impact of the infection. It is important to realize that our understanding of the coronavirus and its impact on our patients is constantly evolving. I encourage all providers to stay current with updates on clinical guidelines. In addition, we should pay attention to the myriad of clinical trials and registries now underway, as they may provide more insight into optimal clinical management in these challenging times.

Most importantly, stay safe!

References
  1. American Academy of Dermatology. Guidance on the use of medications during COVID-19 outbreak. https://assets.ctfassets.net/1ny4yoiyrqia/PicgNuD0IpYd9MSOwab47/5e6d85324e7b5aafed45dde0ac4ea21e/Guidance_on_medications_AHTF_approved_April_15.pdf. Updated April 15, 2020. Accessed July 27, 2020.
  2. National Psoriasis Foundation. NPF forms COVID-19 Task Force. https://www.psoriasis.org/advance/coronavirus. Updated July 7, 2020. Accessed July 27, 2020.
Article PDF
Author and Disclosure Information

From the Department of Dermatology, Ichan School of Medicine at Mount Sinai, New York, New York.

The author reports no conflict of interest.

Correspondence: Jeffrey M. Weinberg, MD, 10 Union Square E, Ste 3C, New York, NY 10003 ([email protected]).

Issue
Cutis - 106(2S)
Publications
Topics
Page Number
6
Sections
Author and Disclosure Information

From the Department of Dermatology, Ichan School of Medicine at Mount Sinai, New York, New York.

The author reports no conflict of interest.

Correspondence: Jeffrey M. Weinberg, MD, 10 Union Square E, Ste 3C, New York, NY 10003 ([email protected]).

Author and Disclosure Information

From the Department of Dermatology, Ichan School of Medicine at Mount Sinai, New York, New York.

The author reports no conflict of interest.

Correspondence: Jeffrey M. Weinberg, MD, 10 Union Square E, Ste 3C, New York, NY 10003 ([email protected]).

Article PDF
Article PDF

After learning about coronavirus disease 2019 (COVID-19) on the news, we were all aware that it would eventually affect our lives and our dermatology practices. However, once the COVID-19 pandemic arrived in the United States, we were under a shelter-in-place order, schools were shut, and most businesses were closed within a few weeks.

As dermatologists, we were considered essential workers, and our offices could remain open. However, as the numbers of cases accelerated in New York City—the global epicenter of the pandemic—and we approached our peak, I closed down my practice, except for emergencies.

One of the first medical challenges dermatologists faced in the early days of the COVID-19 pandemic was the proper management of our psoriasis patients. The major concern was that patients on biologics and other immunomodulatory therapies might be at an increased risk for COVID-19 infection and increased morbidity if affected. I received a multitude of telephone calls from patients taking these therapies who expressed high levels of concern and anxiety and were looking for direction as to whether they should continue their medications.

Early on, several of our professional societies provided guidelines regarding the use of systemic immunosuppressive agents during the COVID-19 pandemic. On April 15, 2020, the American Academy of Dermatology (AAD) advised, “Dermatologists must delicately balance the risk of immunosuppression with the risk of disease flare requiring urgent intervention with patient-specific risks.”1 The AAD strongly recommended that patients should not stop their ongoing systemic immunosuppressive therapy without consulting their physicians. The AAD’s guidance provided specific recommendations for the following groups: (1) patients on systemic immunosuppressive agents who have not tested positive or exhibited signs/symptoms of COVID-19, (2) patients on systemic immunosuppressive agents who have tested positive for COVID-19 or exhibit signs/symptoms of COVID-19, (3) patients who have halted systemic immunosuppressive therapy after testing positive for COVID-19 (in whom it recommended physicians could reinitiate treatment), and (4) patients being considered for systemic immunosuppressive agents.1

The National Psoriasis Foundation (NPF) also recognized the need for additional guidelines for health care providers and patients on managing psoriatic disease during the COVID-19 pandemic. In June 2020, the NPF formed a COVID-19 Task Force, which released its own recommendations for adult and pediatric patients with psoriatic disease.2 Similar to the AAD, the NPF COVID-19 Task Force recommended that patients do not stop biologic or oral therapies for psoriasis during the current health crisis, stating the following: “While some uncertainties remain, initial data suggest that the benefit of continuing treatments for psoriatic diseases outweighs the hypothetical risks associated with immune modulating treatment of poor COVID-19–related outcomes for most patients.” Individuals in high-risk groups were advised to consult their health care providers regarding whether they should continue or alter therapy during the pandemic, and the clinical decision would be guided by the specific treatment regimen; the patient’s age, disease characteristics, and underlying medical conditions; or any particular concerns. Additionally, the task force emphasized that patients with psoriatic disease should continue to follow common sense measures to lower the risk of becoming infected with COVID-19, including practicing physical distancing, wearing face coverings in public settings, and washing their hands regularly.2



We remain in the midst of the COVID-19 pandemic with no true guidance as to the future course and impact of the infection. It is important to realize that our understanding of the coronavirus and its impact on our patients is constantly evolving. I encourage all providers to stay current with updates on clinical guidelines. In addition, we should pay attention to the myriad of clinical trials and registries now underway, as they may provide more insight into optimal clinical management in these challenging times.

Most importantly, stay safe!

After learning about coronavirus disease 2019 (COVID-19) on the news, we were all aware that it would eventually affect our lives and our dermatology practices. However, once the COVID-19 pandemic arrived in the United States, we were under a shelter-in-place order, schools were shut, and most businesses were closed within a few weeks.

As dermatologists, we were considered essential workers, and our offices could remain open. However, as the numbers of cases accelerated in New York City—the global epicenter of the pandemic—and we approached our peak, I closed down my practice, except for emergencies.

One of the first medical challenges dermatologists faced in the early days of the COVID-19 pandemic was the proper management of our psoriasis patients. The major concern was that patients on biologics and other immunomodulatory therapies might be at an increased risk for COVID-19 infection and increased morbidity if affected. I received a multitude of telephone calls from patients taking these therapies who expressed high levels of concern and anxiety and were looking for direction as to whether they should continue their medications.

Early on, several of our professional societies provided guidelines regarding the use of systemic immunosuppressive agents during the COVID-19 pandemic. On April 15, 2020, the American Academy of Dermatology (AAD) advised, “Dermatologists must delicately balance the risk of immunosuppression with the risk of disease flare requiring urgent intervention with patient-specific risks.”1 The AAD strongly recommended that patients should not stop their ongoing systemic immunosuppressive therapy without consulting their physicians. The AAD’s guidance provided specific recommendations for the following groups: (1) patients on systemic immunosuppressive agents who have not tested positive or exhibited signs/symptoms of COVID-19, (2) patients on systemic immunosuppressive agents who have tested positive for COVID-19 or exhibit signs/symptoms of COVID-19, (3) patients who have halted systemic immunosuppressive therapy after testing positive for COVID-19 (in whom it recommended physicians could reinitiate treatment), and (4) patients being considered for systemic immunosuppressive agents.1

The National Psoriasis Foundation (NPF) also recognized the need for additional guidelines for health care providers and patients on managing psoriatic disease during the COVID-19 pandemic. In June 2020, the NPF formed a COVID-19 Task Force, which released its own recommendations for adult and pediatric patients with psoriatic disease.2 Similar to the AAD, the NPF COVID-19 Task Force recommended that patients do not stop biologic or oral therapies for psoriasis during the current health crisis, stating the following: “While some uncertainties remain, initial data suggest that the benefit of continuing treatments for psoriatic diseases outweighs the hypothetical risks associated with immune modulating treatment of poor COVID-19–related outcomes for most patients.” Individuals in high-risk groups were advised to consult their health care providers regarding whether they should continue or alter therapy during the pandemic, and the clinical decision would be guided by the specific treatment regimen; the patient’s age, disease characteristics, and underlying medical conditions; or any particular concerns. Additionally, the task force emphasized that patients with psoriatic disease should continue to follow common sense measures to lower the risk of becoming infected with COVID-19, including practicing physical distancing, wearing face coverings in public settings, and washing their hands regularly.2



We remain in the midst of the COVID-19 pandemic with no true guidance as to the future course and impact of the infection. It is important to realize that our understanding of the coronavirus and its impact on our patients is constantly evolving. I encourage all providers to stay current with updates on clinical guidelines. In addition, we should pay attention to the myriad of clinical trials and registries now underway, as they may provide more insight into optimal clinical management in these challenging times.

Most importantly, stay safe!

References
  1. American Academy of Dermatology. Guidance on the use of medications during COVID-19 outbreak. https://assets.ctfassets.net/1ny4yoiyrqia/PicgNuD0IpYd9MSOwab47/5e6d85324e7b5aafed45dde0ac4ea21e/Guidance_on_medications_AHTF_approved_April_15.pdf. Updated April 15, 2020. Accessed July 27, 2020.
  2. National Psoriasis Foundation. NPF forms COVID-19 Task Force. https://www.psoriasis.org/advance/coronavirus. Updated July 7, 2020. Accessed July 27, 2020.
References
  1. American Academy of Dermatology. Guidance on the use of medications during COVID-19 outbreak. https://assets.ctfassets.net/1ny4yoiyrqia/PicgNuD0IpYd9MSOwab47/5e6d85324e7b5aafed45dde0ac4ea21e/Guidance_on_medications_AHTF_approved_April_15.pdf. Updated April 15, 2020. Accessed July 27, 2020.
  2. National Psoriasis Foundation. NPF forms COVID-19 Task Force. https://www.psoriasis.org/advance/coronavirus. Updated July 7, 2020. Accessed July 27, 2020.
Issue
Cutis - 106(2S)
Issue
Cutis - 106(2S)
Page Number
6
Page Number
6
Publications
Publications
Topics
Article Type
Sections
Citation Override
Cutis. 2020 August;106(2S):6
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
Article PDF Media

Management of Psoriasis With Biologics in Clinical Practice: An Update for 2020

Article Type
Changed
Thu, 08/20/2020 - 00:01

The advent of biologic therapy over the last 2 decades has transformed the treatment of psoriasis; patients who either are not good candidates for or have an inadequate response to traditional treatments (topicals and/or phototherapy) now have numerous options for treatment.1 Patients burdened by extensive disease, recurrent flares, and stubborn treatment areas are ideal candidates for biologics. There are 11 biologics approved by the US Food and Drug Administration (FDA)(Table) for treating moderate to severe plaque psoriasis as supported by grade A evidence. The FDA has authorized 1 new biologic—risankizumab—since the joint guidelines from the American Academy of Dermatology and National Psoriasis Foundation were released for the treatment of psoriasis with biologics.2 This article aims to address updates on recent clinical trial findings (April 2019 to April 2020) regarding biologic therapy initiation and maintenance for adult patients. Prescribers should use this update as guidance for determining the appropriate biologic class based on patient characteristics and for approaching biologic-experienced patients with refractory psoriasis. This update also may serve as a reference for the recommended dosing regimens of the 11 approved biologics.

Using Risankizumab

Risankizumab is a new biologic that selectively targets the IL-23 pathway by binding the p19 subunit of IL-23. It was approved by the FDA in April 2019. Two recent studies have demonstrated the efficacy of risankizumab in disease management.3,4

IMMvent was a double-blind, 2-part, phase 3, randomized controlled trial (RCT) of participants 18 years and older (N=605) with moderate to severe psoriasis (with or without psoriatic arthritis) across 11 countries.3 Inclusion criteria consisted of psoriasis involving at least 10% of the body surface area (BSA), absolute psoriasis area and severity index (PASI) score of 12 or higher, and static physician global assessment (sPGA) score of 3 or higher. Prior biologic treatment did not preclude study entry (excluding risankizumab or adalimumab), and nearly 40% of participants previously had been on a different biologic. Notably, this trial allowed for inclusion of patients with prior malignancy (>5 years prior) and patients who tested positive for exposure to tuberculosis (TB) but were not shown to have active TB (provided appropriate treatment for latent TB was started). Study participants identified as white (81%), Asian (14%), black/African American (4%), or other ethnicity (1%). Part A involved administration of 150 mg risankizumab (n=301) at weeks 0 and 4 or 80 mg adalimumab (n=304) loading dose at week 0 followed by 40 mg at week 1 and 40 mg every other week thereafter until the end of week 15. At week 16 there was a significant difference in proportion of participants achieving 90% or more improvement (PASI-90) with risankizumab (72%) vs adalimumab (47%)(P<.0001) and achieving an sPGA score of 0 or 1 (clear or almost clear) with risankizumab (84%) vs adalimumab (60%)(P<.0001). In part B (weeks 16–44), adalimumab immediate responder (PASI ≥50 to PASI <90) participants were re-randomized to continue adalimumab 40 mg every other week (starting from week 17 and stopping at week 44) or switch to 150 mg risankizumab administered at weeks 16, 20, and 32. Patients taking risankizumab in part A continued the drug, administered at weeks 16 and 28. At week 44, there was a significant difference in percentage of participants achieving PASI-90 with risankizumab (66%) vs adalimumab (21%)(P<.0001).3

IMMhance was another double-blind phase 3 RCT with 2 parts that assessed the clinical efficacy of risankizumab compared to placebo in patients 18 years or older (N=507) across 9 countries with the same inclusion criteria for patients as IMMvent.4 Part A involved administration of 150 mg risankizumab (n=407) or placebo (n=100) at weeks 0 and 4 using a 4:1 random allocation ratio. At week 16, regardless of initial treatment, all participants received 150 mg risankizumab. Treatment results at week 16 showed a significant difference in percentage of participants achieving PASI-90 with risankizumab (73.2%) vs placebo (2.0%)(P<.001) and sPGA score of 0 or 1 with risankizumab (83.5%) vs placebo (7.0%)(P<.001). Furthermore, in part B (weeks 16–104), at week 28 participants on risankizumab with an sPGA score of 0 or 1 were randomized with a 1:2 allocation ratio to continue 150 mg risankizumab or switch to placebo to produce a treatment withdrawal effect. Part B results showed a significant difference in the proportion of participants achieving an sPGA score of 0 or 1 with risankizumab (87.4%) vs placebo (61.3%)(P<.001) at week 52 and at week 104 with risankizumab (81.1%) vs placebo (7.1%)(P<.001). Risankizumab was well tolerated, with the most common adverse events (AEs) being nasopharyngitis (23.4%), upper respiratory tract infection (15.4%), and headache (6.8%). Serious AEs included cancer (2.6%; 2.2 events per 100 patient-years), hepatic events (4.6%) including hepatic cirrhosis (0.2%), and serious infections (1.8%; 1.4 events per 100 patient-years).4



Overall, the strengths of risankizumab with regard to its clinical efficacy and utility in biologic-experienced patients were confirmed in these studies. The inclusion of patients with prior treated malignancy and positive TB tests also was more in line with what one might encounter with real-world practice and, as such, provided valuable data to help aid treatment decisions. These 2 studies provided valuable evidence for the therapeutic benefit and relatively mild safety profile of risankizumab in treatment of moderate to severe psoriasis for patients with and without prior biologic therapy.

 

 

Choosing a Biologic

Refractory psoriasis involves nonresponse (primary failure) or return of disease symptoms after initial improvement (secondary failure) with a biologic. Selecting a biologic for patients who have experienced prior biologic failure is difficult. It is still unknown whether it is more efficacious for patients to try a same-class drug or a biologic targeting a different inflammatory pathway or cytokine. Studies have shown mixed results regarding how to manage patients with biologic failure, with both approaches demonstrating positive outcomes.

One analysis of the Corrona Psoriasis Registry included 144 patients, the majority of whom (89.8%) were biologic experienced, who began secukinumab treatment and returned for a 6-month follow-up (5–9 months).5 Patients enrolled in the registry were 18 years or older, had been diagnosed with psoriasis by a dermatologist, and initiated or switched an FDA-approved systemic agent or biologic within the last 12 months. Of biologic-experienced participants, 37.7% had used 3 or more biologics. More than half of included participants were either male (55%) or obese (53.4%). Comorbidities included hypertension (43.2%), hyperlipidemia (33.9%), anxiety (20.3%), diabetes mellitus (15.3%), cardiovascular disease (14.4%), and depression (13.6%). After 6 months of treatment, there was significant improvement in the involvement of BSA (mean difference, 12.1), investigator global assessment score (1.5), dermatology life quality index (DLQI)(4.8), pain (23.2), itch (−30.8), fatigue (8.8), and work productivity (9.2)(P<.01). Secukinumab therapy displayed notable reduction in symptom severity in this population with difficult-to-treat psoriasis. Its relative success in this cohort provides support for its use in treating patients who have failed other classes of biologics.5

Evidence supporting reduction of pruritus and pain with secukinumab also was notable. The CLEAR phase 3 RCT involved participants treated with 300 mg secukinumab every week for the first 4 weeks and then every 4 weeks thereafter for 48 weeks (n=312), up to 100 weeks (n=277).6 Participants had complete relief of pain (score 0), itching, and scaling at week 16 (69.4%, 49.7%, and 61.2%, respectively), week 52 (67.1%, 48.9%, and 53.3%, respectively), and week 104 (70.9%, 47.4%, and 54.8%, respectively). Reported AEs included candida infections (7.2%), malignant or unspecified tumors (1.5%), and neutropenia (<1%).6

Researchers investigated intraclass switching to brodalumab with prior failure of IL-17 inhibitors. An open-label study involved participants (n=39) with prior failure with secukinumab or ixekizumab therapy.7 Participants were administered 210 mg brodalumab with standard dosing at weeks 0, 1, and 2, and then every 2 weeks thereafter. At week 16, 69% of participants achieved PASI-75, 44% achieved PASI-90, 28% achieved PASI-100, and 62% achieved an sPGA score of 0 or 1. The authors attributed the relative success of brodalumab compared to prior anti–IL-17 agents to inhibition of the IL-17 receptor with brodalumab rather than the IL-17A ligand.7 Brodalumab may be a useful alternative biologic for patients with nonresponse to and secondary failure with biologics, including the IL-17A inhibitors.

Recent findings support effective skin clearance and improved symptom management with ixekizumab and ustekinumab. Of note, ixekizumab was reported to provide rapid improvement in skin lesions and quality of life to a greater extent than guselkumab.

The IXORA-R double-blinded RCT compared the clinical benefit of participants 18 years and older taking standard approved dosages of ixekizumab (n=520) or guselkumab (n=507).8 Patients were included if they had plaque psoriasis for at least 6 months before baseline, an sPGA score of at least 3, PASI score of 12 or higher, 10% or greater BSA, no prior IL-17 inhibitor failure, no use of IL-23 p19 inhibitors, and no use of any biologic within the specified period prior to baseline. At week 12, ixekizumab showed superior clinical improvement measured by the proportion of participants achieving complete skin clearance (ie, PASI-100)(41%) compared to guselkumab (25%)(P<.001). There were more participants taking ixekizumab who reported DLQI of 0 or 1 (no impact of disease on quality of life)(34%) compared to guselkumab (21%)(P<.001) as early on as week 4. The most common AE was upper respiratory tract infection (7%) in both groups. The risk of treatment-emergent AEs (56%), discontinuation because of AEs (2%), and serious AEs (3%) were comparable in both groups. The number of injection-site reactions was higher with ixekizumab (13%) vs guselkumab (3%). The authors concluded that ixekizumab offers the ability to provide rapid relief of symptoms, which is associated with improved DLQI.8



Response to ustekinumab therapy was assessed in a patient cohort enrolled in the Corrona Psoriasis Registry. This study involved 178 participants 18 years and older with psoriasis involvement of 3% or greater BSA who were treated with ustekinumab.9 By their 6-month follow-up visit, 55.6% of participants achieved adequate treatment response (BSA improving to <3% or 75% from enrollment). Increasing patient age was significantly associated with decreased likelihood of achieving a response (odds ratio, 0.981 [95% confidence interval, 0.962-0.999]; P=.049). Ustekinumab is a practical option for psoriasis treatment that seems to yield better results in younger patients.9 This evidence reveals that increased patient age is a characteristic that may contribute to poor treatment response and should be considered when choosing the best fit for biologic therapy.

Final Thoughts

Using evidence-based interventions to treat patients is the cornerstone of ethical and high-quality medical care. This guide sought to provide relevant updates in a variety of both comparator and pivotal trials, with the goal of summarizing clinically relevant information that may be extracted from these trials to guide patient care. It is not an exhaustive review but may be utilized as a reference tool to fine-tune selection criteria in choosing 1 of 11 biologics for the treatment of psoriasis.

References
  1. Pithadia DJ, Reynolds KA, Lee EB, et al. Translating the 2019 AAD-NPF Guidelines of Care for the Management of Psoriasis With Biologics to clinical practice. Cutis. 2019;104(suppl 2):12-16.
  2. Menter A, Strober BE, Kaplan DH, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with biologics [published online February 13, 2019]. J Am Acad Dermatol. 2019;80:1029-1072.
  3. Reich K, Gooderham M, Thaçi D, et al. Risankizumab compared with adalimumab in patients with moderate-to-severe plaque psoriasis (IMMvent): a randomised, double-blind, active-comparator-controlled phase 3 trial. Lancet. 2019;394:576-586.
  4. Blauvelt A, Leonardi CL, Gooderham M, et al. Efficacy and safety of continuous risankizumab therapy vs treatment withdrawal in patients with moderate to severe plaque psoriasis: a phase 3 randomized clinical trial. JAMA Dermatol. 2020;156:649-658.
  5. Strober BE, Germino R, Guana A, et al. US real-world effectiveness of secukinumab for the treatment of psoriasis: 6-month analysis from the Corrona Psoriasis Registry. J Dermatolog Treat. 2020;31:333-341.
  6. Thaçi D, Puig L, Reich K, et al. Secukinumab demonstrates sustained efficacy in clearing skin and improving patient-reported outcomes in patients with moderate-to-severe psoriasis through 2 years of treatment: results from the CLEAR study. J Am Acad Dermatol. 2019;81:1405-1409.
  7. Kimmel G, Chima M, Kim HJ, et al. Brodalumab in the treatment of moderate to severe psoriasis in patients when previous anti-interleukin 17A therapies have failed. J Am Acad Dermatol. 2019;81:857-859.
  8. Blauvelt A, Papp K, Gottlieb A, et al. A head‐to‐head comparison of ixekizumab vs. guselkumab in patients with moderate‐to‐severe plaque psoriasis: 12‐week efficacy, safety and speed of response from a randomized, double‐blinded trial. Br J Dermatol. 2020;182:1348-1358.
  9. Van Voorhees AS, Mason MA, Harrold LR, et al. Characterization of insufficient responders to ustekinumab in patients with moderate-to-severe psoriasis in the US Corrona Psoriasis Registry [published online February 27, 2020]. J Dermatolog Treat. doi:10.1080/09546634.2020.1720586.
Article PDF
Author and Disclosure Information

Ms. Ivanic is from Meharry Medical College, Nashville, Tennessee. Ms. Naderi-Azad is from the University of Toronto Faculty of Medicine, Ontario, Canada. Ms. Walia is from Lake Erie College of Osteopathic Medicine, Pennsylvania. Dr. Han is from the Icahn School of Medicine at Mount Sinai, New York, New York. Dr. Wu is from the Dermatology Research and Education Foundation, Irvine, California.

Ms. Ivanic, Ms. Naderi-Azad, and Ms. Walia report no conflict of interest. Dr. Han is or has been a consultant/advisor, investigator, or speaker for AbbVie; Athenex; Boehringer Ingelheim; Bond Avillion; Bristol-Myers Squibb; Celgene Corporation; Eli Lilly and Company; Janssen Biotech, Inc; LEO Pharma; MC2 Therapeutics; Novartis; Ortho Dermatologics; PellePharm; Pfizer; Regeneron Pharmaceuticals, Inc; Sanofi Genzyme; Sun Pharmaceutical; and UCB. Dr. Wu is or has been a consultant, investigator, or speaker for AbbVie; Almirall; Amgen; Arcutis; Boehringer Ingelheim; Bristol-Myers Squibb; Celgene Corporation; Dermavant; Dermira; Dr. Reddy’s Laboratories; Eli Lilly and Company; Janssen Biotech, Inc; LEO Pharma; Novartis; Regeneron Pharmaceuticals, Inc; Sanofi Genzyme; Sun Pharmaceutical; UCB; and Valeant Pharmaceuticals North America.

Correspondence: Jashin J. Wu, MD ([email protected]).

Issue
Cutis - 106(2S)
Publications
Topics
Page Number
21-24
Sections
Author and Disclosure Information

Ms. Ivanic is from Meharry Medical College, Nashville, Tennessee. Ms. Naderi-Azad is from the University of Toronto Faculty of Medicine, Ontario, Canada. Ms. Walia is from Lake Erie College of Osteopathic Medicine, Pennsylvania. Dr. Han is from the Icahn School of Medicine at Mount Sinai, New York, New York. Dr. Wu is from the Dermatology Research and Education Foundation, Irvine, California.

Ms. Ivanic, Ms. Naderi-Azad, and Ms. Walia report no conflict of interest. Dr. Han is or has been a consultant/advisor, investigator, or speaker for AbbVie; Athenex; Boehringer Ingelheim; Bond Avillion; Bristol-Myers Squibb; Celgene Corporation; Eli Lilly and Company; Janssen Biotech, Inc; LEO Pharma; MC2 Therapeutics; Novartis; Ortho Dermatologics; PellePharm; Pfizer; Regeneron Pharmaceuticals, Inc; Sanofi Genzyme; Sun Pharmaceutical; and UCB. Dr. Wu is or has been a consultant, investigator, or speaker for AbbVie; Almirall; Amgen; Arcutis; Boehringer Ingelheim; Bristol-Myers Squibb; Celgene Corporation; Dermavant; Dermira; Dr. Reddy’s Laboratories; Eli Lilly and Company; Janssen Biotech, Inc; LEO Pharma; Novartis; Regeneron Pharmaceuticals, Inc; Sanofi Genzyme; Sun Pharmaceutical; UCB; and Valeant Pharmaceuticals North America.

Correspondence: Jashin J. Wu, MD ([email protected]).

Author and Disclosure Information

Ms. Ivanic is from Meharry Medical College, Nashville, Tennessee. Ms. Naderi-Azad is from the University of Toronto Faculty of Medicine, Ontario, Canada. Ms. Walia is from Lake Erie College of Osteopathic Medicine, Pennsylvania. Dr. Han is from the Icahn School of Medicine at Mount Sinai, New York, New York. Dr. Wu is from the Dermatology Research and Education Foundation, Irvine, California.

Ms. Ivanic, Ms. Naderi-Azad, and Ms. Walia report no conflict of interest. Dr. Han is or has been a consultant/advisor, investigator, or speaker for AbbVie; Athenex; Boehringer Ingelheim; Bond Avillion; Bristol-Myers Squibb; Celgene Corporation; Eli Lilly and Company; Janssen Biotech, Inc; LEO Pharma; MC2 Therapeutics; Novartis; Ortho Dermatologics; PellePharm; Pfizer; Regeneron Pharmaceuticals, Inc; Sanofi Genzyme; Sun Pharmaceutical; and UCB. Dr. Wu is or has been a consultant, investigator, or speaker for AbbVie; Almirall; Amgen; Arcutis; Boehringer Ingelheim; Bristol-Myers Squibb; Celgene Corporation; Dermavant; Dermira; Dr. Reddy’s Laboratories; Eli Lilly and Company; Janssen Biotech, Inc; LEO Pharma; Novartis; Regeneron Pharmaceuticals, Inc; Sanofi Genzyme; Sun Pharmaceutical; UCB; and Valeant Pharmaceuticals North America.

Correspondence: Jashin J. Wu, MD ([email protected]).

Article PDF
Article PDF

The advent of biologic therapy over the last 2 decades has transformed the treatment of psoriasis; patients who either are not good candidates for or have an inadequate response to traditional treatments (topicals and/or phototherapy) now have numerous options for treatment.1 Patients burdened by extensive disease, recurrent flares, and stubborn treatment areas are ideal candidates for biologics. There are 11 biologics approved by the US Food and Drug Administration (FDA)(Table) for treating moderate to severe plaque psoriasis as supported by grade A evidence. The FDA has authorized 1 new biologic—risankizumab—since the joint guidelines from the American Academy of Dermatology and National Psoriasis Foundation were released for the treatment of psoriasis with biologics.2 This article aims to address updates on recent clinical trial findings (April 2019 to April 2020) regarding biologic therapy initiation and maintenance for adult patients. Prescribers should use this update as guidance for determining the appropriate biologic class based on patient characteristics and for approaching biologic-experienced patients with refractory psoriasis. This update also may serve as a reference for the recommended dosing regimens of the 11 approved biologics.

Using Risankizumab

Risankizumab is a new biologic that selectively targets the IL-23 pathway by binding the p19 subunit of IL-23. It was approved by the FDA in April 2019. Two recent studies have demonstrated the efficacy of risankizumab in disease management.3,4

IMMvent was a double-blind, 2-part, phase 3, randomized controlled trial (RCT) of participants 18 years and older (N=605) with moderate to severe psoriasis (with or without psoriatic arthritis) across 11 countries.3 Inclusion criteria consisted of psoriasis involving at least 10% of the body surface area (BSA), absolute psoriasis area and severity index (PASI) score of 12 or higher, and static physician global assessment (sPGA) score of 3 or higher. Prior biologic treatment did not preclude study entry (excluding risankizumab or adalimumab), and nearly 40% of participants previously had been on a different biologic. Notably, this trial allowed for inclusion of patients with prior malignancy (>5 years prior) and patients who tested positive for exposure to tuberculosis (TB) but were not shown to have active TB (provided appropriate treatment for latent TB was started). Study participants identified as white (81%), Asian (14%), black/African American (4%), or other ethnicity (1%). Part A involved administration of 150 mg risankizumab (n=301) at weeks 0 and 4 or 80 mg adalimumab (n=304) loading dose at week 0 followed by 40 mg at week 1 and 40 mg every other week thereafter until the end of week 15. At week 16 there was a significant difference in proportion of participants achieving 90% or more improvement (PASI-90) with risankizumab (72%) vs adalimumab (47%)(P<.0001) and achieving an sPGA score of 0 or 1 (clear or almost clear) with risankizumab (84%) vs adalimumab (60%)(P<.0001). In part B (weeks 16–44), adalimumab immediate responder (PASI ≥50 to PASI <90) participants were re-randomized to continue adalimumab 40 mg every other week (starting from week 17 and stopping at week 44) or switch to 150 mg risankizumab administered at weeks 16, 20, and 32. Patients taking risankizumab in part A continued the drug, administered at weeks 16 and 28. At week 44, there was a significant difference in percentage of participants achieving PASI-90 with risankizumab (66%) vs adalimumab (21%)(P<.0001).3

IMMhance was another double-blind phase 3 RCT with 2 parts that assessed the clinical efficacy of risankizumab compared to placebo in patients 18 years or older (N=507) across 9 countries with the same inclusion criteria for patients as IMMvent.4 Part A involved administration of 150 mg risankizumab (n=407) or placebo (n=100) at weeks 0 and 4 using a 4:1 random allocation ratio. At week 16, regardless of initial treatment, all participants received 150 mg risankizumab. Treatment results at week 16 showed a significant difference in percentage of participants achieving PASI-90 with risankizumab (73.2%) vs placebo (2.0%)(P<.001) and sPGA score of 0 or 1 with risankizumab (83.5%) vs placebo (7.0%)(P<.001). Furthermore, in part B (weeks 16–104), at week 28 participants on risankizumab with an sPGA score of 0 or 1 were randomized with a 1:2 allocation ratio to continue 150 mg risankizumab or switch to placebo to produce a treatment withdrawal effect. Part B results showed a significant difference in the proportion of participants achieving an sPGA score of 0 or 1 with risankizumab (87.4%) vs placebo (61.3%)(P<.001) at week 52 and at week 104 with risankizumab (81.1%) vs placebo (7.1%)(P<.001). Risankizumab was well tolerated, with the most common adverse events (AEs) being nasopharyngitis (23.4%), upper respiratory tract infection (15.4%), and headache (6.8%). Serious AEs included cancer (2.6%; 2.2 events per 100 patient-years), hepatic events (4.6%) including hepatic cirrhosis (0.2%), and serious infections (1.8%; 1.4 events per 100 patient-years).4



Overall, the strengths of risankizumab with regard to its clinical efficacy and utility in biologic-experienced patients were confirmed in these studies. The inclusion of patients with prior treated malignancy and positive TB tests also was more in line with what one might encounter with real-world practice and, as such, provided valuable data to help aid treatment decisions. These 2 studies provided valuable evidence for the therapeutic benefit and relatively mild safety profile of risankizumab in treatment of moderate to severe psoriasis for patients with and without prior biologic therapy.

 

 

Choosing a Biologic

Refractory psoriasis involves nonresponse (primary failure) or return of disease symptoms after initial improvement (secondary failure) with a biologic. Selecting a biologic for patients who have experienced prior biologic failure is difficult. It is still unknown whether it is more efficacious for patients to try a same-class drug or a biologic targeting a different inflammatory pathway or cytokine. Studies have shown mixed results regarding how to manage patients with biologic failure, with both approaches demonstrating positive outcomes.

One analysis of the Corrona Psoriasis Registry included 144 patients, the majority of whom (89.8%) were biologic experienced, who began secukinumab treatment and returned for a 6-month follow-up (5–9 months).5 Patients enrolled in the registry were 18 years or older, had been diagnosed with psoriasis by a dermatologist, and initiated or switched an FDA-approved systemic agent or biologic within the last 12 months. Of biologic-experienced participants, 37.7% had used 3 or more biologics. More than half of included participants were either male (55%) or obese (53.4%). Comorbidities included hypertension (43.2%), hyperlipidemia (33.9%), anxiety (20.3%), diabetes mellitus (15.3%), cardiovascular disease (14.4%), and depression (13.6%). After 6 months of treatment, there was significant improvement in the involvement of BSA (mean difference, 12.1), investigator global assessment score (1.5), dermatology life quality index (DLQI)(4.8), pain (23.2), itch (−30.8), fatigue (8.8), and work productivity (9.2)(P<.01). Secukinumab therapy displayed notable reduction in symptom severity in this population with difficult-to-treat psoriasis. Its relative success in this cohort provides support for its use in treating patients who have failed other classes of biologics.5

Evidence supporting reduction of pruritus and pain with secukinumab also was notable. The CLEAR phase 3 RCT involved participants treated with 300 mg secukinumab every week for the first 4 weeks and then every 4 weeks thereafter for 48 weeks (n=312), up to 100 weeks (n=277).6 Participants had complete relief of pain (score 0), itching, and scaling at week 16 (69.4%, 49.7%, and 61.2%, respectively), week 52 (67.1%, 48.9%, and 53.3%, respectively), and week 104 (70.9%, 47.4%, and 54.8%, respectively). Reported AEs included candida infections (7.2%), malignant or unspecified tumors (1.5%), and neutropenia (<1%).6

Researchers investigated intraclass switching to brodalumab with prior failure of IL-17 inhibitors. An open-label study involved participants (n=39) with prior failure with secukinumab or ixekizumab therapy.7 Participants were administered 210 mg brodalumab with standard dosing at weeks 0, 1, and 2, and then every 2 weeks thereafter. At week 16, 69% of participants achieved PASI-75, 44% achieved PASI-90, 28% achieved PASI-100, and 62% achieved an sPGA score of 0 or 1. The authors attributed the relative success of brodalumab compared to prior anti–IL-17 agents to inhibition of the IL-17 receptor with brodalumab rather than the IL-17A ligand.7 Brodalumab may be a useful alternative biologic for patients with nonresponse to and secondary failure with biologics, including the IL-17A inhibitors.

Recent findings support effective skin clearance and improved symptom management with ixekizumab and ustekinumab. Of note, ixekizumab was reported to provide rapid improvement in skin lesions and quality of life to a greater extent than guselkumab.

The IXORA-R double-blinded RCT compared the clinical benefit of participants 18 years and older taking standard approved dosages of ixekizumab (n=520) or guselkumab (n=507).8 Patients were included if they had plaque psoriasis for at least 6 months before baseline, an sPGA score of at least 3, PASI score of 12 or higher, 10% or greater BSA, no prior IL-17 inhibitor failure, no use of IL-23 p19 inhibitors, and no use of any biologic within the specified period prior to baseline. At week 12, ixekizumab showed superior clinical improvement measured by the proportion of participants achieving complete skin clearance (ie, PASI-100)(41%) compared to guselkumab (25%)(P<.001). There were more participants taking ixekizumab who reported DLQI of 0 or 1 (no impact of disease on quality of life)(34%) compared to guselkumab (21%)(P<.001) as early on as week 4. The most common AE was upper respiratory tract infection (7%) in both groups. The risk of treatment-emergent AEs (56%), discontinuation because of AEs (2%), and serious AEs (3%) were comparable in both groups. The number of injection-site reactions was higher with ixekizumab (13%) vs guselkumab (3%). The authors concluded that ixekizumab offers the ability to provide rapid relief of symptoms, which is associated with improved DLQI.8



Response to ustekinumab therapy was assessed in a patient cohort enrolled in the Corrona Psoriasis Registry. This study involved 178 participants 18 years and older with psoriasis involvement of 3% or greater BSA who were treated with ustekinumab.9 By their 6-month follow-up visit, 55.6% of participants achieved adequate treatment response (BSA improving to <3% or 75% from enrollment). Increasing patient age was significantly associated with decreased likelihood of achieving a response (odds ratio, 0.981 [95% confidence interval, 0.962-0.999]; P=.049). Ustekinumab is a practical option for psoriasis treatment that seems to yield better results in younger patients.9 This evidence reveals that increased patient age is a characteristic that may contribute to poor treatment response and should be considered when choosing the best fit for biologic therapy.

Final Thoughts

Using evidence-based interventions to treat patients is the cornerstone of ethical and high-quality medical care. This guide sought to provide relevant updates in a variety of both comparator and pivotal trials, with the goal of summarizing clinically relevant information that may be extracted from these trials to guide patient care. It is not an exhaustive review but may be utilized as a reference tool to fine-tune selection criteria in choosing 1 of 11 biologics for the treatment of psoriasis.

The advent of biologic therapy over the last 2 decades has transformed the treatment of psoriasis; patients who either are not good candidates for or have an inadequate response to traditional treatments (topicals and/or phototherapy) now have numerous options for treatment.1 Patients burdened by extensive disease, recurrent flares, and stubborn treatment areas are ideal candidates for biologics. There are 11 biologics approved by the US Food and Drug Administration (FDA)(Table) for treating moderate to severe plaque psoriasis as supported by grade A evidence. The FDA has authorized 1 new biologic—risankizumab—since the joint guidelines from the American Academy of Dermatology and National Psoriasis Foundation were released for the treatment of psoriasis with biologics.2 This article aims to address updates on recent clinical trial findings (April 2019 to April 2020) regarding biologic therapy initiation and maintenance for adult patients. Prescribers should use this update as guidance for determining the appropriate biologic class based on patient characteristics and for approaching biologic-experienced patients with refractory psoriasis. This update also may serve as a reference for the recommended dosing regimens of the 11 approved biologics.

Using Risankizumab

Risankizumab is a new biologic that selectively targets the IL-23 pathway by binding the p19 subunit of IL-23. It was approved by the FDA in April 2019. Two recent studies have demonstrated the efficacy of risankizumab in disease management.3,4

IMMvent was a double-blind, 2-part, phase 3, randomized controlled trial (RCT) of participants 18 years and older (N=605) with moderate to severe psoriasis (with or without psoriatic arthritis) across 11 countries.3 Inclusion criteria consisted of psoriasis involving at least 10% of the body surface area (BSA), absolute psoriasis area and severity index (PASI) score of 12 or higher, and static physician global assessment (sPGA) score of 3 or higher. Prior biologic treatment did not preclude study entry (excluding risankizumab or adalimumab), and nearly 40% of participants previously had been on a different biologic. Notably, this trial allowed for inclusion of patients with prior malignancy (>5 years prior) and patients who tested positive for exposure to tuberculosis (TB) but were not shown to have active TB (provided appropriate treatment for latent TB was started). Study participants identified as white (81%), Asian (14%), black/African American (4%), or other ethnicity (1%). Part A involved administration of 150 mg risankizumab (n=301) at weeks 0 and 4 or 80 mg adalimumab (n=304) loading dose at week 0 followed by 40 mg at week 1 and 40 mg every other week thereafter until the end of week 15. At week 16 there was a significant difference in proportion of participants achieving 90% or more improvement (PASI-90) with risankizumab (72%) vs adalimumab (47%)(P<.0001) and achieving an sPGA score of 0 or 1 (clear or almost clear) with risankizumab (84%) vs adalimumab (60%)(P<.0001). In part B (weeks 16–44), adalimumab immediate responder (PASI ≥50 to PASI <90) participants were re-randomized to continue adalimumab 40 mg every other week (starting from week 17 and stopping at week 44) or switch to 150 mg risankizumab administered at weeks 16, 20, and 32. Patients taking risankizumab in part A continued the drug, administered at weeks 16 and 28. At week 44, there was a significant difference in percentage of participants achieving PASI-90 with risankizumab (66%) vs adalimumab (21%)(P<.0001).3

IMMhance was another double-blind phase 3 RCT with 2 parts that assessed the clinical efficacy of risankizumab compared to placebo in patients 18 years or older (N=507) across 9 countries with the same inclusion criteria for patients as IMMvent.4 Part A involved administration of 150 mg risankizumab (n=407) or placebo (n=100) at weeks 0 and 4 using a 4:1 random allocation ratio. At week 16, regardless of initial treatment, all participants received 150 mg risankizumab. Treatment results at week 16 showed a significant difference in percentage of participants achieving PASI-90 with risankizumab (73.2%) vs placebo (2.0%)(P<.001) and sPGA score of 0 or 1 with risankizumab (83.5%) vs placebo (7.0%)(P<.001). Furthermore, in part B (weeks 16–104), at week 28 participants on risankizumab with an sPGA score of 0 or 1 were randomized with a 1:2 allocation ratio to continue 150 mg risankizumab or switch to placebo to produce a treatment withdrawal effect. Part B results showed a significant difference in the proportion of participants achieving an sPGA score of 0 or 1 with risankizumab (87.4%) vs placebo (61.3%)(P<.001) at week 52 and at week 104 with risankizumab (81.1%) vs placebo (7.1%)(P<.001). Risankizumab was well tolerated, with the most common adverse events (AEs) being nasopharyngitis (23.4%), upper respiratory tract infection (15.4%), and headache (6.8%). Serious AEs included cancer (2.6%; 2.2 events per 100 patient-years), hepatic events (4.6%) including hepatic cirrhosis (0.2%), and serious infections (1.8%; 1.4 events per 100 patient-years).4



Overall, the strengths of risankizumab with regard to its clinical efficacy and utility in biologic-experienced patients were confirmed in these studies. The inclusion of patients with prior treated malignancy and positive TB tests also was more in line with what one might encounter with real-world practice and, as such, provided valuable data to help aid treatment decisions. These 2 studies provided valuable evidence for the therapeutic benefit and relatively mild safety profile of risankizumab in treatment of moderate to severe psoriasis for patients with and without prior biologic therapy.

 

 

Choosing a Biologic

Refractory psoriasis involves nonresponse (primary failure) or return of disease symptoms after initial improvement (secondary failure) with a biologic. Selecting a biologic for patients who have experienced prior biologic failure is difficult. It is still unknown whether it is more efficacious for patients to try a same-class drug or a biologic targeting a different inflammatory pathway or cytokine. Studies have shown mixed results regarding how to manage patients with biologic failure, with both approaches demonstrating positive outcomes.

One analysis of the Corrona Psoriasis Registry included 144 patients, the majority of whom (89.8%) were biologic experienced, who began secukinumab treatment and returned for a 6-month follow-up (5–9 months).5 Patients enrolled in the registry were 18 years or older, had been diagnosed with psoriasis by a dermatologist, and initiated or switched an FDA-approved systemic agent or biologic within the last 12 months. Of biologic-experienced participants, 37.7% had used 3 or more biologics. More than half of included participants were either male (55%) or obese (53.4%). Comorbidities included hypertension (43.2%), hyperlipidemia (33.9%), anxiety (20.3%), diabetes mellitus (15.3%), cardiovascular disease (14.4%), and depression (13.6%). After 6 months of treatment, there was significant improvement in the involvement of BSA (mean difference, 12.1), investigator global assessment score (1.5), dermatology life quality index (DLQI)(4.8), pain (23.2), itch (−30.8), fatigue (8.8), and work productivity (9.2)(P<.01). Secukinumab therapy displayed notable reduction in symptom severity in this population with difficult-to-treat psoriasis. Its relative success in this cohort provides support for its use in treating patients who have failed other classes of biologics.5

Evidence supporting reduction of pruritus and pain with secukinumab also was notable. The CLEAR phase 3 RCT involved participants treated with 300 mg secukinumab every week for the first 4 weeks and then every 4 weeks thereafter for 48 weeks (n=312), up to 100 weeks (n=277).6 Participants had complete relief of pain (score 0), itching, and scaling at week 16 (69.4%, 49.7%, and 61.2%, respectively), week 52 (67.1%, 48.9%, and 53.3%, respectively), and week 104 (70.9%, 47.4%, and 54.8%, respectively). Reported AEs included candida infections (7.2%), malignant or unspecified tumors (1.5%), and neutropenia (<1%).6

Researchers investigated intraclass switching to brodalumab with prior failure of IL-17 inhibitors. An open-label study involved participants (n=39) with prior failure with secukinumab or ixekizumab therapy.7 Participants were administered 210 mg brodalumab with standard dosing at weeks 0, 1, and 2, and then every 2 weeks thereafter. At week 16, 69% of participants achieved PASI-75, 44% achieved PASI-90, 28% achieved PASI-100, and 62% achieved an sPGA score of 0 or 1. The authors attributed the relative success of brodalumab compared to prior anti–IL-17 agents to inhibition of the IL-17 receptor with brodalumab rather than the IL-17A ligand.7 Brodalumab may be a useful alternative biologic for patients with nonresponse to and secondary failure with biologics, including the IL-17A inhibitors.

Recent findings support effective skin clearance and improved symptom management with ixekizumab and ustekinumab. Of note, ixekizumab was reported to provide rapid improvement in skin lesions and quality of life to a greater extent than guselkumab.

The IXORA-R double-blinded RCT compared the clinical benefit of participants 18 years and older taking standard approved dosages of ixekizumab (n=520) or guselkumab (n=507).8 Patients were included if they had plaque psoriasis for at least 6 months before baseline, an sPGA score of at least 3, PASI score of 12 or higher, 10% or greater BSA, no prior IL-17 inhibitor failure, no use of IL-23 p19 inhibitors, and no use of any biologic within the specified period prior to baseline. At week 12, ixekizumab showed superior clinical improvement measured by the proportion of participants achieving complete skin clearance (ie, PASI-100)(41%) compared to guselkumab (25%)(P<.001). There were more participants taking ixekizumab who reported DLQI of 0 or 1 (no impact of disease on quality of life)(34%) compared to guselkumab (21%)(P<.001) as early on as week 4. The most common AE was upper respiratory tract infection (7%) in both groups. The risk of treatment-emergent AEs (56%), discontinuation because of AEs (2%), and serious AEs (3%) were comparable in both groups. The number of injection-site reactions was higher with ixekizumab (13%) vs guselkumab (3%). The authors concluded that ixekizumab offers the ability to provide rapid relief of symptoms, which is associated with improved DLQI.8



Response to ustekinumab therapy was assessed in a patient cohort enrolled in the Corrona Psoriasis Registry. This study involved 178 participants 18 years and older with psoriasis involvement of 3% or greater BSA who were treated with ustekinumab.9 By their 6-month follow-up visit, 55.6% of participants achieved adequate treatment response (BSA improving to <3% or 75% from enrollment). Increasing patient age was significantly associated with decreased likelihood of achieving a response (odds ratio, 0.981 [95% confidence interval, 0.962-0.999]; P=.049). Ustekinumab is a practical option for psoriasis treatment that seems to yield better results in younger patients.9 This evidence reveals that increased patient age is a characteristic that may contribute to poor treatment response and should be considered when choosing the best fit for biologic therapy.

Final Thoughts

Using evidence-based interventions to treat patients is the cornerstone of ethical and high-quality medical care. This guide sought to provide relevant updates in a variety of both comparator and pivotal trials, with the goal of summarizing clinically relevant information that may be extracted from these trials to guide patient care. It is not an exhaustive review but may be utilized as a reference tool to fine-tune selection criteria in choosing 1 of 11 biologics for the treatment of psoriasis.

References
  1. Pithadia DJ, Reynolds KA, Lee EB, et al. Translating the 2019 AAD-NPF Guidelines of Care for the Management of Psoriasis With Biologics to clinical practice. Cutis. 2019;104(suppl 2):12-16.
  2. Menter A, Strober BE, Kaplan DH, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with biologics [published online February 13, 2019]. J Am Acad Dermatol. 2019;80:1029-1072.
  3. Reich K, Gooderham M, Thaçi D, et al. Risankizumab compared with adalimumab in patients with moderate-to-severe plaque psoriasis (IMMvent): a randomised, double-blind, active-comparator-controlled phase 3 trial. Lancet. 2019;394:576-586.
  4. Blauvelt A, Leonardi CL, Gooderham M, et al. Efficacy and safety of continuous risankizumab therapy vs treatment withdrawal in patients with moderate to severe plaque psoriasis: a phase 3 randomized clinical trial. JAMA Dermatol. 2020;156:649-658.
  5. Strober BE, Germino R, Guana A, et al. US real-world effectiveness of secukinumab for the treatment of psoriasis: 6-month analysis from the Corrona Psoriasis Registry. J Dermatolog Treat. 2020;31:333-341.
  6. Thaçi D, Puig L, Reich K, et al. Secukinumab demonstrates sustained efficacy in clearing skin and improving patient-reported outcomes in patients with moderate-to-severe psoriasis through 2 years of treatment: results from the CLEAR study. J Am Acad Dermatol. 2019;81:1405-1409.
  7. Kimmel G, Chima M, Kim HJ, et al. Brodalumab in the treatment of moderate to severe psoriasis in patients when previous anti-interleukin 17A therapies have failed. J Am Acad Dermatol. 2019;81:857-859.
  8. Blauvelt A, Papp K, Gottlieb A, et al. A head‐to‐head comparison of ixekizumab vs. guselkumab in patients with moderate‐to‐severe plaque psoriasis: 12‐week efficacy, safety and speed of response from a randomized, double‐blinded trial. Br J Dermatol. 2020;182:1348-1358.
  9. Van Voorhees AS, Mason MA, Harrold LR, et al. Characterization of insufficient responders to ustekinumab in patients with moderate-to-severe psoriasis in the US Corrona Psoriasis Registry [published online February 27, 2020]. J Dermatolog Treat. doi:10.1080/09546634.2020.1720586.
References
  1. Pithadia DJ, Reynolds KA, Lee EB, et al. Translating the 2019 AAD-NPF Guidelines of Care for the Management of Psoriasis With Biologics to clinical practice. Cutis. 2019;104(suppl 2):12-16.
  2. Menter A, Strober BE, Kaplan DH, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with biologics [published online February 13, 2019]. J Am Acad Dermatol. 2019;80:1029-1072.
  3. Reich K, Gooderham M, Thaçi D, et al. Risankizumab compared with adalimumab in patients with moderate-to-severe plaque psoriasis (IMMvent): a randomised, double-blind, active-comparator-controlled phase 3 trial. Lancet. 2019;394:576-586.
  4. Blauvelt A, Leonardi CL, Gooderham M, et al. Efficacy and safety of continuous risankizumab therapy vs treatment withdrawal in patients with moderate to severe plaque psoriasis: a phase 3 randomized clinical trial. JAMA Dermatol. 2020;156:649-658.
  5. Strober BE, Germino R, Guana A, et al. US real-world effectiveness of secukinumab for the treatment of psoriasis: 6-month analysis from the Corrona Psoriasis Registry. J Dermatolog Treat. 2020;31:333-341.
  6. Thaçi D, Puig L, Reich K, et al. Secukinumab demonstrates sustained efficacy in clearing skin and improving patient-reported outcomes in patients with moderate-to-severe psoriasis through 2 years of treatment: results from the CLEAR study. J Am Acad Dermatol. 2019;81:1405-1409.
  7. Kimmel G, Chima M, Kim HJ, et al. Brodalumab in the treatment of moderate to severe psoriasis in patients when previous anti-interleukin 17A therapies have failed. J Am Acad Dermatol. 2019;81:857-859.
  8. Blauvelt A, Papp K, Gottlieb A, et al. A head‐to‐head comparison of ixekizumab vs. guselkumab in patients with moderate‐to‐severe plaque psoriasis: 12‐week efficacy, safety and speed of response from a randomized, double‐blinded trial. Br J Dermatol. 2020;182:1348-1358.
  9. Van Voorhees AS, Mason MA, Harrold LR, et al. Characterization of insufficient responders to ustekinumab in patients with moderate-to-severe psoriasis in the US Corrona Psoriasis Registry [published online February 27, 2020]. J Dermatolog Treat. doi:10.1080/09546634.2020.1720586.
Issue
Cutis - 106(2S)
Issue
Cutis - 106(2S)
Page Number
21-24
Page Number
21-24
Publications
Publications
Topics
Article Type
Sections
Citation Override
Cutis. 2020 August;106(2S):21-24
Inside the Article

Practice Points

  • Inform patients about current data guiding treatment from clinical trials of biologics.
  • Explain to patients that finding the treatment that is the best fit for them may require trial and error, as everyone responds to treatments differently.
  • Consult with patients about misconceptions and potential fears about biologics and what the protocol is for monitoring safety during treatment.
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
Article PDF Media

Treatment of Psoriasis in Pregnancy

Article Type
Changed
Thu, 11/03/2022 - 22:12

Historically, there have been limited data available on the management of psoriasis in pregnancy. The most comprehensive discussion of treatment guidelines is from 2012.1 In the interim, many biologics have been approved for treating psoriasis, with slow accumulation of pregnancy safety data. The 2019 American Academy of Dermatology–National Psoriasis Foundation guidelines on biologics for psoriasis contain updated information but also highlight the paucity of pregnancy safety data.2 This gap is in part a consequence of the exclusion and disenrollment of pregnant women from clinical trials.3 Additionally, lack of detection through registries contributes; pregnancy capture in registries is low compared to the expected number of pregnancies estimated from US Census data.4 Despite these shortcomings, psoriasis patients who are already pregnant or are considering becoming pregnant frequently are encountered in practice and may need treatment. This article reviews the evidence on commonly used treatments for psoriasis in pregnancy.

Background

For many patients, psoriasis improves during pregnancy5,6 and becomes worse postpartum. In a prospective study, most patients reported improvement in pregnancy corresponding to a significant decrease in affected body surface area (P<.001) by 10 to 20 weeks’ gestation. Most patients also reported worsening of psoriasis postpartum; a significant increase in psoriatic body surface area (P=.001) was observed after delivery.7 Despite these findings, a considerable number of patients also experience stable disease or worsening of disease during pregnancy.

In addition to the maternal disease state, the issue of pregnancy outcomes is paramount. In the inflammatory bowel disease and rheumatology literature, it is established that uncontrolled disease is associated with poorer pregnancy outcomes.8-10 Guidelines vary among societies on the use of biologics in pregnancy generally (eTable 11,2,9,11-24), but some societies recommend systemic agents to achieve disease control during pregnancy.9,25

Assessing the potential interplay between disease severity and outcomes in pregnant women with psoriasis is further complicated by the slowly growing body of literature demonstrating that women with psoriasis have more comorbidities26 and worse pregnancy outcomes.27,28 Pregnant psoriasis patients are more likely to smoke, have depression, and be overweight or obese prior to pregnancy and are less likely to take prenatal vitamins.26 They also have an increased risk for cesarean birth, gestational diabetes, gestational hypertension, and preeclampsia.28 In contrast to these prior studies, a systematic review revealed no risk for adverse outcomes in pregnant women with psoriasis.29

Assessment of Treatments for Psoriasis in Pregnancy

In light of these issues, treatment of psoriasis during pregnancy should be assessed from several vantage points. Of note, the US Food and Drug Administration changed its classification scheme in 2015 to a more narrative format called the Pregnancy and Lactation Labeling Rule.30 Prior classifications, however, provide a reasonable starting point for categorizing the safety of drugs (Table31). Importantly, time of exposure to systemic agents also matters; first-trimester exposure is more likely to affect embryogenesis, whereas second- and third-trimester exposures are more prone to affect other aspects of fetal growth. eTable 2 provides data on the use of oral and topical medications to treat psoriasis in pregnancy.1,8,22,32-45

Topical Agents
Topical steroids are largely understood to be reasonable treatment options, though consideration of potency, formulation, area of application, and use of occlusion is important.1,46 Risk for orofacial cleft has been noted with first-trimester topical steroid exposure, though a 2015 Cochrane review update determined that the relative risk of this association was not significantly elevated.32



The impact of topical calcipotriene and salicylic acid has not been studied in human pregnancies,1 but systemic absorption can occur for both. There is potential for vitamin D toxicity with calcipotriene46; consequently, use during pregnancy is not recommended.1,46 Some authors recommend against topical salicylic acid in pregnancy; others report that limited exposure is permissible.47 In fact, as salicylic acid commonly is found in over-the-counter acne products, many women of childbearing potential likely have quotidian exposure.

Preterm delivery and low birthweight have been reported with oral tacrolimus; however, risk with topical tacrolimus is thought to be low1 because the molecular size likely prohibits notable absorption.47 Evidence for the use of anthralin and coal tar also is scarce. First-trimester coal tar use should be avoided; subsequent use in pregnancy should be restricted given concern for adverse outcomes.1

 

 



Phototherapy
Broadband or narrowband UVB therapy is recommended as second-line therapy in pregnancy. No cases of fetal risk or premature delivery associated with UVB therapy were found in our search.1 Phototherapy can exacerbate melasma47 and decrease folate levels48; as such, some authors recommend folate supplementation in females of childbearing age who are being treated with phototherapy.49 Psoralen, used in psoralen plus UVA therapy, is mutagenic and therefore contraindicated in pregnancy.1

Oral Medications
Both methotrexate, which is a teratogen, abortifacient, and mutagen,1 and systemic retinoids, which are teratogens, are contraindicated in pregnancy.1,47 Acitretin labeling recommends avoiding pregnancy for 3 years posttreatment50 because alcohol intake prolongs the medication’s half-life.22



Apremilast use is not documented in pregnant psoriasis patients51; an ongoing registry of the Organization of Tetralogy Information Specialists has not reported publicly to date.52 Animal studies of apremilast have documented dose-related decreased birthweight and fetal loss.22

Safety data for systemic steroids, used infrequently in psoriasis, are not well established. First-trimester prednisone exposure has been associated with prematurity, low birthweight, and congenital abnormalities.38 A separate evaluation of 1047 children exposed to betamethasone in utero failed to demonstrate significant change in birthweight or head circumference. However, repeat antenatal corticosteroid exposure was associated with attention problems at 2 years of age.39

Data regarding cyclosporine use, derived primarily from organ transplant recipients, suggest elevated risk for prematurity and low birthweight.53,54 A meta-analysis demonstrated that organ transplant recipients taking cyclosporine had a nonsignificantly elevated odds ratio for congenital malformations, prematurity, and low birthweight.42 Cyclosporine use for psoriasis in pregnancy is not well described; in a study, rates of prematurity and low birthweight were both 21%.43 Limited data are available for Janus kinase inhibitors, none of which are approved for psoriasis, though clinical trials in psoriasis and psoriatic arthritis are underway (ClinicalTrials.gov identifiers NCT04246372, NCT03104374, NCT03104400).

Biologics and Small-Molecule Inhibitors
Limited data on biologics in pregnancy exist25 (eTable 3). Placental transport of IgG antibodies, including biologics, increases throughout pregnancy, especially in the third trimester.82 Infants of mothers treated with a biologic with potential for placental transfer are therefore considered by some authors to be immunosuppressed during the first months of life.2

Looking globally across biologics used for psoriasis, limited safety data are encouraging. In a review of PSOLAR (Psoriasis Longitudinal Assessment and Registry), 83 pregnancies with biologic exposure resulted in 59 live births (71%); 18 spontaneous abortions (22%); 6 induced abortions (7%); no congenital abnormalities; and 7 reports of neonatal problems, including respiratory issues, ABO blood group mismatch, hospitalization, and opioid withdrawal.83

Use of tumor necrosis factor (TNF) inhibitors in pregnancy has the most data25 and is considered a reasonable treatment option. Historically, there was concern about the risk for VACTERL syndrome (vertebral defects, anal atresia, cardiac defects, tracheoesophageal fistula, renal anomalies, limb abnormalities) with exposure to a TNF inhibitor,25,84-86 but further reports have alleviated these concerns. Active transplacental transport occurs for adalimumab, infliximab, and golimumab,87 but given structural differences, transport of certolizumab and etanercept is substantially less.88,89 In the CRIB study of placental transfer of certolizumab from mother to infant (N=14), pharmacokinetic data demonstrated no quantifiable certolizumab levels in 13 infants and minimal levels in 1 infant at birth.88 There are fewer data available on the use of other biologics in pregnancy, but for those in which active placental transport is relevant, similar concerns (ie, immunosuppression) might arise (eTable 3).

Concern over biologics largely involves risk for newborn immunosuppression. A case report detailed a Crohn disease patient treated with infliximab who gave birth to an infant who died of disseminated bacille Calmette-Guérin infection at 4.5 months after receiving the vaccine at 3 months.90 This case underscores the importance of delaying live vaccination in infants born to mothers who were treated with a biologic during pregnancy. Authors have provided various data on how long to avoid vaccination; some state as long as 1 year.91

In pregnant females with inflammatory bowel disease treated with a biologic, no correlation was observed among maternal, placental, and infant serum biologic levels and neonatal infection. However, an association between preterm birth and the level of the biologic in maternal and placental (but not infant) serum and preterm birth was observed.92



In another report from the same registry, combination therapy with a TNF inhibitor and another immunomodulator led to an increased risk for infection in infants at 12 months of age, compared to infants exposed to monotherapy89 or exposed to neither agent.93 A strategy to circumvent this potential problem is to avoid treatment with actively transported molecules in the third trimester.

Conclusion

Limited data exist to guide providers who are treating pregnant women with psoriasis. Our understanding of treatment of psoriasis in pregnancy is limited as a consequence of regulations surrounding clinical trials and inadequate detection of pregnancies in registries. Further efforts are necessary to better understand the relationship between psoriasis and pregnancy and how to manage pregnant women with psoriasis.

References
  1. Bae YS, Van Voorhees AS, Hsu S, et al. Review of treatment options for psoriasis in pregnant or lactating women: from the medical board of the National Psoriasis Foundation. J Am Acad Dermatol. 2012;67:459-477.
  2. Menter A, Strober BE, Kaplan DH, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with biologics. J Am Acad Dermatol. 2019;80:1029-1072.
  3. Flood KS, Porter ML, Kimball AB. Use of biologics in pregnancy: limitations stemming from clinical trials and registry experience. J Eur Acad Dermatol Venereol. 2019;33:E276-E277.
  4. Horn EJ, Chambers CD, Menter A, et al. Pregnancy outcomes in psoriasis: why do we know so little? J Am Acad Dermatol. 2009;61:E5-E8.
  5. Raychaudhuri SP, Navare T, Gross J, et al. Clinical course of psoriasis during pregnancy. Int J Dermatol. 2003;42:518-520.
  6. Boyd AS, Morris LF, Phillips CM, et al. Psoriasis and pregnancy: hormone and immune system interaction. Int J Dermatol. 1996;35:169-172.
  7. Murase JE, Chan KK, Garite TJ, et al. Hormonal effect on psoriasis in pregnancy and post partum. Arch Dermatol. 2005;14:601-606.
  8. Götestam Skorpen C, Hoeltzenbein M, Tincani A, et al. The EULAR points to consider for use of antirheumatic drugs before pregnancy, and during pregnancy and lactation. Ann Rheum Dis. 2016;75:795-810.
  9. Nguyen GC, Seow CH, Maxwell C, et al. The Toronto consensus statements for the management of inflammatory bowel disease in pregnancy. Gastroenterology. 2016;150:734-757.
  10. Wise J. Rheumatic diseases should be actively treated in pregnancy, new guidelines say. BMJ. 2016;532:i312.
  11. Puig L, Carrascosa JM, Carretero G, et al. Spanish evidence-based guidelines on the treatment of psoriasis with biologic agents, 2013. part 1: on efficacy and choice of treatment. Actas Dermosifiliogr. 2013;104:694-709.
  12. Girolomoni G, Altomore G, Ayala F, et al. Differential management of mild-to-severe psoriasis with biologic drugs: an Italian Delphi consensus expert panel. J Dermatolog Treat. 2015;26:128-133.
  13. Yeung J, Gooderham MJ, Grewal P, et al. Management of plaque psoriasis with biologic therapies in women of child-bearing potential consensus paper. J Cutan Med Surg. 2020;24:3S-14S.
  14. Louthrenoo W, Kasitanon N, Kathamort W, et al. 2016 updated Thai Rheumatism Association Recommendations for the use of biologic and targeted synthetic disease-modifying anti-rheumatic drugs in patients with rheumatoid arthritis. Int J Rheum Dis. 2017;20:1166-1184.
  15. Flint J, Panchal S, Hurrell A, et al. BSR and BHPR guideline on prescribing drugs in pregnancy and breastfeeding—part I: standard and biologic disease modifying anti-rheumatic drugs and corticosteroids. Rheumatology (Oxford). 2016;55:1693-1697.
  16. van der Woude CJ, Ardizzone S, Bengtson MB, et al. The second European evidenced-based consensus on reproduction and pregnancy in inflammatory bowel disease. J Crohns Colitis. 2015;9:107-124.
  17. Orlando A, Armuzz A, Papi C, et al. The Italian Society of Gastroenterology (SIGE) and the Italian Group for the study of Inflammatory Bowel Disease (IG-IBD) Clinical Practice Guidelines: the use of tumor necrosis factor-alpha antagonist therapy in inflammatory bowel disease. Dig Liver Dis. 2011;43:1-20.
  18. Puchner A, Grochenig HP, Sautner J, et al. Immunosuppressives and biologics during pregnancy and lactation. Wien Klin Wochenschr. 2019;131:29-44.
  19. ACOG Committee opinion no. 776: immune modulating therapies in pregnancy and lactation. Obstet Gynecol. 2019;133:E287-E297.
  20. Lamb CA, Kennedy NA, Raine T, et al. British Society of Gastroenterology consensus guidelines on the management of inflammatory bowel disease in adults. Gut. 2019;68:S1-S106.
  21. Goëb V, Ardizzone M, Arnard L, et al. Recommendations for using TNF-α antagonists and French Clinical Practice Guidelines endorsed by the French National Authority for Health. Joint Bone Spine. 2013;80:574-581.
  22. Rademaker M, Agnew K, Andrews M, et al. Psoriasis in those planning a family, pregnant or breast-feeding. the Australasian Psoriasis Collaboration. Australas J Dermatol. 2018;59:86-100.
  23. Mahadevan U, Robinson C, Bernasko N, et al. Inflammatory bowel disease in pregnancy clinical care pathway: a report from the American Gastroenterological Association IBD Parenthood Project Working Group. Gastroenterology. 2019;156:1508-1524.
  24. Mahadevan U, Cucchiara S, Hyam JS, et al. The London position statement of the World Congress of Gastroenterology on biological therapy for IBD with the European Crohn’s and Colitis Organisation: pregnancy and pediatrics. Am J Gastroenterol. 2011;106:214-223.
  25. Porter ML, Lockwood SJ, Kimball AB. Update on biologic safety for patients with psoriasis during pregnancy. Int J Womens Dermatol. 2017;3:21-25.
  26. Bandoli G, Johnson DL, Jones KL, et al. Potentially modifiable risk factors for adverse pregnancy outcomes in women with psoriasis. Br J Dermatol. 2010;163:334-339.
  27. Lima XT, Janakiraman V, Hughes MD, et al. The impact of psoriasis on pregnancy outcomes. J Invest Dermatol. 2012;132:85-91.
  28. Bröms G, Haerskjold A, Granath F, et al. Effect of maternal psoriasis on pregnancy and birth outcomes: a population-based cohort study from Denmark and Sweden. Acta Derm Venereol. 2018;98:728-734.
  29. Bobotsis R, Gulliver WP, Monaghan K, et al. Psoriasis and adverse pregnancy outcomes: a systematic review of observational studies. Br J Dermatol. 2016;175:464-472.
  30. Blattner CM, Danesh M, Safaee M, et al. Understanding the new FDA pregnancy and lactation labeling rules. Int J Womens Dermatol. 2016;2:5-7.
  31. Pernia S, DeMaagd G. The new pregnancy and lactation labeling rule. P T. 2016;4:713-715.
  32. Chi C-C, Wang S-H, Wojnarowska F, et al. Safety of topical corticosteroids in pregnancy. Cochrane Database Syst Rev. 2015:CD007346.
  33. Chi CC, Wang SH, Kirtschig G. Safety of topical corticosteroids in pregnancy. JAMA Dermatol. 2016;152:934-935.
  34. Dovonex (calcipotriene) Cream, 0.005% [package insert]. Dublin, Ireland: Leo Laboratories, Ltd; March 2015.
  35. Franssen ME, van der Wilt GJ, de Jong PC, et al. A retrospective study of the teratogenicity of dermatological coal tar products. Acta Derm Venereol. 1999;79:390-391.
  36. Garbis H, Elefant E, Bertolotti E, et al. Pregnancy outcome after periconceptional and first-trimester exposure to methoxsalen photochemotherapy. Arch Dermatol. 1995;131:492-493.
  37. Horizon Pharma USA. RAYOS (prednisone). https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/202020s000lbl.pdf https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/202020s000lbl.pdf.
  38. Park-Wyllie L, Mazzotta P, Pastuszak A, et al. Birth defects after maternal exposure to corticosteroids: prospective cohort study and meta-analysis of epidemiological studies. Teratology. 2000;62:385-392.
  39. Crowther CA, Doyle LW, Haslam RR, et al. Outcomes at 2 years of age after repeat doses of antenatal corticosteroids. N Engl J Med. 2007;357:1179-1189.
  40. Palmsten K, Rolland M, Herbert MF, et al. Patterns of prednisone use during pregnancy in women with rheumatoid arthritis: daily and cumulative dose. Pharmacoepidemiol Drug Saf. 2018;27:430-438.
  41. Groth K, Brännström M, Mölne J, et al. Cyclosporine A exposure during pregnancy in mice: effects on reproductive performance in mothers and offspring. Hum Reprod. 2010;25:697-704.
  42. Bar Oz B, Hackman R, Einarson T, et al. Pregnancy outcome after cyclosporine therapy during pregnancy: a meta-analysis. Transplantation. 2001;71:1051-1055.
  43. Paziana K, Del Monaco M, Cardonick E, et al. Ciclosporin use during pregnancy. Drug Saf. 2013;36:279-294.
  44. Lamarque V, Leleu MF, Monka C, et al. Analysis of 629 pregnancy outcomes in transplant recipients treated with Sandimmun. Transplant Proc. 1997;29:2480.
  45. Otezla (apremilast) tablets, for oral use [package insert]. Summit, NJ: Celgene Corporation; June 2017. http://www.accessdata.fda.gov/drugsatfda_docs/label/2017/205437s006lbl.pdf. Accessed July 8, 2020.
  46. Kurizky PS, de Castro Ferreira C, Nogueira LSC, et al. Treatment of psoriasis and psoriatic arthritis during pregnancy and breastfeeding. An Bras Dermatol. 2015;90:367-375.
  47. Murase JE, Heller MM, Butler DC. Safety of dermatologic medications in pregnancy and lactation. J Am Acad Dermatol. 2014;70:401.e1-401.e4.
  48. El-Saie LT, Rabie AR, Kamel MI, et al. Effect of narrowband ultraviolet B phototherapy on serum folic acid levels in patients with psoriasis. Lasers Med Sci. 2011;26:481-485.
  49. Murase JE, Koo JY, Berger TG. Narrowband ultraviolet B phototherapy influences serum folate levels in patients with vitiligo. J Am Acad Dermatol. 2010;62:710-711.
  50. Soriatane (acitretin) capsules [package insert]. Morrisville, NC: Stiefel Laboratories, Inc; April 2011. http://www.accessdata.fda.gov/drugsatfda_docs/label/2011/019821s018mg.pdf. Accessed July 8, 2020.
  51. Kaushik SB, Lebwohl MG. Psoriasis: which therapy for which patient: focus on special populations and chronic infections. J Am Acad Dermatol. 2019;80:43-53.
  52. Help us better understand the effects of Otezla in pregnancy. MotherToBaby website. https://mothertobaby.org/ongoing-study/otezla/. Accessed July 8, 2020.
  53. Bangsgaard N, Rørbye C, Skov L. Treating psoriasis during pregnancy: safety and efficacy of treatments. Am J Clin Dermatol. 2015;16:389-398.
  54. Tyler K. Dermatologic therapy in pregnancy. Clin Obstet Gynecol. 2015;58:112-118.
  55. Luu M, Benzenine E, Doret M, et al. Continuous anti–TNF-α use throughout pregnancy: possible complications for the mother but not for the fetus. a retrospective cohort on the French National Health Insurance Database (EVASION). Am J Gastroenterol. 2018;113:1669-1677.
  56. Bröms G, Granath F, Ekbom A, et al. Low risk of birth defects for infants whose mothers are treated with anti-tumor necrosis factor agents during pregnancy. Clin Gastroenterol Hepatol. 2016;14:234-241.
  57. Mirdamadi K, Salinas T, Vali R, et al. Meta-analysis of pregnancy outcomes after exposure to TNF-α inhibitors during pregnancy for the treatment of arthritic diseases. J Popul Ther Clin Pharmacol. 2018;25:E53-E56.
  58. Shihab Z, Yeomans ND, De Cruz P. Anti-tumour necrosis factor α therapies and inflammatory bowel disease pregnancy outcomes: a meta-analysis. J Crohns Colitis. 2016;10:979-988.
  59. Bröms G, Kieler H, Ekbom A, et al. Anti-TNF treatment during pregnancy and birth outcomes: a population-based study from Denmark, Finland, and Sweden. Pharmacoepidemiol Drug Saf. 2020;29:316-327.
  60. Diav-Citrin O, Otcheretianski-Volodarsky A, Shechtman S, et al. Pregnancy outcome following gestational exposure to TNF-alpha-inhibitors: a prospective, comparative, observational study. Reprod Toxicol. 2014;43:78-84.
  61. FDA determinations for pregnant and nursing women. National Psoriasis Foundation website. http://www.psoriasis.org/pregnancy/fda-determinations. Accessed July 8, 2020.
  62. Østensen M. Safety issues of biologics in pregnant patients with rheumatic diseases. Ann N Y Acad Sci. 2014;1317:32-38.
  63. Chambers CD, Johnson DL, Luo Y, et al. Pregnancy outcome in women treated with adalimumab for the treatment of rheumatoid arthritis: the OTIS Autoimmune Diseases in Pregnancy Project. Arthritis Rheum. 2012;64:2466.
  64. Clowse ME, Wolf DC, Forger F, et al. Pregnancy outcomes after exposure to certolizumab pegol: updated results from a pharmacovigilance safety database. Arthritis Rheumatol. 2018;70:1399-1407.
  65. Carman WJ, Accortt NA, Anthony MS, et al. Pregnancy and infant outcomes including major congenital malformations among women with chronic inflammatory arthritis or psoriasis, with and without etanercept use. Pharmacoepidemiol Drug Saf. 2017;26:1109-1118.
  66. Janssen. SIMPONI (golilumab). https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/125289s0064lbl.pdf.
  67. Yurkon K, Guo CY, Harrison D, et al. Pregnancy outcomes in women with dermatologic conditions exposed to infliximab. J Am Acad Dermatol. 2014;70:AB179.
  68. Watson N, Wu K, Farr P, et al. Ustekinumab exposure during conception and pregnancy in patients with chronic plaque psoriasis: a case series of 10 pregnancies. Br J Dermatol. 2019;180:195-196.
  69. Naureckas S, Slater J, Gearhart N, et al. Pregnancy outcomes in women with psoriasis and psoriatic arthritis exposed to ustekinumab. J Am Acad Dermatol. 2016;74:AB264.
  70. Haycraft K, DiRuggiero D, Rozzo SJ, et al. Outcomes of pregnancies from tildrakizumab phases I to III clinical development program. J Clin Aesthet Dermatol. 2019;12:S27-S28.
  71. Tremfya (guselkumab) injection, for subcutaneous use [package insert]. Horsham, PA: Janssen Biotech, Inc; July 2017. http://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761061s000lbl.pdf. Accessed Juy 8, 2020.
  72. Skyrizi (risankizumab-rzaa) injection, for subcutaneous use [package insert]. Northi Chicago, IL; April 2019. http://www.accessdata.fda.gov/drugsatfda_docs/label/2019/761105s000lbl.pdf. Accessed July 8, 2020.
  73. Siliq (brodalumab) injection, for subcutaneous use [package insert]. Bridgewater, NJ: Valeant Pharmaceuticals North America LLC; February 2017. http://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761032lbl.pdf. Accessed July 8, 2020.
  74. Feldman S, Pangallo B, Xu W, et al. Ixekizumab and pregnancy outcome. J Am Acad Dermatol. 2017;76:AB419.
  75. Clarke DO, Hilbish KG, Waters DG, et al. Assessment of ixekizumab, an interleukin-17A monoclonal antibody, for potential effects on reproduction and development, including immune system function, in cynomolgus monkeys. Reprod Toxicol. 2015;58:160-173.
  76. Warren RB, Reich K, Langley RG, et al. Secukinumab in pregnancy: outcomes in psoriasis, psoriatic arthritis and ankylosing spondylitis from the global safety database. Br J Dermatol. 2018;179:1205-1207.
  77. Nardin C, Colas M, Curie V, et al. Pregnancy after tubal sterilization in a woman treated with biologics for severe psoriasis. Dermatol Ther (Heidelb). 2018;8:323-326.
  78. Xeljanz (tofacitinib) tablets for oral administration [package insert]. New York, NY: Pfizer; November 2012. http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/203214s000lbl.pdf. Accessed July 8, 2020.
  79. Pfizer. Xeljanz (tofacitinib). https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/203214s018lbl.pdf.
  80. Mahadevan U, Dubinsky M, Su C, et al. Outcomes of pregnancies with maternal/paternal exposure in the tofacitinib safety databases for ulcerative colitis. Inflamm Bowel Dis. 2018;24:2494-2500.
  81. Clowse ME, Feldman SR, Isaacs JD, et al. Pregnancy outcomes in the tofacitinib safety databases for rheumatoid arthritis and psoriasis. Drug Saf. 2016;39:755-762.
  82. Malek A, Sager R, Kuhn P, et al. Evolution of maternofetal transport of immunoglobulins during human pregnancy. Am J Reprod Immunol. 1996;36:248-255.
  83. Kimball AB, Crow JA, Ridley K, et al. Pregnancy outcomes in women with moderate to severe psoriasis: the PSOLAR experience. J Am Acad Dermatol. 2014;70(suppl 1):AB179.
  84. Carter JD, Valeriano J, Vasey FB. Tumor necrosis factor-alpha inhibition and VATER association: a causal relationship. J Rheumatol. 2006;33:1014-1017.
  85. Carter JD, Ladhani A, Ricca LR, et al. A safety assessment of tumor necrosis factor antagonists during pregnancy: a review of the Food and Drug Administration database. J Rheumatol. 2009;36:635-641.
  86. Koren G, Inoue M. Do tumor necrosis factor inhibitors cause malformations in humans? J Rheumatol. 2009;36:465-466.
  87. Johansen C, Jimenez-Solem E, Haerskjold A, et al. The use and safety of TNF inhibitors during pregnancy in women with psoriasis: a review. Int J Mol Sci. 2018;19:E1349.
  88. Mariette X, Forger F, Abraham B, et al. Lack of placental transfer of certolizumab pegol during pregnancy: results from CRIB, a prospective, postmarketing, pharmacokinetic study. Ann Rheum Dis. 2018;77:228-233.
  89. Mahadevan U, Wolf DC, Dubinsky M, et al. Placental transfer of anti-tumor necrosis factor agents in pregnant patients with inflammatory bowel disease. Clin Gastroenterol Hepatol. 2013;11:286-292.
  90. Cheent K, Nolan J, Sharig S, et al. Case report: fatal case of disseminated BCG infection in an infant born to a mother taking infliximab for Crohn’s disease. J Crohns Colitis. 2010;4:603-605.
  91. Julsgaard M, Christensen LA, Gibson PR, et al. Concentrations of adalimumab and infliximab in mothers and newborns, and effects on infection. Gastroenterology. 2016;151:110-119.
  92. Mahadevan U, Martin C, Kane SV, et al. Do infant serum levels of biologic agents at birth correlate with risk of adverse outcomes? results from the PIANO registry. Gastroenterology. 2016;150:S91-S92.
  93. Mahadevan U, Martin CF, Sandler RS, et al. PIANO: a 1000 patient prospective registry of pregnancy outcomes in women with IBD exposed to immunomodulators and biologic therapy [AGA abstract 865]. Gastroenterology. 2012;142:S-149.
Article PDF
Author and Disclosure Information

Dr. Flood is from the Department of Dermatology, University of Cincinnati, Ohio. Drs. Savage, Porter, and Kimball are from Beth Israel Deaconess Medical Center, Boston, Massachusetts. Drs. Porter and Kimball are from the Clinical Laboratory for Epidemiology and Applied Research in Skin (CLEARS), Department of Dermatology.

Dr. Flood has received fellowship funding from AbbVie; Janssen Biotech, Inc; and the National Psoriasis Foundation that went directly to her institution. Dr. Savage reports no conflict of interest. Dr. Porter is a consultant and/or investigator for AbbVie; Bristol-Myers Squibb; Eli Lilly and Company; Janssen Biotech, Inc; Novartis; Pfizer; and UCB. She gave a talk on psoriasis and pregnancy for an honorarium for the National Psoriasis Foundation. Dr. Kimball is a consultant and investigator for AbbVie; Bristol-Myers Squibb; Eli Lilly and Company; Janssen Biotech, Inc; Novartis; Pfizer Inc; and UCB. She serves on the University of California, San Diego, OTIS (Organization of Teratology Information Specialists) Pregnancy Study Scientific Advisory Panels for the Apremilast Pregnancy Exposure Registry (Amgen Inc) and the Ustekinumab Pregnancy Exposure Registry (Janssen Biotech, Inc). Her fellowship program receives funding from AbbVie; Janssen Biotech, Inc; and the National Psoriasis Foundation.

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

Correspondence: Alexa B. Kimball, MD, MPH, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215 ([email protected]).

Issue
Cutis - 106(2S)
Publications
Topics
Page Number
15-20, E1-E9
Sections
Author and Disclosure Information

Dr. Flood is from the Department of Dermatology, University of Cincinnati, Ohio. Drs. Savage, Porter, and Kimball are from Beth Israel Deaconess Medical Center, Boston, Massachusetts. Drs. Porter and Kimball are from the Clinical Laboratory for Epidemiology and Applied Research in Skin (CLEARS), Department of Dermatology.

Dr. Flood has received fellowship funding from AbbVie; Janssen Biotech, Inc; and the National Psoriasis Foundation that went directly to her institution. Dr. Savage reports no conflict of interest. Dr. Porter is a consultant and/or investigator for AbbVie; Bristol-Myers Squibb; Eli Lilly and Company; Janssen Biotech, Inc; Novartis; Pfizer; and UCB. She gave a talk on psoriasis and pregnancy for an honorarium for the National Psoriasis Foundation. Dr. Kimball is a consultant and investigator for AbbVie; Bristol-Myers Squibb; Eli Lilly and Company; Janssen Biotech, Inc; Novartis; Pfizer Inc; and UCB. She serves on the University of California, San Diego, OTIS (Organization of Teratology Information Specialists) Pregnancy Study Scientific Advisory Panels for the Apremilast Pregnancy Exposure Registry (Amgen Inc) and the Ustekinumab Pregnancy Exposure Registry (Janssen Biotech, Inc). Her fellowship program receives funding from AbbVie; Janssen Biotech, Inc; and the National Psoriasis Foundation.

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

Correspondence: Alexa B. Kimball, MD, MPH, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215 ([email protected]).

Author and Disclosure Information

Dr. Flood is from the Department of Dermatology, University of Cincinnati, Ohio. Drs. Savage, Porter, and Kimball are from Beth Israel Deaconess Medical Center, Boston, Massachusetts. Drs. Porter and Kimball are from the Clinical Laboratory for Epidemiology and Applied Research in Skin (CLEARS), Department of Dermatology.

Dr. Flood has received fellowship funding from AbbVie; Janssen Biotech, Inc; and the National Psoriasis Foundation that went directly to her institution. Dr. Savage reports no conflict of interest. Dr. Porter is a consultant and/or investigator for AbbVie; Bristol-Myers Squibb; Eli Lilly and Company; Janssen Biotech, Inc; Novartis; Pfizer; and UCB. She gave a talk on psoriasis and pregnancy for an honorarium for the National Psoriasis Foundation. Dr. Kimball is a consultant and investigator for AbbVie; Bristol-Myers Squibb; Eli Lilly and Company; Janssen Biotech, Inc; Novartis; Pfizer Inc; and UCB. She serves on the University of California, San Diego, OTIS (Organization of Teratology Information Specialists) Pregnancy Study Scientific Advisory Panels for the Apremilast Pregnancy Exposure Registry (Amgen Inc) and the Ustekinumab Pregnancy Exposure Registry (Janssen Biotech, Inc). Her fellowship program receives funding from AbbVie; Janssen Biotech, Inc; and the National Psoriasis Foundation.

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

Correspondence: Alexa B. Kimball, MD, MPH, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA 02215 ([email protected]).

Article PDF
Article PDF

Historically, there have been limited data available on the management of psoriasis in pregnancy. The most comprehensive discussion of treatment guidelines is from 2012.1 In the interim, many biologics have been approved for treating psoriasis, with slow accumulation of pregnancy safety data. The 2019 American Academy of Dermatology–National Psoriasis Foundation guidelines on biologics for psoriasis contain updated information but also highlight the paucity of pregnancy safety data.2 This gap is in part a consequence of the exclusion and disenrollment of pregnant women from clinical trials.3 Additionally, lack of detection through registries contributes; pregnancy capture in registries is low compared to the expected number of pregnancies estimated from US Census data.4 Despite these shortcomings, psoriasis patients who are already pregnant or are considering becoming pregnant frequently are encountered in practice and may need treatment. This article reviews the evidence on commonly used treatments for psoriasis in pregnancy.

Background

For many patients, psoriasis improves during pregnancy5,6 and becomes worse postpartum. In a prospective study, most patients reported improvement in pregnancy corresponding to a significant decrease in affected body surface area (P<.001) by 10 to 20 weeks’ gestation. Most patients also reported worsening of psoriasis postpartum; a significant increase in psoriatic body surface area (P=.001) was observed after delivery.7 Despite these findings, a considerable number of patients also experience stable disease or worsening of disease during pregnancy.

In addition to the maternal disease state, the issue of pregnancy outcomes is paramount. In the inflammatory bowel disease and rheumatology literature, it is established that uncontrolled disease is associated with poorer pregnancy outcomes.8-10 Guidelines vary among societies on the use of biologics in pregnancy generally (eTable 11,2,9,11-24), but some societies recommend systemic agents to achieve disease control during pregnancy.9,25

Assessing the potential interplay between disease severity and outcomes in pregnant women with psoriasis is further complicated by the slowly growing body of literature demonstrating that women with psoriasis have more comorbidities26 and worse pregnancy outcomes.27,28 Pregnant psoriasis patients are more likely to smoke, have depression, and be overweight or obese prior to pregnancy and are less likely to take prenatal vitamins.26 They also have an increased risk for cesarean birth, gestational diabetes, gestational hypertension, and preeclampsia.28 In contrast to these prior studies, a systematic review revealed no risk for adverse outcomes in pregnant women with psoriasis.29

Assessment of Treatments for Psoriasis in Pregnancy

In light of these issues, treatment of psoriasis during pregnancy should be assessed from several vantage points. Of note, the US Food and Drug Administration changed its classification scheme in 2015 to a more narrative format called the Pregnancy and Lactation Labeling Rule.30 Prior classifications, however, provide a reasonable starting point for categorizing the safety of drugs (Table31). Importantly, time of exposure to systemic agents also matters; first-trimester exposure is more likely to affect embryogenesis, whereas second- and third-trimester exposures are more prone to affect other aspects of fetal growth. eTable 2 provides data on the use of oral and topical medications to treat psoriasis in pregnancy.1,8,22,32-45

Topical Agents
Topical steroids are largely understood to be reasonable treatment options, though consideration of potency, formulation, area of application, and use of occlusion is important.1,46 Risk for orofacial cleft has been noted with first-trimester topical steroid exposure, though a 2015 Cochrane review update determined that the relative risk of this association was not significantly elevated.32



The impact of topical calcipotriene and salicylic acid has not been studied in human pregnancies,1 but systemic absorption can occur for both. There is potential for vitamin D toxicity with calcipotriene46; consequently, use during pregnancy is not recommended.1,46 Some authors recommend against topical salicylic acid in pregnancy; others report that limited exposure is permissible.47 In fact, as salicylic acid commonly is found in over-the-counter acne products, many women of childbearing potential likely have quotidian exposure.

Preterm delivery and low birthweight have been reported with oral tacrolimus; however, risk with topical tacrolimus is thought to be low1 because the molecular size likely prohibits notable absorption.47 Evidence for the use of anthralin and coal tar also is scarce. First-trimester coal tar use should be avoided; subsequent use in pregnancy should be restricted given concern for adverse outcomes.1

 

 



Phototherapy
Broadband or narrowband UVB therapy is recommended as second-line therapy in pregnancy. No cases of fetal risk or premature delivery associated with UVB therapy were found in our search.1 Phototherapy can exacerbate melasma47 and decrease folate levels48; as such, some authors recommend folate supplementation in females of childbearing age who are being treated with phototherapy.49 Psoralen, used in psoralen plus UVA therapy, is mutagenic and therefore contraindicated in pregnancy.1

Oral Medications
Both methotrexate, which is a teratogen, abortifacient, and mutagen,1 and systemic retinoids, which are teratogens, are contraindicated in pregnancy.1,47 Acitretin labeling recommends avoiding pregnancy for 3 years posttreatment50 because alcohol intake prolongs the medication’s half-life.22



Apremilast use is not documented in pregnant psoriasis patients51; an ongoing registry of the Organization of Tetralogy Information Specialists has not reported publicly to date.52 Animal studies of apremilast have documented dose-related decreased birthweight and fetal loss.22

Safety data for systemic steroids, used infrequently in psoriasis, are not well established. First-trimester prednisone exposure has been associated with prematurity, low birthweight, and congenital abnormalities.38 A separate evaluation of 1047 children exposed to betamethasone in utero failed to demonstrate significant change in birthweight or head circumference. However, repeat antenatal corticosteroid exposure was associated with attention problems at 2 years of age.39

Data regarding cyclosporine use, derived primarily from organ transplant recipients, suggest elevated risk for prematurity and low birthweight.53,54 A meta-analysis demonstrated that organ transplant recipients taking cyclosporine had a nonsignificantly elevated odds ratio for congenital malformations, prematurity, and low birthweight.42 Cyclosporine use for psoriasis in pregnancy is not well described; in a study, rates of prematurity and low birthweight were both 21%.43 Limited data are available for Janus kinase inhibitors, none of which are approved for psoriasis, though clinical trials in psoriasis and psoriatic arthritis are underway (ClinicalTrials.gov identifiers NCT04246372, NCT03104374, NCT03104400).

Biologics and Small-Molecule Inhibitors
Limited data on biologics in pregnancy exist25 (eTable 3). Placental transport of IgG antibodies, including biologics, increases throughout pregnancy, especially in the third trimester.82 Infants of mothers treated with a biologic with potential for placental transfer are therefore considered by some authors to be immunosuppressed during the first months of life.2

Looking globally across biologics used for psoriasis, limited safety data are encouraging. In a review of PSOLAR (Psoriasis Longitudinal Assessment and Registry), 83 pregnancies with biologic exposure resulted in 59 live births (71%); 18 spontaneous abortions (22%); 6 induced abortions (7%); no congenital abnormalities; and 7 reports of neonatal problems, including respiratory issues, ABO blood group mismatch, hospitalization, and opioid withdrawal.83

Use of tumor necrosis factor (TNF) inhibitors in pregnancy has the most data25 and is considered a reasonable treatment option. Historically, there was concern about the risk for VACTERL syndrome (vertebral defects, anal atresia, cardiac defects, tracheoesophageal fistula, renal anomalies, limb abnormalities) with exposure to a TNF inhibitor,25,84-86 but further reports have alleviated these concerns. Active transplacental transport occurs for adalimumab, infliximab, and golimumab,87 but given structural differences, transport of certolizumab and etanercept is substantially less.88,89 In the CRIB study of placental transfer of certolizumab from mother to infant (N=14), pharmacokinetic data demonstrated no quantifiable certolizumab levels in 13 infants and minimal levels in 1 infant at birth.88 There are fewer data available on the use of other biologics in pregnancy, but for those in which active placental transport is relevant, similar concerns (ie, immunosuppression) might arise (eTable 3).

Concern over biologics largely involves risk for newborn immunosuppression. A case report detailed a Crohn disease patient treated with infliximab who gave birth to an infant who died of disseminated bacille Calmette-Guérin infection at 4.5 months after receiving the vaccine at 3 months.90 This case underscores the importance of delaying live vaccination in infants born to mothers who were treated with a biologic during pregnancy. Authors have provided various data on how long to avoid vaccination; some state as long as 1 year.91

In pregnant females with inflammatory bowel disease treated with a biologic, no correlation was observed among maternal, placental, and infant serum biologic levels and neonatal infection. However, an association between preterm birth and the level of the biologic in maternal and placental (but not infant) serum and preterm birth was observed.92



In another report from the same registry, combination therapy with a TNF inhibitor and another immunomodulator led to an increased risk for infection in infants at 12 months of age, compared to infants exposed to monotherapy89 or exposed to neither agent.93 A strategy to circumvent this potential problem is to avoid treatment with actively transported molecules in the third trimester.

Conclusion

Limited data exist to guide providers who are treating pregnant women with psoriasis. Our understanding of treatment of psoriasis in pregnancy is limited as a consequence of regulations surrounding clinical trials and inadequate detection of pregnancies in registries. Further efforts are necessary to better understand the relationship between psoriasis and pregnancy and how to manage pregnant women with psoriasis.

Historically, there have been limited data available on the management of psoriasis in pregnancy. The most comprehensive discussion of treatment guidelines is from 2012.1 In the interim, many biologics have been approved for treating psoriasis, with slow accumulation of pregnancy safety data. The 2019 American Academy of Dermatology–National Psoriasis Foundation guidelines on biologics for psoriasis contain updated information but also highlight the paucity of pregnancy safety data.2 This gap is in part a consequence of the exclusion and disenrollment of pregnant women from clinical trials.3 Additionally, lack of detection through registries contributes; pregnancy capture in registries is low compared to the expected number of pregnancies estimated from US Census data.4 Despite these shortcomings, psoriasis patients who are already pregnant or are considering becoming pregnant frequently are encountered in practice and may need treatment. This article reviews the evidence on commonly used treatments for psoriasis in pregnancy.

Background

For many patients, psoriasis improves during pregnancy5,6 and becomes worse postpartum. In a prospective study, most patients reported improvement in pregnancy corresponding to a significant decrease in affected body surface area (P<.001) by 10 to 20 weeks’ gestation. Most patients also reported worsening of psoriasis postpartum; a significant increase in psoriatic body surface area (P=.001) was observed after delivery.7 Despite these findings, a considerable number of patients also experience stable disease or worsening of disease during pregnancy.

In addition to the maternal disease state, the issue of pregnancy outcomes is paramount. In the inflammatory bowel disease and rheumatology literature, it is established that uncontrolled disease is associated with poorer pregnancy outcomes.8-10 Guidelines vary among societies on the use of biologics in pregnancy generally (eTable 11,2,9,11-24), but some societies recommend systemic agents to achieve disease control during pregnancy.9,25

Assessing the potential interplay between disease severity and outcomes in pregnant women with psoriasis is further complicated by the slowly growing body of literature demonstrating that women with psoriasis have more comorbidities26 and worse pregnancy outcomes.27,28 Pregnant psoriasis patients are more likely to smoke, have depression, and be overweight or obese prior to pregnancy and are less likely to take prenatal vitamins.26 They also have an increased risk for cesarean birth, gestational diabetes, gestational hypertension, and preeclampsia.28 In contrast to these prior studies, a systematic review revealed no risk for adverse outcomes in pregnant women with psoriasis.29

Assessment of Treatments for Psoriasis in Pregnancy

In light of these issues, treatment of psoriasis during pregnancy should be assessed from several vantage points. Of note, the US Food and Drug Administration changed its classification scheme in 2015 to a more narrative format called the Pregnancy and Lactation Labeling Rule.30 Prior classifications, however, provide a reasonable starting point for categorizing the safety of drugs (Table31). Importantly, time of exposure to systemic agents also matters; first-trimester exposure is more likely to affect embryogenesis, whereas second- and third-trimester exposures are more prone to affect other aspects of fetal growth. eTable 2 provides data on the use of oral and topical medications to treat psoriasis in pregnancy.1,8,22,32-45

Topical Agents
Topical steroids are largely understood to be reasonable treatment options, though consideration of potency, formulation, area of application, and use of occlusion is important.1,46 Risk for orofacial cleft has been noted with first-trimester topical steroid exposure, though a 2015 Cochrane review update determined that the relative risk of this association was not significantly elevated.32



The impact of topical calcipotriene and salicylic acid has not been studied in human pregnancies,1 but systemic absorption can occur for both. There is potential for vitamin D toxicity with calcipotriene46; consequently, use during pregnancy is not recommended.1,46 Some authors recommend against topical salicylic acid in pregnancy; others report that limited exposure is permissible.47 In fact, as salicylic acid commonly is found in over-the-counter acne products, many women of childbearing potential likely have quotidian exposure.

Preterm delivery and low birthweight have been reported with oral tacrolimus; however, risk with topical tacrolimus is thought to be low1 because the molecular size likely prohibits notable absorption.47 Evidence for the use of anthralin and coal tar also is scarce. First-trimester coal tar use should be avoided; subsequent use in pregnancy should be restricted given concern for adverse outcomes.1

 

 



Phototherapy
Broadband or narrowband UVB therapy is recommended as second-line therapy in pregnancy. No cases of fetal risk or premature delivery associated with UVB therapy were found in our search.1 Phototherapy can exacerbate melasma47 and decrease folate levels48; as such, some authors recommend folate supplementation in females of childbearing age who are being treated with phototherapy.49 Psoralen, used in psoralen plus UVA therapy, is mutagenic and therefore contraindicated in pregnancy.1

Oral Medications
Both methotrexate, which is a teratogen, abortifacient, and mutagen,1 and systemic retinoids, which are teratogens, are contraindicated in pregnancy.1,47 Acitretin labeling recommends avoiding pregnancy for 3 years posttreatment50 because alcohol intake prolongs the medication’s half-life.22



Apremilast use is not documented in pregnant psoriasis patients51; an ongoing registry of the Organization of Tetralogy Information Specialists has not reported publicly to date.52 Animal studies of apremilast have documented dose-related decreased birthweight and fetal loss.22

Safety data for systemic steroids, used infrequently in psoriasis, are not well established. First-trimester prednisone exposure has been associated with prematurity, low birthweight, and congenital abnormalities.38 A separate evaluation of 1047 children exposed to betamethasone in utero failed to demonstrate significant change in birthweight or head circumference. However, repeat antenatal corticosteroid exposure was associated with attention problems at 2 years of age.39

Data regarding cyclosporine use, derived primarily from organ transplant recipients, suggest elevated risk for prematurity and low birthweight.53,54 A meta-analysis demonstrated that organ transplant recipients taking cyclosporine had a nonsignificantly elevated odds ratio for congenital malformations, prematurity, and low birthweight.42 Cyclosporine use for psoriasis in pregnancy is not well described; in a study, rates of prematurity and low birthweight were both 21%.43 Limited data are available for Janus kinase inhibitors, none of which are approved for psoriasis, though clinical trials in psoriasis and psoriatic arthritis are underway (ClinicalTrials.gov identifiers NCT04246372, NCT03104374, NCT03104400).

Biologics and Small-Molecule Inhibitors
Limited data on biologics in pregnancy exist25 (eTable 3). Placental transport of IgG antibodies, including biologics, increases throughout pregnancy, especially in the third trimester.82 Infants of mothers treated with a biologic with potential for placental transfer are therefore considered by some authors to be immunosuppressed during the first months of life.2

Looking globally across biologics used for psoriasis, limited safety data are encouraging. In a review of PSOLAR (Psoriasis Longitudinal Assessment and Registry), 83 pregnancies with biologic exposure resulted in 59 live births (71%); 18 spontaneous abortions (22%); 6 induced abortions (7%); no congenital abnormalities; and 7 reports of neonatal problems, including respiratory issues, ABO blood group mismatch, hospitalization, and opioid withdrawal.83

Use of tumor necrosis factor (TNF) inhibitors in pregnancy has the most data25 and is considered a reasonable treatment option. Historically, there was concern about the risk for VACTERL syndrome (vertebral defects, anal atresia, cardiac defects, tracheoesophageal fistula, renal anomalies, limb abnormalities) with exposure to a TNF inhibitor,25,84-86 but further reports have alleviated these concerns. Active transplacental transport occurs for adalimumab, infliximab, and golimumab,87 but given structural differences, transport of certolizumab and etanercept is substantially less.88,89 In the CRIB study of placental transfer of certolizumab from mother to infant (N=14), pharmacokinetic data demonstrated no quantifiable certolizumab levels in 13 infants and minimal levels in 1 infant at birth.88 There are fewer data available on the use of other biologics in pregnancy, but for those in which active placental transport is relevant, similar concerns (ie, immunosuppression) might arise (eTable 3).

Concern over biologics largely involves risk for newborn immunosuppression. A case report detailed a Crohn disease patient treated with infliximab who gave birth to an infant who died of disseminated bacille Calmette-Guérin infection at 4.5 months after receiving the vaccine at 3 months.90 This case underscores the importance of delaying live vaccination in infants born to mothers who were treated with a biologic during pregnancy. Authors have provided various data on how long to avoid vaccination; some state as long as 1 year.91

In pregnant females with inflammatory bowel disease treated with a biologic, no correlation was observed among maternal, placental, and infant serum biologic levels and neonatal infection. However, an association between preterm birth and the level of the biologic in maternal and placental (but not infant) serum and preterm birth was observed.92



In another report from the same registry, combination therapy with a TNF inhibitor and another immunomodulator led to an increased risk for infection in infants at 12 months of age, compared to infants exposed to monotherapy89 or exposed to neither agent.93 A strategy to circumvent this potential problem is to avoid treatment with actively transported molecules in the third trimester.

Conclusion

Limited data exist to guide providers who are treating pregnant women with psoriasis. Our understanding of treatment of psoriasis in pregnancy is limited as a consequence of regulations surrounding clinical trials and inadequate detection of pregnancies in registries. Further efforts are necessary to better understand the relationship between psoriasis and pregnancy and how to manage pregnant women with psoriasis.

References
  1. Bae YS, Van Voorhees AS, Hsu S, et al. Review of treatment options for psoriasis in pregnant or lactating women: from the medical board of the National Psoriasis Foundation. J Am Acad Dermatol. 2012;67:459-477.
  2. Menter A, Strober BE, Kaplan DH, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with biologics. J Am Acad Dermatol. 2019;80:1029-1072.
  3. Flood KS, Porter ML, Kimball AB. Use of biologics in pregnancy: limitations stemming from clinical trials and registry experience. J Eur Acad Dermatol Venereol. 2019;33:E276-E277.
  4. Horn EJ, Chambers CD, Menter A, et al. Pregnancy outcomes in psoriasis: why do we know so little? J Am Acad Dermatol. 2009;61:E5-E8.
  5. Raychaudhuri SP, Navare T, Gross J, et al. Clinical course of psoriasis during pregnancy. Int J Dermatol. 2003;42:518-520.
  6. Boyd AS, Morris LF, Phillips CM, et al. Psoriasis and pregnancy: hormone and immune system interaction. Int J Dermatol. 1996;35:169-172.
  7. Murase JE, Chan KK, Garite TJ, et al. Hormonal effect on psoriasis in pregnancy and post partum. Arch Dermatol. 2005;14:601-606.
  8. Götestam Skorpen C, Hoeltzenbein M, Tincani A, et al. The EULAR points to consider for use of antirheumatic drugs before pregnancy, and during pregnancy and lactation. Ann Rheum Dis. 2016;75:795-810.
  9. Nguyen GC, Seow CH, Maxwell C, et al. The Toronto consensus statements for the management of inflammatory bowel disease in pregnancy. Gastroenterology. 2016;150:734-757.
  10. Wise J. Rheumatic diseases should be actively treated in pregnancy, new guidelines say. BMJ. 2016;532:i312.
  11. Puig L, Carrascosa JM, Carretero G, et al. Spanish evidence-based guidelines on the treatment of psoriasis with biologic agents, 2013. part 1: on efficacy and choice of treatment. Actas Dermosifiliogr. 2013;104:694-709.
  12. Girolomoni G, Altomore G, Ayala F, et al. Differential management of mild-to-severe psoriasis with biologic drugs: an Italian Delphi consensus expert panel. J Dermatolog Treat. 2015;26:128-133.
  13. Yeung J, Gooderham MJ, Grewal P, et al. Management of plaque psoriasis with biologic therapies in women of child-bearing potential consensus paper. J Cutan Med Surg. 2020;24:3S-14S.
  14. Louthrenoo W, Kasitanon N, Kathamort W, et al. 2016 updated Thai Rheumatism Association Recommendations for the use of biologic and targeted synthetic disease-modifying anti-rheumatic drugs in patients with rheumatoid arthritis. Int J Rheum Dis. 2017;20:1166-1184.
  15. Flint J, Panchal S, Hurrell A, et al. BSR and BHPR guideline on prescribing drugs in pregnancy and breastfeeding—part I: standard and biologic disease modifying anti-rheumatic drugs and corticosteroids. Rheumatology (Oxford). 2016;55:1693-1697.
  16. van der Woude CJ, Ardizzone S, Bengtson MB, et al. The second European evidenced-based consensus on reproduction and pregnancy in inflammatory bowel disease. J Crohns Colitis. 2015;9:107-124.
  17. Orlando A, Armuzz A, Papi C, et al. The Italian Society of Gastroenterology (SIGE) and the Italian Group for the study of Inflammatory Bowel Disease (IG-IBD) Clinical Practice Guidelines: the use of tumor necrosis factor-alpha antagonist therapy in inflammatory bowel disease. Dig Liver Dis. 2011;43:1-20.
  18. Puchner A, Grochenig HP, Sautner J, et al. Immunosuppressives and biologics during pregnancy and lactation. Wien Klin Wochenschr. 2019;131:29-44.
  19. ACOG Committee opinion no. 776: immune modulating therapies in pregnancy and lactation. Obstet Gynecol. 2019;133:E287-E297.
  20. Lamb CA, Kennedy NA, Raine T, et al. British Society of Gastroenterology consensus guidelines on the management of inflammatory bowel disease in adults. Gut. 2019;68:S1-S106.
  21. Goëb V, Ardizzone M, Arnard L, et al. Recommendations for using TNF-α antagonists and French Clinical Practice Guidelines endorsed by the French National Authority for Health. Joint Bone Spine. 2013;80:574-581.
  22. Rademaker M, Agnew K, Andrews M, et al. Psoriasis in those planning a family, pregnant or breast-feeding. the Australasian Psoriasis Collaboration. Australas J Dermatol. 2018;59:86-100.
  23. Mahadevan U, Robinson C, Bernasko N, et al. Inflammatory bowel disease in pregnancy clinical care pathway: a report from the American Gastroenterological Association IBD Parenthood Project Working Group. Gastroenterology. 2019;156:1508-1524.
  24. Mahadevan U, Cucchiara S, Hyam JS, et al. The London position statement of the World Congress of Gastroenterology on biological therapy for IBD with the European Crohn’s and Colitis Organisation: pregnancy and pediatrics. Am J Gastroenterol. 2011;106:214-223.
  25. Porter ML, Lockwood SJ, Kimball AB. Update on biologic safety for patients with psoriasis during pregnancy. Int J Womens Dermatol. 2017;3:21-25.
  26. Bandoli G, Johnson DL, Jones KL, et al. Potentially modifiable risk factors for adverse pregnancy outcomes in women with psoriasis. Br J Dermatol. 2010;163:334-339.
  27. Lima XT, Janakiraman V, Hughes MD, et al. The impact of psoriasis on pregnancy outcomes. J Invest Dermatol. 2012;132:85-91.
  28. Bröms G, Haerskjold A, Granath F, et al. Effect of maternal psoriasis on pregnancy and birth outcomes: a population-based cohort study from Denmark and Sweden. Acta Derm Venereol. 2018;98:728-734.
  29. Bobotsis R, Gulliver WP, Monaghan K, et al. Psoriasis and adverse pregnancy outcomes: a systematic review of observational studies. Br J Dermatol. 2016;175:464-472.
  30. Blattner CM, Danesh M, Safaee M, et al. Understanding the new FDA pregnancy and lactation labeling rules. Int J Womens Dermatol. 2016;2:5-7.
  31. Pernia S, DeMaagd G. The new pregnancy and lactation labeling rule. P T. 2016;4:713-715.
  32. Chi C-C, Wang S-H, Wojnarowska F, et al. Safety of topical corticosteroids in pregnancy. Cochrane Database Syst Rev. 2015:CD007346.
  33. Chi CC, Wang SH, Kirtschig G. Safety of topical corticosteroids in pregnancy. JAMA Dermatol. 2016;152:934-935.
  34. Dovonex (calcipotriene) Cream, 0.005% [package insert]. Dublin, Ireland: Leo Laboratories, Ltd; March 2015.
  35. Franssen ME, van der Wilt GJ, de Jong PC, et al. A retrospective study of the teratogenicity of dermatological coal tar products. Acta Derm Venereol. 1999;79:390-391.
  36. Garbis H, Elefant E, Bertolotti E, et al. Pregnancy outcome after periconceptional and first-trimester exposure to methoxsalen photochemotherapy. Arch Dermatol. 1995;131:492-493.
  37. Horizon Pharma USA. RAYOS (prednisone). https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/202020s000lbl.pdf https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/202020s000lbl.pdf.
  38. Park-Wyllie L, Mazzotta P, Pastuszak A, et al. Birth defects after maternal exposure to corticosteroids: prospective cohort study and meta-analysis of epidemiological studies. Teratology. 2000;62:385-392.
  39. Crowther CA, Doyle LW, Haslam RR, et al. Outcomes at 2 years of age after repeat doses of antenatal corticosteroids. N Engl J Med. 2007;357:1179-1189.
  40. Palmsten K, Rolland M, Herbert MF, et al. Patterns of prednisone use during pregnancy in women with rheumatoid arthritis: daily and cumulative dose. Pharmacoepidemiol Drug Saf. 2018;27:430-438.
  41. Groth K, Brännström M, Mölne J, et al. Cyclosporine A exposure during pregnancy in mice: effects on reproductive performance in mothers and offspring. Hum Reprod. 2010;25:697-704.
  42. Bar Oz B, Hackman R, Einarson T, et al. Pregnancy outcome after cyclosporine therapy during pregnancy: a meta-analysis. Transplantation. 2001;71:1051-1055.
  43. Paziana K, Del Monaco M, Cardonick E, et al. Ciclosporin use during pregnancy. Drug Saf. 2013;36:279-294.
  44. Lamarque V, Leleu MF, Monka C, et al. Analysis of 629 pregnancy outcomes in transplant recipients treated with Sandimmun. Transplant Proc. 1997;29:2480.
  45. Otezla (apremilast) tablets, for oral use [package insert]. Summit, NJ: Celgene Corporation; June 2017. http://www.accessdata.fda.gov/drugsatfda_docs/label/2017/205437s006lbl.pdf. Accessed July 8, 2020.
  46. Kurizky PS, de Castro Ferreira C, Nogueira LSC, et al. Treatment of psoriasis and psoriatic arthritis during pregnancy and breastfeeding. An Bras Dermatol. 2015;90:367-375.
  47. Murase JE, Heller MM, Butler DC. Safety of dermatologic medications in pregnancy and lactation. J Am Acad Dermatol. 2014;70:401.e1-401.e4.
  48. El-Saie LT, Rabie AR, Kamel MI, et al. Effect of narrowband ultraviolet B phototherapy on serum folic acid levels in patients with psoriasis. Lasers Med Sci. 2011;26:481-485.
  49. Murase JE, Koo JY, Berger TG. Narrowband ultraviolet B phototherapy influences serum folate levels in patients with vitiligo. J Am Acad Dermatol. 2010;62:710-711.
  50. Soriatane (acitretin) capsules [package insert]. Morrisville, NC: Stiefel Laboratories, Inc; April 2011. http://www.accessdata.fda.gov/drugsatfda_docs/label/2011/019821s018mg.pdf. Accessed July 8, 2020.
  51. Kaushik SB, Lebwohl MG. Psoriasis: which therapy for which patient: focus on special populations and chronic infections. J Am Acad Dermatol. 2019;80:43-53.
  52. Help us better understand the effects of Otezla in pregnancy. MotherToBaby website. https://mothertobaby.org/ongoing-study/otezla/. Accessed July 8, 2020.
  53. Bangsgaard N, Rørbye C, Skov L. Treating psoriasis during pregnancy: safety and efficacy of treatments. Am J Clin Dermatol. 2015;16:389-398.
  54. Tyler K. Dermatologic therapy in pregnancy. Clin Obstet Gynecol. 2015;58:112-118.
  55. Luu M, Benzenine E, Doret M, et al. Continuous anti–TNF-α use throughout pregnancy: possible complications for the mother but not for the fetus. a retrospective cohort on the French National Health Insurance Database (EVASION). Am J Gastroenterol. 2018;113:1669-1677.
  56. Bröms G, Granath F, Ekbom A, et al. Low risk of birth defects for infants whose mothers are treated with anti-tumor necrosis factor agents during pregnancy. Clin Gastroenterol Hepatol. 2016;14:234-241.
  57. Mirdamadi K, Salinas T, Vali R, et al. Meta-analysis of pregnancy outcomes after exposure to TNF-α inhibitors during pregnancy for the treatment of arthritic diseases. J Popul Ther Clin Pharmacol. 2018;25:E53-E56.
  58. Shihab Z, Yeomans ND, De Cruz P. Anti-tumour necrosis factor α therapies and inflammatory bowel disease pregnancy outcomes: a meta-analysis. J Crohns Colitis. 2016;10:979-988.
  59. Bröms G, Kieler H, Ekbom A, et al. Anti-TNF treatment during pregnancy and birth outcomes: a population-based study from Denmark, Finland, and Sweden. Pharmacoepidemiol Drug Saf. 2020;29:316-327.
  60. Diav-Citrin O, Otcheretianski-Volodarsky A, Shechtman S, et al. Pregnancy outcome following gestational exposure to TNF-alpha-inhibitors: a prospective, comparative, observational study. Reprod Toxicol. 2014;43:78-84.
  61. FDA determinations for pregnant and nursing women. National Psoriasis Foundation website. http://www.psoriasis.org/pregnancy/fda-determinations. Accessed July 8, 2020.
  62. Østensen M. Safety issues of biologics in pregnant patients with rheumatic diseases. Ann N Y Acad Sci. 2014;1317:32-38.
  63. Chambers CD, Johnson DL, Luo Y, et al. Pregnancy outcome in women treated with adalimumab for the treatment of rheumatoid arthritis: the OTIS Autoimmune Diseases in Pregnancy Project. Arthritis Rheum. 2012;64:2466.
  64. Clowse ME, Wolf DC, Forger F, et al. Pregnancy outcomes after exposure to certolizumab pegol: updated results from a pharmacovigilance safety database. Arthritis Rheumatol. 2018;70:1399-1407.
  65. Carman WJ, Accortt NA, Anthony MS, et al. Pregnancy and infant outcomes including major congenital malformations among women with chronic inflammatory arthritis or psoriasis, with and without etanercept use. Pharmacoepidemiol Drug Saf. 2017;26:1109-1118.
  66. Janssen. SIMPONI (golilumab). https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/125289s0064lbl.pdf.
  67. Yurkon K, Guo CY, Harrison D, et al. Pregnancy outcomes in women with dermatologic conditions exposed to infliximab. J Am Acad Dermatol. 2014;70:AB179.
  68. Watson N, Wu K, Farr P, et al. Ustekinumab exposure during conception and pregnancy in patients with chronic plaque psoriasis: a case series of 10 pregnancies. Br J Dermatol. 2019;180:195-196.
  69. Naureckas S, Slater J, Gearhart N, et al. Pregnancy outcomes in women with psoriasis and psoriatic arthritis exposed to ustekinumab. J Am Acad Dermatol. 2016;74:AB264.
  70. Haycraft K, DiRuggiero D, Rozzo SJ, et al. Outcomes of pregnancies from tildrakizumab phases I to III clinical development program. J Clin Aesthet Dermatol. 2019;12:S27-S28.
  71. Tremfya (guselkumab) injection, for subcutaneous use [package insert]. Horsham, PA: Janssen Biotech, Inc; July 2017. http://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761061s000lbl.pdf. Accessed Juy 8, 2020.
  72. Skyrizi (risankizumab-rzaa) injection, for subcutaneous use [package insert]. Northi Chicago, IL; April 2019. http://www.accessdata.fda.gov/drugsatfda_docs/label/2019/761105s000lbl.pdf. Accessed July 8, 2020.
  73. Siliq (brodalumab) injection, for subcutaneous use [package insert]. Bridgewater, NJ: Valeant Pharmaceuticals North America LLC; February 2017. http://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761032lbl.pdf. Accessed July 8, 2020.
  74. Feldman S, Pangallo B, Xu W, et al. Ixekizumab and pregnancy outcome. J Am Acad Dermatol. 2017;76:AB419.
  75. Clarke DO, Hilbish KG, Waters DG, et al. Assessment of ixekizumab, an interleukin-17A monoclonal antibody, for potential effects on reproduction and development, including immune system function, in cynomolgus monkeys. Reprod Toxicol. 2015;58:160-173.
  76. Warren RB, Reich K, Langley RG, et al. Secukinumab in pregnancy: outcomes in psoriasis, psoriatic arthritis and ankylosing spondylitis from the global safety database. Br J Dermatol. 2018;179:1205-1207.
  77. Nardin C, Colas M, Curie V, et al. Pregnancy after tubal sterilization in a woman treated with biologics for severe psoriasis. Dermatol Ther (Heidelb). 2018;8:323-326.
  78. Xeljanz (tofacitinib) tablets for oral administration [package insert]. New York, NY: Pfizer; November 2012. http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/203214s000lbl.pdf. Accessed July 8, 2020.
  79. Pfizer. Xeljanz (tofacitinib). https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/203214s018lbl.pdf.
  80. Mahadevan U, Dubinsky M, Su C, et al. Outcomes of pregnancies with maternal/paternal exposure in the tofacitinib safety databases for ulcerative colitis. Inflamm Bowel Dis. 2018;24:2494-2500.
  81. Clowse ME, Feldman SR, Isaacs JD, et al. Pregnancy outcomes in the tofacitinib safety databases for rheumatoid arthritis and psoriasis. Drug Saf. 2016;39:755-762.
  82. Malek A, Sager R, Kuhn P, et al. Evolution of maternofetal transport of immunoglobulins during human pregnancy. Am J Reprod Immunol. 1996;36:248-255.
  83. Kimball AB, Crow JA, Ridley K, et al. Pregnancy outcomes in women with moderate to severe psoriasis: the PSOLAR experience. J Am Acad Dermatol. 2014;70(suppl 1):AB179.
  84. Carter JD, Valeriano J, Vasey FB. Tumor necrosis factor-alpha inhibition and VATER association: a causal relationship. J Rheumatol. 2006;33:1014-1017.
  85. Carter JD, Ladhani A, Ricca LR, et al. A safety assessment of tumor necrosis factor antagonists during pregnancy: a review of the Food and Drug Administration database. J Rheumatol. 2009;36:635-641.
  86. Koren G, Inoue M. Do tumor necrosis factor inhibitors cause malformations in humans? J Rheumatol. 2009;36:465-466.
  87. Johansen C, Jimenez-Solem E, Haerskjold A, et al. The use and safety of TNF inhibitors during pregnancy in women with psoriasis: a review. Int J Mol Sci. 2018;19:E1349.
  88. Mariette X, Forger F, Abraham B, et al. Lack of placental transfer of certolizumab pegol during pregnancy: results from CRIB, a prospective, postmarketing, pharmacokinetic study. Ann Rheum Dis. 2018;77:228-233.
  89. Mahadevan U, Wolf DC, Dubinsky M, et al. Placental transfer of anti-tumor necrosis factor agents in pregnant patients with inflammatory bowel disease. Clin Gastroenterol Hepatol. 2013;11:286-292.
  90. Cheent K, Nolan J, Sharig S, et al. Case report: fatal case of disseminated BCG infection in an infant born to a mother taking infliximab for Crohn’s disease. J Crohns Colitis. 2010;4:603-605.
  91. Julsgaard M, Christensen LA, Gibson PR, et al. Concentrations of adalimumab and infliximab in mothers and newborns, and effects on infection. Gastroenterology. 2016;151:110-119.
  92. Mahadevan U, Martin C, Kane SV, et al. Do infant serum levels of biologic agents at birth correlate with risk of adverse outcomes? results from the PIANO registry. Gastroenterology. 2016;150:S91-S92.
  93. Mahadevan U, Martin CF, Sandler RS, et al. PIANO: a 1000 patient prospective registry of pregnancy outcomes in women with IBD exposed to immunomodulators and biologic therapy [AGA abstract 865]. Gastroenterology. 2012;142:S-149.
References
  1. Bae YS, Van Voorhees AS, Hsu S, et al. Review of treatment options for psoriasis in pregnant or lactating women: from the medical board of the National Psoriasis Foundation. J Am Acad Dermatol. 2012;67:459-477.
  2. Menter A, Strober BE, Kaplan DH, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with biologics. J Am Acad Dermatol. 2019;80:1029-1072.
  3. Flood KS, Porter ML, Kimball AB. Use of biologics in pregnancy: limitations stemming from clinical trials and registry experience. J Eur Acad Dermatol Venereol. 2019;33:E276-E277.
  4. Horn EJ, Chambers CD, Menter A, et al. Pregnancy outcomes in psoriasis: why do we know so little? J Am Acad Dermatol. 2009;61:E5-E8.
  5. Raychaudhuri SP, Navare T, Gross J, et al. Clinical course of psoriasis during pregnancy. Int J Dermatol. 2003;42:518-520.
  6. Boyd AS, Morris LF, Phillips CM, et al. Psoriasis and pregnancy: hormone and immune system interaction. Int J Dermatol. 1996;35:169-172.
  7. Murase JE, Chan KK, Garite TJ, et al. Hormonal effect on psoriasis in pregnancy and post partum. Arch Dermatol. 2005;14:601-606.
  8. Götestam Skorpen C, Hoeltzenbein M, Tincani A, et al. The EULAR points to consider for use of antirheumatic drugs before pregnancy, and during pregnancy and lactation. Ann Rheum Dis. 2016;75:795-810.
  9. Nguyen GC, Seow CH, Maxwell C, et al. The Toronto consensus statements for the management of inflammatory bowel disease in pregnancy. Gastroenterology. 2016;150:734-757.
  10. Wise J. Rheumatic diseases should be actively treated in pregnancy, new guidelines say. BMJ. 2016;532:i312.
  11. Puig L, Carrascosa JM, Carretero G, et al. Spanish evidence-based guidelines on the treatment of psoriasis with biologic agents, 2013. part 1: on efficacy and choice of treatment. Actas Dermosifiliogr. 2013;104:694-709.
  12. Girolomoni G, Altomore G, Ayala F, et al. Differential management of mild-to-severe psoriasis with biologic drugs: an Italian Delphi consensus expert panel. J Dermatolog Treat. 2015;26:128-133.
  13. Yeung J, Gooderham MJ, Grewal P, et al. Management of plaque psoriasis with biologic therapies in women of child-bearing potential consensus paper. J Cutan Med Surg. 2020;24:3S-14S.
  14. Louthrenoo W, Kasitanon N, Kathamort W, et al. 2016 updated Thai Rheumatism Association Recommendations for the use of biologic and targeted synthetic disease-modifying anti-rheumatic drugs in patients with rheumatoid arthritis. Int J Rheum Dis. 2017;20:1166-1184.
  15. Flint J, Panchal S, Hurrell A, et al. BSR and BHPR guideline on prescribing drugs in pregnancy and breastfeeding—part I: standard and biologic disease modifying anti-rheumatic drugs and corticosteroids. Rheumatology (Oxford). 2016;55:1693-1697.
  16. van der Woude CJ, Ardizzone S, Bengtson MB, et al. The second European evidenced-based consensus on reproduction and pregnancy in inflammatory bowel disease. J Crohns Colitis. 2015;9:107-124.
  17. Orlando A, Armuzz A, Papi C, et al. The Italian Society of Gastroenterology (SIGE) and the Italian Group for the study of Inflammatory Bowel Disease (IG-IBD) Clinical Practice Guidelines: the use of tumor necrosis factor-alpha antagonist therapy in inflammatory bowel disease. Dig Liver Dis. 2011;43:1-20.
  18. Puchner A, Grochenig HP, Sautner J, et al. Immunosuppressives and biologics during pregnancy and lactation. Wien Klin Wochenschr. 2019;131:29-44.
  19. ACOG Committee opinion no. 776: immune modulating therapies in pregnancy and lactation. Obstet Gynecol. 2019;133:E287-E297.
  20. Lamb CA, Kennedy NA, Raine T, et al. British Society of Gastroenterology consensus guidelines on the management of inflammatory bowel disease in adults. Gut. 2019;68:S1-S106.
  21. Goëb V, Ardizzone M, Arnard L, et al. Recommendations for using TNF-α antagonists and French Clinical Practice Guidelines endorsed by the French National Authority for Health. Joint Bone Spine. 2013;80:574-581.
  22. Rademaker M, Agnew K, Andrews M, et al. Psoriasis in those planning a family, pregnant or breast-feeding. the Australasian Psoriasis Collaboration. Australas J Dermatol. 2018;59:86-100.
  23. Mahadevan U, Robinson C, Bernasko N, et al. Inflammatory bowel disease in pregnancy clinical care pathway: a report from the American Gastroenterological Association IBD Parenthood Project Working Group. Gastroenterology. 2019;156:1508-1524.
  24. Mahadevan U, Cucchiara S, Hyam JS, et al. The London position statement of the World Congress of Gastroenterology on biological therapy for IBD with the European Crohn’s and Colitis Organisation: pregnancy and pediatrics. Am J Gastroenterol. 2011;106:214-223.
  25. Porter ML, Lockwood SJ, Kimball AB. Update on biologic safety for patients with psoriasis during pregnancy. Int J Womens Dermatol. 2017;3:21-25.
  26. Bandoli G, Johnson DL, Jones KL, et al. Potentially modifiable risk factors for adverse pregnancy outcomes in women with psoriasis. Br J Dermatol. 2010;163:334-339.
  27. Lima XT, Janakiraman V, Hughes MD, et al. The impact of psoriasis on pregnancy outcomes. J Invest Dermatol. 2012;132:85-91.
  28. Bröms G, Haerskjold A, Granath F, et al. Effect of maternal psoriasis on pregnancy and birth outcomes: a population-based cohort study from Denmark and Sweden. Acta Derm Venereol. 2018;98:728-734.
  29. Bobotsis R, Gulliver WP, Monaghan K, et al. Psoriasis and adverse pregnancy outcomes: a systematic review of observational studies. Br J Dermatol. 2016;175:464-472.
  30. Blattner CM, Danesh M, Safaee M, et al. Understanding the new FDA pregnancy and lactation labeling rules. Int J Womens Dermatol. 2016;2:5-7.
  31. Pernia S, DeMaagd G. The new pregnancy and lactation labeling rule. P T. 2016;4:713-715.
  32. Chi C-C, Wang S-H, Wojnarowska F, et al. Safety of topical corticosteroids in pregnancy. Cochrane Database Syst Rev. 2015:CD007346.
  33. Chi CC, Wang SH, Kirtschig G. Safety of topical corticosteroids in pregnancy. JAMA Dermatol. 2016;152:934-935.
  34. Dovonex (calcipotriene) Cream, 0.005% [package insert]. Dublin, Ireland: Leo Laboratories, Ltd; March 2015.
  35. Franssen ME, van der Wilt GJ, de Jong PC, et al. A retrospective study of the teratogenicity of dermatological coal tar products. Acta Derm Venereol. 1999;79:390-391.
  36. Garbis H, Elefant E, Bertolotti E, et al. Pregnancy outcome after periconceptional and first-trimester exposure to methoxsalen photochemotherapy. Arch Dermatol. 1995;131:492-493.
  37. Horizon Pharma USA. RAYOS (prednisone). https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/202020s000lbl.pdf https://www.accessdata.fda.gov/drugsatfda_docs/label/2012/202020s000lbl.pdf.
  38. Park-Wyllie L, Mazzotta P, Pastuszak A, et al. Birth defects after maternal exposure to corticosteroids: prospective cohort study and meta-analysis of epidemiological studies. Teratology. 2000;62:385-392.
  39. Crowther CA, Doyle LW, Haslam RR, et al. Outcomes at 2 years of age after repeat doses of antenatal corticosteroids. N Engl J Med. 2007;357:1179-1189.
  40. Palmsten K, Rolland M, Herbert MF, et al. Patterns of prednisone use during pregnancy in women with rheumatoid arthritis: daily and cumulative dose. Pharmacoepidemiol Drug Saf. 2018;27:430-438.
  41. Groth K, Brännström M, Mölne J, et al. Cyclosporine A exposure during pregnancy in mice: effects on reproductive performance in mothers and offspring. Hum Reprod. 2010;25:697-704.
  42. Bar Oz B, Hackman R, Einarson T, et al. Pregnancy outcome after cyclosporine therapy during pregnancy: a meta-analysis. Transplantation. 2001;71:1051-1055.
  43. Paziana K, Del Monaco M, Cardonick E, et al. Ciclosporin use during pregnancy. Drug Saf. 2013;36:279-294.
  44. Lamarque V, Leleu MF, Monka C, et al. Analysis of 629 pregnancy outcomes in transplant recipients treated with Sandimmun. Transplant Proc. 1997;29:2480.
  45. Otezla (apremilast) tablets, for oral use [package insert]. Summit, NJ: Celgene Corporation; June 2017. http://www.accessdata.fda.gov/drugsatfda_docs/label/2017/205437s006lbl.pdf. Accessed July 8, 2020.
  46. Kurizky PS, de Castro Ferreira C, Nogueira LSC, et al. Treatment of psoriasis and psoriatic arthritis during pregnancy and breastfeeding. An Bras Dermatol. 2015;90:367-375.
  47. Murase JE, Heller MM, Butler DC. Safety of dermatologic medications in pregnancy and lactation. J Am Acad Dermatol. 2014;70:401.e1-401.e4.
  48. El-Saie LT, Rabie AR, Kamel MI, et al. Effect of narrowband ultraviolet B phototherapy on serum folic acid levels in patients with psoriasis. Lasers Med Sci. 2011;26:481-485.
  49. Murase JE, Koo JY, Berger TG. Narrowband ultraviolet B phototherapy influences serum folate levels in patients with vitiligo. J Am Acad Dermatol. 2010;62:710-711.
  50. Soriatane (acitretin) capsules [package insert]. Morrisville, NC: Stiefel Laboratories, Inc; April 2011. http://www.accessdata.fda.gov/drugsatfda_docs/label/2011/019821s018mg.pdf. Accessed July 8, 2020.
  51. Kaushik SB, Lebwohl MG. Psoriasis: which therapy for which patient: focus on special populations and chronic infections. J Am Acad Dermatol. 2019;80:43-53.
  52. Help us better understand the effects of Otezla in pregnancy. MotherToBaby website. https://mothertobaby.org/ongoing-study/otezla/. Accessed July 8, 2020.
  53. Bangsgaard N, Rørbye C, Skov L. Treating psoriasis during pregnancy: safety and efficacy of treatments. Am J Clin Dermatol. 2015;16:389-398.
  54. Tyler K. Dermatologic therapy in pregnancy. Clin Obstet Gynecol. 2015;58:112-118.
  55. Luu M, Benzenine E, Doret M, et al. Continuous anti–TNF-α use throughout pregnancy: possible complications for the mother but not for the fetus. a retrospective cohort on the French National Health Insurance Database (EVASION). Am J Gastroenterol. 2018;113:1669-1677.
  56. Bröms G, Granath F, Ekbom A, et al. Low risk of birth defects for infants whose mothers are treated with anti-tumor necrosis factor agents during pregnancy. Clin Gastroenterol Hepatol. 2016;14:234-241.
  57. Mirdamadi K, Salinas T, Vali R, et al. Meta-analysis of pregnancy outcomes after exposure to TNF-α inhibitors during pregnancy for the treatment of arthritic diseases. J Popul Ther Clin Pharmacol. 2018;25:E53-E56.
  58. Shihab Z, Yeomans ND, De Cruz P. Anti-tumour necrosis factor α therapies and inflammatory bowel disease pregnancy outcomes: a meta-analysis. J Crohns Colitis. 2016;10:979-988.
  59. Bröms G, Kieler H, Ekbom A, et al. Anti-TNF treatment during pregnancy and birth outcomes: a population-based study from Denmark, Finland, and Sweden. Pharmacoepidemiol Drug Saf. 2020;29:316-327.
  60. Diav-Citrin O, Otcheretianski-Volodarsky A, Shechtman S, et al. Pregnancy outcome following gestational exposure to TNF-alpha-inhibitors: a prospective, comparative, observational study. Reprod Toxicol. 2014;43:78-84.
  61. FDA determinations for pregnant and nursing women. National Psoriasis Foundation website. http://www.psoriasis.org/pregnancy/fda-determinations. Accessed July 8, 2020.
  62. Østensen M. Safety issues of biologics in pregnant patients with rheumatic diseases. Ann N Y Acad Sci. 2014;1317:32-38.
  63. Chambers CD, Johnson DL, Luo Y, et al. Pregnancy outcome in women treated with adalimumab for the treatment of rheumatoid arthritis: the OTIS Autoimmune Diseases in Pregnancy Project. Arthritis Rheum. 2012;64:2466.
  64. Clowse ME, Wolf DC, Forger F, et al. Pregnancy outcomes after exposure to certolizumab pegol: updated results from a pharmacovigilance safety database. Arthritis Rheumatol. 2018;70:1399-1407.
  65. Carman WJ, Accortt NA, Anthony MS, et al. Pregnancy and infant outcomes including major congenital malformations among women with chronic inflammatory arthritis or psoriasis, with and without etanercept use. Pharmacoepidemiol Drug Saf. 2017;26:1109-1118.
  66. Janssen. SIMPONI (golilumab). https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/125289s0064lbl.pdf.
  67. Yurkon K, Guo CY, Harrison D, et al. Pregnancy outcomes in women with dermatologic conditions exposed to infliximab. J Am Acad Dermatol. 2014;70:AB179.
  68. Watson N, Wu K, Farr P, et al. Ustekinumab exposure during conception and pregnancy in patients with chronic plaque psoriasis: a case series of 10 pregnancies. Br J Dermatol. 2019;180:195-196.
  69. Naureckas S, Slater J, Gearhart N, et al. Pregnancy outcomes in women with psoriasis and psoriatic arthritis exposed to ustekinumab. J Am Acad Dermatol. 2016;74:AB264.
  70. Haycraft K, DiRuggiero D, Rozzo SJ, et al. Outcomes of pregnancies from tildrakizumab phases I to III clinical development program. J Clin Aesthet Dermatol. 2019;12:S27-S28.
  71. Tremfya (guselkumab) injection, for subcutaneous use [package insert]. Horsham, PA: Janssen Biotech, Inc; July 2017. http://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761061s000lbl.pdf. Accessed Juy 8, 2020.
  72. Skyrizi (risankizumab-rzaa) injection, for subcutaneous use [package insert]. Northi Chicago, IL; April 2019. http://www.accessdata.fda.gov/drugsatfda_docs/label/2019/761105s000lbl.pdf. Accessed July 8, 2020.
  73. Siliq (brodalumab) injection, for subcutaneous use [package insert]. Bridgewater, NJ: Valeant Pharmaceuticals North America LLC; February 2017. http://www.accessdata.fda.gov/drugsatfda_docs/label/2017/761032lbl.pdf. Accessed July 8, 2020.
  74. Feldman S, Pangallo B, Xu W, et al. Ixekizumab and pregnancy outcome. J Am Acad Dermatol. 2017;76:AB419.
  75. Clarke DO, Hilbish KG, Waters DG, et al. Assessment of ixekizumab, an interleukin-17A monoclonal antibody, for potential effects on reproduction and development, including immune system function, in cynomolgus monkeys. Reprod Toxicol. 2015;58:160-173.
  76. Warren RB, Reich K, Langley RG, et al. Secukinumab in pregnancy: outcomes in psoriasis, psoriatic arthritis and ankylosing spondylitis from the global safety database. Br J Dermatol. 2018;179:1205-1207.
  77. Nardin C, Colas M, Curie V, et al. Pregnancy after tubal sterilization in a woman treated with biologics for severe psoriasis. Dermatol Ther (Heidelb). 2018;8:323-326.
  78. Xeljanz (tofacitinib) tablets for oral administration [package insert]. New York, NY: Pfizer; November 2012. http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/203214s000lbl.pdf. Accessed July 8, 2020.
  79. Pfizer. Xeljanz (tofacitinib). https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/203214s018lbl.pdf.
  80. Mahadevan U, Dubinsky M, Su C, et al. Outcomes of pregnancies with maternal/paternal exposure in the tofacitinib safety databases for ulcerative colitis. Inflamm Bowel Dis. 2018;24:2494-2500.
  81. Clowse ME, Feldman SR, Isaacs JD, et al. Pregnancy outcomes in the tofacitinib safety databases for rheumatoid arthritis and psoriasis. Drug Saf. 2016;39:755-762.
  82. Malek A, Sager R, Kuhn P, et al. Evolution of maternofetal transport of immunoglobulins during human pregnancy. Am J Reprod Immunol. 1996;36:248-255.
  83. Kimball AB, Crow JA, Ridley K, et al. Pregnancy outcomes in women with moderate to severe psoriasis: the PSOLAR experience. J Am Acad Dermatol. 2014;70(suppl 1):AB179.
  84. Carter JD, Valeriano J, Vasey FB. Tumor necrosis factor-alpha inhibition and VATER association: a causal relationship. J Rheumatol. 2006;33:1014-1017.
  85. Carter JD, Ladhani A, Ricca LR, et al. A safety assessment of tumor necrosis factor antagonists during pregnancy: a review of the Food and Drug Administration database. J Rheumatol. 2009;36:635-641.
  86. Koren G, Inoue M. Do tumor necrosis factor inhibitors cause malformations in humans? J Rheumatol. 2009;36:465-466.
  87. Johansen C, Jimenez-Solem E, Haerskjold A, et al. The use and safety of TNF inhibitors during pregnancy in women with psoriasis: a review. Int J Mol Sci. 2018;19:E1349.
  88. Mariette X, Forger F, Abraham B, et al. Lack of placental transfer of certolizumab pegol during pregnancy: results from CRIB, a prospective, postmarketing, pharmacokinetic study. Ann Rheum Dis. 2018;77:228-233.
  89. Mahadevan U, Wolf DC, Dubinsky M, et al. Placental transfer of anti-tumor necrosis factor agents in pregnant patients with inflammatory bowel disease. Clin Gastroenterol Hepatol. 2013;11:286-292.
  90. Cheent K, Nolan J, Sharig S, et al. Case report: fatal case of disseminated BCG infection in an infant born to a mother taking infliximab for Crohn’s disease. J Crohns Colitis. 2010;4:603-605.
  91. Julsgaard M, Christensen LA, Gibson PR, et al. Concentrations of adalimumab and infliximab in mothers and newborns, and effects on infection. Gastroenterology. 2016;151:110-119.
  92. Mahadevan U, Martin C, Kane SV, et al. Do infant serum levels of biologic agents at birth correlate with risk of adverse outcomes? results from the PIANO registry. Gastroenterology. 2016;150:S91-S92.
  93. Mahadevan U, Martin CF, Sandler RS, et al. PIANO: a 1000 patient prospective registry of pregnancy outcomes in women with IBD exposed to immunomodulators and biologic therapy [AGA abstract 865]. Gastroenterology. 2012;142:S-149.
Issue
Cutis - 106(2S)
Issue
Cutis - 106(2S)
Page Number
15-20, E1-E9
Page Number
15-20, E1-E9
Publications
Publications
Topics
Article Type
Sections
Citation Override
Cutis. 2020 August;106(2S):15-20, E1-E9
Inside the Article

Practice Points

  • Robust safety data often are lacking for the use of topical and systemic agents to treat psoriasis in pregnancy.
  • Professional society guidelines on the use of systemic agents in pregnancy vary among dermatology, gastroenterology, and rheumatology organizations.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Gating Strategy
No Gating
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Scalp Wound Closures in Mohs Micrographic Surgery: A Survey of Staples vs Sutures

Article Type
Changed
Sun, 08/16/2020 - 23:16

Limited data exist comparing staples and sutures for scalp closures during Mohs micrographic surgery (MMS). As a result, the closure method for these scalp wounds is based on surgeon preference without established consensus. The purpose of this study was to survey practicing Mohs surgeons on their scalp wound closure preferences as well as the clinical and economic variables that impact their decisions. Understanding practice habits can guide future trial design, with a goal of creating established criterion for MMS scalp wound closures.

Methods

An anonymous survey was distributed from April 2019 to June 2019 to fellowship-trained Mohs surgeons using an electronic mailing list from the American College of Mohs Surgery (ACMS). The 10-question survey was approved by the University of Kansas institutional review board and the executive committee of the ACMS. Surgeons were asked about their preferred method for scalp wound closure as well as clinical and economic variables that impacted those preferences. Respondents indicated their frequency of using deep sutures, epidermal sutures, and wound undermining on a sliding scale of 0% to 100%. Comparisons were made between practice habits, preferences, and surgeon demographics using t tests. Statistical significance was determined as P<.05.

Results

Sixty-eight ACMS fellowship-trained Mohs surgeons completed the survey. The average age of respondents was 45 years; 69.1% (n=47) of respondents were male, and 76.5% (n=52) practiced in a private setting (Table 1). Regardless of epidermal closure type, deep suture placement was used in an average (standard deviation [SD]) of 88.8% (19.5%) of cases overall, which did not statistically differ between years of Mohs experience or practice setting (Table 2). Wound undermining was performed in an average (SD) of 83.0% (24.3%) of cases overall and was more prevalent in private vs academic settings (87.6% [17.8%] vs 65.7% [35.0%]; P<.01). Epidermal sutures were used in an average (SD) of 27.1% (33.5%) of scalp wound cases overall. Surgeons with less experience (≤5 years) used them more frequently (average [SD], 42.7% [36.2%] of cases) than surgeons with more experience (≥16 years; average [SD], 18.8% [32.6%] of cases; P=.037). There was no significant difference between epidermal suture placement rates and practice setting (average [SD], 18.1% [28.1%] of cases for academic providers vs 30.0% [34.8%] of cases with private providers; P=.210).

Clinical and economic factors that were most important during wound closure were ranked (beginning with most important) as the following: risk of complications, cosmetic outcome, hair preservation, patient comfort during closure, healing time, and closure cost. In all demographic cases, risk of complications was ranked 1 or 2 (1=most important; 6=least important) overall; cost was the least important factor overall (Table 2).

Surgeons perceived staples to be superior for speed of closure and for closing wounds in high-tension areas, whereas sutures were perceived as superior when considering cost of closure and ease of removal (Table 3). Successful healing rate, healing time, hair preservation, overall cosmetic outcome, and lower risk of complications were viewed as equivalent when comparing staples and sutures.



In cases in which surgeons did not use staples for closure, the most important factors for opting to not use them were patient discomfort (52.9% [n=36]), cost (25.0% [n=17]), and worse overall cosmetic outcome (23.5% [n=16])(Table 4). The most frequent locations outside of scalp wounds that physicians considered the use of staples for closure were the back (19.1% [n=13]), thigh (10.3% [n=7]), and shoulder (8.8% [n=6]).

Comment

Epidermal closure with sutures was reportedly used in an average of only 27.1% of scalp wound cases, with clinical factors such as cosmetic outcome, risk of complications, and closure time seen as either equivalent or inferior to staples. Our data suggest that surgeon closure perceptions generally are in agreement with established head and neck literature within different medical specialties that favor staple closures, particularly in high-tension areas.1 Interestingly, the most common reasons given for not using staples included patient discomfort, cost, and worse cosmetic outcomes, which are unsubstantiated with head and neck comparative studies.2-4

Although cost was the least important variable for determining closure type in our surveyed cohort, it is likely that the overall cost of closure is frequently underestimated. A higher material cost is noted with staples; however, the largest determinant of overall cost remains the surgeon’s time, which is reduced by factors of 10 or more when closing with staples.2,3 This difference—coupled with the unchanged cosmetic outcome and complication rates—makes staples more advantageous for high-tension scalp wounds.4 Moreover, the stapling technique is more reproducible than suturing, which requires more surgical skill and experience.



Limitations of this study include a lack of directly comparable data for staple and suture scalp wound closures. In addition, the small cohort of respondents in this preliminary study can serve to guide future studies.

Conclusion

Scalp wounds during MMS were most frequently closed using staples vs sutures, with the perception that these methods are equivalent in complication risk, cosmetic outcome, and overall patient satisfaction. These results agree with comparative literature for head and neck surgery and assist with establishing an epidemiologic baseline for future studies comparing their use during MMS.

References
  1. Ritchie AJ, Rocke LG. Staples versus sutures in the closure of scalp wounds: a prospective, double-blind, randomized trial. Injury. 1989;20:217-218.
  2. Batra J, Bekal RK, Byadgi S, et al. Comparison of skin staples and standard sutures for closing incisions after head and neck cancer surgery: a double-blind, randomized and prospective study. J Maxillofac Oral Surg. 2016;15:243-250.
  3. Kanegaye JT, Vance CW, Chan L, et al. Comparison of skin stapling devices and standard sutures for pediatric scalp lacerations: a randomized study of cost and time benefits. J Pediatr. 1997;130:808-813.
  4. Khan ANGA, Dayan PS, Miller S, et al. Cosmetic outcome of scalp wound closure with staples in the pediatric emergency department: a prospective, randomized trial. Pediatr Emerg Care. 2002;18:171-173.
Article PDF
Author and Disclosure Information

From the Division of Dermatology, University of Kansas Medical Center, Kansas City.

The authors report no conflict of interest.

Correspondence: Brett C. Neill, MD, Division of Dermatology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160 ([email protected]).

Issue
Cutis - 106(2)
Publications
Topics
Page Number
96-99
Sections
Author and Disclosure Information

From the Division of Dermatology, University of Kansas Medical Center, Kansas City.

The authors report no conflict of interest.

Correspondence: Brett C. Neill, MD, Division of Dermatology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160 ([email protected]).

Author and Disclosure Information

From the Division of Dermatology, University of Kansas Medical Center, Kansas City.

The authors report no conflict of interest.

Correspondence: Brett C. Neill, MD, Division of Dermatology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS 66160 ([email protected]).

Article PDF
Article PDF

Limited data exist comparing staples and sutures for scalp closures during Mohs micrographic surgery (MMS). As a result, the closure method for these scalp wounds is based on surgeon preference without established consensus. The purpose of this study was to survey practicing Mohs surgeons on their scalp wound closure preferences as well as the clinical and economic variables that impact their decisions. Understanding practice habits can guide future trial design, with a goal of creating established criterion for MMS scalp wound closures.

Methods

An anonymous survey was distributed from April 2019 to June 2019 to fellowship-trained Mohs surgeons using an electronic mailing list from the American College of Mohs Surgery (ACMS). The 10-question survey was approved by the University of Kansas institutional review board and the executive committee of the ACMS. Surgeons were asked about their preferred method for scalp wound closure as well as clinical and economic variables that impacted those preferences. Respondents indicated their frequency of using deep sutures, epidermal sutures, and wound undermining on a sliding scale of 0% to 100%. Comparisons were made between practice habits, preferences, and surgeon demographics using t tests. Statistical significance was determined as P<.05.

Results

Sixty-eight ACMS fellowship-trained Mohs surgeons completed the survey. The average age of respondents was 45 years; 69.1% (n=47) of respondents were male, and 76.5% (n=52) practiced in a private setting (Table 1). Regardless of epidermal closure type, deep suture placement was used in an average (standard deviation [SD]) of 88.8% (19.5%) of cases overall, which did not statistically differ between years of Mohs experience or practice setting (Table 2). Wound undermining was performed in an average (SD) of 83.0% (24.3%) of cases overall and was more prevalent in private vs academic settings (87.6% [17.8%] vs 65.7% [35.0%]; P<.01). Epidermal sutures were used in an average (SD) of 27.1% (33.5%) of scalp wound cases overall. Surgeons with less experience (≤5 years) used them more frequently (average [SD], 42.7% [36.2%] of cases) than surgeons with more experience (≥16 years; average [SD], 18.8% [32.6%] of cases; P=.037). There was no significant difference between epidermal suture placement rates and practice setting (average [SD], 18.1% [28.1%] of cases for academic providers vs 30.0% [34.8%] of cases with private providers; P=.210).

Clinical and economic factors that were most important during wound closure were ranked (beginning with most important) as the following: risk of complications, cosmetic outcome, hair preservation, patient comfort during closure, healing time, and closure cost. In all demographic cases, risk of complications was ranked 1 or 2 (1=most important; 6=least important) overall; cost was the least important factor overall (Table 2).

Surgeons perceived staples to be superior for speed of closure and for closing wounds in high-tension areas, whereas sutures were perceived as superior when considering cost of closure and ease of removal (Table 3). Successful healing rate, healing time, hair preservation, overall cosmetic outcome, and lower risk of complications were viewed as equivalent when comparing staples and sutures.



In cases in which surgeons did not use staples for closure, the most important factors for opting to not use them were patient discomfort (52.9% [n=36]), cost (25.0% [n=17]), and worse overall cosmetic outcome (23.5% [n=16])(Table 4). The most frequent locations outside of scalp wounds that physicians considered the use of staples for closure were the back (19.1% [n=13]), thigh (10.3% [n=7]), and shoulder (8.8% [n=6]).

Comment

Epidermal closure with sutures was reportedly used in an average of only 27.1% of scalp wound cases, with clinical factors such as cosmetic outcome, risk of complications, and closure time seen as either equivalent or inferior to staples. Our data suggest that surgeon closure perceptions generally are in agreement with established head and neck literature within different medical specialties that favor staple closures, particularly in high-tension areas.1 Interestingly, the most common reasons given for not using staples included patient discomfort, cost, and worse cosmetic outcomes, which are unsubstantiated with head and neck comparative studies.2-4

Although cost was the least important variable for determining closure type in our surveyed cohort, it is likely that the overall cost of closure is frequently underestimated. A higher material cost is noted with staples; however, the largest determinant of overall cost remains the surgeon’s time, which is reduced by factors of 10 or more when closing with staples.2,3 This difference—coupled with the unchanged cosmetic outcome and complication rates—makes staples more advantageous for high-tension scalp wounds.4 Moreover, the stapling technique is more reproducible than suturing, which requires more surgical skill and experience.



Limitations of this study include a lack of directly comparable data for staple and suture scalp wound closures. In addition, the small cohort of respondents in this preliminary study can serve to guide future studies.

Conclusion

Scalp wounds during MMS were most frequently closed using staples vs sutures, with the perception that these methods are equivalent in complication risk, cosmetic outcome, and overall patient satisfaction. These results agree with comparative literature for head and neck surgery and assist with establishing an epidemiologic baseline for future studies comparing their use during MMS.

Limited data exist comparing staples and sutures for scalp closures during Mohs micrographic surgery (MMS). As a result, the closure method for these scalp wounds is based on surgeon preference without established consensus. The purpose of this study was to survey practicing Mohs surgeons on their scalp wound closure preferences as well as the clinical and economic variables that impact their decisions. Understanding practice habits can guide future trial design, with a goal of creating established criterion for MMS scalp wound closures.

Methods

An anonymous survey was distributed from April 2019 to June 2019 to fellowship-trained Mohs surgeons using an electronic mailing list from the American College of Mohs Surgery (ACMS). The 10-question survey was approved by the University of Kansas institutional review board and the executive committee of the ACMS. Surgeons were asked about their preferred method for scalp wound closure as well as clinical and economic variables that impacted those preferences. Respondents indicated their frequency of using deep sutures, epidermal sutures, and wound undermining on a sliding scale of 0% to 100%. Comparisons were made between practice habits, preferences, and surgeon demographics using t tests. Statistical significance was determined as P<.05.

Results

Sixty-eight ACMS fellowship-trained Mohs surgeons completed the survey. The average age of respondents was 45 years; 69.1% (n=47) of respondents were male, and 76.5% (n=52) practiced in a private setting (Table 1). Regardless of epidermal closure type, deep suture placement was used in an average (standard deviation [SD]) of 88.8% (19.5%) of cases overall, which did not statistically differ between years of Mohs experience or practice setting (Table 2). Wound undermining was performed in an average (SD) of 83.0% (24.3%) of cases overall and was more prevalent in private vs academic settings (87.6% [17.8%] vs 65.7% [35.0%]; P<.01). Epidermal sutures were used in an average (SD) of 27.1% (33.5%) of scalp wound cases overall. Surgeons with less experience (≤5 years) used them more frequently (average [SD], 42.7% [36.2%] of cases) than surgeons with more experience (≥16 years; average [SD], 18.8% [32.6%] of cases; P=.037). There was no significant difference between epidermal suture placement rates and practice setting (average [SD], 18.1% [28.1%] of cases for academic providers vs 30.0% [34.8%] of cases with private providers; P=.210).

Clinical and economic factors that were most important during wound closure were ranked (beginning with most important) as the following: risk of complications, cosmetic outcome, hair preservation, patient comfort during closure, healing time, and closure cost. In all demographic cases, risk of complications was ranked 1 or 2 (1=most important; 6=least important) overall; cost was the least important factor overall (Table 2).

Surgeons perceived staples to be superior for speed of closure and for closing wounds in high-tension areas, whereas sutures were perceived as superior when considering cost of closure and ease of removal (Table 3). Successful healing rate, healing time, hair preservation, overall cosmetic outcome, and lower risk of complications were viewed as equivalent when comparing staples and sutures.



In cases in which surgeons did not use staples for closure, the most important factors for opting to not use them were patient discomfort (52.9% [n=36]), cost (25.0% [n=17]), and worse overall cosmetic outcome (23.5% [n=16])(Table 4). The most frequent locations outside of scalp wounds that physicians considered the use of staples for closure were the back (19.1% [n=13]), thigh (10.3% [n=7]), and shoulder (8.8% [n=6]).

Comment

Epidermal closure with sutures was reportedly used in an average of only 27.1% of scalp wound cases, with clinical factors such as cosmetic outcome, risk of complications, and closure time seen as either equivalent or inferior to staples. Our data suggest that surgeon closure perceptions generally are in agreement with established head and neck literature within different medical specialties that favor staple closures, particularly in high-tension areas.1 Interestingly, the most common reasons given for not using staples included patient discomfort, cost, and worse cosmetic outcomes, which are unsubstantiated with head and neck comparative studies.2-4

Although cost was the least important variable for determining closure type in our surveyed cohort, it is likely that the overall cost of closure is frequently underestimated. A higher material cost is noted with staples; however, the largest determinant of overall cost remains the surgeon’s time, which is reduced by factors of 10 or more when closing with staples.2,3 This difference—coupled with the unchanged cosmetic outcome and complication rates—makes staples more advantageous for high-tension scalp wounds.4 Moreover, the stapling technique is more reproducible than suturing, which requires more surgical skill and experience.



Limitations of this study include a lack of directly comparable data for staple and suture scalp wound closures. In addition, the small cohort of respondents in this preliminary study can serve to guide future studies.

Conclusion

Scalp wounds during MMS were most frequently closed using staples vs sutures, with the perception that these methods are equivalent in complication risk, cosmetic outcome, and overall patient satisfaction. These results agree with comparative literature for head and neck surgery and assist with establishing an epidemiologic baseline for future studies comparing their use during MMS.

References
  1. Ritchie AJ, Rocke LG. Staples versus sutures in the closure of scalp wounds: a prospective, double-blind, randomized trial. Injury. 1989;20:217-218.
  2. Batra J, Bekal RK, Byadgi S, et al. Comparison of skin staples and standard sutures for closing incisions after head and neck cancer surgery: a double-blind, randomized and prospective study. J Maxillofac Oral Surg. 2016;15:243-250.
  3. Kanegaye JT, Vance CW, Chan L, et al. Comparison of skin stapling devices and standard sutures for pediatric scalp lacerations: a randomized study of cost and time benefits. J Pediatr. 1997;130:808-813.
  4. Khan ANGA, Dayan PS, Miller S, et al. Cosmetic outcome of scalp wound closure with staples in the pediatric emergency department: a prospective, randomized trial. Pediatr Emerg Care. 2002;18:171-173.
References
  1. Ritchie AJ, Rocke LG. Staples versus sutures in the closure of scalp wounds: a prospective, double-blind, randomized trial. Injury. 1989;20:217-218.
  2. Batra J, Bekal RK, Byadgi S, et al. Comparison of skin staples and standard sutures for closing incisions after head and neck cancer surgery: a double-blind, randomized and prospective study. J Maxillofac Oral Surg. 2016;15:243-250.
  3. Kanegaye JT, Vance CW, Chan L, et al. Comparison of skin stapling devices and standard sutures for pediatric scalp lacerations: a randomized study of cost and time benefits. J Pediatr. 1997;130:808-813.
  4. Khan ANGA, Dayan PS, Miller S, et al. Cosmetic outcome of scalp wound closure with staples in the pediatric emergency department: a prospective, randomized trial. Pediatr Emerg Care. 2002;18:171-173.
Issue
Cutis - 106(2)
Issue
Cutis - 106(2)
Page Number
96-99
Page Number
96-99
Publications
Publications
Topics
Article Type
Sections
Inside the Article

Practice Points

  • Scalp wounds present a unique challenge for closure during Mohs micrographic surgery due to the scalp's tendency to bleed, limited elasticity, and hair-bearing nature.
  • Among fellowship-trained Mohs surgeons, scalp wounds were closed with staples more often than with epidermal sutures.
  • Staples and sutures for scalp wounds were perceived to be equivalent in risk of complications, cosmetic outcome, and overall patient satisfaction.
  • Compared to epidermal sutures, staples were perceived as advantageous in high-tension areas and for speed of closure.
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
Article PDF Media

Product News August 2020

Article Type
Changed
Sun, 08/16/2020 - 23:19

FDA Approves Wynzora Cream for Plaque Psoriasis

MC2 Therapeutics announces US Food and Drug Administration (FDA) approval of Wynzora Cream (calcipotriene 0.005% and betamethasone dipropionate 0.064%) for once-daily treatment of plaque psoriasis in adults.

Wynzora Cream is based on PAD Technology, which enables stability of calcipotriene and betamethasone dipropionate in an aqueous formulation. Key features of PAD Technology formulations are high penetration of active ingredients to the target tissue, improved solubility and stability of active ingredients, high tolerability, and excellent treatment convenience. In the phase 3 trials conducted at multiple sites in the United States and the European Union, Wynzora Cream has demonstrated a combination of clinical efficacy, a favorable safety profile, and high convenience, offering overall better patient satisfaction in the topical treatment of plaque psoriasis in the real-world setting.

WynzoraCream is applied to affected areas once daily for up to 8 weeks and not more than 100 g per week. Patients should stop treatment when the plaque psoriasis is under control, unless a health care provider gives other instructions.

MC2 Therapeutics also has submitted a Marketing Authorization Application in the European Union for Wynzora Cream (50 µg/g calcipotriol and 0.5 mg/g betamethasone [as dipropionate]) for the treatment of plaque psoriasis. For more information, visit www.mc2therapeutics.com.

 

 

If you would like your product included in Product News, please email a press release to the Editorial Office at [email protected].

Article PDF
Issue
Cutis - 106(2)
Publications
Topics
Page Number
103
Sections
Article PDF
Article PDF

FDA Approves Wynzora Cream for Plaque Psoriasis

MC2 Therapeutics announces US Food and Drug Administration (FDA) approval of Wynzora Cream (calcipotriene 0.005% and betamethasone dipropionate 0.064%) for once-daily treatment of plaque psoriasis in adults.

Wynzora Cream is based on PAD Technology, which enables stability of calcipotriene and betamethasone dipropionate in an aqueous formulation. Key features of PAD Technology formulations are high penetration of active ingredients to the target tissue, improved solubility and stability of active ingredients, high tolerability, and excellent treatment convenience. In the phase 3 trials conducted at multiple sites in the United States and the European Union, Wynzora Cream has demonstrated a combination of clinical efficacy, a favorable safety profile, and high convenience, offering overall better patient satisfaction in the topical treatment of plaque psoriasis in the real-world setting.

WynzoraCream is applied to affected areas once daily for up to 8 weeks and not more than 100 g per week. Patients should stop treatment when the plaque psoriasis is under control, unless a health care provider gives other instructions.

MC2 Therapeutics also has submitted a Marketing Authorization Application in the European Union for Wynzora Cream (50 µg/g calcipotriol and 0.5 mg/g betamethasone [as dipropionate]) for the treatment of plaque psoriasis. For more information, visit www.mc2therapeutics.com.

 

 

If you would like your product included in Product News, please email a press release to the Editorial Office at [email protected].

FDA Approves Wynzora Cream for Plaque Psoriasis

MC2 Therapeutics announces US Food and Drug Administration (FDA) approval of Wynzora Cream (calcipotriene 0.005% and betamethasone dipropionate 0.064%) for once-daily treatment of plaque psoriasis in adults.

Wynzora Cream is based on PAD Technology, which enables stability of calcipotriene and betamethasone dipropionate in an aqueous formulation. Key features of PAD Technology formulations are high penetration of active ingredients to the target tissue, improved solubility and stability of active ingredients, high tolerability, and excellent treatment convenience. In the phase 3 trials conducted at multiple sites in the United States and the European Union, Wynzora Cream has demonstrated a combination of clinical efficacy, a favorable safety profile, and high convenience, offering overall better patient satisfaction in the topical treatment of plaque psoriasis in the real-world setting.

WynzoraCream is applied to affected areas once daily for up to 8 weeks and not more than 100 g per week. Patients should stop treatment when the plaque psoriasis is under control, unless a health care provider gives other instructions.

MC2 Therapeutics also has submitted a Marketing Authorization Application in the European Union for Wynzora Cream (50 µg/g calcipotriol and 0.5 mg/g betamethasone [as dipropionate]) for the treatment of plaque psoriasis. For more information, visit www.mc2therapeutics.com.

 

 

If you would like your product included in Product News, please email a press release to the Editorial Office at [email protected].

Issue
Cutis - 106(2)
Issue
Cutis - 106(2)
Page Number
103
Page Number
103
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Article PDF Media

Assessment of Nail Content in the American Academy of Dermatology Patient Education Website

Article Type
Changed
Sun, 08/16/2020 - 23:14

To the Editor:

Patients with skin, hair, or nail concerns often utilize online resources to self-diagnose or learn more about physician-diagnosed conditions. The American Academy of Dermatology (AAD) website offers the public access to informational pages categorized by disease or treatment (https://www.aad.org/public). We sought to evaluate the nail content by searching the Patients and Public section of the AAD website to qualitatively and quantitatively describe mentions of nail conditions. Psoriasis, psoriatic arthritis, atopic dermatitis, and ringworm content also were analyzed and compared to nail content. The analysis was performed on September 7, 2019.

Of the 73 topics listed in the Diseases and Treatments section of the site, 17 (23%) specifically mentioned nail symptoms or pathology (Table). Three additional topics—atopic dermatitis, cellulitis, and neurodermatitis—recommended keeping nails short to prevent injury from scratching. There was 1 mention of obtaining fungal cultures, 2 of nail scraping microscopy, 2 of nail clippings, and 2 of nail-related cancers. There were no mentions of nail biopsies. The total number of unique clinical images across all sections was 300, with 12 of nails. The video library contained 84 videos, of which 6 focused on nail health.



Several sections were reviewed for overall content to understand the typical number of images and videos included for other common conditions. The general psoriasis section included 6 images and 3 videos detailing disease etiology, symptoms, and treatments, without depictions of nails. The psoriatic arthritis section included 8 images and 1 video as well as 1 image of nail pitting. The atopic dermatitis section included 6 images and 6 videos on disease etiology, symptoms, and treatment. The ringworm section, which collectively included all tinea infections, contained 11 images and 1 video on symptoms. In contrast, the only nail-specific section—nail fungus—contained only 2 images and 1 video.

Our study demonstrated that nail content is underrepresented in the public education section of the AAD website. If patients are unable to find nail disease material on the AAD website, they may seek alternative sources that are unreliable. Prior studies have shown that patient Internet resources for subungual melanoma and onychomycosis often are inadequate in quality and readability.1,2

Representative photographs and key information on common nail diseases could be added to improve patient education. The atopic dermatitis section should include text on related nail changes with accompanying images. We also recommend including paronychia information and images as either a separate topic or in the cellulitis section. The contact dermatitis section mentions nail cosmetics as causative factors, but an image of roller-coaster onycholysis may be more helpful.3 Although the alopecia areata section mentions nail changes, this information should be added to the general hair loss section of the site, as many patients may initially seek out the latter category. Herpes simplex may affect nails, and an image showing these changes would be instructive. In addition, pyogenic granulomas and paronychia occur with isotretinoin use.4



Many of the included images were not representative of common clinical findings. The nail lichen planus image showed pitting instead of more typical findings of nail plate atrophy and pterygium. The nail melanoma image showed thickened yellow toenails and the fifth toenail with a thin gray-brown band instead of an isolated wide black band. The nail fungus section included images of superficial onychomycosis and severe onychodystrophy instead of showing more common changes such as distal onycholysis with subungual hyperkeratosis, which is typical of the most common subtype, distal lateral subungual onychomycosis.5 Onychomycosis was referenced again in the ringworm section with 1 image repeated from the nail fungus section and another image that appeared to be a subungual hematoma.

The AAD website offers important patient education resources; however, nail content is underrepresented on this platform. Dermatologists are experts on nail disease, and increased efforts are needed to educate the public about frequently encountered nail signs and symptoms that could signify a serious underlying condition.

After our original search and analysis, new nail topics, images, and videos have been added; therefore, there has been a positive trend toward new nail content being added to site, which will greatly benefit patients.

References
  1. Kang R, Lipner S. Assessment of internet sources on subungual melanoma [published online August 30, 2018]. Melanoma Res. doi:10.1097/CMR.0000000000000508.
  2. Kang R, Lipner S. Evaluation of onychomycosis information on the internet. J Drugs Dermatol. 2019;18:484-487.
  3. Rieder EA, Tosti A. Cosmetically induced disorders of the nail with update on contemporary nail manicures. J Clin Aesthet Dermatol. 2016;9:39-44.
  4. Arias-Santiago S, Husein-ElAhmed H, Aneiros-Cachaza J, et al. Uncommon side effects of isotretinoin therapy: paronychia and pyogenic granuloma. J Am Acad Dermatol. 2011;64:AB37.
  5. Lipner SR, Scher RK. Onychomycosis: clinical overview and diagnosis. J Am Acad Dermatol. 2019;80:835-851.
Article PDF
Author and Disclosure Information

Dr. Zusstone is from MedStar Washington Hospital Center, Washington, DC. Dr. Lipner is from the Department of Dermatology, Weill Cornell Medicine, New York, New York.

Dr. Zusstone reports no conflict of interest. Dr. Lipner is Deputy Chair of the American Academy of Dermatology Public Education Committee.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Ave, New York, NY 10021 ([email protected]).

Issue
Cutis - 106(2)
Publications
Topics
Page Number
93-95
Sections
Author and Disclosure Information

Dr. Zusstone is from MedStar Washington Hospital Center, Washington, DC. Dr. Lipner is from the Department of Dermatology, Weill Cornell Medicine, New York, New York.

Dr. Zusstone reports no conflict of interest. Dr. Lipner is Deputy Chair of the American Academy of Dermatology Public Education Committee.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Ave, New York, NY 10021 ([email protected]).

Author and Disclosure Information

Dr. Zusstone is from MedStar Washington Hospital Center, Washington, DC. Dr. Lipner is from the Department of Dermatology, Weill Cornell Medicine, New York, New York.

Dr. Zusstone reports no conflict of interest. Dr. Lipner is Deputy Chair of the American Academy of Dermatology Public Education Committee.

Correspondence: Shari R. Lipner, MD, PhD, 1305 York Ave, New York, NY 10021 ([email protected]).

Article PDF
Article PDF

To the Editor:

Patients with skin, hair, or nail concerns often utilize online resources to self-diagnose or learn more about physician-diagnosed conditions. The American Academy of Dermatology (AAD) website offers the public access to informational pages categorized by disease or treatment (https://www.aad.org/public). We sought to evaluate the nail content by searching the Patients and Public section of the AAD website to qualitatively and quantitatively describe mentions of nail conditions. Psoriasis, psoriatic arthritis, atopic dermatitis, and ringworm content also were analyzed and compared to nail content. The analysis was performed on September 7, 2019.

Of the 73 topics listed in the Diseases and Treatments section of the site, 17 (23%) specifically mentioned nail symptoms or pathology (Table). Three additional topics—atopic dermatitis, cellulitis, and neurodermatitis—recommended keeping nails short to prevent injury from scratching. There was 1 mention of obtaining fungal cultures, 2 of nail scraping microscopy, 2 of nail clippings, and 2 of nail-related cancers. There were no mentions of nail biopsies. The total number of unique clinical images across all sections was 300, with 12 of nails. The video library contained 84 videos, of which 6 focused on nail health.



Several sections were reviewed for overall content to understand the typical number of images and videos included for other common conditions. The general psoriasis section included 6 images and 3 videos detailing disease etiology, symptoms, and treatments, without depictions of nails. The psoriatic arthritis section included 8 images and 1 video as well as 1 image of nail pitting. The atopic dermatitis section included 6 images and 6 videos on disease etiology, symptoms, and treatment. The ringworm section, which collectively included all tinea infections, contained 11 images and 1 video on symptoms. In contrast, the only nail-specific section—nail fungus—contained only 2 images and 1 video.

Our study demonstrated that nail content is underrepresented in the public education section of the AAD website. If patients are unable to find nail disease material on the AAD website, they may seek alternative sources that are unreliable. Prior studies have shown that patient Internet resources for subungual melanoma and onychomycosis often are inadequate in quality and readability.1,2

Representative photographs and key information on common nail diseases could be added to improve patient education. The atopic dermatitis section should include text on related nail changes with accompanying images. We also recommend including paronychia information and images as either a separate topic or in the cellulitis section. The contact dermatitis section mentions nail cosmetics as causative factors, but an image of roller-coaster onycholysis may be more helpful.3 Although the alopecia areata section mentions nail changes, this information should be added to the general hair loss section of the site, as many patients may initially seek out the latter category. Herpes simplex may affect nails, and an image showing these changes would be instructive. In addition, pyogenic granulomas and paronychia occur with isotretinoin use.4



Many of the included images were not representative of common clinical findings. The nail lichen planus image showed pitting instead of more typical findings of nail plate atrophy and pterygium. The nail melanoma image showed thickened yellow toenails and the fifth toenail with a thin gray-brown band instead of an isolated wide black band. The nail fungus section included images of superficial onychomycosis and severe onychodystrophy instead of showing more common changes such as distal onycholysis with subungual hyperkeratosis, which is typical of the most common subtype, distal lateral subungual onychomycosis.5 Onychomycosis was referenced again in the ringworm section with 1 image repeated from the nail fungus section and another image that appeared to be a subungual hematoma.

The AAD website offers important patient education resources; however, nail content is underrepresented on this platform. Dermatologists are experts on nail disease, and increased efforts are needed to educate the public about frequently encountered nail signs and symptoms that could signify a serious underlying condition.

After our original search and analysis, new nail topics, images, and videos have been added; therefore, there has been a positive trend toward new nail content being added to site, which will greatly benefit patients.

To the Editor:

Patients with skin, hair, or nail concerns often utilize online resources to self-diagnose or learn more about physician-diagnosed conditions. The American Academy of Dermatology (AAD) website offers the public access to informational pages categorized by disease or treatment (https://www.aad.org/public). We sought to evaluate the nail content by searching the Patients and Public section of the AAD website to qualitatively and quantitatively describe mentions of nail conditions. Psoriasis, psoriatic arthritis, atopic dermatitis, and ringworm content also were analyzed and compared to nail content. The analysis was performed on September 7, 2019.

Of the 73 topics listed in the Diseases and Treatments section of the site, 17 (23%) specifically mentioned nail symptoms or pathology (Table). Three additional topics—atopic dermatitis, cellulitis, and neurodermatitis—recommended keeping nails short to prevent injury from scratching. There was 1 mention of obtaining fungal cultures, 2 of nail scraping microscopy, 2 of nail clippings, and 2 of nail-related cancers. There were no mentions of nail biopsies. The total number of unique clinical images across all sections was 300, with 12 of nails. The video library contained 84 videos, of which 6 focused on nail health.



Several sections were reviewed for overall content to understand the typical number of images and videos included for other common conditions. The general psoriasis section included 6 images and 3 videos detailing disease etiology, symptoms, and treatments, without depictions of nails. The psoriatic arthritis section included 8 images and 1 video as well as 1 image of nail pitting. The atopic dermatitis section included 6 images and 6 videos on disease etiology, symptoms, and treatment. The ringworm section, which collectively included all tinea infections, contained 11 images and 1 video on symptoms. In contrast, the only nail-specific section—nail fungus—contained only 2 images and 1 video.

Our study demonstrated that nail content is underrepresented in the public education section of the AAD website. If patients are unable to find nail disease material on the AAD website, they may seek alternative sources that are unreliable. Prior studies have shown that patient Internet resources for subungual melanoma and onychomycosis often are inadequate in quality and readability.1,2

Representative photographs and key information on common nail diseases could be added to improve patient education. The atopic dermatitis section should include text on related nail changes with accompanying images. We also recommend including paronychia information and images as either a separate topic or in the cellulitis section. The contact dermatitis section mentions nail cosmetics as causative factors, but an image of roller-coaster onycholysis may be more helpful.3 Although the alopecia areata section mentions nail changes, this information should be added to the general hair loss section of the site, as many patients may initially seek out the latter category. Herpes simplex may affect nails, and an image showing these changes would be instructive. In addition, pyogenic granulomas and paronychia occur with isotretinoin use.4



Many of the included images were not representative of common clinical findings. The nail lichen planus image showed pitting instead of more typical findings of nail plate atrophy and pterygium. The nail melanoma image showed thickened yellow toenails and the fifth toenail with a thin gray-brown band instead of an isolated wide black band. The nail fungus section included images of superficial onychomycosis and severe onychodystrophy instead of showing more common changes such as distal onycholysis with subungual hyperkeratosis, which is typical of the most common subtype, distal lateral subungual onychomycosis.5 Onychomycosis was referenced again in the ringworm section with 1 image repeated from the nail fungus section and another image that appeared to be a subungual hematoma.

The AAD website offers important patient education resources; however, nail content is underrepresented on this platform. Dermatologists are experts on nail disease, and increased efforts are needed to educate the public about frequently encountered nail signs and symptoms that could signify a serious underlying condition.

After our original search and analysis, new nail topics, images, and videos have been added; therefore, there has been a positive trend toward new nail content being added to site, which will greatly benefit patients.

References
  1. Kang R, Lipner S. Assessment of internet sources on subungual melanoma [published online August 30, 2018]. Melanoma Res. doi:10.1097/CMR.0000000000000508.
  2. Kang R, Lipner S. Evaluation of onychomycosis information on the internet. J Drugs Dermatol. 2019;18:484-487.
  3. Rieder EA, Tosti A. Cosmetically induced disorders of the nail with update on contemporary nail manicures. J Clin Aesthet Dermatol. 2016;9:39-44.
  4. Arias-Santiago S, Husein-ElAhmed H, Aneiros-Cachaza J, et al. Uncommon side effects of isotretinoin therapy: paronychia and pyogenic granuloma. J Am Acad Dermatol. 2011;64:AB37.
  5. Lipner SR, Scher RK. Onychomycosis: clinical overview and diagnosis. J Am Acad Dermatol. 2019;80:835-851.
References
  1. Kang R, Lipner S. Assessment of internet sources on subungual melanoma [published online August 30, 2018]. Melanoma Res. doi:10.1097/CMR.0000000000000508.
  2. Kang R, Lipner S. Evaluation of onychomycosis information on the internet. J Drugs Dermatol. 2019;18:484-487.
  3. Rieder EA, Tosti A. Cosmetically induced disorders of the nail with update on contemporary nail manicures. J Clin Aesthet Dermatol. 2016;9:39-44.
  4. Arias-Santiago S, Husein-ElAhmed H, Aneiros-Cachaza J, et al. Uncommon side effects of isotretinoin therapy: paronychia and pyogenic granuloma. J Am Acad Dermatol. 2011;64:AB37.
  5. Lipner SR, Scher RK. Onychomycosis: clinical overview and diagnosis. J Am Acad Dermatol. 2019;80:835-851.
Issue
Cutis - 106(2)
Issue
Cutis - 106(2)
Page Number
93-95
Page Number
93-95
Publications
Publications
Topics
Article Type
Sections
Inside the Article

Practice Points

  • Patients often utilize online resources to research skin, hair, and nail conditions.
  • Nail signs and symptoms may represent a serious underlying condition, and nail content is underrepresented on the American Academy of Dermatology (AAD) Patients and Public section of the website.
  • There is a need for more information on nail conditions on the AAD website, offering patients a more comprehensive online dermatology resource. Subsequently, there has been a positive trend toward new nail content being added to the site.
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
Article PDF Media

Laser Safety: The Need for Protocols

Article Type
Changed
Sun, 08/16/2020 - 23:00

The use of lasers in dermatology has evolved and expanded since their first cutaneous use in 1963.1 As the fundamental understanding of the interaction of laser energy with biological tissues increased, the need for laser safety became apparent. Since then, lasers of varying wavelengths have been developed, each with its specific chromophore target and specific safety need. Protocols, such as a checklist, that have been shown to reduce adverse events in surgery and in the intensive care unit can be borrowed to decrease risk from laser injury and optimize laser safety in dermatology.2 The safety of the patient, the laser operator, and the other health care providers involved in the delivery of laser therapy led to the first US Food and Drug Administration (FDA) guidelines for laser use in 1984.3

There are 4 regulatory organizations for laser safety in the United States: the American National Standards Institute (ANSI), the Occupational Health and Safety Administration (OSHA), the FDA’s Center for Devices and Radiological Health, and The Joint Commission. The American National Standards Institute is a nonprofit group composed of laser manufacturers, government agencies, professional societies, educational institutions, and consumer and labor groups. It publishes voluntary safety standards and periodic updates (the series is labelled ANSI Z136) for the use of lasers in general (ANSI Z136.1) and for health care use in particular (ANSI Z136.3), including their use in dermatology. Laser hazard classifications also originate from ANSI. The standards of care established by ANSI guidelines are those by which health care providers are judged in health care litigation and are used by the other 3 organizations listed above. The Center for Devices and Radiological Health oversees laser manufacturers and their adherence to safety standards, determines laser hazard classifications such as ANSI, and requires manufacturers to affix a hazard class to the laser when manufactured. The Joint Commission is the accreditation body for health care programs and inspects hospitals and clinics for compliance with ANSI standards. Additionally, the American Society for Laser Medicine and Surgery, the American Academy of Dermatology, and the American Society for Dermatologic Surgery are professional organizations involved in laser operational safety training.3

Laser Principles

The basic principles of lasers include transmission, absorption, scatter, and reflection, all occurring when laser light is applied to biological tissues. The effects of the laser are a function of the target tissue (the chromophore) and the wavelength of light being used.4 In the skin, there are 3 main endogenous chromophores: water, hemoglobin, and melanin. Some experts consider collagen to be a fourth and separate entity as a chromophore. Tattoos are considered exogenous chromophores.3 The basic principles of lasers are important to understand and keep in mind when discussing laser safety, as they are the mechanisms through which unintended consequences can occur.

Laser Safety

Ocular Hazards
Ocular hazards are a notable concern in laser surgery. The eye is uniquely susceptible to laser light, and eye injuries represent a majority of reported injuries, which can occur through direct beam, mirror reflection by surgical instruments, and beam reflection off the skin (4%–7% of light that hits the skin is reflected because of the refractive index between air and the stratum corneum).3 The different wavelengths of lasers affect different parts of the eye. The 3 parts of the eye affected most are the retina, cornea, and lens. Not only is the lens primarily at risk for acute (lenticular burns) and chronic (cataracts) injury from the laser, but secondarily the lens also can concentrate a laser beam onto the retina by a factor of 100,000 (Table 1).3

The use of ocular protective equipment, sometimes referred to as personal protective eyewear (PPE), is essential and is mandated by ANSI and OSHA for all class 3 and class 4 lasers. The eyewear must be labeled with the wavelength and the degree of optical protection—termed the optical density (OD) or filter factor—of each lens and should match the laser being used. Laser manufacturers, as required by ANSI, must provide the wavelength and OD of their lasers, and both can be found on each laser as well as in ANSI Z136.1.3

Vendors supplying PPE generally provide the material, usually glass or polycarbonate; color; visible light transmission, which is the actual amount of light that reaches one’s eye through the lens; filter specifications, which contain the OD at certain wavelengths; and the types of lasers for which each specific PPE is used. It is important to match the laser to the correct PPE. The use of multiple types of lasers in the same office or laser treatment area can present challenges regarding eye safety. Matching the PPE to the laser in use is critical, and therefore all steps to prevent error for patients and personnel should be employed. One recommendation is to place each laser in a separate room with the appropriate PPE hung outside on the door of that room.



When the treatment area is in the periocular region, protection of the patient’s cornea is essential. Leaded eye shields with nonreflective surfaces have been shown to offer the best protection.5 Prior to placement, anesthetic eye drops and lubrication are important for patient comfort and protection from corneal injury.

 

 



Laser-Generated Airborne Contaminants
Other hazards associated with laser use not directly related to the beam are laser-generated airborne contaminants (LGACs), including chemicals, viruses, bacteria, aerosolized blood products, and nanoparticles (<1 µm) known as ultrafine particles (UFPs). According to ANSI, electrosurgical devices and lasers generate the same smoke. The plume (surgical smoke) is known to contain as many as 60 chemicals, including but not limited to carbon monoxide, acrylonitrite, hydrocyanide, benzene, toluene, naphthalene, and formaldehyde. Several are known carcinogens, and others are environmental toxins.6,7



Smoke management is an important consideration for dermatologists and their patients and generally includes respiratory protection via masks and ventilation techniques. However, the practice is not universal, and oversight agencies such as OSHA and the National Institute for Occupational Safety and Health (NIOSH) provide guidelines only; they do not enforce. As such, smoke management is voluntary and not widely practiced. In a 2014 survey of 997 dermatologic surgeons who were asked if smoke management is used in their practice, 77% of respondents indicated no smoke management was used.6

The Surgical Plume: Composition
A 2014 study from the University of California, San Diego Department of Dermatology analyzed surgical smoke.6 The researchers placed the smoke collection probe 16 to 18 inches above the electrocautery site, which approximates the location of the surgeon’s head during the procedure. Assessing smoke composition, they found high levels of carcinogens and irritants. Two compounds found in their assay—1,3-butadiene and benzene—also are found in secondhand cigarette smoke. However, the concentrations in the plume were 17-fold higher for 1,3-butadiene and 10-fold higher for benzene than those found in secondhand cigarette smoke. The risk from chronic, long-term exposure to these airborne contaminants is notable, as benzene (a known carcinogen as determined by the US Department of Health and Human Services) is known to cause leukemia. For example, a busy Mohs surgeon can reach the equivalent of as many as 50 hours of continuous smoke exposure over the course of a year.6

The Surgical Plume: Particle Concentration
Ultrafine particles can bypass conventional filtering systems (surgical masks and N95 respirators) because of their extremely small size, which allows them to pass further into the lungs and all the way to the alveolar spaces. Geographic regions with high UFPs have been shown to have higher overall mortality rates, as well as higher rates of reactive airway disease, cardiovascular disease, and lung cancer. A 2016 study by Chuang et al7 published in JAMA Dermatology looked at the UFPs in the surgical plume from laser hair removal (LHR) procedures. The plume of LHR has a distinct odor and easily discernible particulates. The investigators measured the UFPs at the level of the laser practitioner and the patient’s face during LHR with a smoke evacuator turned on and again with it turned off for 30 seconds, and then compared them to UFPs measured in the treatment room, the waiting room, and outside the building. There were substantial increases in UFPs from the LHR procedure, especially for the laser practitioner, when the smoke evacuator was off. The ambient baseline particle count, as measured in the clinic waiting area, began at 15,300 particles per cubic centimeter (PPC), and once the LHR procedure began (smoke evacuator on), there was a greater than 8-fold PPC increase above baseline (15,300 PPC to 129,376 PPC) in UFPs measured for the laser practitioner. Importantly, during LHR when the smoke evacuator was turned off for 30 seconds, there was a more than 28-fold increase (15,300 PPC to 435,888 PPC) over baseline to the practitioner (Figure).7


Ultrafine particle (UFP) counts during laser hair removal. Data from Chuang et al.7 PPC indicates particles per cubic centimeter

The Surgical Plume: Viruses, Bacteria, and Aerosolized Blood Products
Viruses and bacteria are thought to be transmissible via the plume, and proviral human immunodeficiency virus DNA has been found in the plume as well as evacuator equipment used to reduce plume exposure.8 A study from 1988 found that CO2 laser users treating verrucae had human papillomavirus in the laser plume.9 A comparison study of CO2 laser users treating verrucae had an increased incidence of nasopharyngeal human papillomavirus infection when compared to a control group, and the plume also contained aerosolized blood.10 The American National Standards Institute, OSHA, and NIOSH all agree that LGAC control from lasers is necessary through respiratory protection and ventilation, but none of these organizations provides specific equipment recommendations. The American Society for Laser Medicine and Surgery has published a position statement on laser plume.11



The Surgical Plume: Smoke Management
Many virus particles and UFPs are less than 0.1 µm in size. It is important to note that neither surgical masks nor high-filtration masks, such as the N95 respirator, filter particles smaller than 0.1 µm. The first line of defense in smoke management is the local exhaust ventilation (LEV) system, which includes wall suction and/or a smoke evacuator. The smoke evacuator is considered the more important of the two. General filtration, such as wall suction, is a low-flow system and is really used for liquids. It can be used as a supplement to the smoke evacuator to control small amounts of plume if fitted with an in-line filter. There are 2 types of LEV filters: ultralow particulate air filters filter particles larger than 0.1µm, whereas high-efficiency particulate air filters filter particles larger than 0.3 µm. The ultralow particulate filters are used in most of the newer LEVs in use today and filter 0.1-µm particles at 99.99% efficiency.3

Of utmost importance when using a smoke evacuator system is suction tip placement. Placing the suction tip 1 cm from the tissue damage site has been shown to be 98.6% effective at removing laser plume. If moved to 2 cm, effectiveness decreases to less than 50%.11 Proper management recommendations based on current evidence suggest that use of a smoke evacuator and an approved fit-tested N95 respirator might provide maximum protection.6 In addition to plume exposure, tissue splatter can occur, especially during ablative (CO2) and tattoo laser therapy, which should prompt consideration of a face shield.11 There are several vendors and models available online, and a simple Internet search for surgical tissue splatter face shields will provide multiple options.

 

 



The standard surgical mask is not NIOSH approved and only effectively (99%) filters particles larger than 5 µm (vs 25% efficacy for 0.3-µm particles). Its main purpose is to protect the patient from the wearer.12



High-filtration masks, which capture particles as small as 0.1 µm, should be used instead. The surgical N95 respirator is a NIOSH-certified respirator and is recommended for use in cases when smoke management is necessary. The FDA does not test or certify these masks; it only clears them after reviewing manufacturer test data. Technically, to be called a surgical mask, it must be cleared by the FDA.12 The 95 of N95 indicates filter efficiency ratings of 95% when testing the filter efficiency using particles of approximately 0.3 µm in diameter (Table 2).13 Because 77% of surgical smoke particles are smaller than 1.1 µm, surgical masks and N95 respirators are never sufficient as stand-alone protection.14 An LEV system is much more important for safe surgical smoke management. However, recommendations call for the use of a smoke evacuator and a high-filtration mask together to obtain the most protection available.14



Fire Hazards
Fire hazards constitute another area of concern for the laser user and are seen with class 4 lasers. There usually are 2 types of fire hazards: electrical fires inside the laser (often faulty wiring) and flash fires (laser beam contacts flammable material). Flammable materials (eg, hair, hair products, makeup, fabrics, plastic, alcohol, chlorhexidine, aluminum chloride, elastic strap on safety goggles, gauze, drapes) should be identified and removed prior to laser use. CO2 and erbium:YAG lasers tend to pose the worst risk for flash fires.15

Precautions for fire control in the laser room should include fire extinguishers and/or fire extinguisher blankets, a water basin, and fire-resistant drapes available as needed. Flammable material such as gauze should be kept wet, or a nonflammable version should be used.3

Additional Safety Considerations
Whenever lasers are being used, it is important to cover any windows in the laser treatment area (LTA) to prevent the laser beam from passing through the glass window. Laser-blocking window covers are a requirement and are available from several vendors. Covers that block every laser class are available and come as a shade or a flat cover that is attached with Velcro or magnets. They also come with “Laser in Use” warning signs for additional safety. Access to the LTA when the laser is in use should be controlled and appropriate warning signs placed on the door to prevent inadvertent entry without proper PPE. Locking the door to the LTA while using the laser is an additional safety measure and can be included on a checklist.

For the dermatologist, the skin is a primary focus, and similar to the eye, can be at risk for injury. The most common type of injury resembles a sunburn, such as those seen in the UVB range, that appears as redness and sometimes blistering,15 which is an important consideration, and attention should be given to all those in the laser room.

Checklists

Checklists are ubiquitous throughout many occupations and many medical specialties. Their usefulness in preventing adverse events is well established. Any patient-provider encounter in which a series of sequential actions is required is a perfect situation for a checklist. In dermatologic laser surgery where the eye is uniquely susceptible to injury, a laser safety checklist is essential. Additionally, there are issues with LGACs and fire that are important to consider. Having protocols (ie, a checklist) in place that address these safety issues has been shown to reduce adverse outcomes.2 There are a number of templates available from various sources that can be customized to the laser treatment area. We provide a modifiable example (Table 3).

Conclusion

Laser usage in dermatologic surgery has increased. According to surveys from the American Society for Dermatologic Surgery, in 2012 there were approximately 2 million laser/light/energy-based procedures performed. By 2017, there were 3.27 million, up from 2.79 million in 2016, representing an approximate 1-year increase of 17%.16 Lasers have allowed interventions for skin, vascular, and aesthetic conditions that were once untreatable. As their use increases in number and broadens in scope, there also has been an increase in litigation alleging malpractice for misuse of the laser.17 Adverse events, which include photochemical or thermal injuries to the skin, pigmentation issues, scarring, plume-related issues, and fires, do occur. One solution to reduce the chance of an adverse outcome is to implement a checklist. Research using checklists has shown that adverse events are reduced when checklists are created and implemented properly. Improving checklist compliance also improves patient outcomes.17 The American National Standards Institute, in their ANSI Z136 series, and the World Health Organization provide checklist templates. We include our checklist for use in laser surgery (Table 3). Understanding that each laser treatment area is unique, the templates can serve as a starting point and can then be customized to suit the needs of each dermatologist.

References
  1. Goldman L, Blaney DJ, Kindel DJ, et al. Effect of the laser beam on the skin. J Invest Dermatol. 1963;40:121-122.
  2. Daggett C, Daggett A. The surgical check list revisited. Int J Surg Res Pract. 2017;4:051.
  3. Pritzker RN, Rohrer TE. Laser safety: standards and guidelines. In: Nouri K, ed. Handbook of Lasers in Dermatology. London, England: Springer; 2014:11-28.
  4. Husain Z, Alster TS. The role of lasers and intense pulsed light technology in dermatology. Clin Cosmet Investig Dermatol. 2016;9:29-40.
  5. Ries WR, Clymer MA, Reinisch L. Laser safety features of eye shields. Lasers Surg Med. 1996;18:309-315.
  6. Oganesyan G, Eimputh S, Kim SS, et al. Surgical smoke detection in dermatologic surgery. Dermatol Surg. 2014;40:1373-1377.
  7. Chuang GS, Farinelli W, Christiani DC, et al. Gaseous and particulate content of laser hair removal plume. JAMA Dermatol. 2016;152:1320-1326.
  8. Baggish MS, Poiesz BJ, Joret D, et al. Presence of human immunodeficiency DNA in laser smoke. Lasers Surg Med. 1991;11:197-203.
  9. Garden JM, O’Banion MK, Shelnitz LS, et al. Papilloma virus in the vapor of carbon dioxide laser-treated verrucae. JAMA. 1988;259:1199-1202.
  10. Gloster HM Jr, Roenigk RK. Risk of acquiring human papillomavirus from the plume produced by the carbon dioxide laser in the treatment of warts. J Am Acad Dermatol. 1995;32:436-441.
  11. American Society for Laser Medicine and Surgery. ASLMS laser and energy device plume position statement. http://www.aslms.org/for-professionals/professional-resources/safety-and-complications/aslms-laser-and-energy-device-plume-position-statement. Accessed October 4, 2019.
  12. A comparison of surgical masks, surgical N95 respirators, and industrial N95 respirators. OH&S website. https://ohsonline.com/Articles/2014/05/01/Comparison-Respiratory.aspx?Page=3. Published May 1, 2014. Accessed October 4, 2019.
  13. 3M Infection Prevention N95 particulate respirators, 1860/1860s and 1870. Frequently Asked Questions. http://multimedia.3m.com/mws/media/323208O/n95-particulate-respirators-1860-1860s-1870-faqs.pdf. Accessed October 4, 2019.
  14. Lewin JM, Brauer JA, Ostad A. Surgical smoke and the dermatologist. J Am Acad Dermatol. 2011;65:636-641.
  15. Arefiev K, Warycha M, Whiting D, et al. Flammability of topical preparations and surgical dressings in cutaneous and laser surgery: a controlled simulation study. J Am Acad Dermatol. 2012;67:700-705.
  16. ASDS survey on dermatologic procedures. American Society for Dermatologic Surgery website. https://www.asds.net/Medical-Professionals/Practice-Resources/ASDS-Survey-on-Dermatologic-Procedures. Accessed October 4, 2019.
  17. Jalian HR, Jalian CA, Avram MM. Common causes of injury and legal action in laser surgery. JAMA Dermatol. 2013;149:188-193.
Article PDF
Author and Disclosure Information

From the Department of Dermatology, Tulane University School of Medicine, New Orleans, Louisiana. Dr. C. Daggett also is from Brookwood Baptist Health, Birmingham, Alabama. Dr. McBurney also is from Sanova Dermatology, Lafayette, Louisiana.

Drs. A. Daggett, C. Daggett, and McBurney report no conflict of interest. Dr. Murina is on the speaker’s bureau for AbbVie, Celgene Corporation, and Novartis and has participated in the advisory board for Celgene Corporation. She also has research grants from Regeneron Pharmaceuticals, Inc.

Correspondence: Caroline Daggett, MD ([email protected]).

Issue
Cutis - 106(2)
Publications
Topics
Page Number
87-92
Sections
Author and Disclosure Information

From the Department of Dermatology, Tulane University School of Medicine, New Orleans, Louisiana. Dr. C. Daggett also is from Brookwood Baptist Health, Birmingham, Alabama. Dr. McBurney also is from Sanova Dermatology, Lafayette, Louisiana.

Drs. A. Daggett, C. Daggett, and McBurney report no conflict of interest. Dr. Murina is on the speaker’s bureau for AbbVie, Celgene Corporation, and Novartis and has participated in the advisory board for Celgene Corporation. She also has research grants from Regeneron Pharmaceuticals, Inc.

Correspondence: Caroline Daggett, MD ([email protected]).

Author and Disclosure Information

From the Department of Dermatology, Tulane University School of Medicine, New Orleans, Louisiana. Dr. C. Daggett also is from Brookwood Baptist Health, Birmingham, Alabama. Dr. McBurney also is from Sanova Dermatology, Lafayette, Louisiana.

Drs. A. Daggett, C. Daggett, and McBurney report no conflict of interest. Dr. Murina is on the speaker’s bureau for AbbVie, Celgene Corporation, and Novartis and has participated in the advisory board for Celgene Corporation. She also has research grants from Regeneron Pharmaceuticals, Inc.

Correspondence: Caroline Daggett, MD ([email protected]).

Article PDF
Article PDF

The use of lasers in dermatology has evolved and expanded since their first cutaneous use in 1963.1 As the fundamental understanding of the interaction of laser energy with biological tissues increased, the need for laser safety became apparent. Since then, lasers of varying wavelengths have been developed, each with its specific chromophore target and specific safety need. Protocols, such as a checklist, that have been shown to reduce adverse events in surgery and in the intensive care unit can be borrowed to decrease risk from laser injury and optimize laser safety in dermatology.2 The safety of the patient, the laser operator, and the other health care providers involved in the delivery of laser therapy led to the first US Food and Drug Administration (FDA) guidelines for laser use in 1984.3

There are 4 regulatory organizations for laser safety in the United States: the American National Standards Institute (ANSI), the Occupational Health and Safety Administration (OSHA), the FDA’s Center for Devices and Radiological Health, and The Joint Commission. The American National Standards Institute is a nonprofit group composed of laser manufacturers, government agencies, professional societies, educational institutions, and consumer and labor groups. It publishes voluntary safety standards and periodic updates (the series is labelled ANSI Z136) for the use of lasers in general (ANSI Z136.1) and for health care use in particular (ANSI Z136.3), including their use in dermatology. Laser hazard classifications also originate from ANSI. The standards of care established by ANSI guidelines are those by which health care providers are judged in health care litigation and are used by the other 3 organizations listed above. The Center for Devices and Radiological Health oversees laser manufacturers and their adherence to safety standards, determines laser hazard classifications such as ANSI, and requires manufacturers to affix a hazard class to the laser when manufactured. The Joint Commission is the accreditation body for health care programs and inspects hospitals and clinics for compliance with ANSI standards. Additionally, the American Society for Laser Medicine and Surgery, the American Academy of Dermatology, and the American Society for Dermatologic Surgery are professional organizations involved in laser operational safety training.3

Laser Principles

The basic principles of lasers include transmission, absorption, scatter, and reflection, all occurring when laser light is applied to biological tissues. The effects of the laser are a function of the target tissue (the chromophore) and the wavelength of light being used.4 In the skin, there are 3 main endogenous chromophores: water, hemoglobin, and melanin. Some experts consider collagen to be a fourth and separate entity as a chromophore. Tattoos are considered exogenous chromophores.3 The basic principles of lasers are important to understand and keep in mind when discussing laser safety, as they are the mechanisms through which unintended consequences can occur.

Laser Safety

Ocular Hazards
Ocular hazards are a notable concern in laser surgery. The eye is uniquely susceptible to laser light, and eye injuries represent a majority of reported injuries, which can occur through direct beam, mirror reflection by surgical instruments, and beam reflection off the skin (4%–7% of light that hits the skin is reflected because of the refractive index between air and the stratum corneum).3 The different wavelengths of lasers affect different parts of the eye. The 3 parts of the eye affected most are the retina, cornea, and lens. Not only is the lens primarily at risk for acute (lenticular burns) and chronic (cataracts) injury from the laser, but secondarily the lens also can concentrate a laser beam onto the retina by a factor of 100,000 (Table 1).3

The use of ocular protective equipment, sometimes referred to as personal protective eyewear (PPE), is essential and is mandated by ANSI and OSHA for all class 3 and class 4 lasers. The eyewear must be labeled with the wavelength and the degree of optical protection—termed the optical density (OD) or filter factor—of each lens and should match the laser being used. Laser manufacturers, as required by ANSI, must provide the wavelength and OD of their lasers, and both can be found on each laser as well as in ANSI Z136.1.3

Vendors supplying PPE generally provide the material, usually glass or polycarbonate; color; visible light transmission, which is the actual amount of light that reaches one’s eye through the lens; filter specifications, which contain the OD at certain wavelengths; and the types of lasers for which each specific PPE is used. It is important to match the laser to the correct PPE. The use of multiple types of lasers in the same office or laser treatment area can present challenges regarding eye safety. Matching the PPE to the laser in use is critical, and therefore all steps to prevent error for patients and personnel should be employed. One recommendation is to place each laser in a separate room with the appropriate PPE hung outside on the door of that room.



When the treatment area is in the periocular region, protection of the patient’s cornea is essential. Leaded eye shields with nonreflective surfaces have been shown to offer the best protection.5 Prior to placement, anesthetic eye drops and lubrication are important for patient comfort and protection from corneal injury.

 

 



Laser-Generated Airborne Contaminants
Other hazards associated with laser use not directly related to the beam are laser-generated airborne contaminants (LGACs), including chemicals, viruses, bacteria, aerosolized blood products, and nanoparticles (<1 µm) known as ultrafine particles (UFPs). According to ANSI, electrosurgical devices and lasers generate the same smoke. The plume (surgical smoke) is known to contain as many as 60 chemicals, including but not limited to carbon monoxide, acrylonitrite, hydrocyanide, benzene, toluene, naphthalene, and formaldehyde. Several are known carcinogens, and others are environmental toxins.6,7



Smoke management is an important consideration for dermatologists and their patients and generally includes respiratory protection via masks and ventilation techniques. However, the practice is not universal, and oversight agencies such as OSHA and the National Institute for Occupational Safety and Health (NIOSH) provide guidelines only; they do not enforce. As such, smoke management is voluntary and not widely practiced. In a 2014 survey of 997 dermatologic surgeons who were asked if smoke management is used in their practice, 77% of respondents indicated no smoke management was used.6

The Surgical Plume: Composition
A 2014 study from the University of California, San Diego Department of Dermatology analyzed surgical smoke.6 The researchers placed the smoke collection probe 16 to 18 inches above the electrocautery site, which approximates the location of the surgeon’s head during the procedure. Assessing smoke composition, they found high levels of carcinogens and irritants. Two compounds found in their assay—1,3-butadiene and benzene—also are found in secondhand cigarette smoke. However, the concentrations in the plume were 17-fold higher for 1,3-butadiene and 10-fold higher for benzene than those found in secondhand cigarette smoke. The risk from chronic, long-term exposure to these airborne contaminants is notable, as benzene (a known carcinogen as determined by the US Department of Health and Human Services) is known to cause leukemia. For example, a busy Mohs surgeon can reach the equivalent of as many as 50 hours of continuous smoke exposure over the course of a year.6

The Surgical Plume: Particle Concentration
Ultrafine particles can bypass conventional filtering systems (surgical masks and N95 respirators) because of their extremely small size, which allows them to pass further into the lungs and all the way to the alveolar spaces. Geographic regions with high UFPs have been shown to have higher overall mortality rates, as well as higher rates of reactive airway disease, cardiovascular disease, and lung cancer. A 2016 study by Chuang et al7 published in JAMA Dermatology looked at the UFPs in the surgical plume from laser hair removal (LHR) procedures. The plume of LHR has a distinct odor and easily discernible particulates. The investigators measured the UFPs at the level of the laser practitioner and the patient’s face during LHR with a smoke evacuator turned on and again with it turned off for 30 seconds, and then compared them to UFPs measured in the treatment room, the waiting room, and outside the building. There were substantial increases in UFPs from the LHR procedure, especially for the laser practitioner, when the smoke evacuator was off. The ambient baseline particle count, as measured in the clinic waiting area, began at 15,300 particles per cubic centimeter (PPC), and once the LHR procedure began (smoke evacuator on), there was a greater than 8-fold PPC increase above baseline (15,300 PPC to 129,376 PPC) in UFPs measured for the laser practitioner. Importantly, during LHR when the smoke evacuator was turned off for 30 seconds, there was a more than 28-fold increase (15,300 PPC to 435,888 PPC) over baseline to the practitioner (Figure).7


Ultrafine particle (UFP) counts during laser hair removal. Data from Chuang et al.7 PPC indicates particles per cubic centimeter

The Surgical Plume: Viruses, Bacteria, and Aerosolized Blood Products
Viruses and bacteria are thought to be transmissible via the plume, and proviral human immunodeficiency virus DNA has been found in the plume as well as evacuator equipment used to reduce plume exposure.8 A study from 1988 found that CO2 laser users treating verrucae had human papillomavirus in the laser plume.9 A comparison study of CO2 laser users treating verrucae had an increased incidence of nasopharyngeal human papillomavirus infection when compared to a control group, and the plume also contained aerosolized blood.10 The American National Standards Institute, OSHA, and NIOSH all agree that LGAC control from lasers is necessary through respiratory protection and ventilation, but none of these organizations provides specific equipment recommendations. The American Society for Laser Medicine and Surgery has published a position statement on laser plume.11



The Surgical Plume: Smoke Management
Many virus particles and UFPs are less than 0.1 µm in size. It is important to note that neither surgical masks nor high-filtration masks, such as the N95 respirator, filter particles smaller than 0.1 µm. The first line of defense in smoke management is the local exhaust ventilation (LEV) system, which includes wall suction and/or a smoke evacuator. The smoke evacuator is considered the more important of the two. General filtration, such as wall suction, is a low-flow system and is really used for liquids. It can be used as a supplement to the smoke evacuator to control small amounts of plume if fitted with an in-line filter. There are 2 types of LEV filters: ultralow particulate air filters filter particles larger than 0.1µm, whereas high-efficiency particulate air filters filter particles larger than 0.3 µm. The ultralow particulate filters are used in most of the newer LEVs in use today and filter 0.1-µm particles at 99.99% efficiency.3

Of utmost importance when using a smoke evacuator system is suction tip placement. Placing the suction tip 1 cm from the tissue damage site has been shown to be 98.6% effective at removing laser plume. If moved to 2 cm, effectiveness decreases to less than 50%.11 Proper management recommendations based on current evidence suggest that use of a smoke evacuator and an approved fit-tested N95 respirator might provide maximum protection.6 In addition to plume exposure, tissue splatter can occur, especially during ablative (CO2) and tattoo laser therapy, which should prompt consideration of a face shield.11 There are several vendors and models available online, and a simple Internet search for surgical tissue splatter face shields will provide multiple options.

 

 



The standard surgical mask is not NIOSH approved and only effectively (99%) filters particles larger than 5 µm (vs 25% efficacy for 0.3-µm particles). Its main purpose is to protect the patient from the wearer.12



High-filtration masks, which capture particles as small as 0.1 µm, should be used instead. The surgical N95 respirator is a NIOSH-certified respirator and is recommended for use in cases when smoke management is necessary. The FDA does not test or certify these masks; it only clears them after reviewing manufacturer test data. Technically, to be called a surgical mask, it must be cleared by the FDA.12 The 95 of N95 indicates filter efficiency ratings of 95% when testing the filter efficiency using particles of approximately 0.3 µm in diameter (Table 2).13 Because 77% of surgical smoke particles are smaller than 1.1 µm, surgical masks and N95 respirators are never sufficient as stand-alone protection.14 An LEV system is much more important for safe surgical smoke management. However, recommendations call for the use of a smoke evacuator and a high-filtration mask together to obtain the most protection available.14



Fire Hazards
Fire hazards constitute another area of concern for the laser user and are seen with class 4 lasers. There usually are 2 types of fire hazards: electrical fires inside the laser (often faulty wiring) and flash fires (laser beam contacts flammable material). Flammable materials (eg, hair, hair products, makeup, fabrics, plastic, alcohol, chlorhexidine, aluminum chloride, elastic strap on safety goggles, gauze, drapes) should be identified and removed prior to laser use. CO2 and erbium:YAG lasers tend to pose the worst risk for flash fires.15

Precautions for fire control in the laser room should include fire extinguishers and/or fire extinguisher blankets, a water basin, and fire-resistant drapes available as needed. Flammable material such as gauze should be kept wet, or a nonflammable version should be used.3

Additional Safety Considerations
Whenever lasers are being used, it is important to cover any windows in the laser treatment area (LTA) to prevent the laser beam from passing through the glass window. Laser-blocking window covers are a requirement and are available from several vendors. Covers that block every laser class are available and come as a shade or a flat cover that is attached with Velcro or magnets. They also come with “Laser in Use” warning signs for additional safety. Access to the LTA when the laser is in use should be controlled and appropriate warning signs placed on the door to prevent inadvertent entry without proper PPE. Locking the door to the LTA while using the laser is an additional safety measure and can be included on a checklist.

For the dermatologist, the skin is a primary focus, and similar to the eye, can be at risk for injury. The most common type of injury resembles a sunburn, such as those seen in the UVB range, that appears as redness and sometimes blistering,15 which is an important consideration, and attention should be given to all those in the laser room.

Checklists

Checklists are ubiquitous throughout many occupations and many medical specialties. Their usefulness in preventing adverse events is well established. Any patient-provider encounter in which a series of sequential actions is required is a perfect situation for a checklist. In dermatologic laser surgery where the eye is uniquely susceptible to injury, a laser safety checklist is essential. Additionally, there are issues with LGACs and fire that are important to consider. Having protocols (ie, a checklist) in place that address these safety issues has been shown to reduce adverse outcomes.2 There are a number of templates available from various sources that can be customized to the laser treatment area. We provide a modifiable example (Table 3).

Conclusion

Laser usage in dermatologic surgery has increased. According to surveys from the American Society for Dermatologic Surgery, in 2012 there were approximately 2 million laser/light/energy-based procedures performed. By 2017, there were 3.27 million, up from 2.79 million in 2016, representing an approximate 1-year increase of 17%.16 Lasers have allowed interventions for skin, vascular, and aesthetic conditions that were once untreatable. As their use increases in number and broadens in scope, there also has been an increase in litigation alleging malpractice for misuse of the laser.17 Adverse events, which include photochemical or thermal injuries to the skin, pigmentation issues, scarring, plume-related issues, and fires, do occur. One solution to reduce the chance of an adverse outcome is to implement a checklist. Research using checklists has shown that adverse events are reduced when checklists are created and implemented properly. Improving checklist compliance also improves patient outcomes.17 The American National Standards Institute, in their ANSI Z136 series, and the World Health Organization provide checklist templates. We include our checklist for use in laser surgery (Table 3). Understanding that each laser treatment area is unique, the templates can serve as a starting point and can then be customized to suit the needs of each dermatologist.

The use of lasers in dermatology has evolved and expanded since their first cutaneous use in 1963.1 As the fundamental understanding of the interaction of laser energy with biological tissues increased, the need for laser safety became apparent. Since then, lasers of varying wavelengths have been developed, each with its specific chromophore target and specific safety need. Protocols, such as a checklist, that have been shown to reduce adverse events in surgery and in the intensive care unit can be borrowed to decrease risk from laser injury and optimize laser safety in dermatology.2 The safety of the patient, the laser operator, and the other health care providers involved in the delivery of laser therapy led to the first US Food and Drug Administration (FDA) guidelines for laser use in 1984.3

There are 4 regulatory organizations for laser safety in the United States: the American National Standards Institute (ANSI), the Occupational Health and Safety Administration (OSHA), the FDA’s Center for Devices and Radiological Health, and The Joint Commission. The American National Standards Institute is a nonprofit group composed of laser manufacturers, government agencies, professional societies, educational institutions, and consumer and labor groups. It publishes voluntary safety standards and periodic updates (the series is labelled ANSI Z136) for the use of lasers in general (ANSI Z136.1) and for health care use in particular (ANSI Z136.3), including their use in dermatology. Laser hazard classifications also originate from ANSI. The standards of care established by ANSI guidelines are those by which health care providers are judged in health care litigation and are used by the other 3 organizations listed above. The Center for Devices and Radiological Health oversees laser manufacturers and their adherence to safety standards, determines laser hazard classifications such as ANSI, and requires manufacturers to affix a hazard class to the laser when manufactured. The Joint Commission is the accreditation body for health care programs and inspects hospitals and clinics for compliance with ANSI standards. Additionally, the American Society for Laser Medicine and Surgery, the American Academy of Dermatology, and the American Society for Dermatologic Surgery are professional organizations involved in laser operational safety training.3

Laser Principles

The basic principles of lasers include transmission, absorption, scatter, and reflection, all occurring when laser light is applied to biological tissues. The effects of the laser are a function of the target tissue (the chromophore) and the wavelength of light being used.4 In the skin, there are 3 main endogenous chromophores: water, hemoglobin, and melanin. Some experts consider collagen to be a fourth and separate entity as a chromophore. Tattoos are considered exogenous chromophores.3 The basic principles of lasers are important to understand and keep in mind when discussing laser safety, as they are the mechanisms through which unintended consequences can occur.

Laser Safety

Ocular Hazards
Ocular hazards are a notable concern in laser surgery. The eye is uniquely susceptible to laser light, and eye injuries represent a majority of reported injuries, which can occur through direct beam, mirror reflection by surgical instruments, and beam reflection off the skin (4%–7% of light that hits the skin is reflected because of the refractive index between air and the stratum corneum).3 The different wavelengths of lasers affect different parts of the eye. The 3 parts of the eye affected most are the retina, cornea, and lens. Not only is the lens primarily at risk for acute (lenticular burns) and chronic (cataracts) injury from the laser, but secondarily the lens also can concentrate a laser beam onto the retina by a factor of 100,000 (Table 1).3

The use of ocular protective equipment, sometimes referred to as personal protective eyewear (PPE), is essential and is mandated by ANSI and OSHA for all class 3 and class 4 lasers. The eyewear must be labeled with the wavelength and the degree of optical protection—termed the optical density (OD) or filter factor—of each lens and should match the laser being used. Laser manufacturers, as required by ANSI, must provide the wavelength and OD of their lasers, and both can be found on each laser as well as in ANSI Z136.1.3

Vendors supplying PPE generally provide the material, usually glass or polycarbonate; color; visible light transmission, which is the actual amount of light that reaches one’s eye through the lens; filter specifications, which contain the OD at certain wavelengths; and the types of lasers for which each specific PPE is used. It is important to match the laser to the correct PPE. The use of multiple types of lasers in the same office or laser treatment area can present challenges regarding eye safety. Matching the PPE to the laser in use is critical, and therefore all steps to prevent error for patients and personnel should be employed. One recommendation is to place each laser in a separate room with the appropriate PPE hung outside on the door of that room.



When the treatment area is in the periocular region, protection of the patient’s cornea is essential. Leaded eye shields with nonreflective surfaces have been shown to offer the best protection.5 Prior to placement, anesthetic eye drops and lubrication are important for patient comfort and protection from corneal injury.

 

 



Laser-Generated Airborne Contaminants
Other hazards associated with laser use not directly related to the beam are laser-generated airborne contaminants (LGACs), including chemicals, viruses, bacteria, aerosolized blood products, and nanoparticles (<1 µm) known as ultrafine particles (UFPs). According to ANSI, electrosurgical devices and lasers generate the same smoke. The plume (surgical smoke) is known to contain as many as 60 chemicals, including but not limited to carbon monoxide, acrylonitrite, hydrocyanide, benzene, toluene, naphthalene, and formaldehyde. Several are known carcinogens, and others are environmental toxins.6,7



Smoke management is an important consideration for dermatologists and their patients and generally includes respiratory protection via masks and ventilation techniques. However, the practice is not universal, and oversight agencies such as OSHA and the National Institute for Occupational Safety and Health (NIOSH) provide guidelines only; they do not enforce. As such, smoke management is voluntary and not widely practiced. In a 2014 survey of 997 dermatologic surgeons who were asked if smoke management is used in their practice, 77% of respondents indicated no smoke management was used.6

The Surgical Plume: Composition
A 2014 study from the University of California, San Diego Department of Dermatology analyzed surgical smoke.6 The researchers placed the smoke collection probe 16 to 18 inches above the electrocautery site, which approximates the location of the surgeon’s head during the procedure. Assessing smoke composition, they found high levels of carcinogens and irritants. Two compounds found in their assay—1,3-butadiene and benzene—also are found in secondhand cigarette smoke. However, the concentrations in the plume were 17-fold higher for 1,3-butadiene and 10-fold higher for benzene than those found in secondhand cigarette smoke. The risk from chronic, long-term exposure to these airborne contaminants is notable, as benzene (a known carcinogen as determined by the US Department of Health and Human Services) is known to cause leukemia. For example, a busy Mohs surgeon can reach the equivalent of as many as 50 hours of continuous smoke exposure over the course of a year.6

The Surgical Plume: Particle Concentration
Ultrafine particles can bypass conventional filtering systems (surgical masks and N95 respirators) because of their extremely small size, which allows them to pass further into the lungs and all the way to the alveolar spaces. Geographic regions with high UFPs have been shown to have higher overall mortality rates, as well as higher rates of reactive airway disease, cardiovascular disease, and lung cancer. A 2016 study by Chuang et al7 published in JAMA Dermatology looked at the UFPs in the surgical plume from laser hair removal (LHR) procedures. The plume of LHR has a distinct odor and easily discernible particulates. The investigators measured the UFPs at the level of the laser practitioner and the patient’s face during LHR with a smoke evacuator turned on and again with it turned off for 30 seconds, and then compared them to UFPs measured in the treatment room, the waiting room, and outside the building. There were substantial increases in UFPs from the LHR procedure, especially for the laser practitioner, when the smoke evacuator was off. The ambient baseline particle count, as measured in the clinic waiting area, began at 15,300 particles per cubic centimeter (PPC), and once the LHR procedure began (smoke evacuator on), there was a greater than 8-fold PPC increase above baseline (15,300 PPC to 129,376 PPC) in UFPs measured for the laser practitioner. Importantly, during LHR when the smoke evacuator was turned off for 30 seconds, there was a more than 28-fold increase (15,300 PPC to 435,888 PPC) over baseline to the practitioner (Figure).7


Ultrafine particle (UFP) counts during laser hair removal. Data from Chuang et al.7 PPC indicates particles per cubic centimeter

The Surgical Plume: Viruses, Bacteria, and Aerosolized Blood Products
Viruses and bacteria are thought to be transmissible via the plume, and proviral human immunodeficiency virus DNA has been found in the plume as well as evacuator equipment used to reduce plume exposure.8 A study from 1988 found that CO2 laser users treating verrucae had human papillomavirus in the laser plume.9 A comparison study of CO2 laser users treating verrucae had an increased incidence of nasopharyngeal human papillomavirus infection when compared to a control group, and the plume also contained aerosolized blood.10 The American National Standards Institute, OSHA, and NIOSH all agree that LGAC control from lasers is necessary through respiratory protection and ventilation, but none of these organizations provides specific equipment recommendations. The American Society for Laser Medicine and Surgery has published a position statement on laser plume.11



The Surgical Plume: Smoke Management
Many virus particles and UFPs are less than 0.1 µm in size. It is important to note that neither surgical masks nor high-filtration masks, such as the N95 respirator, filter particles smaller than 0.1 µm. The first line of defense in smoke management is the local exhaust ventilation (LEV) system, which includes wall suction and/or a smoke evacuator. The smoke evacuator is considered the more important of the two. General filtration, such as wall suction, is a low-flow system and is really used for liquids. It can be used as a supplement to the smoke evacuator to control small amounts of plume if fitted with an in-line filter. There are 2 types of LEV filters: ultralow particulate air filters filter particles larger than 0.1µm, whereas high-efficiency particulate air filters filter particles larger than 0.3 µm. The ultralow particulate filters are used in most of the newer LEVs in use today and filter 0.1-µm particles at 99.99% efficiency.3

Of utmost importance when using a smoke evacuator system is suction tip placement. Placing the suction tip 1 cm from the tissue damage site has been shown to be 98.6% effective at removing laser plume. If moved to 2 cm, effectiveness decreases to less than 50%.11 Proper management recommendations based on current evidence suggest that use of a smoke evacuator and an approved fit-tested N95 respirator might provide maximum protection.6 In addition to plume exposure, tissue splatter can occur, especially during ablative (CO2) and tattoo laser therapy, which should prompt consideration of a face shield.11 There are several vendors and models available online, and a simple Internet search for surgical tissue splatter face shields will provide multiple options.

 

 



The standard surgical mask is not NIOSH approved and only effectively (99%) filters particles larger than 5 µm (vs 25% efficacy for 0.3-µm particles). Its main purpose is to protect the patient from the wearer.12



High-filtration masks, which capture particles as small as 0.1 µm, should be used instead. The surgical N95 respirator is a NIOSH-certified respirator and is recommended for use in cases when smoke management is necessary. The FDA does not test or certify these masks; it only clears them after reviewing manufacturer test data. Technically, to be called a surgical mask, it must be cleared by the FDA.12 The 95 of N95 indicates filter efficiency ratings of 95% when testing the filter efficiency using particles of approximately 0.3 µm in diameter (Table 2).13 Because 77% of surgical smoke particles are smaller than 1.1 µm, surgical masks and N95 respirators are never sufficient as stand-alone protection.14 An LEV system is much more important for safe surgical smoke management. However, recommendations call for the use of a smoke evacuator and a high-filtration mask together to obtain the most protection available.14



Fire Hazards
Fire hazards constitute another area of concern for the laser user and are seen with class 4 lasers. There usually are 2 types of fire hazards: electrical fires inside the laser (often faulty wiring) and flash fires (laser beam contacts flammable material). Flammable materials (eg, hair, hair products, makeup, fabrics, plastic, alcohol, chlorhexidine, aluminum chloride, elastic strap on safety goggles, gauze, drapes) should be identified and removed prior to laser use. CO2 and erbium:YAG lasers tend to pose the worst risk for flash fires.15

Precautions for fire control in the laser room should include fire extinguishers and/or fire extinguisher blankets, a water basin, and fire-resistant drapes available as needed. Flammable material such as gauze should be kept wet, or a nonflammable version should be used.3

Additional Safety Considerations
Whenever lasers are being used, it is important to cover any windows in the laser treatment area (LTA) to prevent the laser beam from passing through the glass window. Laser-blocking window covers are a requirement and are available from several vendors. Covers that block every laser class are available and come as a shade or a flat cover that is attached with Velcro or magnets. They also come with “Laser in Use” warning signs for additional safety. Access to the LTA when the laser is in use should be controlled and appropriate warning signs placed on the door to prevent inadvertent entry without proper PPE. Locking the door to the LTA while using the laser is an additional safety measure and can be included on a checklist.

For the dermatologist, the skin is a primary focus, and similar to the eye, can be at risk for injury. The most common type of injury resembles a sunburn, such as those seen in the UVB range, that appears as redness and sometimes blistering,15 which is an important consideration, and attention should be given to all those in the laser room.

Checklists

Checklists are ubiquitous throughout many occupations and many medical specialties. Their usefulness in preventing adverse events is well established. Any patient-provider encounter in which a series of sequential actions is required is a perfect situation for a checklist. In dermatologic laser surgery where the eye is uniquely susceptible to injury, a laser safety checklist is essential. Additionally, there are issues with LGACs and fire that are important to consider. Having protocols (ie, a checklist) in place that address these safety issues has been shown to reduce adverse outcomes.2 There are a number of templates available from various sources that can be customized to the laser treatment area. We provide a modifiable example (Table 3).

Conclusion

Laser usage in dermatologic surgery has increased. According to surveys from the American Society for Dermatologic Surgery, in 2012 there were approximately 2 million laser/light/energy-based procedures performed. By 2017, there were 3.27 million, up from 2.79 million in 2016, representing an approximate 1-year increase of 17%.16 Lasers have allowed interventions for skin, vascular, and aesthetic conditions that were once untreatable. As their use increases in number and broadens in scope, there also has been an increase in litigation alleging malpractice for misuse of the laser.17 Adverse events, which include photochemical or thermal injuries to the skin, pigmentation issues, scarring, plume-related issues, and fires, do occur. One solution to reduce the chance of an adverse outcome is to implement a checklist. Research using checklists has shown that adverse events are reduced when checklists are created and implemented properly. Improving checklist compliance also improves patient outcomes.17 The American National Standards Institute, in their ANSI Z136 series, and the World Health Organization provide checklist templates. We include our checklist for use in laser surgery (Table 3). Understanding that each laser treatment area is unique, the templates can serve as a starting point and can then be customized to suit the needs of each dermatologist.

References
  1. Goldman L, Blaney DJ, Kindel DJ, et al. Effect of the laser beam on the skin. J Invest Dermatol. 1963;40:121-122.
  2. Daggett C, Daggett A. The surgical check list revisited. Int J Surg Res Pract. 2017;4:051.
  3. Pritzker RN, Rohrer TE. Laser safety: standards and guidelines. In: Nouri K, ed. Handbook of Lasers in Dermatology. London, England: Springer; 2014:11-28.
  4. Husain Z, Alster TS. The role of lasers and intense pulsed light technology in dermatology. Clin Cosmet Investig Dermatol. 2016;9:29-40.
  5. Ries WR, Clymer MA, Reinisch L. Laser safety features of eye shields. Lasers Surg Med. 1996;18:309-315.
  6. Oganesyan G, Eimputh S, Kim SS, et al. Surgical smoke detection in dermatologic surgery. Dermatol Surg. 2014;40:1373-1377.
  7. Chuang GS, Farinelli W, Christiani DC, et al. Gaseous and particulate content of laser hair removal plume. JAMA Dermatol. 2016;152:1320-1326.
  8. Baggish MS, Poiesz BJ, Joret D, et al. Presence of human immunodeficiency DNA in laser smoke. Lasers Surg Med. 1991;11:197-203.
  9. Garden JM, O’Banion MK, Shelnitz LS, et al. Papilloma virus in the vapor of carbon dioxide laser-treated verrucae. JAMA. 1988;259:1199-1202.
  10. Gloster HM Jr, Roenigk RK. Risk of acquiring human papillomavirus from the plume produced by the carbon dioxide laser in the treatment of warts. J Am Acad Dermatol. 1995;32:436-441.
  11. American Society for Laser Medicine and Surgery. ASLMS laser and energy device plume position statement. http://www.aslms.org/for-professionals/professional-resources/safety-and-complications/aslms-laser-and-energy-device-plume-position-statement. Accessed October 4, 2019.
  12. A comparison of surgical masks, surgical N95 respirators, and industrial N95 respirators. OH&S website. https://ohsonline.com/Articles/2014/05/01/Comparison-Respiratory.aspx?Page=3. Published May 1, 2014. Accessed October 4, 2019.
  13. 3M Infection Prevention N95 particulate respirators, 1860/1860s and 1870. Frequently Asked Questions. http://multimedia.3m.com/mws/media/323208O/n95-particulate-respirators-1860-1860s-1870-faqs.pdf. Accessed October 4, 2019.
  14. Lewin JM, Brauer JA, Ostad A. Surgical smoke and the dermatologist. J Am Acad Dermatol. 2011;65:636-641.
  15. Arefiev K, Warycha M, Whiting D, et al. Flammability of topical preparations and surgical dressings in cutaneous and laser surgery: a controlled simulation study. J Am Acad Dermatol. 2012;67:700-705.
  16. ASDS survey on dermatologic procedures. American Society for Dermatologic Surgery website. https://www.asds.net/Medical-Professionals/Practice-Resources/ASDS-Survey-on-Dermatologic-Procedures. Accessed October 4, 2019.
  17. Jalian HR, Jalian CA, Avram MM. Common causes of injury and legal action in laser surgery. JAMA Dermatol. 2013;149:188-193.
References
  1. Goldman L, Blaney DJ, Kindel DJ, et al. Effect of the laser beam on the skin. J Invest Dermatol. 1963;40:121-122.
  2. Daggett C, Daggett A. The surgical check list revisited. Int J Surg Res Pract. 2017;4:051.
  3. Pritzker RN, Rohrer TE. Laser safety: standards and guidelines. In: Nouri K, ed. Handbook of Lasers in Dermatology. London, England: Springer; 2014:11-28.
  4. Husain Z, Alster TS. The role of lasers and intense pulsed light technology in dermatology. Clin Cosmet Investig Dermatol. 2016;9:29-40.
  5. Ries WR, Clymer MA, Reinisch L. Laser safety features of eye shields. Lasers Surg Med. 1996;18:309-315.
  6. Oganesyan G, Eimputh S, Kim SS, et al. Surgical smoke detection in dermatologic surgery. Dermatol Surg. 2014;40:1373-1377.
  7. Chuang GS, Farinelli W, Christiani DC, et al. Gaseous and particulate content of laser hair removal plume. JAMA Dermatol. 2016;152:1320-1326.
  8. Baggish MS, Poiesz BJ, Joret D, et al. Presence of human immunodeficiency DNA in laser smoke. Lasers Surg Med. 1991;11:197-203.
  9. Garden JM, O’Banion MK, Shelnitz LS, et al. Papilloma virus in the vapor of carbon dioxide laser-treated verrucae. JAMA. 1988;259:1199-1202.
  10. Gloster HM Jr, Roenigk RK. Risk of acquiring human papillomavirus from the plume produced by the carbon dioxide laser in the treatment of warts. J Am Acad Dermatol. 1995;32:436-441.
  11. American Society for Laser Medicine and Surgery. ASLMS laser and energy device plume position statement. http://www.aslms.org/for-professionals/professional-resources/safety-and-complications/aslms-laser-and-energy-device-plume-position-statement. Accessed October 4, 2019.
  12. A comparison of surgical masks, surgical N95 respirators, and industrial N95 respirators. OH&S website. https://ohsonline.com/Articles/2014/05/01/Comparison-Respiratory.aspx?Page=3. Published May 1, 2014. Accessed October 4, 2019.
  13. 3M Infection Prevention N95 particulate respirators, 1860/1860s and 1870. Frequently Asked Questions. http://multimedia.3m.com/mws/media/323208O/n95-particulate-respirators-1860-1860s-1870-faqs.pdf. Accessed October 4, 2019.
  14. Lewin JM, Brauer JA, Ostad A. Surgical smoke and the dermatologist. J Am Acad Dermatol. 2011;65:636-641.
  15. Arefiev K, Warycha M, Whiting D, et al. Flammability of topical preparations and surgical dressings in cutaneous and laser surgery: a controlled simulation study. J Am Acad Dermatol. 2012;67:700-705.
  16. ASDS survey on dermatologic procedures. American Society for Dermatologic Surgery website. https://www.asds.net/Medical-Professionals/Practice-Resources/ASDS-Survey-on-Dermatologic-Procedures. Accessed October 4, 2019.
  17. Jalian HR, Jalian CA, Avram MM. Common causes of injury and legal action in laser surgery. JAMA Dermatol. 2013;149:188-193.
Issue
Cutis - 106(2)
Issue
Cutis - 106(2)
Page Number
87-92
Page Number
87-92
Publications
Publications
Topics
Article Type
Sections
Inside the Article

Practice Points

  • Laser therapy has evolved and expanded since its first cutaneous use in 1963.
  • The 4 regulatory agencies for laser safety in the United States establish standards and guidelines, but implementation is voluntary.
  • Ocular hazards, laser-generated airborne contaminants, fires, and unintended laser beam injuries constitute the main safety concerns.
  • Safety protocols with a laser checklist can reduce adverse outcomes.
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
Article PDF Media