topical treatment – Skin Therapy Letter https://www.skintherapyletter.com Written by Dermatologists for Dermatologists Thu, 13 Mar 2025 22:01:27 +0000 en-US hourly 1 https://wordpress.org/?v=6.8.1 Pediatric Hidradenitis Suppurativa: An Overview https://www.skintherapyletter.com/hidradenitis-suppurativa/pediatric-overview/ Mon, 20 Jan 2025 19:20:44 +0000 https://www.skintherapyletter.com/?p=15694 Jordanna Roesler, MD1; Allison Gregory, MD, FRCPC1,3; Wingfield Rehmus, MD, MPH1-3

1Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC, Canada
2Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
3Division of Dermatology, BC Children’s Hospital, Vancouver, BC, Canada

Conflicts of interest: The authors declare that there are no conflicts of interest.
Funding sources: None.

Abstract:
Hidradenitis suppurativa (HS) is a chronic, recurring inflammatory skin disease that significantly impacts the quality of life of patients.1 HS is more common in adults and adolescents, although true incidence rates may be underestimated due to a lack of earlier recognition of HS in children.2 Pediatric HS is a challenging clinical entity to diagnose and manage. Although considered uncommon, treatment of pediatric HS can drastically improve psychosocial well-being and should be considered in children presenting with recurring painful skin nodules, abscesses, scarring and sinus tracts. Multiple comorbidities are associated with pediatric HS, including depression, anxiety, inflammatory bowel disease, metabolic syndrome, and obesity.3 Medical management of pediatric HS poses a unique challenge given the paucity of literature surrounding efficacy and long-term treatment outcomes in pediatric patients. The purpose of this article is to discuss the epidemiology, pathogenesis, comorbidities, and management of pediatric HS.

Keywords: childhood hidradenitis, early onset hidradenitis suppurativa, hidradenitis suppurativa in children, inflammatory disorders, pediatric dermatology

Introduction

Hidradenitis suppurativa (HS) is a chronic disease involving the follicular unit that typically presents with inflammatory intertriginous lesions.4 Depending on severity, cutaneous involvement can manifest as painful nodules, abscesses, sinus tracts, and/or hypertrophic scarring.5 HS usually presents in adolescents and adults, and is considered uncommon in children, with an estimated prevalence of less than 2% in prepubescent children.6 A recent cross-sectional analysis reported 96.8% of pediatric patients with HS were ≥10 years old, with the highest prevalence reported in patients aged 15-17 years old.7 Some have noted that delays in care for pediatric patients may reflect an under-recognition of pediatric HS.4 In the adult population, women are more commonly affected by HS in comparison to men. Similarly, pediatric HS is more commonly reported in girls, although the exact prevalence is unknown.8 Unfortunately, most literature on pediatric HS is limited to small case series, case studies, or extrapolation from adult studies.9 More pediatric focused research is needed to better understand disease burden, prevalence, and treatment.

Pathogenesis

The pathogenesis of HS specific to pediatric patients is not well understood and primarily relies on extrapolation from basic sciences and adults with HS. HS pathophysiology is complex and involves environmental, immunologic, and genetic factors. HS is considered a disorder of follicular occlusion, in which hair follicle dysregulation and inflammation play key roles.10 As affected hair follicles become occluded and eventually rupture, bacteria and keratin enter the surrounding dermis, promoting an inflammatory state and subsequent lesion formation. Many patients with HS have a positive family history, which has prompted genetic studies.11 Gene mutations that alter antimicrobial peptides and cytokines have been demonstrated in patients with HS.12 Heterozygous mutations in gamma‐secretase (γ‐S), a protease involved in follicular keratinization regulation have been identified in autosomal dominant forms, supporting a genetic link.12,13 Gamma-secretase deficiencies have also been associated with impaired sebaceous gland formation and follicle disintegration in mice studies.14 Some research suggests that patients with early-onset HS appear more likely to have a positive family history.15 From an immunologic standpoint, both the innate and adaptive immune system play important roles. Decreased expression of antimicrobial peptides may facilitate superficial colonization by bacteria and promote ongoing inflammation through pro-inflammatory cytokines.16 Pro-inflammatory cytokines involved in HS include but are not limited to interleukin (IL)-1, IL-10, IL-17, IL-22, IL-23, and tumor necrosis factor (TNF)-alpha.9,16 Other factors that can promote HS pathogenesis and impact disease severity include microbial dysbiosis, microbial colonization, mechanical friction, and hormones.17 In addition, sinus tracts develop a psoriasiform lining, which tries to recapitulate the epidermis, shedding keratin and causing further inflammation. Hence, persistent lesions still exist despite systemic therapy and deroofing is often curative and essential to include in full-spectrum care.

Clinical Features and Diagnosis

Pediatric HS is a clinical diagnosis based on its typical morphology of deep nodules, cysts, sinus tracts, and fibrotic scars in intertriginous areas. A cross-sectional study assessing the clinical features of children <18 years old (mean age of 15.3 years) with HS reported a similar presenting clinical spectrum to adult-onset disease.18 Typical sites include those abundant with apocrine glands, such as the axillae, inframammary area, groin, and perianal region. Drainage from involved sites is a commonly reported symptom.19 There are currently no guidelines regarding investigations for HS in pediatric patients or adults. Laboratory investigations or skin biopsy are unnecessary for diagnosis, but imaging may be considered for operative planning when assessing sinus tracts.18 Ultimately, given the lack of research and consensus, there are currently no screening guidelines for investigating potential comorbidities in pediatric patients with HS. The Hurley staging system is often used to categorize patients into three disease groups based on their level of severity.20 Stage I includes abscess formation (single or multiple), without sinus tract(s) or scarring, Stage II includes those with recurrent abscesses with sinus tracts and scarring present, and Stage III encompasses diffuse involvement, with multiple abscesses and interconnected sinus tracts.20 The Sartorius scoring system is typically reserved for clinical trials and is not commonly used in clinical practice.8 Another useful scoring system is the International Hidradenitis Suppurativa Severity Score System (IHS4) which is a validated, dynamic assessment of HS severity that encompasses counting nodules, abscesses, and draining sinus tracts/fistulas.21 The Hidradenitis Suppurativa Quality Of Life (HiSQOL) scoring system may also be useful for capturing impactful areas of HS such as pain, odor, and drainage, which are not measured by the Dermatology Life Quality Index (DLQI) and should be considered by treatment providers.

Associated Comorbidities

Multiple comorbidities have been associated with pediatric HS, including more hormonal imbalances in comparison to adult populations, with manifestations including acne, premature adrenarche, adrenal hyperplasia, metabolic syndrome, and obesity.6 Although the overall association between early-onset HS and premature adrenarche and hormonal imbalance remains unclear, assessing for precocious puberty in children presenting with HS may be an important consideration depending on the clinical presentation. From a database of 870 pediatric patients, an elevated body mass index (BMI) and obesity were higher in comparison to reference population standards, as was the prevalence of smoking.18 Aside from metabolic syndrome, inflammatory bowel disease (IBD) and spondyloarthropathy have also been shown to be associated with HS.9 Patients with Down syndrome have been shown in multiple studies to have an earlier onset of HS although the mechanism behind this remains unknown.9 A detailed history, including inquiring about a family history of HS and associated comorbid symptoms and a physical examination should be completed. From a psychosocial perspective, HS can drastically impact quality of life and is associated with significant psychological distress.8 Painful, inflammatory lesions can limit children’s ability to play, exercise, or attend school which can contribute to obesity and further worsening of disease.6 Furthermore, social stigma surrounding HS can negatively affect psychosocial well-being, especially during the adolescent period. Overall, higher rates of anxiety and depression have been reported in pediatric-aged HS patients compared to those without HS.9 A cross-sectional study recently examined the quality of life impacts of HS in 25 pediatric patients aged 12-17 years of age.22 They found that 32% of patients had positive screening results for depression on the Patient Health Questionnaire-2, a depression screening tool.22 The Skindex-Teen questionnaire, an adolescent quality of life questionnaire for skin disease was also used, which demonstrated a higher average score in patients with more moderate-severe HS.22 Overall, clinicians should have a high level of suspicion for psychological comorbidities when treating pediatric patients with HS.

Treatment

Management of HS in the pediatric population is limited given the lack of information surrounding long-term outcomes. Determining the appropriate treatment involves weighing the biopsychosocial impact on the child, disease severity, and side effects of medications or procedures. In general, treatment of HS includes topical or systemic medications and surgical modalities depending on the severity. Lifestyle modifications are typically encouraged for all patients and include smoking cessation, weight management, and avoidance of triggers. Patient and family education should emphasize that HS is a chronic disease without a cure, with treatment focusing on disease and symptom management.

For Hurley Stage I disease, conservative management with topical treatment, such as clindamycin 1% solution, azelaic acid 15%, resorcinol 15%, or combination treatment with clindamycin/ benzoyl peroxide is recommended.6 Of note, resorcinol is the only topical treatment with studies completed for HS in adults and is a medication that must be compounded. Topical antiseptics and clindamycin are considered safe for use but may be ineffective for more moderate or severe HS.23 For non-prescription treatments, laser hair removal has been effective via the Hidradenitis Suppurativa Clinical Response (HiSCR) response in patients with mild-to-moderate disease.24 Supplementation with 100 mg of oral zinc has also been shown to improve HS.25 Concurrent supplementation with 4 mg of copper should be considered to prevent copper deficiency.25 For those where topical treatments fail or children with Hurley Stage II disease, systemic medications can be explored. Systemic antibiotics such as doxycycline, clindamycin with rifampicin, metronidazole, and erythromycin are appropriate for use in children with more severe disease.6 Counselling regarding potential tooth discoloration and enamel hypoplasia should be done for patients under 8 years old receiving tetracycline antibiotics.23 However, antibiotics are not a feasible long-term solution. If there is recurrence after treatment, adalimumab or secukinumab should be considered. Oral finasteride demonstrated improvement in resistant cases from a small pediatric case series, however potential side effects include transient sexual dysfunction in males, and pediatric safety data is lacking, particularly for prepubertal males.26 Systemic retinoids used for the treatment of HS include acitretin and isotretinoin, although these have considerable risks and isotretinoin tends to be more effective in milder, folliculocentric subtypes. The long-lasting teratogenic effects of acitretin make it unsuitable for patients with childbearing potential and isotretinoin in children under 12 years of age has been reported to cause premature epiphyseal closure.27 Importantly, all patients of childbearing potential should be counselled surrounding teratogenic effects where applicable.

In terms of biologics, adalimumab is currently the only approved choice in North America for pediatric patients older than 12 years of age who weigh at least 30 kg.28 Safety data surrounding the use of adalimumab in pediatric patients for HS is limited, although adalimumab has been used effectively in pediatric patients for other inflammatory diseases including Crohn’s disease, psoriasis, and juvenile idiopathic arthritis.29 Secukinumab, an IL-17 inhibitor, is both Health Canada and US FDA approved for treatment of adults with moderate-to-severe HS. Based on clinical studies in adults, it may be a therapeutic option for first- or second-line off-label treatment of pediatric HS patients.30,31 Overall, dermatologists should have a low threshold to treat systemically and preventatively, as HS is typically a progressive disease that can become less responsive to biologic therapy as time passes and severity increases. Surgical modalities may be another option for older children. Depending on the extent of disease, wide excision and/or minimally invasive deroofing can be considered. A recent cross-sectional study found that surgical excision and deroofing were reported as useful for all 23 pediatric patients assessed, while those treated with simple excision had zero responders in 7 cases treated with simple excision.32 However, a surgical approach is more invasive and carries the risk of infection, scarring, and recurrence.9 A retrospective review of 11 patients under 18 years old with a total of 23 operative sites reported an overall complication rate of 87% and a 7% reoperation rate.33 Remission after a single procedure was reported in 57% of included sites.33 However, it is crucial to combine both medical preventative treatments with surgical therapy, as success rates are much higher with a combination approach.

Conclusion

Pediatric HS is an understudied and underrecognized disease with significant biopsychosocial impacts. Unfortunately, diagnosis is often delayed given the wide variety of presentations in early disease. Clinicians should consider associated comorbidities such as metabolic syndrome, inflammatory bowel disease, and anxiety and depression. Early recognition, diagnosis, and management are essential in improving quality of life and managing symptoms for children and adolescents with HS. Further research focused on long-term outcomes, associated comorbidities, and medical management is needed to improve our understanding and treatment of pediatric hidradenitis suppurativa.

References





    1. van Straalen KR, Prens EP, Gudjonsson JE. Insights into hidradenitis suppurativa. J Allergy Clin Immunol. 2022 Apr;149(4):1150-61.

    2. Seivright J, Collier E, Grogan T, et al. Pediatric hidradenitis suppurativa: epidemiology, disease presentation, and treatments. J Dermatolog Treat. 2022 Jun;33(4):2391-3.

    3. Tiri H, Jokelainen J, Timonen M, et al. Somatic and psychiatric comorbidities of hidradenitis suppurativa in children and adolescents. J Am Acad Dermatol. 2018 Sep;79(3):514-9.

    4. Liy-Wong C, Kim M, Kirkorian AY, et al. Hidradenitis suppurativa in the pediatric population: an international, multicenter, retrospective, cross-sectional study of 481 pediatric patients. JAMA Dermatol. 2021 Apr 1;157(4):385-91.

    5. Revuz J. Hidradenitis suppurativa. J Eur Acad Dermatol Venereol. 2009 Sep; 23(9):985-98.

    6. Liy-Wong C, Pope E, Lara-Corrales I. Hidradenitis suppurativa in the pediatric population. J Am Acad Dermatol. 2015 Nov;73(5 Suppl 1):S36-41.

    7. Garg A, Wertenteil S, Baltz R, et al. Prevalence estimates for hidradenitis suppurativa among children and adolescents in the United States: a gender- and age-adjusted population analysis. J Invest Dermatol. 2018 Oct;138(10):2152-6.

    8. Scheinfeld N. Hidradenitis suppurativa in prepubescent and pubescent children. Clin Dermatol. 2015 May-Jun;33(3):316-9.

    9. Choi E, Ooi XT, Chandran NS. Hidradenitis suppurativa in pediatric patients. J Am Acad Dermatol. 2022 Jan;86(1):140-7.

    10. Vinkel C, Thomsen SF. Hidradenitis suppurativa: causes, features, and current treatments. J Clin Aesthet Dermatol. 2018 Oct;11(10):17-23.

    11. Ingram JR. The genetics of hidradenitis suppurativa. Dermatol Clin. 2016 Jan;34(1):23-8.

    12. Duchatelet S, Miskinyte S, Delage M, et al. Low prevalence of GSC gene mutations in a large cohort of predominantly Caucasian patients with hidradenitis suppurativa. J Invest Dermatol. 2020 Oct;140(10):2085-8.

    13. Pink AE, Simpson MA, Desai N, et al. γ-Secretase mutations in hidradenitis suppurativa: new insights into disease pathogenesis. J Invest Dermatol. 2013 Mar;133(3):601-7.

    14. Pan Y, Lin MH, Tian X, et al. gamma-secretase functions through Notch signaling to maintain skin appendages but is not required for their patterning or initial morphogenesis. Dev Cell. 2004 Nov;7(5):731-43.

    15. Deckers IE, van der Zee HH, Boer J, et al. Correlation of early-onset hidradenitis suppurativa with stronger genetic susceptibility and more widespread involvement. J Am Acad Dermatol. 2015 Mar;72(3):485-8.

    16. Kelly G, Sweeney CM, Tobin AM, et al. Hidradenitis suppurativa: the role of immune dysregulation. Int J Dermatol. 2014 Oct;53(10):1186-96.

    17. Frew JW, Hawkes JE, Krueger JG. A systematic review and critical evaluation of inflammatory cytokine associations in hidradenitis suppurativa. F1000Res. 2018 Dec 13;7:1930.

    18. Garcovich S, Fania L, Caposiena D, et al. Pediatric hidradenitis suppurativa: a cross-sectional study on clinical features and treatment approaches. J Cutan Med Surg. 2022 Mar-Apr;26(2):127-34.

    19. Ballard K, Shuman VL. Hidradenitis suppurativa. 2024 May 6. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan. PMID: 30521288.

    20. Vazquez BG, Alikhan A, Weaver AL, et al. Incidence of hidradenitis suppurativa and associated factors: a population-based study of Olmsted County, Minnesota. J Invest Dermatol. 2013 Jan;133(1):97-103.

    21. Zouboulis CC, Prens EP, Sayed CJ, et al. International Hidradenitis Suppurativa Severity Scoring System (IHS4) as a holistic measure of hidradenitis suppurativa disease severity compared with Hurley staging: a post hoc analysis of the SUNRISE and SUNSHINE phase 3 trials of secukinumab. J Eur Acad Dermatol Venereol. 2024 Jun;38(6):e496-9.

    22. McAndrew R, Lopes FCPS, Sebastian K, et al. Quality of life in hidradenitis suppurativa: a cross-sectional study of a pediatric population. J Am Acad Dermatol. 2021 Mar;84(3):829-30.

    23. Goldburg SR, Strober BE, Payette MJ. Hidradenitis suppurativa: current and emerging treatments. J Am Acad Dermatol. 2020 May;82(5):1061-82.

    24. Fabbrocini G, França K, Lotti T, et al. Intralesional diode laser 1064 nm for the treatment of hidradenitis suppurativa: a report of twenty patients. Open Access Maced J Med Sci. 2018 Jan 7;6(1):31-4.

    25. Johnston LA, Alhusayen R, Bourcier M, et al. Practical guidelines for managing patients with hidradenitis suppurativa: an update. J Cutan Med Surg. 2022 Sep-Oct;26(2_suppl):2S-24S.

    26. Randhawa HK, Hamilton J, Pope E. Finasteride for the treatment of hidradenitis suppurativa in children and adolescents. JAMA Dermatol. 2013 Jun;149(6):732-5.

    27. Luthi F, Eggel Y, Theumann N. Premature epiphyseal closure in an adolescent treated by retinoids for acne: an unusual cause of anterior knee pain. Joint Bone Spine. 2012 May;79(3):314-6.

    28. Sachdeva M, Kim P, Mufti A, et al. Biologic use in pediatric patients with hidradenitis suppurativa: a systematic review. J Cutan Med Surg. 2022 Mar-Apr;26(2):176-80.

    29. Horneff G, Seyger MMB, Arikan D, et al. Safety of adalimumab in pediatric patients with polyarticular juvenile idiopathic arthritis, enthesitis-related arthritis, psoriasis, and crohn’s disease. J Pediatr. 2018 Oct;201:166-75.

    30. Chung CS, Park SE, Hsiao JL, et al. A review of hidradenitis suppurativa in special populations: considerations in children, pregnant and breastfeeding women, and the elderly. Dermatol Ther (Heidelb). 2024 Sep;14(9):2407-25.

    31. Kimball AB, Jemec GBE, Alavi A, et al. Secukinumab in moderate-to-severe hidradenitis suppurativa (SUNSHINE and SUNRISE): week 16 and week 52 results of two identical, multicentre, randomised, placebo-controlled, double-blind phase 3 trials. Lancet. 2023 Mar 4;401(10378):747-61. Erratum in: Lancet. 2024 Feb 17;403(10427):618.

    32. Riis PT, Saunte DM, Sigsgaard V, et al. Clinical characteristics of pediatric hidradenitis suppurativa: a cross-sectional multicenter study of 140 patients. Arch Dermatol Res. 2020 Dec;312(10):715-24.

    33. Ge S, Ngaage LM, Orbay H, et al. Surgical management of pediatric hidradenitis suppurativa: a case series and review of the literature. Ann Plast Surg. 2020 May;84(5):570-4.




Purchase Article PDF for $1.99

]]>
A Practical Guide to Advanced Topical Drug Delivery Systems in Dermatology https://www.skintherapyletter.com/dermatology/topical-drug-delivery-guide/ Fri, 22 Sep 2023 15:41:29 +0000 https://www.skintherapyletter.com/?p=14785 Andrei Metelitsa, MD, FRCPC1,2; Isabelle Delorme, MD3; Daniel O’Sullivan, MPharm4; Rami Zeinab, PhD4; Mark Legault, PhD4; Melinda Gooderham, MD, FRCPC5,6

1Beacon Dermatology, Calgary, AB, Canada
2Division of Dermatology, University of Calgary, Calgary, AB, Canada
3Dermatologie Clinic, Drummondville, QC, Canada
4Bausch Health Canada Inc, Laval, QC, Canada
5SKiN Centre for Dermatology, Peterborough, ON, Canada
6Queen’s University, Kingston, ON, Canada

Conflict of interest: The authors declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article:
Isabelle Delorme has been a member of advisory boards for AbbVie, Bausch Health, Eli-Lilly, Janssen, Novartis, Sanofi-Genzyme. She has been a part of clinical trials for AbbVie, Amgen, Anaptys Bio, Arcutis, Bausch Health, BMS, Celgene, Dermira, Devonian, Eli-Lilly, Galderma, Glenmark Pharmaceutical, Innovaderm Research, Janssen, Leo Pharma, Novartis, Regeneron. She has recieved honoraria from AbbVie, Amgen, Avene, Celgene, Eli-Lilly, Janssen, Novartis, UCB Pharma. She has been part of speaker’s bureaus for AbbVie, Celgene, Bausch Health, Eli-Lilly, Janssen, Medexus Inc., Novartis, Sanofi Genzyme. Melinda Gooderham has been an investigator, speaker and/or advisor for – AbbVie, Amgen, Akros, Arcutis, Aslan, Bausch Health, BMS, Boehringer Ingelheim, Celgene, Dermira, Dermavant, Eli Lilly, Galderma, GSK, Incyte, Janssen, Kyowa Kirin, Leo Pharma, MedImmune, Merck, Novartis, Pfizer, Regeneron, Roche, Sanofi Genzyme, Sun Pharma, and UCB. Andrei Metelitsa has been a consultant for Bausch Health, Galderma, Leo Pharma and Pfizer.
Daniel O’Sullivan, Rami Zeinab and Mark Legault are employees of Bausch Health Canada.

Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article. This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Abstract:
Dermatological diseases such as atopic dermatitis, acne, and psoriasis result in significant morbidity and decreased quality of life. The first line of treatment for such diseases is often topical medications. While topical delivery allows active drug to be delivered directly to the target site, the skin is a virtually impermeable barrier that impedes delivery of large molecules. Thus, the formulation and delivery system are integral elements of topical medications. Patients also have preferences for the properties of topical formulations and these preferences can positively or negatively impact adherence. Therefore, the choice of topical formulation is a key consideration. Recent developments in drug delivery systems have produced enhanced topical treatments that improve efficacy, safety, and patient acceptability. Awareness of the delivery system in which drugs are formulated is critical as this can have profound implications on treatment success. This paper provides an overview and clinical commentary on advances in topical delivery systems and their impact on dermatological practice.

Keywords: acne, corticosteroid, dermatitis, dermatology, halobetasol, psoriasis, retinoid, tazarotene, topical, vehicle, Duobrii, Arazlo, Bryhali


Introduction

Topical therapies are often the first line of treatment for common skin diseases such as atopic dermatitis (AD), acne vulgaris (AV), and psoriasis (PsO).1 Prescribing effective therapy is crucial for optimal treatment outcomes.2 However, the composition of topical treatments is complex and should be taken into consideration when choosing a topical treatment in terms of both “formulation” and “drug delivery system” wherein the formulation includes the active drug and vehicle while the delivery system refers to technologies affecting the therapeutic action of the drug (including potency, stability, dispersion, and penetration). Both formulation and delivery system should be considered for each patient on the basis of the potency of the active molecule, patient preference, skin type, skin condition, and affected site.3 Several recent reviews have discussed topical vehicles, ingredients, and their effects on the skin,4-10 however, there is a gap in reviewing advanced delivery systems currently in clinical use. This paper provides an up-to-date review of advances in topical delivery systems with a focus on those in clinical use for the treatment of AD, AV, and PsO.

Atopic Dermatitis, Acne Vulgaris and Psoriasis

Skin conditions are a common cause of disease globally,11 with AD, AV, and PsO amongst those with the highest disease burden on patients12. These conditions disrupt the normal skin function,13 trigger abnormal inflammatory responses, alter skin microbiome, and increase susceptibility to irritants/allergens.5-7 AD is a chronic, relapsing disease, presenting as inflamed skin with intense itching.14,15 AV is due to inflammation of the sebaceous follicles, resulting in increased sebum production and a favorable environment for bacterial growth.16 PsO is a chronic disease that presents with characteristic scaling, redness, and thickening of the skin. All of these skin diseases can have important detrimental effects on short and long-term psychosocial and physical health, with substantial morbidity and reduced quality of life.15,17,18 As such, early treatment with the most efficacious therapies available is critical.

Factors to Consider with Topical Drug Delivery Systems

Applying treatment topically provides several advantages for managing skin diseases. It enables delivery of drug directly to the target site, minimizing systemic exposure.19 The skin provides for a regulated, constant delivery rate and circumvents first-pass metabolism.20 However, delivering drugs to targets beneath the epidermis can be challenging.21 The skin is a multi-layered defense barrier constructed to withstand the penetration of external compounds.22 The stratum corneum is very effective in performing this role and minimizes the ingress and egress of molecules/ chemicals.23 As a result, the skin is impermeable to almost all compounds with a molecular weight greater than 500 Daltons.22,23 As diffusion is the principal mechanism by which a drug penetrates the skin,22 formulations must be optimized to maintain solubility, stability, dispersion, and penetration of the active ingredient(s) while ensuring they are sustained at the target site for sufficient time, as well as limit systemic exposure3.

The effectiveness of a topical medication depends not only on pharmacodynamic factors but also on patient preferences, adherence, and the location of disease.2,24 Patient preferences are related to the formulation and drug delivery system whose attributes include moisturization, absorption, greasiness, stickiness, as well as ease and frequency of use.25 These attributes influence adherence to the therapeutic regimen which is a major determinant of treatment success.25 Teixeira et al.26 reported that adherence was impacted not only by patient preferences, but also the body area affected. For example, adherence was higher for patients using gel/cream vs. ointment formulations when the body area affected was large. However, this was reversed with smaller areas. As such, delivery systems are no longer only considered ‘carriers’ of active ingredients, but also integral to the formulation. Hence, drug delivery system research and development have been the main drivers of recent advancements in topical formulations.

Advanced Topical Drug Delivery Systems

Over the years, advanced drug delivery systems have been developed to enhance effectiveness, tolerability, safety, and patient acceptability of topical formulations. Some of these advanced delivery systems are integrated into approved products utilized in clinical practice in Canada (Figures 1 & 2). As such, it is important for healthcare providers to be aware of these systems and their profound implications on treatment outcomes. The following summary provides an overview of some recent advances in drug delivery systems and their clinical application (Table 1).

A Practical Guide to Advanced Topical Drug Delivery Systems in Dermatology - image
Figure 1. Advanced topical drug delivery systems in use in dermatology
API, Active pharmaceutical ingredient
A Practical Guide to Advanced Topical Drug Delivery Systems in Dermatology - image
Figure 2. Timeline of approved advanced topical drug delivery systems in the USA and Canada
AD, atopic dermatitis; AV, acne vulgaris; PET, Polymeric emulsion technology; PsO, psoriasis.
Note: Timeline is based on the Food and Drug Administration (USA) and Health Canada (Canada) approval for the specific product.

Table 1. Advanced topical drug delivery systems in clinical use

Delivery System Description Mechanism of Action Features Products (Disease)
Microencapsulation

Microspheres with a coating of inert, natural, or synthetic polymeric materials around solid or liquid micronized drug particles29

Enhances the stability of the API and allows for controlled release27

  • Protects drug from degradation27
  • Increases penetration of the epidermis by API27
  • Allows targeted delivery of the API27
  • Allows controlled release of API over time
  • Minimizes systemic and local side effects27
  • Enhances drug stability and shelf life28
  • Twyneo® (AV)
  • Amzeeq® (AV)
Microsponges

Uniform, spherical, and porous polymeric delivery system37

Release API onto skin surface in a controlled manner in response to rubbing, elevated temperature or changes in pH39

  • Reduces irritation, mutagenicity and allergenicity39
  • Reduces skin oiliness through sebum absorption5
  • Produces an elegant, patientacceptable formulations39
  • Stable over a range of pHs (1-11) and temperatures (up to 130C)71
  • Compatible with most vehicles and ingredients and self-sterilizing71
  • Size (5-300 μm) limits passage through the stratum corneum39
  • Retin-A Micro® (AV)
Polymeric emulsion technology / Prismatrex™

Particles, moisturizers and hydrating ingredients are encapsulated together within the same oil droplets that are evenly distributed throughout a 3D mesh matrix52

Penetrate through stratum corneum and accumulate in hair follicles13

  • More rapid, controlled, and even release of oil droplets, humectants and excipients from vehicle onto the skin5
  • Provides an occlusive layer to prevent moisture loss and improve skin hydration52
  • Allows more efficient delivery of the API into dermal layers5
  • Improved tolerability and reduced incidence of side effects52
  • Allows for fixed dose combinations, reducing frequency of application and increasing patient adherence5
  • Higher patient acceptance than with previous formulations52
  • Arazlo® (AV)
  • Bryhali® (corticosteroid responsive dermatoses/PsO)
  • Duobrii® (PsO)

Abbreviations: API, Active pharmaceutical ingredient; AD, atopic dermatitis; AV, acne vulgaris; PsO, psoriasis

Microencapsulation

Description of Delivery System

Microencapsulation is a technique that involves entrapping active ingredient(s) in a microcapsule, creating a barrier between the ingredients and the skin.27 Once applied, microcapsules allow controlled release of active ingredients over time.28 This offers advantages for topical delivery of agents (Table 1) where active ingredients are protected against degradation and maintained at the target site for an extended time while limiting local adverse reactions.28,29

Clinical Application Highlights

Encapsulation is the technology behind two topical treatments approved for treatment of moderate to severe AV in the US. The first formulation is a foam-based delivery system that optimizes the topical delivery of minocycline (minocycline topical foam 4%, Amzeeq®).30 Minocycline was previously not available topically due to its instability and systemic side effects.31 Such limitations were mitigated with microencapsulation. In phase 232,33 and 334 clinical trials, microencapsulated minocycline significantly reduced AV lesions and severity compared to placebo; with minimal adverse effects. The second is a cream-based formulation combining tretinoin 0.1% and benzoyl peroxide (BPO) 3% (Twyneo®).35 Tretinoin and BPO are individually entrapped within silica-based microcapsules preventing the degradation of tretinoin by BPO.28 The active ingredients are released over time, providing a consistent drug concentration at the affected site.36 Results from clinical trials show that patients who received this encapsulated combination therapy had improved treatment outcomes compared with patients who received vehicle only. The treatment was well tolerated.36

Microsponges

Description of Delivery System

Microsponges are microscopic, uniform, spherical, porous delivery systems.37 Some features of micropsonges are summarized in Table 1. Their large surface area allows a range of substances to be incorporated into gels, creams, liquids, or powders.38 They can absorb skin secretions, therefore reducing the oiliness of the skin.38,39 When applied, the release of drug is controlled through diffusion, rubbing, moisture, pH, friction, or ambient skin temperature, producing a controlled release and reducing side effects.40 Microsponge polymers possess the ability to load a spectrum of active ingredients and provide the benefits of enhanced effectiveness, mildness, and tolerability to a wide range of skin therapies.41 The microsponge system is stable over a range of pH and temperatures, compatible with most vehicles and ingredients, self-sterilizing as average pore size prevents bacterial penetration, and has a higher payload42 (50-60%) vs. conventional topical drugs and microencapsulation. However, due to their size (5-300 μm), passage through the stratum corneum is limited.39

Clinical Application Highlights

Early topical formulations of tretinoin had high concentrations of active ingredients in alcohol-based solutions leading to skin dryness and irritation.43,44 The innovation of microsponges led to the first alcohol-free, topical retinoid delivery system (tretinoin gel, Retin-A Micro®).45 This microsponge gel contains tretinoin in concentrations of 0.04%, 0.06%, 0.08%, and 0.1%. Tretinoin is entrapped in patented methyl methacrylate/glycol dimethacrylate copolymer porous microspheres (Microsponges® system), within a carbomer-based gel. In the vehicle-controlled clinical trials, tretinoin gel was significantly more effective than vehicle in reducing the severity of acne lesions; and significantly superior to vehicle in the investigator’s global evaluation of the clinical response.46-48 Microsponge delivery of tretinoin has also been associated with decreased irritation compared to earlier alcoholbased cream (Retin-A® cream, 0.1%).49

Polymeric Emulsion Technology

Description of Delivery System

In polymeric emulsion technology (PET), active ingredients are encapsulated within oil droplets, together with moisturizing/ hydrating ingredients (light mineral oil, diethyl sebacate).50 The oil droplets are uniformly dispersed within an oil-in-water emulsion and separated by a three-dimensional mesh matrix.51 Recently, PET has been combined with optimized selection of excipients and emollients to produce a patented delivery system called Prismatrex™. This novel technology allows simultaneous and uniform dispersion of active ingredients onto the skin at lower doses than conventional formulations to achieve comparable therapeutic effect, while providing enhanced hydration and moisturization.52 Also, the technology allows many attributes of patient preferences to be met. By combining water-soluble moisturizing components within the matrix, a lotion can have a pleasant feel; the use of moisturizers over alcohol-based components creates a lowirritancy/ low-drying formulation; and the mesh network breaking down upon contact with the salts on the skin surface results in quick release and absorption, leaving behind minimal greasy or sticky residue.50,51

Clinical Application Highlights

Prismatrex™ is used in recent formulations containing retinoids, corticosteroids, or combination of both. Topical corticosteroids are a mainstay in the treatment of AD and PsO. Due to the defective skin barrier in AD and PsO, formulations with moisturizing effects in a patient-preferred format offer additional therapeutic advantages over previous formulations.53 Halobetasol propionate (HP) 0.01% lotion (Bryhali®), formulated with Prismatrex™, provides several improvements compared to the older 0.05% cream formulation. For example, a reduced concentration of drug while maintaining comparable efficacy to the cream formulation, and a safety profile that allows extended use up to 8 weeks.54 Furthermore, the formulation is non-greasy and aesthetically pleasing, providing a patient preferred treatment option.55

Topical retinoids are a cornerstone in the treatment of AV. However, dryness, erythema, and peeling are important side effects that can impact treatment adherence.56 Prismatrex™ has been used to formulate the third-generation retinoid, tazarotene (TAZ) 0.045% lotion (Arazlo®). The small particle size with this formulation allows better access to the pilosebaceous unit.57 Also, this novel technology allows for uniform distribution of TAZ on the skin along with moisturizing ingredients and emollients, therefore, TAZ may be delivered at a lower and potentially less irritating concentration than the previous 0.1% cream formulation.58 Two identical phase 3, vehicle-controlled studies demonstrated statistically superior efficacy for TAZ 0.045% lotion vs. placebo in once-daily treatment of moderate to severe AV.59

A fixed-combination lotion containing both HP 0.01% and TAZ 0.045% (Duobrii®, HP/TAZ) is formulated using Prismatrex™ technology and indicated for PsO treatment. Higher tissue permeation efficiency of both HP (vs. HP 0.05% cream) and TAZ (vs. TAZ 0.1% cream, Tazorac®) has been achieved with the combination using Prismatrex™ technology.50 The delivery of both active ingredients with anti-inflammatory and anti-proliferative properties into a lotion confers a synergistic effect.50,52 Also, it has demonstrated improved efficacy, tolerability, and maintenance of therapeutic effect compared with monotherapy with either of the active ingredients.50,52,60-62 HP/TAZ’s acute and long-term efficacy (including maintenance of efficacy after cessation of treatment), tolerability, and safety have been demonstrated in phase 2 and 3 vehicle-controlled trials and an open-label extension study.63 Importantly, HP/TAZ is associated with low incidences of adverse events that may be of concern with corticosteroid and retinoid monotherapy.64 Skin atrophy occurs in up to 5% of patients treated with topical corticosteroids.64 In HP/TAZ clinical trials, incidences of skin atrophy were rare and generally resolved by the end of the study despite 8 weeks of daily application.63-64 This may be because TAZ increases the number and activity of dermal fibroblasts and stimulates collagen and elastin production.65 Irritation, pain, and retinoid dermatitis may be a challenge with retinoid monotherapy. Incidences of these AEs were lower with HP/TAZ, than with TAZ monotherapy, potentially due to the anti-inflammatory properties of HP.64 Clinical trials have also shown that the majority of participants who achieve clear skin with HP/TAZ, experience prolonged maintenance of therapeutic effect after treatment cessation.60 Taken together, HP/TAZ provides a treatment option for PsO that is efficacious, safe, combines the beneficial effects of corticosteroids and retinoids, delivers moisturizing and hydrating ingredients, yields longer remission, and is acceptable to patients.

Real World Clinical Commentary

A challenge frequently encountered by dermatologists when managing AD, AV, and PsO is balancing sustained disease suppression with the avoidance of local/systemic side effects. To address this concern, advanced drug delivery systems, such as Primsmatrex™, allow for more efficient delivery of lower doses of active ingredients without compromising efficacy. However, this must also be balanced with overall patient acceptance. Patients consistently cite the following preferences for treatment options: non-greasy applications, reduced skin irritation, spreadable and easily absorbed,36 easily applied in certain body sites (e.g., scalp); easy to wash off with minimal residue, and fast skin responses.66 Novel delivery systems often enhance utilization of existing ingredients making them more acceptable to patients, resulting in therapeutic advances. For example, in acne, the innovative drug delivery systems used in Retin-A Micro®, Arazlo® and Twyneo® have significantly improved tolerability of existing topical retinoid ingredients without sacrificing the overall efficacy of these molecules. While patients are advised to maintain a dosing schedule of every second day during the initial phases of treatment to avoid potential irritation, these agents are well tolerated given the nature of the formulations.

The microencapsulation of minocycline foam allows delivery in a topical formulation as opposed to an oral format, which significantly enhances the safety profile. In psoriasis, where availability of topical agents is quite limited, the Prismatrex™ technology in HP/TAZ provides an excellent new treatment option which combines two effective drugs and is a welcome addition to our therapeutic algorithm. In clinical practice, this formulation is effective, can be less irritating to the skin than TAZ alone and is less likely to induce steroid-induced atrophy as compared to HP alone. Another advantage to these advanced topicals is that they result in improved adherence and therefore better treatment outcomes in the long run. Overall, when choosing the optimal therapeutic agent for treatment of AD, AV, or PsO the dermatologist now has the option to select a formulation that uses an advanced drug delivery system to maximize the chances of achieving successful treatment outcomes. Research in advanced delivery systems is vast, with a wide range of technologies showing promise for future clinical use, such as nanoparticles67, ethosomes68, niosomes69, and liposomes70.

Conclusion

Topical treatment is a cornerstone in managing AD, AV, and PsO;19 however, the delivery of active drugs through the dermal barrier remains a challenge. Furthermore, patient acceptability is a major contributing factor to the effectiveness and adherence to topical treatments. Advances in topical drug formulations and delivery systems address many limitations seen with older formulations. These advances allow for efficient and uniform delivery of active ingredients to target sites, greater patient acceptability, and enhanced treatment outcomes.

Acknowledgements

Medical writing assistance for this manuscript was provided by KTP (Knowledge Translation Partners), Montreal, Canada, funded by Bausch Health Canada.

References



  1. Nguyen T, Zuniga R. Skin conditions: new drugs for managing skin disorders. FP Essent. 2013 Apr;407:11-6. PMID: 23600334.

  2. Wiechers JW. The barrier function of the skin in relation to percutaneous absorption of drugs. Pharm Weekbl Sci. 1989 Dec 15;11(6):185-98.

  3. Rosen J, Landriscina A, Friedman AJ. Principles and approaches for optimizing therapy with unique topical vehicles. J Drugs Dermatol. 2014 Dec;13(12): 1431-5.

  4. Barnes TM, Mijaljica D, Townley JP, et al. Vehicles for drug delivery and cosmetic moisturizers: review and comparison. Pharmaceutics. 2021 Nov 26; 13(12):2012.

  5. Baldwin H, Webster G, Stein Gold L, et al. 50 years of topical retinoids for acne: evolution of treatment. Am J Clin Dermatol. 2021 May;22(3):315-27.

  6. Danby SG, Draelos ZD, Gold LFS, et al. Vehicles for atopic dermatitis therapies: more than just a placebo. J Dermatolog Treat. 2022 Mar;33(2):685-98.

  7. Segaert S, Calzavara-Pinton P, de la Cueva P, et al. Long-term topical management of psoriasis: the road ahead. J Dermatolog Treat. 2022 Feb; 33(1):111-20.

  8. Mayba JN, Gooderham MJ. A guide to topical vehicle formulations. J Cutan Med Surg. 2018 Mar/Apr;22(2):207-12.

  9. Ramadon D, McCrudden MTC, Courtenay AJ, et al. Enhancement strategies for transdermal drug delivery systems: current trends and applications. Drug Deliv Transl Res. 2022 Apr;12(4):758-91.

  10. Praestegaard M, Steele F, Crutchley N. Polyaphron dispersion technology, a novel topical formulation and delivery system combining drug penetration, local tolerability and convenience of application. Dermatol Ther (Heidelb). 2022 Oct;12(10):2217-31.

  11. Flohr C, Hay R. Putting the burden of skin diseases on the global map. Br J Dermatol. 2021 Feb;184(2):189-90.

  12. Pulsipher KJ, Szeto MD, Rundle CW, et al. Global burden of skin disease representation in the literature: bibliometric analysis. JMIR Dermatol. 2021 Aug 31;4(2):e29282.

  13. Makhmalzade BS, Chavoshy F. Polymeric micelles as cutaneous drug delivery system in normal skin and dermatological disorders. J Adv Pharm Technol Res. 2018 Jan-Mar;9(1):2-8.

  14. Ramos Campos EV, Proença PLF, Doretto-Silva L, et al. Trends in nanoformulations for atopic dermatitis treatment. Expert Opin Drug Deliv. 2020 Nov;17(11):1615-30.

  15. Drucker AM. Making sense of measures of clinical signs for atopic dermatitis. Br J Dermatol. 2017 Nov;177(5):1158-9.

  16. Fabbrocini G, Rossi AB, Thouvenin MD, et al. Fragility of epidermis: acne and post-procedure lesional skin. J Eur Acad Dermatol Venereol. 2017 Sep; 31 Suppl 6:3-18.

  17. Layton AM, Thiboutot D, Tan J. Reviewing the global burden of acne: how could we improve care to reduce the burden? Br J Dermatol. 2021 Feb;184(2):219-25.

  18. World Health Organization. Global report on psoriasis. World Health Organization. 2016. Available from: https://apps.who.int/iris/handle/10665/204417. Accessed August 1, 2023.

  19. Nolan BV, Feldman SR. Dermatologic medication adherence. Dermatol Clin. 2009 Apr;27(2):113-20.

  20. Mohammed YH, Moghimi HR, Yousef SA, et al. Efficacy, safety and targets in topical and transdermal active and excipient delivery. Percutaneous Penetration Enhancers Drug Penetration Into/Through the Skin. 2017 Jan 25:369-91. Available from: doi: 10.1007/978-3-662-53270-6_23.

  21. Lane ME, Santos P, Watkinson AC, et al. Passive skin permeation enhancement. In: Topical and transdermal drug delivery; John Wiley & Sons: Hoboken, NJ, USA. 2012; pp.23-42.

  22. Bos JD, Meinardi MM. The 500 Dalton rule for the skin penetration of chemical compounds and drugs. Exp Dermatol. 2000 Jun;9(3):165-9.

  23. Del Rosso JQ, Levin J. The clinical relevance of maintaining the functional integrity of the stratum corneum in both healthy and disease-affected skin. J Clin Aesthet Dermatol. 2011 Sep;4(9):22-42.

  24. Brown KK, Rehmus WE, Kimball AB. Determining the relative importance of patient motivations for nonadherence to topical corticosteroid therapy in psoriasis. J Am Acad Dermatol. 2006 Oct;55(4):607-13.

  25. Eastman WJ, Malahias S, Delconte J, et al. Assessing attributes of topical vehicles for the treatment of acne, atopic dermatitis, and plaque psoriasis. Cutis. 2014 Jul;94(1):46-53.

  26. Teixeira A, Teixeira M, Almeida V, et al. Does the vehicle matter? Real-world evidence on adherence to topical treatment in psoriasis. Pharmaceutics. 2021 Sep 23;13(10):1539.

  27. Casanova F, Santos L. Encapsulation of cosmetic active ingredients for topical application–a review. J Microencapsul. 2016 Feb;33(1):1-17.

  28. Green LJ, Lain E, Prunty T, et al. Enhancing topical pharmacotherapy for acne and rosacea: vehicle choices and outcomes. J Clin Aesthet Dermatol. 2022 May;15(5):36-40.

  29. Ciriminna R, Sciortino M, Alonzo G, et al. From molecules to systems: sol-gel microencapsulation in silica-based materials. Chem Rev. 2011 Feb 9;111(2):765-89.

  30. AMZEEQ® (minocycline) topical foam [prescribing information]. Revised: January 2021. Vyne Pharmaceuticals Inc., Bridgewater, NJ. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/212379s003lbl.pdf. Accessed August 1, 2023.

  31. Foamix Pharmaceuticals. Press release dated October 18, 2019. Foamix receives FDA approval of Amzeeq™ topical minocycline treatment for millions of moderate to severe acne sufferers. Available from: https://www.prnewswire.com/news-releases/foamix-receives-fda-approval-of-amzeeqtopical-minocycline-treatment-for-millions-of-moderate-to-severe-acnesufferers-300941412.html. Accessed August 1, 2023

  32. Shemer A, Shiri J, Mashiah J, et al. Topical minocycline foam for moderate to severe acne vulgaris: phase 2 randomized double-blind, vehicle-controlled study results. J Am Acad Dermatol. 2016 Jun;74(6):1251-2.

  33. Gold LS, Dhawan S, Weiss J, et al. A novel topical minocycline foam for the treatment of moderate-to-severe acne vulgaris: results of 2 randomized, double-blind, phase 3 studies. J Am Acad Dermatol. 2019 Jan;80(1):168-77.

  34. Raoof TJ, Hooper D, Moore A, et al. Efficacy and safety of a novel topical minocycline foam for the treatment of moderate to severe acne vulgaris: a phase 3 study. J Am Acad Dermatol. 2020 Apr;82(4):832-7.

  35. TWYNEO® (tretinoin and benzoyl peroxide) cream [prescribing information]. Revised: July 2021. Sol-Gel Technologies Inc., Whippany, NJ. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/214902s000lbl. pdf. Accessed August 1, 2023.

  36. Del Rosso J, Sugarman J, Green L, et al. Efficacy and safety of microencapsulated benzoyl peroxide and microencapsulated tretinoin for the treatment of acne vulgaris: results from two phase 3 double-blind, randomized, vehiclecontrolled studies. J Am Acad Dermatol. 2023 Jun 24:S0190-9622(23)01182-9.

  37. Vyas SP, Khar RK. Controlled drug delivery: concepts and advances. 2nd ed. New Delhi: Vallabh Prakashan: Delhi, India. 2012 ; pp.1-53. Access from: https://www.vallabhprakashan.com/cdd.aspx

  38. Kaity S, Maiti S, Ghosh AK, et al. Microsponges: a novel strategy for drug delivery system. J Adv Pharm Technol Res. 2010 Jul;1(3):283-90.

  39. Mahant S, Kumar S, Nanda S, et al. Microsponges for dermatological applications: perspectives and challenges. Asian J Pharm Sci. 2020 May;15(3):273-91.

  40. Embil K, Nacht S. The microsponge delivery system (MDS): a topical delivery system with reduced irritancy incorporating multiple triggering mechanisms for the release of actives. J Microencapsul. 1996 Sep-Oct;13(5):575-88.

  41. Pattani A PS, Patravale V. Microsponges: a path-breaking cosmetic innovation. Household and Personal Care Today. 2008;2:45-9.

  42. Valluru R, Ravi G, Bose SP, et al. Microsponges-a comprehensive review: success and challenges. Indo-American Journal of Pharmaceutical Research. 2019 Sep;9(7):3056-67.

  43. Kircik LH, Draelos ZD, Berson DS. Polymeric emulsion technology applied to tretinoin. J Drugs Dermatol. 2019 Apr 1;18(4):s148-54.

  44. Ceilley RI. Advances in topical delivery systems in acne: new solutions to address concentration dependent irritation and dryness. Skinmed. 2011 Jan-Feb;9(1):15-21.

  45. Latter G, Grice JE, Mohammed Y, et al. Targeted topical delivery of retinoids in the management of acne vulgaris: current formulations and novel delivery systems. Pharmaceutics. 2019 Sep 24;11(10):490.

  46. Nyirady J, Grossman RM, Nighland M, et al. A comparative trial of two retinoids commonly used in the treatment of acne vulgaris. J Dermatolog Treat. 2001 Sep;12(3):149-57

  47. Egan N, Loesche MC, Baker MM. Randomized, controlled, bilateral (split-face) comparison trial of the tolerability and patient preference of adapalene gel 0.1% and tretinoin microsphere gel 0.1% for the treatment of acne vulgaris. Cutis. 2001 Oct;68(4 Suppl):20-4.

  48. Thiboutot D, Gold MH, Jarratt MT, et al. Randomized controlled trial of the tolerability, safety, and efficacy of adapalene gel 0.1% and tretinoin microsphere gel 0.1% for the treatment of acne vulgaris. Cutis. 2001 Oct; 68(4 Suppl):10-9.

  49. Nyirady J, Nighland M, Payonk G, et al. A comparative evaluation of tretinoin gel microsphere, 0.1%, versus tretinoin cream, 0.025%, in reducing facial shine. Cutis. 2000 Aug;66(2):153-6.

  50. Tanghetti EA, Stein Gold L, Del Rosso JQ, et al. Optimized formulation for topical application of a fixed combination halobetasol/tazarotene lotion using polymeric emulsion technology. J Dermatolog Treat. 2021 Jun;32(4):391-8.

  51. Tanghetti EA, Gold LS, Del Rosso JQ, et al. Novel lotion formulation using polymeric emulsion technology for improved skin moisturization and drug permeation in patients with psoriasis. Presented at: Winter Clinical Dermatology Conference, January 17-22, 2020, Kohala Coast, Hi.

  52. Lebwohl MG, Tanghetti EA, Stein Gold L, et al. Fixed-combination halobetasol propionate and tazarotene in the treatment of psoriasis: narrative review of mechanisms of action and therapeutic benefits. Dermatol Ther (Heidelb). 2021 Aug;11(4):1157-74.

  53. Trookman NS, Rizer RL, Ford RO, et al. The stratum corneum and atopic dermatitis: moisturizing advantages of a novel desonide hydrogel treatment. Presented at: 66th Annual Meeting of the American Academy of Dermatology; February 1–5, 2008; San Antonio, TX. AB51.

  54. Kerdel FA, Draelos ZD, Tyring SK, et al. A phase 2, multicenter, double-blind, randomized, vehicle-controlled clinical study to compare the safety and efficacy of a halobetasol propionate 0.01% lotion and halobetasol propionate 0.05% cream in the treatment of plaque psoriasis. J Dermatolog Treat. 2019 Jun;30(4):333-9.

  55. Green LJ, Kerdel FA, Cook-Bolden FE, et al. Safety and efficacy of a once-daily halobetasol propionate 0.01% lotion in the treatment of moderate-to-severe plaque psoriasis: results of two phase 3 randomized controlled trials. J Drugs Dermatol. 2018 Oct 1;17(10):1062-9.

  56. Dreno B, Thiboutot D, Gollnick H, et al. Large-scale worldwide observational study of adherence with acne therapy. Int J Dermatol. 2010 Apr;49(4):448-56.

  57. ARAZLO™ (tazarotene 0.045%) lotion [ product monograph ]. Last revised: July 7, 2021. Bausch Health, Canada Inc., Laval, QC. Available from: https:// bauschhealth.ca/wp-content/uploads/2021/07/Arazlo-PM-E-2021-07-08.pdf. Accessed August 1, 2023.

  58. Tanghetti EA, Kircik LH, Green LJ, et al. A phase 2, multicenter, double-blind, randomized, vehicle-controlled clinical study to compare the safety and efficacy of a novel tazarotene 0.045% lotion and tazarotene 0.1% cream in the treatment of moderate-to-severe acne vulgaris. J Drugs Dermatol. 2019 Jun 1;18(6):542.

  59. Tanghetti EA, Werschler WP, Lain T, et al. Tazarotene 0.045% lotion for oncedaily treatment of moderate-to-severe acne vulgaris: results from two phase 3 trials. J Drugs Dermatol. 2020 Jan 1;19(1):70-7.

  60. Pariser DM, Green LJ, Stein Gold L, et al. Halobetasol 0.01%/tazarotene 0.045% lotion in the treatment of moderate-to-severe plaque psoriasis: maintenance of therapeutic effect after cessation of therapy. J Drugs Dermatol. 2018 Jul 1;17(7):723-6.

  61. Lebwohl MG, Stein Gold L, Papp K, et al. Long-term safety and efficacy of a fixed-combination halobetasol propionate 0.01%/tazarotene 0.045% lotion in moderate-to-severe plaque psoriasis: phase 3 open-label study. J Eur Acad Dermatol Venereol. 2021 May;35(5):1152-60.

  62. Gold LS, Lebwohl MG, Bhatia N, et al. Long-term management of moderate-tosevere plaque psoriasis: maintenance of treatment success following cessation of fixed combination halobetasol propionate 0.01% and tazarotene 0.045% (HP/TAZ) lotion. SKIN The Journal of Cutaneous Medicine. 2019 Nov; 3:S31. https://jofskin.org/index.php/skin/article/view/758

  63. DUOBRII™ (halobetasol propionate and tazarotene) lotion [ product monograph ]. Last revised: June 8, 2020. Bausch Health, Canada Inc., Laval, QC. Available from: https://pdf.hres.ca/dpd_pm/00056991.PDF. Accessed August 1, 2023.

  64. Lebwohl M. Strategies to optimize efficacy, duration of remission, and safety in the treatment of plaque psoriasis by using tazarotene in combination with a corticosteroid. J Am Acad Dermatol. 2000 Aug;43(2 Pt 3):S43-6.

  65. Zasada M, Budzisz E. Retinoids: active molecules influencing skin structure formation in cosmetic and dermatological treatments. Postepy Dermatol Alergol. 2019 Aug;36(4):392-7.

  66. Sevimli Dikicier B. Topical treatment of acne vulgaris: efficiency, side effects, and adherence rate. J Int Med Res. 2019 Jul;47(7):2987-92.

  67. Ferreira KCB, Valle A, Paes CQ, et al. Nanostructured lipid carriers for the formulation of topical anti-inflammatory nanomedicines based on natural substances. Pharmaceutics. 2021 Sep 13;13(9).

  68. Paiva-Santos AC, Silva AL, Guerra C, et al. Ethosomes as nanocarriers for the development of skin delivery formulations. Pharm Res. 2021 Jun;38(6):947-70.

  69. Madane VB, Aloorkar NH, Mokale VJ. Niosomes as an ideal drug delivery system. J NanoSci Res Rep. 2021 Aug;3(3):1-9. doi.org/10.47363/JNSRR/2021(3)123.

  70. Carita AC, Eloy JO, Chorilli M, et al. Recent advances and perspectives in liposomes for cutaneous drug delivery. Curr Med Chem. 2018 Feb 13;25(5):606-35.

  71. Vyas A, Kumar Sonker A, et al. Carrier-based drug delivery system for treatment of acne. ScientificWorldJournal. 2014 2014:276260.


Purchase Article PDF for $1.99

]]>
Managing Psoriasis with Topical Agents – Where Do We Stand? https://www.skintherapyletter.com/psoriasis/managing-with-topical-agents-where-do-we-stand/ Wed, 26 Jul 2023 09:43:06 +0000 https://www.skintherapyletter.com/?p=14512 80%) and do not require systemic treatment, these cases can still be particularly challenging to treat as topical therapies present limitations, including efficacy and administration, leading to poor long-term treatment compliance and unsatisfactory treatment responses. The intent of this paper is to provide physicians with a clinically relevant review of the currently available and newly developed topical therapies...]]> Sofianne Gabrielli, MSc1; Charles Lynde, MD2-4; Natalie Cunningham, MD5; Pierre-Luc Dion, MD6; Christina Han, MD7; Sameh Hanna, MD2,8; Ian Landells, MD9; Andrei Metelitsa, MD10-12; Marni Wiseman, MD13,14; Geeta Yadav, MD15-17; Zeinah AlHalees, MD18; Elena Netchiporouk, MD, MSc18

Affiliations


1Faculty of Medicine, McGill University, Montreal, QC, Canada

2Probity Medical Research Inc., Waterloo, ON, Canada

3Division of Dermatology, Western University, London, ON, Canada

4Lynde Institute for Dermatology, Markham, ON, Canada

5Division of Clinical Dermatology & Cutaneous Science, Department of Medicine, Dalhousie University and IWK Health Centre, NS, Canada

6Division of Dermatology, Université Laval, Québec, QC, Canada

7Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC, Canada

8Division of Dermatology, University of Toronto; Dermatology on Bloor, Toronto, ON, Canada

9Memorial University of Newfoundland and Nexus Clinical Research, St John’s, NL, Canada

10Beacon Dermatology, Calgary, AB, Canada

11Division of Dermatology, Department of Medicine, University of Calgary, Calgary, AB, Canada

12Probity Medical Research, Calgary, AB, Canada

13Section of Dermatology, Department of Medicine, University of Manitoba, Winnipeg, MB, Canada

14SKiNWISE Dermatology, Winnipeg, MB, Canada

15Division of Dermatology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada

16FACET Dermatology, Toronto, ON, Canada

17Division of Dermatology, Women’s College Hospital, Toronto, ON, Canada

18Division of Dermatology, McGill University Health Center, Montreal, QC, Canada


Conflicts of Interest


SG: None.

CL: AbbVie, Altius, Amgen, Aralez, Arcutis, Bausch Health, Bayer, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Cipher, Dermavant, Eli Lilly, Fresnius Kabi, GSK, Innovaderm, Intega Skin, Janssen, Kyowa, La Roche Posay, Leo, L’Oreal, Medexus, Merck, P&G, Pediapharm, Regeneron, Roche, Sanofi Genzyme, Sentrex, Teva, Tribute, UCB, Valeant, Viatris, Volo Health.

NC: Advisor, consultant, and/or speaker for AbbVie, Arcutis, Bausch Health, Bristol Myers Squibb, Eli Lilly, Galderma, Janssen, Leo Pharma, L’Oreal, Novartis, Pfizer, Sanofi Genzyme, Sun Pharma, UCB.

PLD: Advisor, consultant, and/or speaker for AbbVie, Amgen, Aralez, Arcutis, Bristol Myers Squibb, Eli Lilly, GSK, Janssen, Leo, Novartis, Pfizer, Sanofi Genzyme, Sun Pharma, UCB, and Vichy.

CH: AbbVie, Arcutis, Amgen, Bausch Health, Celgene, Galderma, Janssen, Leo, Lilly, Novartis, Sanofi Genzyme, Sun Pharma, UCB, Xyon.

SH: AbbVie, Akros, Allergan, Altius Healthcare, Amgen, Aralez, Arcutis, Bausch Health, Bristol Myers Squibb, Boehringer Ingelheim, Biopharma, Caliway, Celgene, Coherus, Concert Pharma, Cutanea, Dermira, Galapagos, Galderma, Glenmark, Incyte, Janssen, Leo, Lilly, Lumenis, Merz, Novartis, Pedia-Pharm, Pfizer, Prollenium, Regeneron, Revanesse, Reistone, Sandoz, Sanofi, Sun Pharma, UCB, Vichy.

IL: AbbVie, Amgen, Boehringer Ingelheim, Celgene, Eli Lilly, Galderma, GSK-Stiefel, Janssen, LEO Pharma, Merck, Novartis, Pfizer, Sanofi Genzyme, and Valeant.

AM: Abbvie, Amgen, Bausch Health, Bristol Myers Squibb, Eli Lilly, Janssen, Leo, Novartis, Sun Pharma, UCB.

MCW: AbbVie, Amgen, Bausch Health, Celgene, Cipher, Eli Lilly, Galderma, Janssen, Leo, Merck, Novartis, Pfizer, Sanofi Genzyme, SUN Pharma, UCB, and Valeant.

GY: AbbVie, Amgen, Aralez, Arcutis, Bausch Health, Bioderma, Bristol Myers Squibb, Byrdie, Galderma, Incyte, Janssen, Johnson & Johnson, Leo, Medexus, Novartis, Pfizer, Sanofi-Regeneron, Sun Pharma, UCB.

ZH: None.

EN: Advisory board/speaker/consultant and/or received investigator-initiated educational and/or research funding from AbbVie, Bausch Health, Beiersdorf, Boehringer Ingelheim, Bristol Myers Squibb, Eli Lilly, Galderma, Janssen, Leo, Medexus, Novartis, Pfizer, Sanofi Genzyme, Sun Pharma, and UCB.


Abstract

Psoriasis vulgaris is a chronic, immune-mediated inflammatory skin disease affecting 2-4% of the Canadian population. Lifelong management is required. While most psoriasis vulgaris cases are mild-to-moderate (>80%) and do not require systemic treatment, these cases can still be particularly challenging to treat as topical therapies present limitations, including efficacy and administration, leading to poor long-term treatment compliance and unsatisfactory treatment responses. The intent of this paper is to provide physicians with a clinically relevant review of the currently available and newly developed topical therapies for psoriasis, the practice guidelines for topical management of mild-to-moderate psoriasis, and the common pitfalls and mitigation strategies to encourage long-term treatment compliance.

PDF Download


Introduction

Psoriasis is a common immune-mediated skin disease affecting ~2-4% of the population in North America.1 In nearly one-third of cases, disease begins during the first 2 decades of life and follows a chronic and persistent course, resulting in high cumulative lifetime disability.2 Psoriasis is divided into 4 major clinical forms, including plaque psoriasis, guttate psoriasis, erythrodermic psoriasis and pustular psoriasis.3 Plaque psoriasis represents >90% of cases and will be the focus of this review. Classically, plaque psoriasis affects the trunk as well as the extensor surfaces of the elbows and knees; it can also affect other body sites, giving rise to regional variants such as scalp, face, intertriginous, palmoplantar, genital and nail psoriasis.3

Because psoriasis is often localized to visible and/or special body sites (knees, elbows, trunk and scalp) and is commonly associated with pain and itch, it often causes significant physical and psychological burdens.4 Stigmatization is common and contributes to poor health-related quality of life (HRQoL), elevated risk of multiple health comorbidities, and increased barrier to treatment.5 Indeed, a recent Global Burden of Disease (GBD) study ranked psoriasis as the second contributor to all skin-related Disability Adjusted Life Years (DALYs).6 The GBD study highlighted an increase in prevalence and morbidity of psoriasis globally, with North America and Europe being particularly affected, emphasizing the significant burden that psoriasis imposes on both individuals and society as a whole.6

Mild, moderate, and severe psoriasis are defined as plaques affecting <3%, between 3-10%, and >10% of body surface area (BSA), respectively.1 Much progress has been realized in regards to managing severe disease with the approval of numerous advanced systemic therapies, including biologics and small molecules.7 Unfortunately, in contrast to severe disease, there has been very limited advancement in the management of mild-to-moderate disease, which affects >80% of the psoriatic patient population.8,9 Mild psoriasis is typically managed with lifelong skin-directed topical therapies.10 Topical therapies are also often used to treat severe psoriasis vulgaris, either as monotherapy or as an adjunct to phototherapy and/or systemic therapy.10

The most commonly used topical therapies in psoriasis include corticosteroids, vitamin D3 analogues, retinoids, calcineurin inhibitors, keratolytics and tar. Multiple fixed-ingredient combination products have become commercially available in the last 20 years with the aim to overcome challenges related to lack of efficacy, compliance and adverse events.11

Our objective is to review: 1) the currently available and newly developed topical therapies, both single and fixed-dose combination products; 2) clinical practice guidelines specific to the topical treatment of psoriasis; and 3) common pitfalls and mitigation strategies when managing psoriasis with topical therapies. While we recognize the importance of behavioural modification and skin care in the management of psoriasis vulgaris of all severities, we refer the reader to the review article by Ko et al. on the topic.12

Literature Search

This narrative literature review included studies that examined currently available therapies for psoriasis from 2010 to present. The review was conducted using the PubMed and Embase databases with the following search terms: (psoriasis) AND [(corticosteroids) OR (topical corticosteroids) OR (topical corticosteroids AND salicylic acid) OR (topical corticosteroids AND coal tar) OR (calcipotriol) OR (calcitriol) OR (tacalcitol) OR (pimecrolimus) OR (tacrolimus) OR (tazarotene) OR (retinoids) OR (corticosteroid* AND calcipotriol) OR (corticosteroid* AND calcitriol) OR (corticosteroids AND tazarotene) OR (roflumilast) OR (tapiranof) OR (topical treatment)] as either keywords or MeSH terms. Clinicaltrials.gov website was also searched for ongoing Phase II and III clinical trials. References of identified manuscripts were manually extracted to identify additional articles. Only articles published in English were considered. Articles were included if they reported on treatment of psoriasis in humans, regardless of study type. All publications were independently assessed by SG and EN first by screening titles, then abstract, followed by full-length manuscripts. Any discrepancies were discussed and resolved.

Topical Therapies for Psoriasis – Our Current Toolbox

Psoriasis is a chronic disease that requires a life-long treatment. Most patients are managed with topical treatments, therefore, it is important to recognize all presently available therapeutic options, taking into account their respective efficacy, safety profile, and usage considerations. Currently, available treatments that will be discussed include topical corticosteroids (TCS), vitamin D3 analogues, calcineurin inhibitors (TCI), tar-based preparations, retinoids, and combination therapies.13 These therapies can be used to induce skin clearance and to maintain disease control.14 Monotherapies with dithranol and salicylic acid will not be discussed, as their clinical use is limited. A clinical timeline of topical therapies for psoriasis is provided in Figure 1. This section will first discuss efficacy and safety considerations of monotherapies followed by combination treatments. Novel therapies, established or previously published guidelines for topical medications use and general recommendations to improve compliance are discussed in following sections.

Managing Psoriasis with Topical Agents - Where Do We Stand? - image
Figure 1. Clinical timeline of topical therapies for psoriasis

Monotherapies

Topical Corticosteroids (TCS)

TCS have been used to treat psoriasis for over 60 years.15 TCS exert broad anti-inflammatory, immunosuppressive, and antimitotic effects.16 The most recent systematic review that we have identified focusing on TCS efficacy and safety in psoriasis dates back to 2012.15 It included 50 randomized controlled trials (RCTs), of which only 11 were placebo controlled. Potent and ultra-potent TCS were used in different formulations to induce and/or maintain disease control. Only 5 studies used the Psoriasis Area and Severity Index (PASI) assessment tool to measure efficacy outcomes. The mean percentage change in the PASI from baseline to 4-8 weeks ranged from 45-60%. However, the overall reported efficacy rating varied greatly depending on the study. An added benefit of occlusion was suggested (1 study), whereas no efficacy difference was established between different vehicles (2 studies). Three studies confirmed that weekend (or episodic) TCS treatment after achieving skin clearance was valuable to prevent plaque recurrence in 30-40% of patients at 6 months.

Since this publication, we identified 2 additional RCTs.17,18 Desoximetasone 0.25% spray twice daily (BID) demonstrated statistically significant clinical success compared to the vehicle as measured by Physician Global Assessment (PGA) score 0/1 at 4 weeks (39% vs. 7%, respectively).17 Efficacy was better among compliant patients who received reminders compared to those that were not reminded, suggesting that compliance is an important barrier to treatment success.18-20 Another RCT compared once daily (QD) halobetasol propionate 0.05% cream vs. halobetasol propionate 0.01% lotion suggesting similar efficacy at 2 weeks.21

Despite corticosteroid-sparing alternatives and combination therapies (discussed below) being commercially available, TCS remain widely used because of the low cost of some products as well as their versatility. They are available in a wide range of vehicles and potencies, ranging from I (ultra-high potency) to VII (low potency). Available vehicles include creams, lotions, foams, gels, ointments, shampoo, sprays, and solutions22 (Table 1). Typically, potent to ultra-potent TCS (class I-II) (e.g., clobetasol propionate) are used for thick plaques on the trunk/limbs or special sites, such as the scalp and palmoplantar regions; moderate potency TCS (class III-IV) (e.g., betamethasone valerate; triamcinolone acetonide) are suitable for thinner plaques on the trunk/limbs, whereas mild potency TCS (class VI-VII, e.g., desonide; hydrocortisone) are recommended for intertriginous areas, genitals and/or face.22 Milder potency preparations are also usually preferred to manage psoriasis in pediatric or pregnant patients.23,24 The choice of the vehicle is made by the treating physician together with the patient. While some vehicles are preferred based on the anatomic site being treated and desired potency, patient preference is of utmost importance since this enhances treatment compliance. An RCT of vehicle preference among various TCS preparations assessed adherence to treatment and improvement of HRQoL among patients with psoriasis between spray, cream, ointment, gel, lotion, foam, and solution.25 It was found that patient preference was highly variable, with less messy products favoured.25 There was no overwhelming agreement on the effect of TCS vehicles in terms of efficacy, hence treatment should be individualized.

Table 1. Topical Corticosteroid Classes of Potency

Class Selected Preparation
I (ultra-high potency)
  • Augmented betamethasone dipropionate 0.05% ointment, and lotion
  • Clobetasol propionate 0.05% cream, ointment and lotion, and solution (shampoo, spray aerosol)
  • Halobetasol propionate 0.05% cream and ointment
II (high potency)
  • Amcinonide 0.1% ointment
  • Augmented betamethasone dipropionate 0.05% cream
  • Betamethasone dipropionate 0.05% cream and ointment
  • Desoximetasone 0.25% ointment
  • Desoximetasone 0.05% gel
  • Fluocinomide 0.05% cream, ointment, and gel
  • Halobetasol propionate 0.01% lotion
  • Mometasone furoate 0.1% ointment
III (mid-potency)
  • Amcinonide 0.1% cream
  • Betamethasone valerate 0.1% ointment 
  • Desoximetasone 0.05% cream and ointment 
  • Fluocinonide 0.05% cream
  • Triamcinolone acetonide 0.5% cream
IV
  • Hydrocortisone valerate 0.2% ointment
  • Mometasone furoate 0.1% cream and lotion
  • Triamcinolone acetonide 0.1% cream and ointment
V
  • Betamethasone dipropionate 0.05% lotion
  • Betamethasone valerate 0.1% cream
  • Desonide 0.05% ointment
  • Hydrocortisone valerate 0.2% cream
  • Prednicarbate 0.1% cream and ointment
VI (low potency)
  • Betamethasone valerate 0.1% lotion
  • Desonide 0.05% cream
VII
  • Hydrocortisone 0.5% ointment
  • Hydrocortisone 1% cream, ointment, and lotion 
  • Hydrocortisone 2.5% cream

List of products obtained from Health Canada’s Drug Product Database on April 25th, 2023.

Adverse events (AEs) with TCS use are generally rare, but may occur with prolonged and/or inappropriate use. These include local AEs such as skin atrophy, telangiectasia, striae, poor wound healing and infections. When used on the face or acne-prone skin, acne exacerbation or de novo periorificial dermatitis/folliculitis may occur.26 Furthermore, even class VII TCS used on eyelids for prolonged periods can lead to cataracts and/or glaucoma.27 Systemic AEs related to hypothalamic-pituitary axis (HPA) suppression are exceedingly rare.28,29 Two systematic reviews/meta-analyses evaluated TCS safety in psoriasis. Literature identified was reassuring, with <5% risk of skin atrophy30 and <5% rate of HPA suppression when TCS were used long-term.31 TCS withdrawal reactions, such as Red Skin Syndrome or topical steroid addiction, have also been reported with discontinuation of prolonged, frequent use of moderate to high potency TCS.32 These severe reactions are rare and are more likely to occur with use on special areas, such as the face and the genitals. A recent review article concluded that TCS are generally safe and effective when used correctly for short periods of time or with short breaks in longer treatments.32 Beside AEs, additional TCS related limitations include potential tachyphylaxis and corticophobia. Whether tachyphylaxis truly exists is debated among experts, as diminished efficacy over time may be related to low adherence to treatment among patients, especially when long-term treatment is required.33 Corticophobia among patients and health care providers remains omnipresent and is a major barrier for treatment efficacy beyond inherent limitations associated with the drugs of this class.34

Vitamin D3 Analogues

The introduction of Vitamin D3 analogues ~30 years ago was met with much enthusiasm due to their steroid-sparing effect.35 Commercially available vitamin D3 analogues in Canada are calcipotriol and calcitriol ointments. Vitamin D3 analogues may be used as monotherapy, as an adjunct to TCS, or as fixed-dose combination therapy. They work by regulating gene transcription, modulating keratinocyte proliferation, and differentiation.36 The mechanism of action also involves the inhibition of T cell proliferation and downstream inflammatory mediators.36

There are several systematic reviews published assessing the efficacy of vitamin D3 analogues compared to vehicle or TCS.37-41 As monotherapy, 1 review reported treatment success (defined as >90% reduction in the PASI score) with vitamin D3 analogues ranging from 4-40% after 6-12 weeks of therapy.37 Another reported a decrease in the PASI ranging from 27.8-60.4% with calcipotriol monotherapy.38 Further, BID use of vitamin D3 analogues was found to be at least as effective as TCS and more effective than placebo at 8 weeks.38,39 A systematic review focused on efficacy of vitamin D3 analogues in pediatric psoriasis patients found 5 studies reporting improvement in PASI from baseline ranging from 17.3-94%, 1 study reporting improvement in Psoriasis Scalp Severity Index (PSSI) of 32.1%, and 1 study reporting 100% clearance of skin lesions.40

In the last 10 years, only 2 new double-blind, vehicle-controlled phase III RCTs evaluated the efficacy and safety of calcipotriol 0.005% foam BID for mild-to-severe psoriasis (defined as plaque psoriasis involving 2-20% BSA) compared to vehicle.42 Both studies demonstrated significant treatment success, defined as the Investigator’s Static Global Assessment (ISGA) scores of 0/1 at 8 weeks. The primary outcome was achieved in 15% vs. 7% of calcipotriol vs. vehicle patients in the first study and 28% vs. 6% in the second study.42

The use of vitamin D3 analogues in psoriasis has been shown to be safe and well-tolerated, with less AEs than TCS. AEs are generally comparable to the vehicle, with application site reactions occurring in less than 2% of subjects.42 Specifically, these include stinging, burning, and peeling of the skin.37,43 Calcitriol may cause less irritation in sensitive areas compared to treatment with calcipotriol.44 When used appropriately (maximum recommended dose of 100 g per week of calcipotriene or 200 g per week of calcitriol), the risk of hypercalcemia is very low.43 Hypercalcemia risk was studied in 3 studies, occurring at a rate <1%.37

Retinoids

Vitamin A and its naturally occurring and synthetic derivatives are referred to as retinoids.45 They were introduced as a treatment for cutaneous disorders in the 1960s and, with the development of safer synthetic retinoids, have become widely used.45 There are many topical retinoids used in dermatology, however, only tazarotene has been studied and indicated for psoriasis. While tazarotene lotion 0.045% is the only formulation commercially available in Canada, it is indicated for acne and used off-label for psoriasis. Tazarotene binds and modulates activity of retinoic acid receptors (RAR)-β and -γ, thereby decreasing inflammation and keratinocyte proliferation.46

There were 2 systematic reviews assessing the efficacy of topical retinoids in psoriasis. The first contained 4 studies comparing tazarotene 0.05% or 0.1% gel or cream to placebo, finding tazarotene to be more effective in improving symptoms in the short-term (6-12 weeks).39 However, the more recent systematic review comparing the same interventions found that symptom clearance as measured by Investigator’s Global Assessment (IGA) ranged from 5.5-6.2% with tazarotene 0.05% and 0.1% cream at 12 weeks, which was not better than placebo.47 In the last 10 years, there have been no new published RCTs assessing the efficacy of topical retinoid monotherapy in psoriasis. Most clinical trials have focused on combination therapy of tazarotene and TCS.

The most common AEs associated with use of tazarotene are cutaneous local irritations such as peeling, erythema, itching, and burning at the site of application.46 Cutaneous absorption of topical retinoids is limited, and there are no known systemic toxicities. However, as retinoids are teratogenic, women of childbearing age must use appropriate contraception.46

Calcineurin Inhibitors (TCI)

Topical calcineurin inhibitors (TCI) have been approved since the early 2000s for the treatment of mild-to-moderate atopic dermatitis. While not indicated for psoriasis, TCI are often used off-label for facial and intertriginous psoriasis to avoid TCS-related AEs.48 The available formulations are pimecrolimus 1% cream and tacrolimus 0.1% and 0.03% ointments. TCI bind to immunophilins, which lead to a decreased release of interleukin (IL)-2 and interferon (INF)-γ and thereby decreased T cell proliferation.49

Systematic reviews assessing the efficacy of TCI for psoriasis confirmed tacrolimus superiority to placebo, TCS and calcitriol in treating facial and intertriginous psoriasis with treatment duration of 8 weeks.50,51 However, pimecrolimus was inferior to standard psoriasis treatments.50,52 One systematic review looked at the synergistic effect of TCI and TCS, which found that there was no additional benefit by combining these agents as opposed to TCS alone.53

In the last 10 years, there have been 2 new RCTs published assessing the efficacy of TCI in psoriasis. The first studied the use of pimecrolimus 1% cream in the treatment of intertriginous psoriasis compared to placebo, finding that 71.4% in the treatment group reported an IGA of 0/1 at 8 weeks.54 The second assessed tacrolimus 0.1% ointment in the treatment of nail psoriasis on 1 hand, using the other hand as a control. At 12 weeks, there was statistically significant improvement in the treated hand as evaluated by the Nail Psoriasis Severity Index (NAPSI) score.55

AEs for topical TCI include skin irritation and discoloration to the site of application.56 In 2006, a black box warning was issued for a potential link with skin cancer and lymphoma.56 However, as a result of subsequent large-scale studies disproving this association,57 Health Canada lifted the black box warning in 2021.58 The systematic reviews and RCTs found similar rates of AEs between treatment and placebo groups,51,54 however patients should be counselled regarding transient burning sensation when prescribed tacrolimus ointment to improve treatment adherence.

Tar

Historically, coal tar was considered a classic anti-psoriatic therapy and was used as a first-line agent for more than 2,000 years to treat psoriasis and other skin diseases.59 Recent studies shed light into the mechanism of action of tar, suggesting modulation of epidermal differentiation and anti-inflammatory effects are likely achieved through activation of the aryl hydrocarbon receptor (AHR).59 The efficacy of coal tar or its distillate, liquor carbonis detergens (LCD), in treating psoriasis was seldom formally evaluated. The limited available data suggests inferior efficacy to other commercially available agents. Specifically, a Cochrane Review (last updated in 2013), identified tar (including LCD) as generally less effective than TCS and vitamin D3 analogue monotherapies.41 Safety has been another important concern. As the “crude” word suggests, coal tar contains >10,000 organic compounds, including carcinogenic chemicals, such as benzene.59 However, carcinogenic potential has not been proven.60 Over-the-counter tar-containing products are available in different formats including lotions, creams, ointments, and shampoos, however, its application can be messy by staining hair, skin, nails, and clothing with a very unpleasant odour.61 While it can be compounded with TCS and other active ingredients to enhance effectiveness and penetration, currently it is primarily used in shampoos for the treatment of scalp psoriasis.61

Combination Therapies

Combination therapies were developed to improve treatment efficacy as it provides 2 mechanisms of action simultaneously and may have additive or synergistic effects.62 Further, they may decrease AEs related to each ingredient alone and are therefore better tolerated than monotherapy.62 Specifically, vitamin D3 analogues and retinoids decrease the risk of skin atrophy, whereas TCS decreases the irritation associated with vitamin D3 analogues and retinoids. Additionally, most fixed-dose combination topical therapies are prescribed to be applied QD as opposed to BID, thereby potentially improving long-term compliance. Commonly prescribed fixed-dose combination topical therapies include TCS and salicylic acid, TCS and vitamin D3 analogues (commercially available since 2001 as ointment, 2012 as gel, and 2016 as aerosol) as well as TCS and retinoids (commercially available as lotion since 2020).

Topical Corticosteroids and Keratolytics

Keratolytic agents, such as salicylic acid and urea, can improve the efficacy of TCS, especially for thicker plaques, by enhancing penetration and improving skin barrier. They are commercially available in combination as salicylic acid 3.0% and betamethasone dipropionate 0.05% ointment and salicylic acid 2.0% and betamethasone dipropionate 0.05% lotion. Additional alternatives can be compounded. There were several RCTs assessing combination therapy of salicylic acid and TCS showing superiority to monotherapy of either salicylic acid or the TCS alone.53,63 However, 2 RCTs compared combination therapy of TCS and salicylic acid to calcipotriol monotherapy, with no clinical difference.64 HRQoL was however improved with the combination therapy and was preferred by patients.63 Only 1 RCT assessed urea in combination with TCS, which found a greater percentage of patients with an improved clinical score compared to monotherapy (47% vs. 33%).63 Similarly, recent data showed that even simple moisturizers containing lipid-ceramides improve the efficacy of TCS.65,66 In the last 10 years, there have been no newly published RCTs assessing the efficacy of topical salicylic acid and TCS combination therapy in psoriasis. Although rare, there is a risk of salicylic acid toxicity with topical application.67

Calcipotriol and Betamethasone Dipropionate Fixed-Dose Combination

Calcipotriol 50 μg/g and betamethasone dipropionate 0.5 mg/g (Cal/BD), available in ointment, gel, and foam formulations in Canada, allows for both anti-inflammatory and anti-proliferative effects. A large Cochrane systematic review published in 2013 assessed the efficacy of combination therapies in psoriasis. Cal/BD was superior to placebo41,68 and monotherapy (Cal or BD alone) in all but 1 RCT as early as after 2-8 weeks of treatment.41

In the last 10 years, there have been many new RCTs assessing the efficacy of Cal/BD for psoriasis focusing on the newer foam formulation, which when used QD for 4-12 weeks demonstrated a significant decrease in PASI from baseline of ~70%, superior to vehicle or either monotherapy.69-72 Further, approximately half of participants achieved an IGA of 0/1.73 Two studies compared Cal/BD to betamethasone valerate 0.1% dressing, showing in the first trial no significant efficacy difference at 4 weeks with Cal/BD ointment74 and the second trial demonstrated superiority of the Cal/BD foam (at 4 weeks).75 Several additional RCTs re-iterated the superiority of Cal/BD therapy vs. vitamin D3 analogue monotherapy.76,70,77-80

Several RCTs focused on Cal/BD vehicle.71,72,81 RCTs comparing different vehicles have shown better efficacy and superior HRQoL improvement with the use of Cal/BD foam compared to gel or ointment formulations71,72,81 and cream compared to suspension.82 However, RCTs assessing vehicle preference as determined by patients did not find any significant preferences when comparing gel vs. foam, gel vs. ointment, and ointment vs. topical suspension, determining that individual patient preference should dictate treatment.83-85 One real-world study found that patients using gel reported greater satisfaction compared to ointment due to ease of use.86

One RCT focused on psoriasis relapse prevention following clinical clearance, defined as PGA score 0 or 1 (clear or almost clear).87 In this study, both treatment arms received Cal/BD QD for 4 weeks initially to achieve skin clearance and were subsequently randomized into Cal/BD or vehicle biweekly as maintenance treatment. Patients that applied Cal/BD proactively experienced 3.1 relapses per year vs. 4.8 relapses (defined as PGA score 2 or higher) seen in the vehicle group. Median time to first relapse was also longer in the proactive management group (56 vs. 30 days) suggesting that proactive approach may be an interesting alternative to reactive approach for interested patients and could potentially be cost-effective.87 As expected, both active treatment and proactive maintenance with Cal/BD were well-tolerated and no cases of skin atrophy were reported in either group.87

Tazarotene and Halobetasol Fixed-Dose Combination

The only combination treatment of retinoid with TCS commercially available in Canada is halobetasol 0.01%/tazarotene 0.045% (HP/TAZ) lotion. A systematic review published in 2012 included 7 studies assessing the combination of retinoids (in general) with TCS, supporting superiority of combination as opposed to retinoid monotherapy at 4 weeks.88 A recently published systematic review of 5 RCTs demonstrated treatment success, defined as at least 2-grade improvement from baseline in the IGA score and IGA score of 0 or 1 (clear or almost clear), of 32.8-52.5% for HP/TAZ compared to 33.3-34% for HP alone and 18.6% for TAZ alone with treatment duration of 2-8 weeks.89

There have been several new RCTs published in the last 10 years assessing efficacy and safety of retinoids and TCS combination therapy. A combination tazarotene 0.05%/betamethasone diproprionate 0.05% (TAZ/BD) applied QD was superior to either agent used as monotherapy in 2 studies.90,91 Treatment success of QD topical HP/TAZ lotion measured by the proportion of patients achieving IGA 0/1 ranged from 31.3-57.8% with treatment durations of 8-12 weeks,92-99 which was significantly more effective than vehicle or either ingredient alone. The most frequent AEs reported were dermatitis, pruritus, pain, and irritation,92,93,98 occurring in 6-20.8% of study participants.91,97 A single RCT analyed the sensitization and irritation potential of HP/TAZ lotion with treatment duration of 4-6 weeks, finding that the topical did not induce contact sensitization and caused only minimal skin irritation, but significantly less than tazarotene alone.100

Topical Therapies for Psoriasis – The Pipeline

As reviewed above, our current toolbox of topical therapy options is limited to TCS and a handful of other agents, such as vitamin D3 analogues, retinoids, tar or their combination. While the marketing of steroid-sparing monotherapies and fixed-dose combinations with TCS represents a major step forward in the management of psoriasis, these treatment options possess limitations in terms of efficacy, AEs, cost, patient satisfaction, and real-world adherence. Hence, there remains an unmet need for new topical therapies.101

There is an exciting topical therapy pipeline in psoriasis (Table 2), roflumilast and tapirnarof will be discussed in this section as phase III RCT data was recently published. Roflumilast is a phosphodiesterase-4 (PDE-4) inhibitor that has been developed into 0.3% cream and foam formulations. Crisaborole, a topical PDE-4 inhibitor, was approved in 2016 for atopic dermatitis in adults and children.102 Its use has been investigated for psoriasis in phase II RCTs, however, despite demonstrating efficacy and safety, results were not published. Rather, focus has shifted to roflumilast, a more potent PDE-4 inhibitor by 25-300X based on in vitro studies.102 PDE-4 inhibition suppresses the breakdown of cyclic adenosine monophosphate (cAMP), decreasing the presence of proinflammatory cytokines involved in the pathogenesis of psoriasis, similar to apremilast used systemically or topical crisaborole.103 Phase I and II RCT data have demonstrated that roflumilast cream QD was superior to the vehicle when used for 2-8 weeks.104-106 Phase III RCT data regarding roflumilast 0.3% cream efficacy and safety has been recently published.107 DERMIS-1 and DERMIS-2 were parallel double-blind RCTs including 439 and 442 patients, respectively. Patients aged ≥2 years with psoriasis affecting 2-20% BSA were recruited and randomized 2:1 into either roflumilast 0.3% cream or vehicle applied QD for 8 weeks. The primary outcome was IGA 0/1 response plus ≥2 grade improvement from baseline, which was achieved in 37.5-42.4% of roflumilast-treated patients vs. 6.1-6.9% vehicle-treated patients. Improvement of PASI ≥75% from baseline (PASI75) was achieved in 39.0-41.6% vs. 5.3-7.6% of roflumilast-treated vs. vehicle-treated patients, respectively. It was also shown to be effective for the treatment of intertriginous psoriasis (68.1-71.2% vs. 13.8-18.5%). The incidence of AEs was comparable to the vehicle, with the most commonly reported events being diarrhea and headache in the roflumilast group. Further, AE profiles were similar in individuals aged 12–17 years relative to adults.107 Currently, roflumilast is approved by the US Food and Drug Administration (FDA) and Health Canada.

Table 2. Topical antipsoriatic agents undergoing clinical trials

Product Class Phase Clinical Trial Number and Status*
BOS-475 0.5%, 1%, or 2% cream Targets BD2 domain of bromodomain containing protein I

NCT03960450,NCT04221906124,125 – Studies completed

SAN021 serum containing 10% East Indian sandalwood oil PDE-4 inhibitor II

NCT03000608126 – Study completed

Crisaborole ointment 0.3%, 0.5%, 1%, 2%, or 5% PDE-4 inhibitor I

NCT01258088, NCT00763204, NCT00762658127-129 – Studies completed

Crisaborole ointment 0.5%, 2%, or 5% PDE-4 inhibitor II

NCT01300052, NCT00759161, NCT00755196, NCT01029405130-133  – Studies completed

LAS41004 ointment (bexarotene/betamethasone dipropionate) Fixed combination retinoid and topical corticosteroid II

NCT02180464, NCT01360944, NCT01283698, NCT01119339, NCT02111499, NCT01462643134-139 – Studies completed

PH-10 0.002%, 0.005%, or 0.01% rose bengal aqueous hydrogel Rose bengal disodium II

NCT01247818, NCT02322086, NCT00941278140-142 – Studies completed

SNA-120 (CT 327/pegcantratinib) 0.5% ointment

TrkA receptor antagonist II

NCT03448081, NCT03322137143,144 – Studies completed

Roflumilast 0.3% foam for scalp and body psoriasis PDE-4 inhibitor II

NCT04128007145 – Study complete

Roflumilast 0.3% cream PDE-4 inhibitor II

NCT03764475, NCT04746911, NCT04655313146-148 – Studies completed

Roflumilast 0.3% foam for scalp and body psoriasis

PDE-4 inhibitor III

NCT05028582149 – Study completed

Roflumilast 0.3% cream PDE-4 inhibitor III

NCT04286607, NCT05763083150,151 – Actively recruiting

M518101 (pefcalcitol) ointment

Vitamin D3 analogue III

NCT01908595, NCT01989429, NCT01878461, NCT01873677152–155 – Study completed

MC2-01 cream (calcipotriene 0.05%/betamethasone dipropionate 0.064%) Fixed combination vitamin D3 analogue and corticosteroid III

NCT03462927156 – Study completed

Tapinarof (DMVT-505) 1% cream Aryl hydrocarbon receptor agonist III

NCT04053387157 – Study completed

NCT05172726158 – Actively recruiting

NCT05680740, NCT05789576159,160 – Active, not yet recruiting

*Last update per ClinicalTrials.gov as of April 25, 2023

Tapinarof is an AHR-modulating agent that acts as an anti-inflammatory compound. It has a similar mechanism of action to tar, which also activates AHRs, however it does not contain carcinogenic chemical compounds.108 Tapinarof is able to regulate innate and adaptive immune responses, affecting Th17 and regulatory T cells. It also has an important role in the development and maintenance of the skin barrier and upregulating barrier genes, such as filaggrin. Lastly, tapinarof inhibits the migration of T cells, decreasing the presence of proinflammatory cytokines, such as tumor necrosis factor (TNF)-α, INF-γ, IL-2, IL-13 and IL-17A.103 Phase III RCT data has been recently published. In 2 parallel double-blind RCTs including >600 patients each, adults with mild-to-severe psoriasis (PGA 2-4, BSA 3-20%) were recruited and randomized 2:1 into either tapiranof 1% cream or vehicle applied QD for 12 weeks. The primary endpoint was IGA 0/1 and 2-point reduction from baseline at 12 weeks, which was achieved in 35.4-40.2% of tapinarof-treated patients vs. 6.0-6.3% of vehicle-treated patients.109 While it was generally well-tolerated, increased rates of pruritus, contact dermatitis and folliculitis were seen in the active treatment group. A second RCT found that significantly more participants achieved a 75% reduction in the PASI score from baseline (PASI75) with tapinarof (50.4%) compared to calcipotriol (38.5%) and placebo (13.9%) when used QD for 12 weeks.110 Tapinarof has been recently approved by the FDA.

Treatment Guidelines for Topical Antipsoriatic Agents

Treatment guidelines for severe psoriasis and psoriatic arthritis are beyond the scope of this manuscript. This section will focus on reviewing guidelines specific to the treatment of mild-to-moderate psoriasis or treatment focused on topical agents.

Current guidelines for the treatment of mild-to-moderate psoriasis recommend topical therapies which include monotherapy with TCS, vitamin D3 analogues, TCI, retinoids, anthralin, and tar as well as combination therapies as first-line options. In Canada specifically, treatment guidelines were initially published in 2011. At this point, Grade A recommendation for first-line topical therapies included TCS or vitamin D3 analogues (i.e. calcipotriol) monotherapy or Cal/BD fixed-dose combination therapy. The treatment guidelines noted that additional topical therapy options were superior to placebo (e.g., retinoids alone or in combination with TCS, 15% LCD) and may be used on a case-by-case basis.10,111 An update of these treatment guidelines was published in 2016 adding topical calcitriol as an additional first-line topical therapy option for mild psoriasis.10,111

The American Academy of Dermatology (AAD) and National Psoriasis Foundation (NPF) have put forth the most recent guidelines in North America in 2020 focusing on topical therapies.14 The AAD-NPF guidelines do not mention specific recommendations for first-line topical therapy options, but rather provide guidance for use within each class. For TCS, Grade A recommendations include using class I to V agents for up to 4 weeks for body psoriasis (excluding intertriginous areas) and class I to VII agents for scalp psoriasis. The use of TCS for prolonged periods (>12 weeks) may be done under the supervision of a physician (Grade C). However, gradual reduction in frequency of TCS is suggested upon clinical improvement, but without defined tapering protocol. Following clinical improvement, maintenance of response can be achieved by using a steroid-sparing agent (e.g., vitamin D3 analogues or TCI) or by using TCS intermittently (e.g., biweekly, this is also known as proactive approach). Additional recommendations made in regards to TCS use were as follows: the use of emollient was suggested to reduce itching and desquamation and to prevent relapse after TCS discontinuation (Grade B). As well, topical salicylic acid alone or in combination with TCS was recommended as an alternative to TCS monotherapy to achieve clear skin (8-16 weeks of treatment, Grade B).

AAD/NPF guidelines recommended vitamin D3 analogue monotherapy and/or in combination with TCS (e.g., Cal/BD fixed-dose combinations) to induce clearance of scalp psoriasis (4-12 weeks treatment, Grade A), facial psoriasis (up to 8 weeks treatment, Grade B, caution to favour class VI-VII TCS agents) or body psoriasis (up to 52 weeks treatment, Grade A). Topical retinoids (e.g., tazarotene) were recommended either as monotherapy, fixed-dose combination (e.g., HP/Taz) or in combination with narrowband ultraviolet light phototherapy (NB-UVB) (Grade B) for plaque psoriasis and nail psoriasis. However, HP/Taz was preferred (Grade A) to induce clear skin (8-16 weeks treatment) due to better efficacy and tolerability.

The off-label use of TCI (e.g., tacrolimus and pimecrolimus) was recommended by AAD/NPF guidelines for facial and inverse psoriasis to achieve clinical improvement (Grade B recommendation) and to maintain response (Grade C recommendation). They also suggested a combination of tacrolimus/6% salicylic acid for body psoriasis (Grade B recommendation).

Grade B recommendation was also stated for short contact anthralin use (≤2 hours per day, up to 8-12 weeks treatment) and Goeckerman therapy (coal tar and NB-UVB) for mild-to-moderate plaque psoriasis.14 Coal tar preparations received Grade A recommendation as well.

Real-world Limitations with Topical Treatments and Strategies to Improve Compliance

As highlighted above, all current topical therapies come with limitations. Patient compliance is certainly among the most important barriers to success. Adherence rates with current topical therapies are low, estimated to range from 50-70% in general.101 The compliance rates for TCS are even more variable and in some instances are thought to be as low 8%, due to prevalent corticophobia among dermatology patients.101,112 Adherence is an important concept that must be evaluated in patients as it is directly associated with better clinical outcomes. A recent RCT demonstrated that a decrease in adherence rate of 10% was associated with a 1-point increase in disease severity.113

Various interventions were studied to improve compliance. Three RCTs integrated reminders in the forms of BID telephone calls, text messages, or smartphone application to remind and motivate patients to use their topical therapy.20,114,115 In all studies, adherence improved and almost doubled compared to non-interventional arms (65% vs. 38% adherence).115 This translated into significantly better clinical outcomes, such as reduction in PGA.20,115 Another RCT developed a web-based application to educate patients with videos, graphics, and text.116 While knowledge was improved, this did not translate to increased treatment adherence.116 Four RCTs approached adherence by offering more clinical support, such as teaching from nursing staff or internet-based reporting.117-120 Compared to standard of care, these clinical trials demonstrated that additional support resulted in greater clinical outcomes as early as 4 weeks, which were sustained at 3 months.117-120

Vehicle selection is an important component of efficacy and adherence. Good vehicles can accelerate barrier restoration and enhance efficacy of active agents by promoting penetration and sustained drug release.121 As discussed above, RCTs assessing patient satisfaction have found that treatment preferences are heterogeneous and may even change over time.83,122 Factors that may influence preferences included age, sex, comorbidities, disease duration, and prior treatments.122 Therefore, a vehicle should be selected to maximize efficacy and meet the diverse needs of the patient while considering bodily location of psoriasis, probability of improvement, and delivery method. An additional very important attribute for a topical therapy to improve patient adherence is convenience. While it may be patient-specific, an agent that does not need to be applied often (QD or less often), is universal (e.g., same product that can be applied anywhere on the skin), is cosmetically acceptable (texture, colour, and odour) and is affordable will likely promote higher patient adherence and thereby achieve better clinical success rates.

As discussed in the guideline review section above, the first aim of psoriasis treatment is to achieve clear/almost clear skin with a topical agent of choice (combined physician/patient decision for agent selection). Prior to fixed-dose combination topical therapies, in order to increase efficacy while mitigating AEs, different strategies were used. These included rotational treatment where patients alternated between 2 agents, usually a TCS and a corticosteroid-sparing molecule;123 or a sequential treatment approach where a superpotent agent (usually TCS class I-II) was used initially with subsequent step down to either a milder TCS, steroid-sparing molecule or a rotational treatment. However, nowadays fixed-dose combination topical therapies are more popular for their additive efficacy, simplicity, and convenience.123 Once acceptable control is achieved, discussion of relapse prevention is important.

Because psoriasis is chronic and likely to recur upon discontinuation of the topical therapy, it is important to educate the patient about the chronicity of the disease and its treatment at the initial and subsequent visits. Two approaches following initial improvement of psoriasis are commonly used in clinical practice to maintain response over longer-term: the proactive and reactive approaches. Combined physician-patient decision-making may opt for either a proactive approach which consists of using the same agent to achieve clear skin (or another topical) intermittently (e.g., biweekly) to psoriasis-prone areas in order to prevent recurrence, or a reactive approach where all treatments are discontinued upon clinical resolution and restarted promptly with first signs of disease recurrence.

Conclusion

The vast majority of our psoriasis patients have a mild-to-moderate disease requiring topical therapies life-long. Consequently, the availability of safe, effective, and convenient products is essential to achieve and maintain clear/almost clear skin and promote long term treatment adherence. In this review, we provided clinicians an up to date safety and efficacy data of commercially available topical products as well as imminent pipeline topicals. North American guidelines for topical treatment of mild-to-moderate psoriasis are summarized as well as clinical tips are provided.

References



  1. Papp KA, Gniadecki R, Beecker J, et al. Psoriasis Prevalence and Severity by Expert Elicitation. Dermatol Ther. 2021; 11: 1053–1064.

  2. Salman A, Yucelten AD, Sarac E, et al. Impact of psoriasis in the quality of life of children, adolescents and their families: a cross-sectional study. An Bras Dermatol. 2018; 93: 819–823.

  3. Sarac G, Koca TT, Baglan T. A brief summary of clinical types of psoriasis. North Clin Istanb. 2016; 3: 79–82.

  4. Pearl RL, Wan MT, Takeshita J, et al. Stigmatizing attitudes toward persons with psoriasis among laypersons and medical students. J Am Acad Dermatol. 2019; 80: 1556–1563.

  5. Alpsoy E, Polat M, Yavuz IH, et al. Internalized Stigma in Pediatric Psoriasis: A Comparative Multicenter Study. Ann Dermatol. 2020; 32: 181–188.

  6. Mehrmal S, Uppal P, Nedley N, et al. The global, regional, and national burden of psoriasis in 195 countries and territories, 1990 to 2017: A systematic analysis from the Global Burden of Disease Study 2017. J Am Acad Dermatol. 2021; 84: 46–52.

  7. Papp KA, Lebwohl MG. Onset of Action of Biologics in Patients With Moderate-to-Severe Psoriasis. J Drugs Dermatol. 2018; 17: 247–250.

  8. Armstrong AW, Mehta MD, Schupp CW, et al. Psoriasis Prevalence in Adults in the United States. JAMA Dermatol. 2021; 157: 940–946.

  9. Menter A, Gottlieb A, Feldman SR, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis: Section 1. Overview of psoriasis and guidelines of care for the treatment of psoriasis with biologics. J Am Acad Dermatol. 2008; 58: 826–850.

  10. Canadian Psoriasis Guidelines Addendum Committee. 2016 addendum to the Canadian guidelines for the management of plaque psoriasis 2009. J Cutan Med Surg. 2016; 20: 375–431.

  11. Gustafson CJ, Watkins C, Hix E, et al. Combination therapy in psoriasis: an evidence-based review. Am J Clin Dermatol. 2013; 14: 9–25.

  12. Ko S-H, Chi C-C, Yeh M-L, et al. Lifestyle changes for treating psoriasis. Cochrane Database Syst Rev. 2019; 7: CD011972.

  13. Armstrong AW, Read C. Pathophysiology, Clinical Presentation, and Treatment of Psoriasis: A Review. JAMA. 2020; 323: 1945–1960.

  14. Elmets CA, Korman NJ, Prater EF, et al. Joint AAD–NPF Guidelines of care for the management and treatment of psoriasis with topical therapy and alternative medicine modalities for psoriasis severity measures. Journal of the American Academy of Dermatology. 2021; 84: 432–470.

  15. Castela E, Archier E, Devaux S, et al. Topical corticosteroids in plaque psoriasis: a systematic review of efficacy and treatment modalities. Journal of the European Academy of Dermatology and Venereology. 2012; 26: 36–46.

  16. Gabros S, Nessel TA, Zito PM. Topical corticosteroids, https://europepmc.org/books/nbk532940 (accessed 11 March 2023).

  17. Saleem MD, Negus D, Feldman SR. Topical 0.25% desoximetasone spray efficacy for moderate to severe plaque psoriasis: a randomized clinical trial. J Dermatolog Treat. 2018; 29: 32–35.

  18. Hogue L, Cardwell LA, Roach C, et al. Psoriasis and Atopic Dermatitis ‘Resistant’ to Topical Treatment Responds Rapidly to Topical Desoximetasone Spray. J Cutan Med Surg. 2019; 23: 157–163.

  19. Okwundu N, Cardwell L, Cline A, et al. Is topical treatment effective for psoriasis in patients who failed topical treatment? J Dermatolog Treat. 2021; 32: 41–44.

  20. Okwundu N, Cardwell LA, Cline AE, et al. Adherence to topical treatment can improve treatment-resistant moderate psoriasis. Cutis. 2020; 105: 89–91;E2;E3.

  21. Kerdel FA, Draelos ZD, Tyring SK, et al. A phase 2, multicenter, double-blind, randomized, vehicle-controlled clinical study to compare the safety and efficacy of a halobetasol propionate 0.01% lotion and halobetasol propionate 0.05% cream in the treatment of plaque psoriasis. J Dermatolog Treat. 2019; 30: 333–339.

  22. Brazzini B, Pimpinelli N. New and established topical corticosteroids in dermatology: clinical pharmacology and therapeutic use. Am J Clin Dermatol. 2002; 3: 47–58.

  23. Shah KN. Diagnosis and treatment of pediatric psoriasis: current and future. Am J Clin Dermatol. 2013; 14: 195–213.

  24. Murase JE, Heller MM, Butler DC. Safety of dermatologic medications in pregnancy and lactation. Journal of the American Academy of Dermatology. 2014; 70: 401.e1–401.e14.

  25. Felix K, Unrue E, Inyang M, et al. Patients preferences for different corticosteroid vehicles are highly variable. J Dermatolog Treat. 2020; 31: 147–151.

  26. Tolaymat L, Hall MR. Perioral Dermatitis. In: StatPearls. Treasure Island (FL): StatPearls Publishing, 2022.

  27. Daniel BS, Orchard D. Ocular side–effects of topical corticosteroids: what a dermatologist needs to know. Australas J Dermatol. 2015; 56: 164–169.

  28. Hengge UR, Ruzicka T, Schwartz RA, et al. Adverse effects of topical glucocorticosteroids. J Am Acad Dermatol. 2006; 54: 1–15; quiz 16–8.

  29. Abraham A, Roga G. Topical steroid-damaged skin. Indian J Dermatol. 2014; 59: 456–459.

  30. Castela E, Archier E, Devaux S, et al. Topical corticosteroids in plaque psoriasis: a systematic review of risk of adrenal axis suppression and skin atrophy. J Eur Acad Dermatol Venereol. 2012; 26 Suppl 3: 47–51.

  31. Broersen LHA, Pereira AM, Jørgensen JOL, et al. Adrenal Insufficiency in Corticosteroids Use: Systematic Review and Meta-Analysis. J Clin Endocrinol Metab. 2015; 100: 2171–2180.

  32. Health Canada. Health Product InfoWatch – July 2022, https://www.canada.ca/en/health-canada/services/drugs-health-products/medeffect-canada/health-product-infowatch/july-2022.html (2022, accessed 7 May 2023).

  33. Feldman SR. Tachyphylaxis to topical corticosteroids: the more you use them, the less they work? Clin Dermatol. 2006; 24: 229–30; discussion 230.

  34. Svendsen MT, Andersen KE, Andersen F, et al. Psoriasis patients’ experiences concerning medical adherence to treatment with topical corticosteroids. Psoriasis (Auckl). 2016; 6: 113–119.

  35. Lebwohl MG. The evolution of vitamin D analogues for the treatment of psoriasis. Arch Dermatol. 1995; 131: 1323.

  36. Lung BE, Mowery ML, Komatsu DEE. Calcitriol, https://europepmc.org/books/nbk526025 (accessed 4 March 2023).

  37. Devaux S, Castela A, Archier E, et al. Topical vitamin D analogues alone or in association with topical steroids for psoriasis: a systematic review. J Eur Acad Dermatol Venereol. 2012; 26 Suppl 3: 52–60.

  38. Ashcroft DM, Po AL, Williams HC, et al. Systematic review of comparative efficacy and tolerability of calcipotriol in treating chronic plaque psoriasis. BMJ. 2000; 320: 963–967.

  39. Naldi L, Rzany B. Psoriasis (chronic plaque). BMJ Clin Evid; 2009, https://www.ncbi.nlm.nih.gov/pubmed/19445765 (2009).

  40. Kravvas G, Gholam K. Use of topical therapies for pediatric psoriasis: A systematic review. Pediatr Dermatol. 2018; 35: 296–302.

  41. Mason AR, Mason J, Cork M, et al. Topical treatments for chronic plaque psoriasis. Cochrane Database of Systematic Reviews. Epub ahead of print 2013. DOI: 10.1002/14651858.cd005028.pub3.

  42. Feldman SR, Matheson R, Bruce S, et al. Efficacy and safety of calcipotriene 0.005% foam for the treatment of plaque-type psoriasis: results of two multicenter, randomized, double-blind, vehicle-controlled, phase III clinical trials. Am J Clin Dermatol. 2012; 13: 261–271.

  43. Kircik L. Efficacy and safety of topical calcitriol 3 microg/g ointment, a new topical therapy for chronic plaque psoriasis. J Drugs Dermatol. 2009; 8: s9–16.

  44. Ortonne JP, Humbert P, Nicolas JF, et al. Intra-individual comparison of the cutaneous safety and efficacy of calcitriol 3 microg g(-1) ointment and calcipotriol 50 microg g(-1) ointment on chronic plaque psoriasis localized in facial, hairline, retroauricular or flexural areas. Br J Dermatol. 2003; 148: 326–333.

  45. Beckenbach L, Baron JM, Merk HF, et al. Retinoid treatment of skin diseases. Eur J Dermatol. 2015; 25: 384–391.

  46. Heath MS, Sahni DR, Curry ZA, et al. Pharmacokinetics of tazarotene and acitretin in psoriasis. Expert Opin Drug Metab Toxicol. 2018; 14: 919–927.

  47. Samarasekera EJ, Sawyer L, Wonderling D, et al. Topical therapies for the treatment of plaque psoriasis: systematic review and network meta-analyses. Br J Dermatol. 2013; 168: 954–967.

  48. Guenther L, Lynde C, Poulin Y. Off-Label Use of Topical Calcineurin Inhibitors in Dermatologic Disorders. J Cutan Med Surg. 2019; 23: 27S–34S.

  49. Safarini OA, Patel J. Calcineurin inhibitors, https://europepmc.org/article/nbk/nbk558995 (2020).

  50. Dattola A, Silvestri M, Bennardo L, et al. Update of calcineurin inhibitors to treat inverse psoriasis: A systematic review. Dermatol Ther. 2018; 31: e12728.

  51. Beck KM, Yang EJ, Sanchez IM, et al. Treatment of Genital Psoriasis: A Systematic Review. Dermatol Ther. 2018; 8: 509–525.

  52. Wang C, Lin A. Efficacy of topical calcineurin inhibitors in psoriasis. J Cutan Med Surg. 2014; 18: 8–14.

  53. Hendriks AGM, Keijsers RRMC, de Jong EMGJ, et al. Combinations of classical time-honoured topicals in plaque psoriasis: a systematic review. J Eur Acad Dermatol Venereol. 2013; 27: 399–410.

  54. Gribetz C, Ling M, Lebwohl M, et al. Pimecrolimus cream 1% in the treatment of intertriginous psoriasis: a double-blind, randomized study. J Am Acad Dermatol. 2004; 51: 731–738.

  55. De Simone C, Maiorino A, Tassone F, et al. Tacrolimus 0.1% ointment in nail psoriasis: a randomized controlled open-label study. J Eur Acad Dermatol Venereol. 2013; 27: 1003–1006.

  56. Carr WW. Topical calcineurin inhibitors for atopic dermatitis: review and treatment recommendations. Paediatr Drugs. 2013; 15: 303–310.

  57. Arana A, Pottegård A, Kuiper JG, et al. Long-Term Risk of Skin Cancer and Lymphoma in Users of Topical Tacrolimus and Pimecrolimus: Final Results from the Extension of the Cohort Study Protopic Joint European Longitudinal Lymphoma and Skin Cancer Evaluation (JOELLE). Clin Epidemiol. 2021; 13: 1141–1153.

  58. Canada LP. The Product Monograph for Protopic® has been updated. PR Newswire, https://www.newswire.ca/news-releases/the-product-monograph-for-protopic-r-has-been-updated-830094216.html (2021, accessed 30 March 2023).

  59. Heymann WR. ‘Tar smarts’ may have a new meaning for atopic dermatitis and psoriasis. Journal of the American Academy of Dermatology. 2019; 80: 56–57.

  60. Roelofzen JHJ, Aben KKH, Oldenhof UTH, et al. No Increased Risk of Cancer after Coal Tar Treatment in Patients with Psoriasis or Eczema. J Invest Dermatol. 2010; 130: 953–961.

  61. Ávalos-Viveros M, Esquivel-García R, García-Pérez M, et al. Updated view of tars for psoriasis: what have we learned over the last decade? Int J Dermatol. 2023; 62: 290–301.

  62. Yan R, Jiang S, Wu Y, et al. Topical calcipotriol/betamethasone dipropionate for psoriasis vulgaris: A systematic review. Indian J Dermatol Venereol Leprol. 2016; 82: 135–144.

  63. Jacobi A, Mayer A, Augustin M. Keratolytics and emollients and their role in the therapy of psoriasis: a systematic review. Dermatol Ther. 2015; 5: 1–18.

  64. Mason J, Mason AR, Cork MJ. Topical preparations for the treatment of psoriasis: a systematic review. Br J Dermatol. 2002; 146: 351–364.

  65. Li X, Yang Q, Zheng J, et al. Efficacy and safety of a topical moisturizer containing linoleic acid and ceramide for mild-to-moderate psoriasis vulgaris: A multicenter randomized controlled trial. Dermatol Ther. 2020; 33: e14263.

  66. Liu M, Li X, Chen X-Y, et al. Topical application of a linoleic acid-ceramide containing moisturizer exhibit therapeutic and preventive benefits for psoriasis vulgaris: a randomized controlled trial. Dermatol Ther. 2015; 28: 373–382.

  67. Madan RK, Levitt J. A review of toxicity from topical salicylic acid preparations. J Am Acad Dermatol. 2014; 70: 788–792.

  68. Zhao Y, Asahina A, Asawanonda P, et al. Systematic review and practical guidance on the use of topical calcipotriol and topical calcipotriol with betamethasone dipropionate as long-term therapy for mild-to-moderate plaque psoriasis. J Dermatol. 2021; 48: 940–960.

  69. Leonardi C, Bagel J, Yamauchi P, et al. Efficacy and Safety of Calcipotriene Plus Betamethasone Dipropionate Aerosol Foam in Patients With Psoriasis Vulgaris–a Randomized Phase III Study (PSO-FAST). J Drugs Dermatol. 2015; 14: 1468–1477.

  70. Lebwohl M, Tyring S, Bukhalo M, et al. Fixed Combination Aerosol Foam Calcipotriene 0.005% (Cal) Plus Betamethasone Dipropionate 0.064% (BD) is More Efficacious than Cal or BD Aerosol Foam Alone for Psoriasis Vulgaris: A Randomized, Double-blind, Multicenter, Three-arm, Phase 2 Study. J Clin Aesthet Dermatol. 2016; 9: 34–41.

  71. Koo J, Tyring S, Werschler WP, et al. Superior efficacy of calcipotriene and betamethasone dipropionate aerosol foam versus ointment in patients with psoriasis vulgaris – A randomized phase II study. Journal of Dermatological Treatment. 2016; 27: 120–127.

  72. Paul C, Stein Gold L, Cambazard F, et al. Calcipotriol plus betamethasone dipropionate aerosol foam provides superior efficacy vs. gel in patients with psoriasis vulgaris: randomized, controlled PSO-ABLE study. Journal of the European Academy of Dermatology and Venereology. 2017; 31: 119–126.

  73. Stein Gold L, Paul C, Romiti R. Efficacy and safety of fixed-dose combination calcipotriol/betamethasone dipropionate foam for the treatment of psoriasis. J Eur Acad Dermatol Venereol. 2021; 35 Suppl 1: 10–19.

  74. Ortonne J-P, Esposito M, Chimenti S, et al. Betamethasone valerate dressing is non-inferior to calcipotriol-betamethasone dipropionate ointment in the treatment of patients with mild-to-moderate chronic plaque psoriasis: results of a randomized assessor-blinded multicentre trial. J Eur Acad Dermatol Venereol. 2014; 28: 1226–1234.

  75. Queille-Roussel C, Rosen M, Clonier F, et al. Efficacy and Safety of Calcipotriol Plus Betamethasone Dipropionate Aerosol Foam Compared with Betamethasone 17-Valerate-Medicated Plaster for the Treatment of Psoriasis. Clin Drug Investig. 2017; 37: 355–361.

  76. Langley RGB, Gupta A, Papp K, et al. Calcipotriol plus betamethasone dipropionate gel compared with tacalcitol ointment and the gel vehicle alone in patients with psoriasis vulgaris: a randomized, controlled clinical trial. Dermatology. 2011; 222: 148–156.

  77. Queille-Roussel C, Hoffmann V, Ganslandt C, et al. Comparison of the antipsoriatic effect and tolerability of calcipotriol-containing products in the treatment of psoriasis vulgaris using a modified psoriasis plaque test. Clin Drug Investig. 2012; 32: 613–619.

  78. Fulmali S, Thurka S, Sultana R, et al. A comparative study on efficacy of treatment regimens with Calcipotriol/Betamethasone Dipropionate Vs Calcipotriol ointments in Plaque type Psoriasis patients at Tertiary Care Hospital, Telangana. Asian Journal of Pharmacy and Pharmacology. 2022; 8: 58–65.

  79. Liu L, Zhang C, Wang J, et al. Comparison of safety and efficacy between calcipotriol plus betamethasone dipropionate gel and calcipotriol scalp solution as long-term treatment for scalp psoriasis in Chinese patients: a national, multicentre, prospective, randomized, active-controlled phase 4 trial. Eur J Dermatol. 2020; 30: 580–590.

  80. Menter A, Gold LS, Bukhalo M, et al. Calcipotriene plus betamethasone dipropionate topical suspension for the treatment of mild to moderate psoriasis vulgaris on the body: a randomized, double-blind, vehicle-controlled trial. J Drugs Dermatol. 2013; 12: 92–98.

  81. Queille-Roussel C, Olesen M, Villumsen J, et al. Efficacy of an innovative aerosol foam formulation of fixed combination calcipotriol plus betamethasone dipropionate in patients with psoriasis vulgaris. Clin Drug Investig. 2015; 35: 239–245.

  82. Gold LS, Green L, Dhawan S, et al. A Phase 3, Randomized Trial Demonstrating the Improved Efficacy and Patient Acceptability of Fixed Dose Calcipotriene and Betamethasone Dipropionate Cream. Journal of Drugs in Dermatology. 2021; 20: 420–425.

  83. Hong C-H, Papp KA, Lophaven KW, et al. Patients with psoriasis have different preferences for topical therapy, highlighting the importance of individualized treatment approaches: randomized phase IIIb PSO-INSIGHTFUL study. J Eur Acad Dermatol Venereol. 2017; 31: 1876–1883.

  84. Sandoval LF, Huang KE, Harrison J, et al. Calcipotriene 0.005%–betamethasone dipropionate 0.064% ointment versus topical suspension in the treatment of plaque psoriasis: a randomized pilot study of patient preference. Cutis. 2014; 94: 304–309.

  85. Iversen L, Jakobsen HB. Patient Preferences for Topical Psoriasis Treatments are Diverse and Difficult to Predict. Dermatol Ther. 2016; 6: 273–285.

  86. Lambert J, Hol CW, Vink J. Real-life effectiveness of once-daily calcipotriol and betamethasone dipropionate gel vs. ointment formulations in psoriasis vulgaris: final analysis of the 52-week PRO-long study. J Eur Acad Dermatol Venereol. 2015; 29: 2349–2355.

  87. Lebwohl M, Kircik L, Lacour J-P, et al. Twice-weekly topical calcipotriene/betamethasone dipropionate foam as proactive management of plaque psoriasis increases time in remission and is well tolerated over 52 weeks (PSO-LONG trial). Journal of the American Academy of Dermatology. 2021; 84: 1269–1277.

  88. Hendriks AGM, Keijsers RRMC, de Jong EMGJ, et al. Efficacy and safety of combinations of first-line topical treatments in chronic plaque psoriasis: a systematic literature review. J Eur Acad Dermatol Venereol. 2013; 27: 931–951.

  89. Ramachandran V, Bertus B, Bashyam AM, et al. Treating Psoriasis With Halobetasol Propionate and Tazarotene Combination: A Review of Phase II and III Clinical Trials. Ann Pharmacother. 2020; 54: 872–878.

  90. Chen H, Sun J, Yang H, et al. Fixed combination of tazarotene and betamethasone dipropionate for treatment of psoriasis vulgaris: The result of a phase 3, multicenter, randomized controlled trial. J Dermatol. 2020; 47: 728–734.

  91. He C, Jin H, Liu X, et al. Tazarotene/Betamethasone Dipropionate Cream in Patients with Plaque Psoriasis: Results of a Prospective, Multicenter, Observational Study. Dermatology. 2021; 237: 603–610.

  92. Lebwohl MG, Stein Gold L, Papp K, et al. Long-term safety and efficacy of a fixed-combination halobetasol propionate 0.01%/tazarotene 0.045% lotion in moderate-to-severe plaque psoriasis: phase 3 open-label study. J Eur Acad Dermatol Venereol. 2021; 35: 1152–1160.

  93. Alexis AF, Desai SR, Han G, et al. Fixed-Combination Halobetasol Propionate and Tazarotene Lotion for Psoriasis in Patients With Skin of Color. J Drugs Dermatol. 2021; 20: 744.

  94. Halobetasol 0.01%/Tazarotene 0.045% Lotion in the Treatment of Moderate-to-Severe Plaque Psoriasis: Maintenance of Therapeutic Effect After Cessation of Therapy. J Drugs Dermatol. 2019; 18: 815–820.

  95. Stein Gold L, Kircik LH, Pariser D, et al. Rapid Onset of Action in Patients With Moderate-to-Severe Plaque Psoriasis With Halobetasol 0.01%/Tazarotene 0.045% Fixed Combination. J Drugs Dermatol. 2018; 17: 863–868.

  96. Pariser DM, Green LJ, Stein Gold L, et al. Halobetasol 0.01%/Tazarotene 0.045% Lotion in the Treatment of Moderate-to-Severe Plaque Psoriasis: Maintenance of Therapeutic Effect After Cessation of Therapy. J Drugs Dermatol. 2018; 17: 723–726.

  97. Gold LS, Lebwohl MG, Sugarman JL, et al. Safety and efficacy of a fixed combination of halobetasol and tazarotene in the treatment of moderate-to-severe plaque psoriasis: results of 2 phase 3 randomized controlled trials. J Am Acad Dermatol. 2018; 79: 287–293.

  98. Sugarman JL, Gold LS, Lebwohl MG, et al. A Phase 2, Multicenter, Double-Blind, Randomized, Vehicle Controlled Clinical Study to Assess the Safety and Efficacy of a Halobetasol/Tazarotene Fixed Combination in the Treatment of Plaque Psoriasis. J Drugs Dermatol. 2017; 16: 197–204.

  99. Kircik LH, Papp KA, Stein Gold L, et al. Assessing the Synergistic Effect of a Fixed Combination Halobetasol Propionate 0.01% and Tazarotene 0.045% Lotion in Moderate-to-Severe Plaque Psoriasis. J Drugs Dermatol. 2019; 18: 279–284.

  100. Del Rosso JQ, Kircik L, Lin T, et al. Halobetasol 0.01%/Tazarotene 0.045% Fixed-combination Lotion in the Treatment of Plaque Psoriasis: Sensitization and Irritation Potential. J Clin Aesthet Dermatol. 2019; 12: 11–15.

  101. Pinter A, van de Kerkhof P. The role of topical therapies along the psoriasis patient journey: An overview from the Symposium ‘Tailoring topical psoriasis treatments to patients’ needs and expectations’ of the 30th EADV Congress 2021. J Eur Acad Dermatol Venereol. 2023; 37 Suppl 1: 3–8.

  102. Milakovic M, Gooderham MJ. Phosphodiesterase-4 Inhibition in Psoriasis. Psoriasis (Auckl). 2021; 11: 21–29.

  103. Psomadakis CE, Han G. New and Emerging Topical Therapies for Psoriasis and Atopic Dermatitis. J Clin Aesthet Dermatol. 2019; 12: 28–34.

  104. Lebwohl MG, Papp KA, Stein Gold L, et al. Trial of Roflumilast Cream for Chronic Plaque Psoriasis. N Engl J Med. 2020; 383: 229–239.

  105. Stein Gold L, Alonso-Llamazares J, Draelos ZD, et al. Effect of Roflumilast Cream (ARQ-151) on Itch and Itch-Related Sleep Loss in Adults with Chronic Plaque Psoriasis: Patient-Reported Itch Outcomes of a Phase 2b Trial. Am J Clin Dermatol. 2023; 24: 305–313.

  106. Snape SD, Wigger-Alberti W, Goehring UM. A phase I randomized trial to assess the effect on skin infiltrate thickness and tolerability of topical phosphodiesterase inhibitors in the treatment of psoriasis vulgaris using a modified psoriasis plaque test. Br J Dermatol. 2016; 175: 479–486.

  107. Lebwohl MG, Kircik LH, Moore AY, et al. Effect of Roflumilast Cream vs Vehicle Cream on Chronic Plaque Psoriasis: The DERMIS-1 and DERMIS-2 Randomized Clinical Trials. JAMA; 328. Epub ahead of print 20 September 2022. DOI: 10.1001/jama.2022.15632.

  108. Bissonnette R, Stein Gold L, Rubenstein DS, et al. Tapinarof in the treatment of psoriasis: A review of the unique mechanism of action of a novel therapeutic aryl hydrocarbon receptor–modulating agent. J Am Acad Dermatol. 2021; 84: 1059–1067.

  109. Lebwohl MG, Stein Gold L, Strober B, et al. Phase 3 Trials of Tapinarof Cream for Plaque Psoriasis. N Engl J Med. 2021; 385: 2219–2229.

  110. Cai L, Chen G-H, Lu Q-J, et al. A double-blind, randomized, placebo- and positive-controlled phase III trial of 1% benvitimod cream in mild-to-moderate plaque psoriasis. Chin Med J. 2020; 133: 2905–2909.

  111. Papp K, Gulliver W, Lynde C, et al. Canadian guidelines for the management of plaque psoriasis: overview. J Cutan Med Surg. 2011; 15: 210–219.

  112. Svendsen MT, Andersen F, Hansen J, et al. Medical adherence to topical corticosteroid preparations prescribed for psoriasis: A systematic review. J Dermatolog Treat. 2017; 28: 32–39.

  113. Carroll CL, Feldman SR, Camacho FT, et al. Better medication adherence results in greater improvement in severity of psoriasis. Br J Dermatol. 2004; 151: 895–897.

  114. Balato N, Megna M, Di Costanzo L, et al. Educational and motivational support service: a pilot study for mobile-phone-based interventions in patients with psoriasis. Br J Dermatol. 2013; 168: 201–205.

  115. Svendsen MT, Andersen F, Andersen KH, et al. A smartphone application supporting patients with psoriasis improves adherence to topical treatment: a randomized controlled trial. Br J Dermatol. 2018; 179: 1062–1071.

  116. Hawkins SD, Barilla S, Feldman SR. Web app based patient education in psoriasis – a randomized controlled trial. Dermatol Online J; 23, https://www.ncbi.nlm.nih.gov/pubmed/28541882 (2017).

  117. Svendsen MT, Feldman SR, Mejldal A, et al. Regular support provided by dermatological nurses improves outcomes in patients with psoriasis treated with topical drugs: a randomized controlled trial. Clin Exp Dermatol. 2022; 47: 2208–2221.

  118. Alinia H, Moradi Tuchayi S, Smith JA, et al. Long-term adherence to topical psoriasis treatment can be abysmal: a 1-year randomized intervention study using objective electronic adherence monitoring. Br J Dermatol. 2017; 176: 759–764.

  119. Reich K, Zschocke I, Bachelez H, et al. A Topical Treatment Optimization Programme (TTOP) improves clinical outcome for calcipotriol/betamethasone gel in psoriasis: results of a 64-week multinational randomized phase IV study in 1790 patients (PSO-TOP). British Journal of Dermatology. 2017; 177: 197–205.

  120. Caldarola G, De Simone C, Moretta G, et al. Role of personalized medication training in improving efficacy and adherence to a topical therapy in psoriatic patients. J Dermatolog Treat. 2017; 28: 722–725.

  121. Danby SG, Draelos ZD, Gold LFS, et al. Vehicles for atopic dermatitis therapies: more than just a placebo. J Dermatolog Treat. 2022; 33: 685–698.

  122. Florek AG, Wang CJ, Armstrong AW. Treatment preferences and treatment satisfaction among psoriasis patients: a systematic review. Arch Dermatol Res. 2018; 310: 271–319.

  123. Koo K, Jeon C, Bhutani T. Beyond monotherapy: a systematic review on creative strategies in topical therapy of psoriasis. J Dermatolog Treat. 2017; 28: 702–708.

  124. Case Medical Research. Study to evaluate the safety and antipsoriatic efficacy of BOS-475 in a psoriasis plaque test. Case Medical Research. Epub ahead of print 8 January 2020. DOI: 10.31525/ct1-nct04221906.

  125. Kernel Networks Inc. Study to evaluate the safety, tolerability, and pharmacokinetics of single and repeat topical administration of BOS-475 in healthy subjects and patients with psoriasis. Case Medical Research. Epub ahead of print 23 May 2019. DOI: 10.31525/ct1-nct03960450.

  126. A Trial of a Botanical Drug (EISO) for Treatment of Mild-to-Moderate Plaque Psoriasis, https://clinicaltrials.gov/ct2/show/results/NCT03000608?term=SAN021&cond=psoriasis&draw=2&rank=2 (accessed 25 April 2023).

  127. Safety Study of Ointment for the Treatment of Plaque-type Psoriasis – Full Text View – ClinicalTrials.gov, https://clinicaltrials.gov/ct2/show/NCT01258088?term=Crisaborole&cond=psoriasis&draw=2&rank=5 (accessed 25 April 2023).

  128. A Phase I Study to Assess Novel Cream in a Psoriasis Plaque Test, https://clinicaltrials.gov/ct2/show/NCT00763204?term=Crisaborole&cond=psoriasis&draw=2&rank=6 (accessed 25 April 2023).

  129. A Phase I Study to Assess Novel Ointment in a Psoriasis Plaque Test, https://clinicaltrials.gov/ct2/show/NCT00762658?term=Crisaborole&cond=psoriasis&draw=2&rank=7 (accessed 25 April 2023).

  130. AN2728 Topical Ointment to Treat Mild-to-Moderate Plaque-Type Psoriasis, https://clinicaltrials.gov/ct2/show/NCT01300052?term=Crisaborole&cond=psoriasis&draw=2&rank=1 (accessed 25 April 2023).

  131. Safety and Efficacy Study of A Novel Ointment to Treat Plaque Type Psoriasis, https://clinicaltrials.gov/ct2/show/NCT00759161?term=Crisaborole&cond=psoriasis&draw=2&rank=2 (accessed 25 April 2023).

  132. Safety and Efficacy Study of A Novel Ointment to Treat Plaque Type Psoriasis, https://clinicaltrials.gov/ct2/show/NCT00755196?term=Crisaborole&cond=psoriasis&draw=2&rank=3 (accessed 25 April 2023).

  133. Safety and Efficacy Study of a Novel Ointment to Treat Plaque Type Psoriasis – Full Text View – ClinicalTrials.Gov, https://clinicaltrials.gov/ct2/show/NCT01029405?term=Crisaborole&cond=psoriasis&draw=2&rank=4 (accessed 25 April 2023).

  134. A Intra-individual Comparison to Investigate the Efficacy and the Safety of LAS41004 Formulation in Mild to Moderate Psoriasis – No Study Results Posted – ClinicalTrials.Gov, https://clinicaltrials.gov/ct2/show/results/NCT02180464?term=LAS41004&cond=psoriasis&draw=2&rank=1 (accessed 25 April 2023).

  135. Exploratory Study to Investigate Efficacy of LAS41004 in a Psoriasis Plaque Test, https://clinicaltrials.gov/ct2/show/results/NCT01360944?term=LAS41004&cond=psoriasis&draw=2&rank=2 (accessed 25 April 2023).

  136. Clinical Trial to Investigate Efficacy of LAS41004 in Psoriasis, https://clinicaltrials.gov/ct2/show/results/NCT01283698?term=LAS41004&cond=psoriasis&draw=2&rank=3 (accessed 25 April 2023).

  137. Study to Investigate Dose-related Efficacy of LAS41004 in the Treatment of Psoriasis, https://clinicaltrials.gov/ct2/show/results/NCT01119339?term=LAS41004&cond=psoriasis&draw=2&rank=4 (accessed 25 April 2023).

  138. Efficacy, Tolerability and Safety of LAS41004 Formulations in a Psoriasis Plaque Test, https://clinicaltrials.gov/ct2/show/NCT02111499?term=LAS41004&cond=psoriasis&draw=2&rank=5 (accessed 25 April 2023).

  139. Efficacy and Tolerability of LAS41004 Formulations in a Non-occlusive Psoriasis Plaque Test – Full Text View – ClinicalTrials.Gov, https://clinicaltrials.gov/ct2/show/NCT01462643?term=LAS41004&cond=psoriasis&draw=2&rank=6 (accessed 25 April 2023).

  140. Randomized Study of PH-10 for Psoriasis, https://clinicaltrials.gov/ct2/show/results/NCT01247818?term=PH-10&cond=Psoriasis&draw=2&rank=1 (accessed 25 April 2023).

  141. A Phase 2 Study of Cellular and Immunologic Changes in the Skin of Subjects Receiving PH-10, https://clinicaltrials.gov/ct2/show/NCT02322086?term=PH-10&cond=Psoriasis&draw=2&rank=3 (accessed 25 April 2023).

  142. An Efficacy and Safety Study of PH-10 Aqueous Hydrogel for the Treatment of Plaque Psoriasis, https://clinicaltrials.gov/ct2/show/NCT00941278?term=PH-10&cond=Psoriasis&draw=2&rank=4 (accessed 25 April 2023).

  143. Safety, Tolerability and Efficacy of SNA-120 for Treatment of Pruritus and Psoriasis in Subjects Treated With Calcipotriene, https://clinicaltrials.gov/ct2/show/study/NCT03448081?term=SNA-120&cond=psoriasis&draw=2&rank=1 (accessed 25 April 2023).

  144. Safety, Efficacy, and Tolerability of SNA-120 in Subjects With Pruritus Associated With Psoriasis Vulgaris, https://clinicaltrials.gov/ct2/show/results/NCT03322137?term=SNA-120&cond=psoriasis&draw=2&rank=2 (accessed 25 April 2023).

  145. Case Medical Research. Safety and efficacy of ARQ-154 foam in adolescent and adult subjects with scalp and body psoriasis. Case Medical Research. Epub ahead of print 16 October 2019. DOI: 10.31525/ct1-nct04128007.

  146. Long-Term Safety of ARQ-151 Cream in Adult Subjects With Chronic Plaque Psoriasis, https://clinicaltrials.gov/ct2/show/NCT03764475?term=Roflumilast&cond=psoriasis&phase=1&draw=2&rank=3 (accessed 25 April 2023).

  147. Maximal Usage Pharmacokinetics and Safety of ARQ-151 in Children With Plaque Psoriasis (ARQ-151-216), https://clinicaltrials.gov/ct2/show/NCT04746911?term=Roflumilast&cond=psoriasis&phase=1&draw=2&rank=6 (accessed 25 April 2023).

  148. Maximal Usage Pharmacokinetics and Safety of ARQ-151 in Children With Plaque Psoriasis (ARQ-151-215), https://clinicaltrials.gov/ct2/show/NCT04655313?term=Roflumilast&cond=psoriasis&phase=1&draw=2&rank=7 (accessed 25 April 2023).

  149. Topical Roflumilast to Treat Scalp and Body Psoriasis (ARRECTOR) – No Study Results Posted – ClinicalTrials.Gov, https://clinicaltrials.gov/ct2/show/results/NCT05028582?term=Roflumilast&cond=psoriasis&phase=2&draw=2&rank=3 (accessed 25 April 2023).

  150. Open-Label Extension Trial of PDE4 Inhibition With Roflumilast for the Management of Plaque Psoriasis – Full Text View – ClinicalTrials.Gov, https://clinicaltrials.gov/ct2/show/NCT04286607?term=Roflumilast&cond=psoriasis&phase=2&draw=2&rank=4 (accessed 25 April 2023).

  151. Bioequivalence Study of Two Treatments for the Treatment of Plaque Psoriasis, https://clinicaltrials.gov/ct2/show/NCT05763082?term=Roflumilast&cond=psoriasis&phase=2&draw=2&rank=5 (accessed 25 April 2023).

  152. Long Term Study to Evaluate Safety and Efficacy of M518101 in Subjects With Plaque Psoriasis, https://clinicaltrials.gov/ct2/show/results/NCT01908595?term=M518101&cond=psoriasis&draw=2&rank=1 (accessed 25 April 2023).

  153. Efficacy Study Comparing Topical M518101 and Vitamin D3 in Adult Psoriasis Patients, https://clinicaltrials.gov/ct2/show/results/NCT01989429?term=M518101&cond=psoriasis&draw=2&rank=2 (accessed 25 April 2023).

  154. Evaluate the Efficacy and Safety of M518101 in Subjects With Plaque Psoriasis, https://clinicaltrials.gov/ct2/show/results/NCT01878461?term=M518101&cond=psoriasis&draw=2&rank=3 (accessed 25 April 2023).

  155. Study to Evaluate the Efficacy and Safety of M518101 in Subjects With Plaque Psoriasis, https://clinicaltrials.gov/ct2/show/results/NCT01873677?term=M518101&cond=psoriasis&draw=2&rank=4 (accessed 25 April 2023).

  156. A Maximal Use Trial Evaluating the Pharmacokinetic Profile of MC2-01 Cream, https://clinicaltrials.gov/ct2/show/results/NCT03462927?term=MC2-01&cond=psoriasis&draw=2&rank=2 (accessed 25 April 2023).

  157. Long Term Extension Study of Tapinarof for Plaque Psoriasis in Adults (3003), https://clinicaltrials.gov/ct2/show/results/NCT04053387?term=Tapinarof&cond=psoriasis&draw=2&rank=3 (accessed 25 April 2023).

  158. Tapinarof for the Treatment of Plaque Psoriasis in Pediatric Subjects, https://clinicaltrials.gov/ct2/show/NCT05172726?term=Tapinarof&cond=psoriasis&draw=2&rank=7 (accessed 25 April 2023).

  159. A Study to Investigate Efficacy and Safety of VTAMA (Tapinarof) Cream, 1% in Intertriginous Plaque Psoriasis, https://clinicaltrials.gov/ct2/show/NCT05680740?term=Tapinarof&cond=psoriasis&draw=2&rank=1 (accessed 25 April 2023).

  160. A Study to Investigate Efficacy and Safety of VTAMA® (Tapinarof) Cream, 1% in Plaque Psoriasis in the Head and Neck Region, https://clinicaltrials.gov/ct2/show/NCT05789576?term=Tapinarof&cond=psoriasis&draw=2&rank=2 (accessed 25 April 2023).


]]>
Update on Drugs & Devices: May – June 2023 https://www.skintherapyletter.com/drug-updates/may-june-2023/ Thu, 15 Jun 2023 17:31:51 +0000 https://www.skintherapyletter.com/?p=14413 Targeted alkali thermolysis (TAT) technology

Trade Name: Brella™ 3-miniute
SweatControl Patch™

Company: Candesant Biomedical

Approval Dates/Comments: In April 2023, the US FDA granted clearance to this single-use disposable patch to reduce excessive underarm sweating in adults with primary axillary hyperhidrosis (PAH). The product uses the company’s novel, patented TAT technology, which is based on the scientific principle of heat production when sodium comes in contact with water – the interaction generates a targeted amount of heat that triggers microthermal injury to the sweat gland, leading to temporary inactivation. Brella™ SweatControl Patch contains a sodium sheet with an adhesive overlay, whereby heat is generated when water in the sweat touches the sodium sheet. Treatment is administered in a doctor’s office and available by prescription only, with results lasting 3-4 months. The regulatory decision of Brella™ was based on the results of SAHARA, a randomized, double-blind, sham-controlled, multicenter clinical study in 110 adults with PAH. Subjects were given bilateral treatment with Brella™ or a sham patch for up to 3 minutes for each underarm; patients were evaluated weekly for 12 weeks post-treatment. The study met its primary and secondary endpoints, and the patch was well tolerated with no serious or severe adverse events reported.


Retifanlimab-dlwr for IV infusion

Trade Name: Zynyz
Company: Incyte Corporation

Approval Dates/Comments: The FDA granted accelerated approval for retifanlimab-dlwr, a humanized monoclonal antibody targeting programmed death receptor-1 (PD-1), in March 2023 for the treatment of adults with metastatic or recurrent locally advanced Merkel cell carcinoma (MCC). This regulatory decision was based on tumor response rate and duration of response. Continued approval may be contingent on verification and description of clinical benefit in confirmatory trials. Safety and efficacy were evaluated in PODIUM-201 (NCT03599713), an open-label, multiregional, single-arm study evaluating 65 patients with metastatic or recurrent locally advanced MCC who had not received prior systemic therapy for advanced disease. The major efficacy outcome measures were objective response rate (ORR) and duration of response (DOR). The ORR was 52% (95% CI: 40, 65) with complete response rate of 18%; 26 patients (76%) had a DOR ≥6 months and 21 (62%) had a DOR ≥12 months. The safety population consisted of 105 patients with MCC. The most common (≥10%) adverse reactions were fatigue, musculoskeletal pain, pruritus, diarrhea, rash, pyrexia, and nausea. Serious adverse reactions occurred in 22% of patients receiving retifanlimab.


Spesolimab IV for IV infusion

Trade Name: Spevigo®
Company: Boehringer Ingelheim

Approval Dates/Comments: Health Canada granted marketing authorization in March 2023 for spesolimab as first-in-class treatment for generalized pustular psoriasis (GPP) flares in adults. Spesolimab is a novel, humanized selective antibody that blocks the activation of the interleukin-36 receptor (IL-36R), a signalling pathway in the immune system shown to be involved in the pathogenesis of many autoinflammatory disorders, including GPP.


CRISPR-based therapy

Trade Name: NTLA-2002
Company: Intellia Therapeutics

Approval Dates/Comments: In March 2023, the FDA granted Regenerative Medicine Advanced Therapy designation to NTLA-2002 for the treatment of hereditary angioedema (HAE). NTLA-2002 is an in vivo CRISPR-based investigational therapy designed to inactivate the target gene, kallikrein B1 (KLKB1), to potentially prevent life-threatening attacks in people with HAE.


Purchase Article PDF for $1.99

]]>
Update on Drugs & Devices: March – April 2023 https://www.skintherapyletter.com/drug-updates/mar-apr-2023/ Thu, 30 Mar 2023 19:37:28 +0000 https://www.skintherapyletter.com/?p=14242 Abrocitinib tablet

Trade Name: Cibinqo®
Company: Pfizer Inc.

Approval Dates/Comments: The US FDA granted a label extension for oral abrocitinib in February 2023, expanding the approved indications to include adolescents (12 to <18 years of age) with refractory, moderate-to-severe atopic dermatitis whose disease is not adequately controlled with other systemic drug products, including biologics, or when use of those therapies is inadvisable. Abrocitinib was previously approved only for the treatment of adults 18 years of age and older.


Lanadelumab-flyo SC injection

Trade Name: Takhzyro®
Company: Takeda

Approval Dates/Comments: In February 2023, the FDA approved the expanded use of lanadelumab-flyo, a human monoclonal antibody that targets plasma kallikrein, for prophylaxis to prevent attacks of hereditary angioedema (HAE) in pediatric patients 2 to <12 years of age. Prior to this label extension, the only approved routine prophylaxis therapeutic options for children 6 to <12 years of age required dosing every 3 to 4 days, and children with HAE 2 to <6 years of age had no approved prophylaxis treatment, therefore this regulatory decision makes lanadelumab-flyo the first prophylaxis therapy for this age group.


Berdazimer gel, 10.3%

Trade Name: SB206
Company: Novan, Inc.

Approval Dates/Comments: A New Drug Application was submitted to the FDA in January 2023 seeking marketing approval for berdazimer gel, 10.3% for the topical treatment of molluscum contagiosum (MC). Berdazimer gel, 10.3% is in position to become a first-in-class topical treatment for MC. The active ingredient, berdazimer sodium, is a novel compound that releases nitric oxide and demonstrates antiviral activity. If approved, this drug would be the first effective and safe topical treatment indicated for MC with the convenience of self-application. Results from the pivotal Phase 3 B-SIMPLE4 clinical study evaluating berdazimer gel for the treatment of MC (NCT04535531) demonstrated a statistically significant improvement in the primary efficacy endpoint (p<0.0001), complete clearance of all lesions at week 12, and treatment was found to be well tolerated, with mild application site pain and mild to moderate erythema reported as the most common adverse events.


Methotrexate oral solution, 2 mg/mL

Trade Name: Jylamvo®
Company: Therakind

Approval Dates/Comments: In November 2022, the FDA approved this preparation of methotrexate (MTX) solution. The on-label indications are shared by the oral tablet formulations of MTX, which include severe, treatment‑refractory, disabling psoriasis which does not respond sufficiently to other forms of treatment (e.g., phototherapy, psoralen and ultraviolet A radiation [PUVA] therapy, and retinoids), adults with severe psoriatic arthritis, and adults with mycosis fungoides (cutaneous T-cell lymphoma) as a single agent or part of a combination chemotherapy regimen.


Anticancer plasmid DNA vaccine

Trade Name: ITI-3000
Company: Immunomic Therapeutics

Approval Dates/Comments: The FDA granted Fast Track designation in November 2022 to the ITI-3000 program for the treatment of Merkel cell carcinoma (MCC). ITI-3000, a plasmid DNA (pDNA) vaccine, is being investigated in a Phase 1, open label, first-in-human study to evaluate the safety, tolerability, and immunogenicity of 4 mg of ITI-3000 in patients with Merkel polyomavirus-positive MCC.

Purchase Article PDF for $1.99

]]>
Racial/Ethnic Variations in the Skin Barrier of Canadians: Implications for Skincare Recommendations Promoting a Healthy Skin Barrier and Mitigation of Atopic Dermatitis https://www.skintherapyletter.com/atopic-dermatitis/racial-ethnic-variations-skincare-recommendations/ Tue, 13 Dec 2022 23:52:57 +0000 https://www.skintherapyletter.com/?p=13954 Tan J MD FRCPC1, Alexis AF MD MPH FAAD2, Beach R MD FRCPC3, Andriessen A PhD4, Li M MD FRCPC5, Claveau J MD FRCPC DABD6, Asiniwasis R MD FRCPC7 

Affiliations


1Fellow of the Royal College of Physicians and Surgeons of Canada, Adjunct Professor, Schulich School of Medicine and Dentistry, Department of Medicine, Western University, Windsor, ON, Canada, President, Windsor Clinical Research Inc., Founder, The Healthy Image Centre, Windsor, ON, Canada.

2Professor of Clinical Dermatology, Weill Cornell Medical College, New York, NY, USA.

3Founder, DermAtelier on Avenue Medical & Cosmetic Dermatology, Assistant professor, Division of Dermatology, Department of Medicine, University of Toronto, Toronto, On, Canada.

4Radboud UMC Nijmegen, Andriessen Consultants, Malden, The Netherlands.

5Division of Dermatology, University of Toronto, Toronto, Ontario, Canada; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Women’s College Hospital, Toronto, Ontario, Canada.

6Fellow of the Royal College of Physicians of Canada; Diplomate of the American Board of Dermatology; Associate Professor, Department of Medicine, Division of Dermatology, Laval University; Director Melanoma and Skin Cancer Clinic, Le Centre Hospitalier Universitaire de Québec, Hôtel-Dieu de Québec, Quebec City, QC, Canada.

7Division of Dermatology, University of Saskatchewan, Regina, SK, Canada.


Abstract

Background: Skin barrier differences and variations in the presentation of common dermatoses such as xerosis and atopic dermatitis (AD) have been reported in racial/ethnic Canadian patients. This review discusses skin barrier differences and explores the role of ceramide-containing skin care in promoting a healthy skin barrier and mitigating AD.
Methodology: A literature review and panel discussions followed by an online review were used to adopt five statements and recommendations to promote a healthy skin barrier in various racial/ethnic Canadian AD populations.
Results: The multifactorial pathogenesis of AD includes genetic and environmental factors that may vary among racial/ethnic and geographic populations. Studies comparing ethnic groups have reported variations in transepidermal water loss, skin lipid levels, and stratum corneum pH. However, these studies frequently have flaws. The panel agreed that essential skincare principles apply to all AD-affected patients regardless of racial/ethnic background.
Conclusion: Robust comparative studies are needed to help clinicians to tailor patient education and recommend routine skincare with gentle cleansers and moisturizers containing lipids for AD management regardless of disease severity and prescription treatment.

Acknowledgments: All authors participated in all the steps of the project, selection of the literature, and the review of the manuscript. All authors read and approved the final version of the manuscript.

Disclosures:
The authors disclosed receipt of an unrestricted educational grant from CeraVe Canada for support with the research of this work. The authors also received consultancy fees for their work on this project.

Keywords: Racial/ethnic skin barrier variations, skincare, atopic dermatitis

PDF Download


Background

Genetic and environmental factors influence the structure and function of the stratum corneum (SC) barrier.1 Approximately 30% of Canadians are estimated to be part of a fast-growing racial/ethnic population by 2031.2 However, morphology and descriptions of dermatoses are based on White patients and the historic assumption that most residents of Canada and the United States are of Northern European descent.2

Differences in the skin barrier properties and function and the presentation of common dermatoses such as xerosis and atopic dermatitis (AD) have been observed in subjects with richly pigmented skin compared to White subjects.1-6 Several studies have investigated SC differences between racial/ethnic skin, comparing SC properties of self-identified Black, White, and Asian skin.1 In one such study, White subjects had an intermediate barrier strength as evidenced by tape strippings, and Asians have been demonstrated to require the least number of tape strippings to disrupt the SC barrier.1 This finding indicates a weaker barrier strength and slower recovery from barrier damage in the Asian population, supporting the observation of sensitive skin seen in Asians.1

There are significant disparities in the prevalence and treatment of skin conditions across Canadian populations.2-6 The burden of AD is higher in racial/ethnic populations, and numerous barriers to treatment exist, including systemic and institutional racism, poverty, crowded housing conditions on reserves, access and cost of basic skincare regimens, and clean water access.2-4 Promoting a healthy skin barrier remains a particular challenge for Indigenous groups, who lack access to appropriate treatments and skincare.2-6

This review discusses skin barrier differences in various racial/ethnic Canadian populations and explores the role of ceramide-containing skin care in promoting a healthy skin barrier and mitigating AD.

Methods

A group of dermatologists assembled during the Dermatology Update conference on November 13, 2021, in Montreal, Quebec. The panel (advisors) [four Canadian dermatologists and one dermatologist from the US] reviewed skin barrier differences in various racial/ethnic Canadian groups exploring dermatology issues related to skin barrier integrity. Additionally, the advisors determined the relevance of skincare-containing ceramides comprising cleansers and moisturizers for these populations to promote a healthy skin barrier and mitigate AD. Finally, the advisors looked into patient and clinician education tools to promote a healthy skin barrier in various racial/ethnic Canadian populations.

The project used a modified Delphi process comprising face-to-face discussions followed by an online follow-up.7-9

Literature Review

Structured literature searches on PubMed and Google Scholar as secondary source of the English-language literature (2010 – September 2021) were conducted before the meeting on September 21 and 22, 2021. We searched for guidelines, consensus papers, clinical studies, and reviews describing skin barrier properties in various racial/ethnic Canadian populations and current best-practice in promoting a healthy skin barrier and mitigation of AD using ceramides containing non-prescription skincare cleansers and moisturizers. Excluded were papers with no original data (unless a review article was deemed relevant), or not dealing with racial/ethnic Canadian or skincare, and publication language other than English.

The Nomenclature Used for the Searches

Searches were performed for the main ethnic Canadian groups [Black, White, Asian and Indigenous populations] and ethnic regions in Canada.2-6 Indigenous is a preferred term within Canadian communities. It is an umbrella term that covers Aboriginal, Metis, and Inuit populations. The publications collected a range of demographic data, including ethnic origin. Demographic factors referred to the quantitative data relating to the study population and its composition, which allowed portions of the population to be broken down into subgroups for closer examination.2-6 Further searches included associations between these demographic factors and the biophysical nature of racial/ethnic skin, skin care practices, and AD treatment product use.

Search Terms

The searches explored present clinical guidelines, treatment options, and therapeutic approaches addressing racial/ethnic Canadian populations using the following terms:
Racial/ethnic Canadian populations AND AD prone skin, OR Black, White, Asian and Indigenous populations in Canada AND AD, OR racial/ethnic Canadians AND skin barrier physiology OR skin barrier function/dysfunction OR racial/ethnic Canadians AND depletion of stratum corneum lipids, OR racial/ethnic Canadians AND AD prevention, OR racial/ethnic Canadians AND AD treatment, OR Black, White, Asian and Indigenous populations in Canada AND mitigation of AD, OR racial/ethnic Canadian populations with AD/AD prone skin AND skincare, OR Black, White, Asian and Indigenous people in Canada AND cleansers OR moisturizers OR emollients OR ceramides OR ce¬ramide containing skincare OR racial/ethnic Canadian AD populations AND skincare efficacy OR safety OR tolerability OR skin irritation

The searches were performed by a dermatologist and a physician/scientist (reviewers). After selection, the publicatiodns were manually reviewed for additional resources.

Priority was given to studies on SC barrier function and the benefits of skincare using cleansers and moisturizers in racial/ethnic Canadian populations with AD or AD-prone skin.

The searches yielded 248 papers, and after excluding 173 articles [duplicates, poor quality, not about Canadian racial/ethnic AD populations or skincare], 75 remained, comprising 4 epidemiology, 4 quality of life (QoL) studies, 20 guidelines, consensus papers and systematic reviews, 19 reviews, 24 clinical studies, and 4 others.

Role of the Panel

The advisors used the literature review results, clinical experience, and expertise to adopt statements and recommendations. The results were integrated into the summary statements presented and discussed during the face-to-face meeting. For example, in a workshop, advisors divided into three groups to create a final set of summary statements about Canadians’ racial/ethnic differences in SC barrier structure and function and skincare for this population, working with 12 draft messages. The final five statements integrate the combined output from the workshop groups and post-meeting online reviews from individual advisors.

Results

Statement 1: The properties and conditions of the skin vary with body site and can be influenced by factors such as skin type, ethnicity, gender, or lifestyle.
Epidemiological data indicate a higher prevalence and severity of AD in racial/ethnic Canadian populations.6,10-13

A three months population survey of all children aged 2-12 years in the community in the First Nations reserve of Natuashish, Labrador, Canada, showed that of 182 examined children, 30 (16.5%) mainly (20/30) had moderate to severe AD.6 IgE levels in children with and without AD had average values at least ten-fold higher than other populations.6

A systematic review and meta-analysis extracted 21 studies [1990 to 2020] from three medical databases [Pubmed, Embase, and Web of Science] to examine the prevalence of AD, clinical manifestation, and risk factors among children and adolescents in the Arctic.10 The cumulative AD incidence was 23%, and the 1-year prevalence was 19%, with the highest incidence in Arctic Scandinavia, lower Greenland, and Russia.10 The review indicated that the risk for AD in indigenous children living in rural Arctic areas seems slightly lower.10 Although the systematic review looked at the Arctic regions and included indigenous peoples, it did not mention Canadians.

A further study [2018] showed an AD prevalence of 20.5%, with the highest prevalence recorded among grade-1 Inuit children at 25%, compared to 15.4% among mixed ethnicity and 14.3% among non-Inuit children.11 The variations in prevalence and risk factors of asthma, allergic rhinitis, and AD among the different ethnicities living in the same subarctic environment may be related to genetic, gene-environment interaction, or lifestyle factors.11

An international study of asthma and allergies using written questionnaires included 8334 adolescents aged 13 to 14 in Vancouver, Saskatoon, Winnipeg, Hamilton, and Halifax, Canada.12 Although AD was significantly more prevalent in Winnipeg (1.31; 1.01-1.69) and Vancouver (1.28; 1.04-1.58), the highest prevalence rates of allergic rhinoconjunctivitis or AD were not observed in the same regions as the highest prevalence rates of wheezing, suggesting dissimilar risk factors.12

A cross-sectional study in Europe and Canada on AD patient-reported burden of disease showed a substantial impact (pruritus, pain, loss of sleep, higher levels of anxiety and depression) which was highest in those with severe AD.13

A similar high burden of AD has been shown in studies from other countries.14-18

Statement 2: The literature suggests racial/ethnic variations in ceramide content, SC structure, and filaggrin mutations. Racial/ethnic differences in barrier structure and function have been observed between Black, White, Asian, and Indigenous populations. Differences in TEWL have also been reported, but data are conflicting, and further research is needed.

The multifactorial pathogenesis of AD includes genetic and environmental factors that may vary among racial/ethnic and geographic populations.19 Genetic and immunophenotypic differences between racial/ ethnic AD populations, such as lower rates of filaggrin gene mutations, have been described among Black populations.20-33 Studies involving small groups of East Asian and African American patients have identified differences in cytokine expression compared to European-American patients.20-33 A literature review on clinical and molecular features of AD found differences in filaggrin (FLG) loss-of-function mutations across various ethnic groups with AD.29 The authors noted that studies in European American compared to Asian American AD populations have consistently shown a higher prevalence of FLG loss-of-function mutations in up to 50% of European and 27% of Asian American patients, respectively.29,30 However, the association between FLG loss-of-function mutations and AD development in populations of African descent is unclear, and other genes may be involved in skin barrier dysfunction.30

A higher prevalence and persistence of AD has been noted in African American children and racial/ethnic disparities in health care utilization and access to therapies.22-30

However, most of the information on racial/ethnic and geographic AD population variations originates from the US and may only be partially applicable to Canadians.

Statement 3: Data on racial/ethnic differences in skin barrier structure and function are limited but suggest variations in some characteristics relevant to skincare.

A healthy skin barrier function depends on the complex interplay among SC pH, desquamation rate, and the appropriate ratio of intrinsic lipids.37-40 The lipids comprise approximately twenty percent of the volume of the healthy stratum corneum (SC) and are composed of CERs (40–50%), cholesterols (20-33%), and free fatty acids (7–13%).37-39 Further lipids include cholesterol-3-sulfate (0-7 %) and cholesteryl esters (0-20 %).37-40

The slightly acidic surface of healthy skin is required to mature and maintain the SC barrier, inhibiting the growth of pathogenic microorganisms.39 Skin acidification plays an important role in SC barrier maturation and the activation of enzymes involved in the extracellular processing of SC lipids.39 The SC pH influences barrier homeostasis, integrity and cohesion, and antimicrobial defense mechanisms.39

It is unclear why specific changes in CER composition do not seem to affect a healthy SC and why deficiency of specific CER species and alterations in fatty acid composition occur in certain skin diseases such as AD.41-45

There is some evidence that the skin barrier in Black skin contains fewer CERs and that the skin barrier in Asian skin is most vulnerable to disruption.1,19 A less cohesive skin barrier in Asian skin might help explain differences in trans-epidermal water loss in this population.1,19 The advisors suggested that studies correlating skin barrier structure to dysfunction in Asian skin (perhaps involving tape stripping) could provide insights. Skin barrier differences (lipids, less cohesive skin barrier) may contribute to ethnic differences in the prevalence of xerosis, pruritus, and AD.

Some individuals with AD may produce inadequate amounts of certain CERs.31,41-45 Many with AD or AD-prone skin exhibit baseline increases in TEWL even within their unaffected, normal-appearing skin.31,41-45 Racial and ethnic differences have been reported in the SC barrier function, including CERs content and TEWL.45

Conventional moisturizers contain occlusives, humectants, and emulsions.39 Newer classes of moisturizers designed to restore skin barrier defects include distinct ratios of lipids that resemble physiological compositions, such as CERs, cholesterol, and essential fatty acids.37-40

CER-containing moisturizers were found to benefit AD patients when used as mono, adjunctive, and maintenance treatment.19,37-39,46-52 Guidelines, algorithms, and consensus papers agreed that the use of moisturizers that contain lipids, such as CERs (or their precursors) reduces pruritus, helps control xerosis, and improve the dysfunctional skin barrier in AD patients.34-39,53

Other ingredients in moisturizers (i.e., virgin coconut oil, glycyrrhetinic acid, V. vinifera, shea butter, mineral water and hyaluronic acid) have also been recommended.54-59

A Canadian study including 47 patients with inflammatory dermatosis, applied thermal water and hyaluronic acid-containing moisturizer for 4 weeks as an adjunct to treatment and found a markedly improved skin condition.59

A systematic review of 92 randomized controlled trials on the efficacy and safety of moisturizers for AD showed that those containing a mixture of substances (urea, glycerin or glycyrrhetinic acid, ceramides) seem to have greater effectiveness than basic emollients.62

Additionally, regular moisturizer use improves pruritus frequently caused by AD.63
As the mainstay of treatment, moisturizers should be liberally applied both in AD-prone skin and AD.34-39, 53, 60-62 The moisturizer should be used at least twice daily directly after bathing and more frequently during acute flare-ups.34-39, 53 Further moisturizers must be suitable for the patient’s skin type, climate, humidity, and environmental conditions.36-39,53-55

The advisors agreed that focusing too much on minor ethnic variations in the skin barrier of AD-affected patients could interfere with essential skincare principles that apply to all skin types. Instead, concentrating on similarities while acknowledging the differences may be more helpful.

A Canadian algorithm for topical treatment of mild-to-moderate AD for adults and pediatric patients and US guidelines for topical treatment of AD include education and avoiding triggers.34,35 Routine skincare with gentle cleansers and moisturizers is considered an integral part of AD management regardless of disease severity and prescription treatment (Table 1).34-38

Table 1: Cleanser and moisturizer use

Cleansers
  • Use nonsoap cleansers (e.g., syndets, aqueous solutions), that are less allergenic, nonirritating, and fragrance-free with a pH between 4–6.
  • Soap-based cleansers should be avoided because they can cause xerosis and skin irritation.
  • Antiseptic-containing cleansers are not recommended due to the limited duration of action of antiseptics and limited clinical data regarding their effectiveness. 
  • Consider a bleach bath for specific cases such as infections.
  • After bathing, gently pad the skin with a soft towel, avoiding rubbing. Next, apply moisturizer while the skin is still moist (within 3 min).
Moisturizers
  • A moisturizer should be used at least twice daily and more frequently during acute flare-ups.
  • Consider patient tolerance and preferences for a moisturizer to enhance treatment adherence.
  • Cream-type moisturizers containing lipids are suitable, and during winter, higher lipid contents are preferred. 
  • During acute flare-ups, moisturizers should be used more frequently in conjunction with anti-inflammatory treatment and continued as maintenance therapy.

Statement 4: Skin barrier differences between racial/ethnic populations may contribute to variations in the prevalence and severity of atopic dermatitis, xerosis, and, pruritus. Environmental issues and disparities in access to care may also play a role.

Although some authors reported a direct relationship between the severity of AD and the degree of SC lipid depletion41-45, the evidence demonstrating an association between CER depletion and AD is inconclusive.19 Other factors may play a role in SC lipid depletion, and the reduced CER could be an epiphenomenon of AD.19

Epidemiological data indicate a higher prevalence and severity of AD in racial/ethnic Canadian populations; while studies do not support the assumption that skin barrier differences are a factor.26-31 It is presumed that the impact of the cold, dry climate throughout parts of Canada may play a role in skin barrier dysfunction amongst these populations at large.

Delays in diagnosis or underestimation of severity may occur in patients with richly pigmented skin due to knowledge gaps in recognizing morphologic features of AD across the spectrum of skin complexions and racial/ethnic populations.19,64-66 Patients with richly pigmented skin may present with variations in the appearance of erythema (Figure 1). AD lesions may appear reddish-brown, violaceous, gray, or hyperchromic rather than bright red (Figure 2). Perifollicular accentuation, papules, scaling, lichenification, and pigmentary changes may be more prominent (Figure 3 and Figure 4). As a consequence, patients with SOC may present with a more advanced stage of AD severely impacting their QoL.19

Racial/Ethnic Variations in the Skin Barrier of Canadians: Implications for Skincare Recommendations Promoting a Healthy Skin Barrier and Mitigation of Atopic Dermatitis - image
Figure 1: In richly pigmented skin the appearance of erythema may vary.
Photo courtesy of Dr. Rao
Racial/Ethnic Variations in the Skin Barrier of Canadians: Implications for Skincare Recommendations Promoting a Healthy Skin Barrier and Mitigation of Atopic Dermatitis - image
Figure 2: AD lesions may appear reddish-brown, violaceous, gray, or hyperchromic rather than bright red.
Photo courtesy of Dr. Rao
Racial/Ethnic Variations in the Skin Barrier of Canadians: Implications for Skincare Recommendations Promoting a Healthy Skin Barrier and Mitigation of Atopic Dermatitis - image
Figure 3: Perifollicular accentuation, papules, scaling, lichenification, and pigmentary changes may be more prominent. Photo courtesy of Dr. Rao
Racial/Ethnic Variations in the Skin Barrier of Canadians: Implications for Skincare Recommendations Promoting a Healthy Skin Barrier and Mitigation of Atopic Dermatitis - image
Figure 4: Perifollicular accentuation, papules, scaling, lichenification, and pigmentary changes may be more prominent. Photo courtesy of Dr. Rao

Canadian Indigenous children and young adults continue to face higher rates of health disparities than their non-Indigenous counterparts.2 In dermatology, this includes a high burden of AD and secondary skin infections.2,3 Environmental factors and disparities in access to care could be a particular challenge for Indigenous groups, who frequently lack access to appropriate treatments.2,3 A systematic review of the pediatric dermatology literature reported on systemic [finances, wait times, geography], sociocultural [culture beliefs and communication], and individual barriers [patient beliefs and health knowledge] to diagnosis, treatment, and maintenance approaches of AD and other skin conditions.67 The identified barriers are interesting to explore further in Canadian AD populations. However, further research is needed to obtain insight into any interventions’ impact on overcoming these barriers.

Awareness amongst AD patients and caretakers, specifically Indigenous groups, on the cause of AD, general treatment principles, available treatments and the role of moisturizers, and adherence to moisturizer regimens are inconsistent.68

Statement 5: Cultural perceptions of healthy skin impact the choice of skincare.

First Nations people have been using medicinal plants for AD treatment. Natural Indigenous medicinal discoveries [safrole, salicylic acid, and ascorbic acid derived from Sassafras albidum, genus Salix trees, and Sassafras officinale] by the Iroquoian and Algonquian-speaking Peoples of North America for AD and other dermatologic conditions are mentioned in the European literature.71 Further examples are Western red cedar’s known principal active compound, β-thujaplicin, has shown efficacy in AD.70 Another active principal compound (7-hydroxymatairesinol) of White spruce may offer benefits due to its anti-inflammatory activity.70 Plants and algae such as hazel may also have benefits; however, studies need to confirm this.71

The effect of traditional treatments and natural remedies for AD may be of interest in managing racial/ethnic Canadian AD populations.70,71 However, such AD treatments may result in adverse effects such as postinflammatory hyperpigmentation or keloid scarring at a higher rate than evidence-based treatment.19

Optimal management of AD is multipronged and includes patient education, prescription treatment, and skincare promoting a healthy skin barrier.68,72-74

Nurse practitioner or physician assistant interventions may significantly increase correct and frequent moisturizer use, reducing AD.76

The choice of skincare should be supported by evidence but is mainly a personal and individual choice.34,36-38,61,75

It is important to note that there are variations in skincare norms across diverse populations; therefore, these cultural variations when providing skincare recommendations need to be considered.19 Integrating evidence-based recommendations for skin care in a culturally competent manner that aligns with the patient’s norms/preferences is key to successful outcomes across diverse populations.2,19 More research is needed to guide culturally appropriate recommendations better.

Limitations

A detailed discussion on genetic factors of racial/ethnic Canadian AD populations is outside the scope of the review. There is an overall lack of robust studies focusing on the prevention, treatment, and maintenance of AD in racial/ethnic Canadian AD populations.

Conclusions

The multifactorial pathogenesis of AD includes genetic and environmental factors that may vary among racial/ethnic and geographic populations. Available data suggest that skincare strategies to improve AD patients’ outcomes should consider racial/ethnic differences, integrating recommendations for skin care in a culturally competent manner that aligns with the patient’s norms and preferences. Future robust comparative studies will help clinicians to tailor patient education and recommend routine skincare with gentle cleansers and moisturizers as an integral part of AD management.

 

References



  1. Muizzuddin N, Hellemans L, Van Overloop L, et al. Structural and functional differences in barrier properties of African American, Caucasian and East Asian skin. J Dermatol Sci. 2010;59(2):123-8. doi: 10.1016/j. jdermsci.2010.06.003. PubMed PMID: 20654785.

  2. Ogunyemi B, Miller-Monthrope Y. The state of ethnic dermatology in Canada. J Cutan Med Surg. 2017;21(5):464-466. Doi: 10.1177/1203475417711110

  3. Naetahe Asiniwasis R, Heck E, Amir Ali A et al. Atopic dermatitis and skin infections are a poorly documented crisis in Canada’s Indigenous pediatric population: It’s time to start the conversation. Pediatr Dermatol. 2021 Nov;38 Suppl 2:188-189. doi: 10.1111/pde.14759.

  4. Allan B, Smylie J. First Peoples, second class treatment: The role of racism in the health and well-being of Indigenous peoples in Canada. Wellesley Institute; 2015. wellesleyinstitute.com/wp-content/uploads/2015/02/Summary-First-Peoples-Second-Class-Treatment-Final.pdf

  5. Eglington T, Asiniwasis R, Pandey M et al. Saskatchewan rural communities and skin diseases: a health practitioner survey on dermatologic conditions seen in Saskatchewan’s remote indigenous communities. SK research showcase. 2020 skresearchshowcase.com/indigenous-health-research-e-poster-1/saskatchewan-rural-communities-and-skin-diseases%3A-a-health-practitioner-survey-on-dermatologic-conditions-seen-in-saskatchewan%27s-remote-indigenous-communities

  6. Forsey RGP. Prevalence of childhood eczema and food sensitization in the First Nations reserve of Natuashish, Labrador, Canada. BMC Pediatr. 2014; 14(1): 76. https://doi.org/10.1186/1471-2431-14-76

  7. Trevelyan EG, Robinson N. Delphi methodology in health research: how to do it? Eur J Integrative Med. 2015;7(4):423-428.

  8. Brouwers M, Kho ME, Browman GP, et al.; AGREE Next Steps Consortium. AGREE II: advancing guideline development, reporting and evaluation in healthcare. Can Med Association J. 2010,182:E839-42.

  9. Smith Begolka W, Elston DM, Beutner KR. American Academy of Dermatology evidence-based guideline development process: responding to new challenges and establishing transparency. J Am Acad Dermatol. 2011 Jun;64(6):e105-12. doi: 10.1016/j.jaad.2010.10.029.

  10. Andersson AM, Halling AS, Loft N et al. Atopic dermatitis among children and adolescents in the Arctic region – a systematic review and meta-analysis. J Eur Acad Dermatol Venerol. 2021;35(8):1642-1654. Doi.org/10.1111/jdv.17276

  11. Ahmed A, Hakim A, Becker A. Evaluation of eczema, asthma, allergic rhinitis and allergies among the Grade-1 children of Iqaluit. Allergy Asthma Clin Immunol. 2018;14:9. Doi:10.1186/s13223-018-0232-2

  12. Wang HY, Pizzichini MMM, Becker AB et al. (2010). Disparate geographic prevalences of asthma, allergic rhinoconjunctivitis, and atopic eczema among adolescents in five Canadian cities. Ped Allergy Immunol. 2010;21:867–877.

  13. De Bruin-Weller M, Gadkari A, Auziere EL et al. The patient-reported disease burden in adults with atopic dermatitis: a cross-sectional study in Europe and Canada. J Eur Acad Dermatol Venerol. 2019;34(5):1026-1036.

  14. Andersen L, Nyeland ME, Nyberg, F. Higher self-reported severity of atopic dermatitis is associated with poorer self-reported Health-Related Quality of Life in France, Germany, the UK, and the USA. Br J Dermatol. 2020 May;182(5):1176-1183.

  15. Cheng CM, Hsu JW, Huang KL et al. Risk of developing major depressive disorder and anxiety disorders among adolescents and adults with atopic dermatitis: A Nationwide longitudinal study. J Affective Disorders. 2015; 178, 60–65.

  16. Chrostowska-Plak D, Reich A, Szepietowski JC. (2012). Relationship Between Ich and Psychological Status of Patients with Atopic Dermatitis. J Eur Acad Dermatol Venereol. 2012;27:239–242.

  17. Halvorsen JA, Lien L, Dalgard Fet al. Suicidal ideation, mental health problems, and social function in adolescents with eczema: A population-based study. J Invest Dermatol 2014;134(7):1847–1854.

  18. Silverberg JI. Public Health Burden and Epidemiology of Atopic Dermatitis. Dermatol Clin; 2017;35:283–289.

  19. Alexis AF, Woolery-Lloyd H, Williams K, Andriessen A, Desai S, Han G, et al. Racial/ethnic variations in skin barrier: Implications for skin care recommendations in skin of color. J Drugs Dermatol 2021;20(9):1-7. doi:10.36849/JDD.6312]

  20. Fishbein AB, Silverberg JI, Wilson EJ, Ong PY. Update on atopic dermatitis: Diagnosis, severity assessment and treatment selection. J Allergy Clin Immunol Pract. 2020;8(1):91-101. doi: 10.1016/j.jaip.2019.06.044.

  21. Shaw TE, Currie GP, Koudelka CW, Simpson EL. Eczema prevalence in the United States: data from the 2003 National survey of children’s health. J Invest Dermatol. 2011;131(1):67-73. Doi: 10.1038/jid.2010.251.

  22. Janumpally SR, Feldman SR, Gupta AK, Fleischer AB. In the United States, black and Asian/Pacific Islanders are more likely than whites to seek medical care for atopic dermatitis. Arch Dematol. 2002;138(5):634-7.

  23. Wan J, Oganisian A, Spieker AJ, et al. Racial/ethnic variation in use of ambulatory and emergency care for atopic dermatitis among US children. J Investig Dermatol. 2019;139(9):1906-1913. PMCID: PMC8320361

  24. Kim Y, Blomberg M, Rifs-Shiman SL, Camargo C, Gold DR, Thyssen JP, et al. Racial/ethnic differences in incidence and persistence of childhood atopic dermatitis. J Invest Dermatol. 2019;139(4):827-834. Doi: 10.1016/j. jid.2018.10.029.

  25. Bell MA, Whang KA, Thomas J, Aguh C, Kwatra SG. Racial and ethnic disparities in access to emerging and frontline therapies in common dermatological conditions: A cross-sectional study. J Natl Med Assoc. 2020;112(6):650-653.

  26. Margolis DJ, Apter AJ, Gupta J, et al. The persistence of atopic dermatitis and fillagrin (FLG) mutations in a US longitudinal cohort. J Allergy Clin Immunol. 2012;130(4):912-7. PMCID: PMC3462287

  27. Leung DY. Atopic dermatitis: Age and race do matter! J Allergy Clin Immunol. 2015;136(5):1265-7. DOI: 10.1016/j.jaci.2015.09.011

  28. Sanyal RD, Pavel AB, Glickman J, Chan TC, Zheng X, Zhang N, et al. Atopic dermatitis in African American patients is TH2/TH22-skewed with TH1/TH17 attenuation. Ann Allergy Asthma Immunol. 2019 Jan;122(1):99-110.e6. doi: 10.1016/j.anai.2018.08.024. PMID: 30223113.

  29. Brunner PM, Gutterman-Yassky E. Racial differences in atopic dermatitis. Ann Allergy Asthma Immunol. 2019;122(5):449-455.

  30. Kaufman BP, Guttman-Yassky E, Alexis AF. Atopic dermatitis in diverse racial and ethnic group-Variations in epidemiology, genetics, clinical presentation, and treatment. Experi Dermatol. 2018;27:340-357.

  31. Muizzuddin N, Hellemans L, Van Overloop L, et al. Structural and functional differences in barrier properties of African American, Caucasian and East Asian skin. J Dermatol Sci. 2010;59(2):123-8. doi: 10.1016/j. jdermsci.2010.06.003.

  32. Wu, K.K., Nguyen, K.B., Sandhu, J.K., & Armstrong, A.W. (2019). Does Location Matter? Geographic Variations in Healthcare Resource Use for Atopic Dermatitis in the United States. J Dermatol Treatment. DOI: 10.1080/09546634.2019.1656796

  33. Chiesa Fuxench ZC, Block J, Boguniewicz M, et al. Atopic Dermatitis in America Study: a cross-sectional study examining the prevalence and disease burden of atopic dermatitis in the US adult population. Journal of Investigative Dermatology. October 2018. doi:10.1016/j.jid.2018.08.028

  34. Lynde CW, Bergman J, Fiorillo L, et al. Clinical insights about topical treatment of mild-to-moderate pediatric and adult atopic dermatitis. J Cutan Med Surg. 2019 May/Jun;23(3_suppl):3S-13S.

  35. Eichenfield LF, Tom WL, Berger TG, et al. Guidelines of care for the management of atopic dermatitis: section 2. Management and treatment of atopic dermatitis with topical therapies. J Am Acad Dermatol. 2014;71:116–132.[PMC free article] [PubMed] [Google Scholar]

  36. van Zuuren EJ, Fedorowicz Z, Christensen R, Lavrijsen A, Arents BWM. Emollients and moisturisers for eczema. Cochrane Database Syst Rev. 2017;2:CD012119. doi:10.1002/14651858.CD012119

  37. Schachner LA, Blume-Peytavi U, Andriessen A, et al. Expert consensus on ceramides containing skincare in newborns and infants and potential mitigation of atopic dermatitis. Ital J Dermatol Venerol. 2022(1). doi:10.23736/S2784-8671.21.07172-3.

  38. Schachner LA, Andriessen A, Benjamin L, et al. A Consensus about the Importance of Ceramide containing Skincare for Normal and Sensitive Skin conditions in Neonates and Infants. J Drugs Dermatol. 2020;19(8):769-776. DOI: 10.36849/jdd.2020.5252 PMID: 32845590

  39. Lynde CW, Tan J, Skotnicki S, Andriessen A, et al. Clinical insights about the role of skin pH in inflammatory dermatological conditions. J Drugs Dermatol. 2019;18(12)S-1:1-16.

  40. t’Kindt R, Jorge L, Dumont E, et al. Profiling and characterizing skin ceramides using reversed-phase liquid chromatography-quadrupole time-of-flight mass spectrometry. Anal Chem 2012;84:403–411.

  41. Sahle FF, Gebre-Mariam T, Dobner B, Wohlrab J, Neubert RH. Skin diseases associated with the depletion of stratum corneum lipids and stratum corneum lipid substitution therapy. Skin Pharmacol. Physiol 2015;28:42–55.

  42. Rerknimitr P, Otsuka A, Nakashima C, Kabashima K. Skin barrier function and atopic dermatitis. Curr Dermatol rep. 2018;7(4):209-220. doi:10.1007/s13671-018-0232-y

  43. Kim D, Lee NR, Park S-Y, et al. As in atopic dermatitis, non-lesional skin in allergic contact dermatitis displays abnormalities in barrier function and ceramide content. J Invest Dermatol. 2017;137(3):748-750. doi:10.1016/j.jid.2016.10.034

  44. Ito S, Ishikawa J, Naoe A, Yoshida H, et al. Ceramide synthase 4 is highly expressed in involved skin of patients with atopic dermatitis. J Eur Acad Dermatol Venereol 2017;31:135–141.

  45. Strugar TL, Kuo A, et al. Connecting the dots: From skin barrier dysfunction to allergic sensitization, and the role of moisturizers in repairing the skin barrier. J Drugs Dermatol. 2019;18(6):581.

  46. Kircik LH, Del Rosso JQ. Nonsteroidal treatment of atopic dermatitis in pediatric patients with a ceramide-dominant topical emulsion formulated with an optimized ratio of physiological lipids. J Clin Aesthet Dermatol. 2011;4(12):25-31.

  47. Lynde CW, Andriessen A. A cohort study on a ceramide-containing cleanser and moisturizer used for atopic dermatitis. Cutis 2014;93(4)207-2013.

  48. Draelos ZD, Baalbaki NH, Cook S, Raab S, Colon G. The effect of a ceramide-containing product on stratum corneum lipid levels in dry legs. J Drugs Dermatol. 2020;19(4)372-376.

  49. Zeichner JA, Del Rosso JQ. Multivesicular emulsion ceramide-containing moisturizers: an evaluation of their role in the management of common skin disorders. J Clin Aesthet Dermatol. 2016;9(12):26-32. PMID: 28210396 | PMCID: PMC5300724

  50. Lueangarun S, Tragulplaingam P, Sugkraroek S. The 24-hr, 28-day, and 7-day post-moisturizing efficacy of ceramides 1, 3, 6-II containing moisturizing cream on skin dryness and barrier disruption in senile xerosis treatment. Dermatol Ther. 2019;32(6)e13090. https://doi.org/10.1111/dth.13090.

  51. Danby SG, Andrew PV, Cork MJ et al. An investigation of the skin barrier restoring effects of a cream containing ceramides in a multivesicular emulsion in people with dry, eczema-prone, skin: The RESTORE study phase I. Dermatol Ther (Heidelb) 2020;(6) https://doi.org/10.1007/s13555-020-00426-3

  52. Danby SG, Andrew PV, Cork MJ et al. An investigation of the skin barrier restoring effects of a cream containing ceramides in a multivesicular emulsion in people with dry, eczema-prone, skin: The RESTORE study phase 2. J Am Acad Dermatol. 2020;83(6) Suppl, AB71; December 1, 2020. DOI: https://doi.org/10.1016/j.jaad.2020.06.373

  53. Del Rosso JQ, Harper J, Kircik L, et al. Consensus Recommendations on Adjunctive Topical Management of Atopic Dermatitis. J Drugs Dermatol. 2018;17:1070-1076.

  54. Rubel D, Thirumoorthy T, Soebaryo RW, Asia-Pacific Consensus Group for Atopic Dermatitis et al. Consensus guidelines for the management of atopic dermatitis: an Asia-Pacific perspective. J Dermatol. 2013;40:160–171.

  55. Chow S, Seow CS, Dizon MV, et al. Asian Academy of Dermatology and Venereology. A clinician’s reference guide for the management of atopic dermatitis in Asians. Asia Pac Allergy. 2018;8:e41.

  56. Comite Nacional de Dermatologia Atopic dermatitis: National consensus 2013. Arch Argent Pediatr. 2014;112:293–294.

  57. Wollenberg A, Barbarot S, Bieber T, European Dermatology Forum (EDF) The European Academy of Dermatology and Venereology (EADV) The European Academy of Allergy and Clinical Immunology (EAACI) et al. Consensus-based European guidelines for treatment of atopic eczema (atopic dermatitis) in adults and children: part I. J Eur Acad Dermatol Venereol. 2018;32:657–682.

  58. Wollenberg A, Barbarot S, Bieber T, European Dermatology Forum (EDF) The European Academy of Dermatology and Venereology (EADV) The European Academy of Allergy and Clinical Immunology (EAACI) et al. Consensus-based European guidelines for treatment of atopic eczema (atopic dermatitis) in adults and children: part II. J Eur Acad Dermatol Venereol. 2018;32:820–878.

  59. Hong CH, Schachter J, Sutton AB et al. 89% Vichy mineralizing water with hyaluronic acid is a well-tolerated adjunct treatment that helps restore skin barrier function in dry skin-related inflammatory dermatoses and post-procedure skin care: A Canadian study. J Cosmetic Dermatol. 2021;(3): doi.org/10.1111/jocd.14116

  60. Mayba JN, Gooderham MJ. Review of atopic dermatitis and topical therapies. J Cutan Med Surg 2016;12: doi.org.10.1177/1203475416685077

  61. Weinstein M, Barber K, Bergman J et al. Atopic dermatitis: A practical guide to management. Eczema Society of Canada 2020 (1). https://eczemahelp.ca/wp-content/uploads/hcp-resources/ESC-Atopic-Dermatitis-A-Practical-Guide-to-Management-HCP-Guideline-2020-PUBLISHED-1.pdf

  62. Micali G, Paterno V, Cannarella R et al. Evidence-based treatment of atopic dermatitis with topical moisturizers. G Ital Dermatol Venerol. 2018;153(3):396-402.

  63. Nowak D. Diagnosis and treatment of pruritus. Can Fam Physician. 2017;63(12:918-924

  64. Pandya AG, Alexis AF, Berger TG, Wintroub BU. Increasing racial and ethnic diversity in dermatology: A call to action. J Am Acad Dermatol. 2016 Mar;74(3):584–7.

  65. Lu JD, Tiwana S, Das P, Siddiqi J, Khosa F. Gender and racial underrepresentation in academic dermatology positions in the United States: a retrospective, cross-sectional study from 2007 to 2018. J Am Acad Dermatol. 2020 Nov;83(5):1513–6

  66. Onasanya J, Liu C. Dermatology education in skin of color: where we are and where we go. Can Med Educ J. 2021;12(6):124-125.

  67. Toy J, Gregory A, Rehmus W. Barriers to healthcare access in pediatric dermatology: A systematic review. Ped Dermatol. 2021;38(2):13-19

  68. Ansiniwasis R, Sajic D, Skotnicki S. Atopic dermatitis: The skin barrier and the role of ceramides. Skin Therapy Letter. 2011;7(5)

  69. Colantonio S, Rivers JK. Botanicals with dermatologic properties derived from First Nations healing: Part 1-trees. J Cutan Med Surg 2017;(2): doi.org/10.1177/1203475417690306

  70. Colantonio S, Rivers JK. Botanicals with dermatologic properties derived from First Nations healing: Part 2-Plants and Algae. J Cutan Med Surg. 2016;(12): doi.org/10.1177/1203475416683390

  71. Perlmuttler J, Cogan R, Wisman MC. Treatment of atopic dermatitis, dermatophytes, and syphilis by Indigenous peoples prior to 1850. J Cutan Med Surg. 2021;(11): doi.org/10.1177/12034754211058403.

  72. Howe W. Treatment of atopic dermatitis. UpToDate 2022(6) https://www.uptodate.com/contents/treatment-of-atopic-dermatitis-eczema

  73. Thomas R, Landells I, Lynde C. Canadian consensus on skin barrier repair therapy in atopic dermatitis. J Cutan Med Surg 2012 doi.org/10.2310/7750.2012.S1PEDIA

  74. Pohar R, McCormack S. Emollient Treatments for Atopic Dermatitis: A Review of Clinical Effectiveness, Cost-Effectiveness, and Guidelines. CADTH Rapid Response Report. Can Agency Drugs Technologies Health 2019 (1) ncbi.nlm.nih.gov/books/NBK544513.

  75. Noreen Heer N, Rippke F, Weber TM, Hebert AA. Daily moisturization fora topic dermatitis: Importance, recommendations, and moisturizer choices. J Nurse Pract. 2021;17(8):920-925.

  76. Lynde CW, Bergman J, Fiorillo L, et al. Clinical insights about topical treatment of mild-to-moderate pediatric and adult atopic dermatitis. J Cutan Med Surg. 2019 May/Jun;23(3_suppl):3S-13S.


]]>
CaSMO Management of Cutaneous Toxicities Associated with Immune Checkpoint Inhibitors: A Practical Primer https://www.skintherapyletter.com/supplement/casmo-management/ Tue, 06 Sep 2022 17:53:11 +0000 https://www.skintherapyletter.com/?p=13515 Maxwell B Sauder MD, FRCPC, DABD1, Joel Claveau MD FRCPC, DABD2, Marcus Butler MD3, Anneke Andriessen PhD4, Tarek Hijal MD, FRCPC5, Charles W Lynde MD, FRCPC6

1Diplomate of the American Board of Dermatology, Fellow of the Royal College of Physicians of Canada, Associate Professor, Department of Medicine University of Toronto, Onco-dermatologist, Princess Margaret Cancer Centre, Director, Toronto, ON, Canada.
2Fellow of the Royal College of Physicians of Canada, Diplomate of the American Board of Dermatology, Associate Professor, Department of Medicine, Division of Dermatology, Laval University, Director Melanoma and Skin Clinic, Le Centre Hospitalier Universitaire de Québec, Hôtel-Dieu de Québec, Quebec City, QC, Canada.
3Medical Oncologist, Medical Oncology Disease Site Lead for Melanoma/Skin Oncology, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre. Assistant Professor, Department of Medicine, University of Toronto, Associate Member, Department of Immunology, University of Toronto, Toronto, ON, Canada
4Radboud UMC, Nijmegen and Andriessen Consultants, Malden, The Netherlands.
5Fellow of the Royal College of Physicians of Canada, Associate Professor, Department of Oncology, McGill University; Director, Division of Radiation Oncology, McGill University Health Centre, Montreal, QC, Canada.
6Fellow of the Royal College of Physicians of Canada, Diplomate of the American Board of Dermatology, Associate Professor, Department of Medicine University of Toronto, Toronto, ON, Canada, Lynderm Research, Markham, ON, Canada.

Abstract
The Canadian skin management in oncology (CaSMO) project was developed to improve cancer patients’ (both active and survivors) quality of life by offering tools to healthcare professionals for preventing, reducing and/or managing cutaneous adverse events (cAEs).
Immunotherapy, specifically immune checkpoint inhibitors (ICIs), for cancer patients is both increasing in indications and use. Similarly, the incidence and onset of cutaneous immunotherapy-related adverse events (cirAEs) vary based on the class and dose of immunotherapy administered, the type of cancer, and factors related to the patients. Dermatologists will increasingly see side effects from immunotherapy.

The CaSMO advisors developed a practical primer on prevention, identification, and treatment, including skincare for cirAEs, focusing on isolated pruritus, psoriasiform eruptions, lichenoid eruptions, eczematous eruptions, and bullous pemphigoid.
A modified Delphi approach was used for the cirAEs practical primer’s development. Recommendations given by the CaSMO advisers are based on information from the guidelines, algorithms, consensus papers, and systematic reviews coupled with their clinical experience and resulting from discussions.

According to the CaSMO group, the management of cirAEs starts with physician awareness and patient education on the occurrence of toxicities, preventive measures, and skincare using gentle cleansers, moisturizers, and sunscreen started before immunotherapy begins and ongoing thereafter as part of the lifestyle.

Keywords: Immunotherapy, cutaneous adverse events, immune checkpoint inhibitor, skin toxicity, psoriasis, pruritus, morbilliform, eczema, bullous pemphigoid

Disclosures
The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this manuscript. This work was supported by an unrestricted educational grant from La Roche-Posay Canada. All authors contributed to the manuscript, reviewed it and agree with its content and publication.

PDF Download


Introduction

In 2021 an estimated 229,200 Canadians were diagnosed with cancer.1 In Canada, of the four most commonly diagnosed types of cancer (lung, breast, colorectal, and prostate), excluding non-melanoma skin cancer, lung cancer is the most lethal and expected to cause more deaths than the other types combined.2,3

Immunotherapy activates the body’s immune system to fight cancer. Immune checkpoint inhibitors (ICIs) prevent the deactivating signal by blocking the T-cell shut-off receptors and ligands from binding to each other, thereby disrupting signaling so that T cells remain active and can then recognize and attack cancer cells.4,5 ICIs are approved for treatment across almost every type of solid-organ tumors, many hematologic malignancies, and in many instances, first-line therapy.4,5 In 2019, in the United States, approximately 40% of oncology patients were eligible for treatment with immunotherapy.6 ICIs may be administered as a single agent (anti-CTLA-4 (ipilimumab), anti-PD-1 (pembrolizumab, nivolumab), anti-PD-L1 (atezolizumab, durvalumab, and avelumab)) or as combination immunotherapy (ipilimumab and nivolumab) and even as a combination with other agents.4-6

Oncology patients treated with immunotherapy may experience cutaneous immunotherapy-related adverse events (cirAEs) at any time during and after the cancer treatment, severely impacting the quality of life (QoL) and potentially cancer treatment outcomes.7-24 The incidence and onset of cirAEs vary depending on the drug class, the type of cancer, and factors related to the patient.25 Adverse skin reactions occur in 14%–47% of patients treated with ICIs, ranging from mild and localized to debilitating and widespread in 1%–3% of patients and even potentially causing death.15,16,26-30 Nivolumab-related cirAEs, including isolated pruritus, occurred in approximately 13–20% of patients, of which 0.7% of patients reported grade three or higher cAEs.29-33 Pembrolizumab-induced cirAEs occurred in 9%–27% of patients, of which 1%−4% of patients reported grade three or higher cirAEs.34,35

Frequently occurring cirAEs include pruritus without primary dermatologic findings (isolated pruritus), lichen planus or lichenoid drug eruptions, psoriasiform reactions, eczematoid eruptions, morbilliform eruptions, and bullous diseases, primarily bullous pemphigoid.7-24  

The cirAEs may indicate a therapeutic response. A multicenter retrospective study on patients with non-small-cell lung carcinoma (NSCLC) receiving nivolumab showed that AEs and cirAEs were a strong predictor of survival outcomes, and those presenting with more than 2 AEs showed more benefit than patients with fewer AEs.14  Vitiligo occurs in some patients with melanoma receiving ICIs (Figure 1), and this is significantly associated with both progression-free-survival (PFS) and overall survival (OS) which indicated a two to four times lower risk of disease progression and death.32,36

CaSMO Management of Cutaneous Toxicities Associated with Immune Checkpoint Inhibitors: A Practical Primer - image
Figure 1: Vitiligo
Photo property of Joel Claveau

Major guidelines10-12 recommend the use of systemic steroids for grade 2 or higher “rash” contrary to evidence showing the use of systemic steroids reduces the anticancer effect of ICIs.33  A secondary analysis of a randomized trial on stage III melanoma patients with cirAEs showed that patients who received treatment with systemic steroids for 30 days or longer had lower OS than those without steroid treatment.33  Another multicenter retrospective analysis of patients treated with anti-PD-1 therapy showed that high-dose glucocorticoids used to treat AEs were associated with poorer PFS and OS.35

Scope of the Canadian Skin Management in Oncology Project

The Canadian skin management in oncology (CaSMO) project was developed to improve cancer patients’ (both active and survivors) QoL by offering tools for preventing and managing cAEs.37-39 A general management algorithm to reduce the incidence of all cutaneous toxicities and maintain healthy skin using general measures and over-the-counter agents38 and an algorithm to reduce and treat acute radiation dermatitis39 were previously published. These algorithms aim to support all healthcare providers treating oncology patients, including physicians, nurses, pharmacists, and other medical providers.38,39 The next step in the project was to develop a practical primer on prevention, identification, and treatment for cirAEs, focusing on isolated pruritus, psoriasiform eruptions, lichenoid eruptions, eczematous eruptions, and bullous pemphigoid.

Methods

For the CaSMO cirAEs practical primer’s development, the advisors used a modified Delphi approach following the AGREE II instrument.40,41 The modified Delphi method is a communication technique for interactive decision-making for medical projects.41 The technique was adapted from face-to-face meetings to a virtual meeting to discuss the outcome of literature searches and reach a consensus on the practical primer based on the selected literature. The virtual discussion was followed by a virtual follow-up, replacing a questionnaire.41 The process entailed preparing the project, selecting the advisors, and conducting systematic literature searches followed by two steps.

Step 1: Virtual panel meeting on November 20, 2021, to review the results of the systematic literature review addressing cirAEs prevention, treatment, and maintenance of cirAEs and to discuss and adopt a practical primer using evidence coupled with the panels’ experience and opinion.

Step 2: Online process to fine-tune the practical primer and prepare and review the publication.

Literature Review

The literature review included guidelines, consensus papers, publications on the prevention and management of cirAEs, and clinical and other research studies published in English from January 2010 to September 2021. Excluded were articles with no original data (unless a review article was deemed relevant), not dealing with cirAEs, and publication language other than English.

A dermatologist and a physician-scientist conducted the searches on September 26 and 27, 2021, on PubMed and Google Scholar as a secondary source of the English-language literature using the terms: cirAEs; prevention and treatment of isolated pruritus, psoriasis, lichen planus, eczematous eruptions, bullous eruptions, QoL of patients with cirAEs; skincare and prescription treatment for the prevention, treatment, and maintenance of cirAEs; adjunctive skincare use; education of staff and patients; communication strategies; adherence; concordance; efficacy; safety; tolerability; skin irritation.

The results of the searches were evaluated independently by two reviewers who resolved discrepancies by discussion. The searches yielded one hundred and six publications. Ninety-four papers remained after excluding duplicates (n = 12), and articles deemed not relevant (other subjects, low quality). Seven guidelines, eight systematic literature reviews, 16 reviews, and 37 clinical studies addressed cirAEs. Case reports were included as they provide valuable information in this fast-developing field. Moreover, cirAEs possess complex issues that are difficult to capture in randomized controlled trials (RCT). Ten consensus papers or algorithms and two clinical studies addressed cirAEs, treatment, and skincare. Finally, fourteen other publications addressed various subjects (epidemiology studies, methodology, and general treatment of cirAEs or similar dermatological conditions). Two reviewers who graded the clinical publications evaluated the literature search results. Grading included study type and quality (RCT of high quality = A, B, or C) and level of evidence (level 1 to level 4) using the pre-established criteria.42  The lack of studies on cirAEs treatment with prescription medications and skincare made grading irrelevant. However, the guidelines, systematic literature reviews, and consensus papers provided valuable information.

Immunotherapy and Associated Cutaneous Adverse Events

Immunotherapy

Immunotherapy uses immune checkpoint inhibitors, such as cytotoxic T-lymphocyte-antigen-4 (CTLA-4), inhibitors ipilimumab, programmed cell death receptor-1 (PD-1) inhibitors pembrolizumab, nivolumab, cemiplimab, and programmed death-ligand 1 (PD-L1) inhibitors atezolizumab, durvalumab, and avelumab (Table 1).4,5-11,43,44

Table 1: Immunotherapy classes, molecules, and indications

Drug Class Name Oncologic Indications
CTLA-4 inhibitors Ipilimumab Melanoma, renal cell, colorectal
Tremelimumab Not FDA approved; orphan drug designation for mesothelioma
PD-1 inhibitors Nivolumab Melanoma, lung, head and neck, Hodgkin's disease, bladder, colorectal, hepatocellular, renal cell
Pembrolizumab Melanoma, lung, head and neck, Hodgkin's disease, primary mediastinal large B-cell lymphoma, bladder, colorectal, gastric, cervical, hepatocellular, Merkel cell, renal cell
Cemiplimab Squamous cell carcinoma, basal cell carcinoma
PD-L1 inhibitors Avelumab Merkel cell, bladder, renal cell
Atezolizumab Bladder, lung, breast
Durvalumab Bladder, lung
Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), ipilimumab, programmed cell death 1 (PD-1) pembrolizumab, nivolumab, and programmed death-ligand 1 (PD-L1) atezolizumab, durvalumab, and avelumab.4,6,8-10

CTLA-4 impinges on many features of T-cell biology.43,44  The CTLA-4 signaling pathway and the PD-1/PD-L1/PD-L2 signaling pathway are critical checkpoints that are used to down-regulate the body’s immune system that tumors often exploit and activate, thereby evading the body’s immune system.43,44 CTLA-4 dampens T-cell responses via cell-intrinsic and extrinsic pathways. Intrinsic events include, among others, inhibition of protein translation, recruitment of phosphatases, activation of ubiquitin ligases, inhibition of cytokine receptor signaling, and inhibition of lipid microdomain formation on the surface of T-cells. Cell extrinsic events include the competition for CD28 in binding to its ligands CD80/86, the removal of CD80/86, the release of suppressive indoleamine-dioxygenase, and the modulation of Treg function. Antibodies to CTLA-4 may facilitate T-cell tumor entry and alter the movement in tumors and rates of egress.43,44

cirAEs can present as almost any dermatologic condition, but most are inflammatory dermatoses.7-11,26 The median time to onset of cirAEs is four weeks but has a broad range from 2 to 150 weeks, including beyond treatment discontinuation.15,17,45

cirAEs of any grade have been reported at 30-40% of patients receiving PD-1/PD-L1 and approximately 50% of patients treated with ipilimumab.31  Maculopapular rash and pruritus are common cirAEs related to these treatments.31

A systematic literature review on cancer treatment with pembrolizumab and nivolumab observed that the most frequently occurring cirAEs were inflammatory dermatitis, pruritus, and vitiligo.46 The rate of any-grade cirAEs with pembrolizumab and nivolumab was reported at 16.7% (RR=2.6) and 14.3% (RR=2.5), respectively.46 The cirAEs may be associated with pruritus and comprised of erythematous macules, papules, and plaques, predominantly low-grade and localized to the trunk and extremities.46 Challenges with reports of cirAEs are accurate descriptions and diagnoses of the specific dermatoses as many are simply reported as “rash”. Additional cirAEs comprised pruritus [incidence pembrolizumab and nivolumab was 20.2% (RR=49.9) and 13.2% (RR=34.5) respectively] and vitiligo [incidence, 8.3% (RR=17.5) and 7.5% (RR=14.6) respectively].45 The researchers noted that knowledge of cirAEs is critical in delivering optimal cancer treatment, maintaining therapeutic agent dose, and health-related QoL.46

The advisors agreed that cirAEs are likely an indicator of ICIs treatment efficacy and should be managed with topical treatment and judicious, ideally steroid-sparing systemic treatment where possible.33,35-38

Approaches to the most common immunotherapy-related cAEs

The advisors discussed and selected the most common cirAEs: isolated pruritus, psoriasiform eruptions, lichenoid eruptions, eczematous eruptions, morbilliform (maculopapular) eruptions, and bullous pemphigoid eruptions.7-25,27-30

According to the advisors, the management of cirAEs starts with patient education on the occurrence of cirAEs, preventive measures, and skincare using gentle cleansers, moisturizers, and sunscreen started before ICIs treatment begins and ongoing during survival as part of the lifestyle.9-25

Diagnosis of cirAEs

When diagnosing the cirAEs, other etiologies such as an infection, an effect of other agents, or other skin conditions should be ruled out.8-11 

Biopsies play a role in accurately diagnosing the cirAEs when morphology is not clear. Histopathologically, cirAEs can be categorized into four broad groups, group 1: Inflammatory skin disorders, which reflect acute, subacute, or chronic inflammation of various patterns associated with variable epidermal changes, including psoriasiform or lichenoid interface chronic dermatitis.16,50-53 Group 2: ICIs-related bullous skin lesions such as bullous pemphigoid or dermatitis herpetiformis, group 3: Keratinocyte alteration—Grover’s disease54/acantholytic dyskeratosis, and group 4: Immune-reaction mediated by alteration of melanocytes such as regression of nevi, tumoural melanosis, and vitiligo.29,36,48-52 For this review, we will only be focusing on the selected cirAEs.

When assessing the severity of cirAEs, grading systems’ trial limitations need to be considered.9 The Common Terminology Criteria for Adverse Events (CTCAE) classification is typically used; however, in the case of clinical trials, clinicians should be cognisant of the version the trial is using as different versions of CTCAE have different definitions and categorizations.26

A general approach to prevention or reduction of cirAEs as well as initial management of the previously published CaSMO algorithm is displayed (Figure 2) for reference.38  

CaSMO Management of Cutaneous Toxicities Associated with Immune Checkpoint Inhibitors: A Practical Primer - image
Figure 2a and 2b: CaSMO algorithm for preventing or reducing cAEs using skincare
Permission for the reproduction of the algorithm was obtained.13

Patient and Caregiver Education

Patient and caregiver education and information should occur before initiating therapy and continue throughout treatment and survivorship.9-11,37,38,47 The patient should be informed that immunotherapy works differently than traditional chemotherapy and that therapeutic response and related cirAEs differ in timing, presentation, and response to treatment.48-57

Patients should be informed that cirAEs may continue even after discontinuation of the ICIs treatment.48-57 Additionally, patients should be educated to inform all healthcare providers that they are receiving or have received ICIs therapy and to report any changes in health status.54 Patients and caregivers need to be informed that cirAEs can often be managed effectively, especially when they are identified early. In addition, education should be provided on good hygiene, skincare, sun avoidance and protection, the safe handling of medications, and infection control is essential in preventing and supporting optimal management of cirAEs.38,48-57

Severe Cutaneous Adverse Reactions (SCARs)

As described in the CaSMO algorithm, any drug eruption should be assessed if they are serious or life-threatening. Severe cutaneous adverse reactions (SCARs) require prompt clinical, urgent referral and triage.9-11,37-39,47  Symptoms that raise suspicion of SCARs include fever, widespread rash, skin pain, skin sloughing, facial or upper-extremity edema, pustules, blisters, or erosions.9-11,38 SCARs include Stevens-Johnson syndrome (SJS), toxic epidermal necrolysis (TEN), overlap SJS-TEN, acute generalized exanthematous pustulosis (AGEP), and drug reaction with eosinophilia and systemic symptoms (DRESS).7,8,12-25,30,31,34,38,45-61

SJS-TEN occurs in up to 2% of patients treated with ICIs exceedingly rare.31,43  SJS-TEN is characterized by extensive epidermal loss (<10% SJS, >30% TEN, 10-30% SJS-TEN overlap) with mucous membrane erosions.31,37-39,43-49  Classically, this is an acute reaction; however, a slower and more indolent presentation may occur.31,52 ICIs-related DRESS is rare and is mostly reported in case studies. Common DRESS features are fever, eosinophilia, lymphocyte activation, multiorgan involvement, and reactivation of herpes viruses, especially HHV6.60,61 Patients usually develop two or three features of symptoms followed by a stepwise development of other symptoms.60,61

AGEP is a rare, severe, potentially fatal, acute cirAEs with systemic involvement, including abnormal liver function and acute respiratory distress.62  Patients with AGEP may develop multiple organ failure or sepsis. We found one case report on AGEP caused by a combination of PD-1 and chemotherapy therapy.62

SCARs tend to occur more commonly with combination ICI therapy. Management and treatment of SCARs vary and is beyond the scope of this paper.31,59 Permanent discontinuation of ICIs treatment is recommended for grade 4 cirAEs.31,43,47

Isolated Pruritus

Immunotherapy-related isolated pruritus occurs frequently and has a significant impact on QoL (Figure 3).7-25,31,46,55-57  Diagnostic criteria for pruritus have been described as core symptoms, including duration of pruritus (more than six weeks) and a history of signs of scratching.63,64 Associated criteria comprise a range of clinical manifestations, symptoms, functions, and emotions. The function includes the extent of impaired QoL, sleep loss, and days of absence from work. Emotions include possible psychological reactions such as, depression, anxiety, anger, and helplessness.64

CaSMO Management of Cutaneous Toxicities Associated with Immune Checkpoint Inhibitors: A Practical Primer - image
Figure 3: Pruritus without primary dermatologic finding
TDC – RA – 77-year-old male with metastatic non-small cell lung cancer treated with pembrolizumab (PD1 inhibitor) and binimetinib (MEK inhibitor) experiencing generalized pruritus with excoriated papules and nodules. a. left arm; b. back
Photo property of MB Sauder.

Isolated pruritus should be a diagnosis of exclusion once other causes such as pre-bullous pemphigoid, bites, or other drug reactions are ruled out.

A stepwise approach includes starting with over-the-counter topical agents, topical prescription agents, systemic drugs, and phototherapy.63,64 General principles before starting topical and/or systemic treatment comprise skincare using gentle cleansers and moisturizers, especially in the presence of xerosis.63,48

The CaSMO algorithm educates on general measures such as avoiding skin irritants, scented products, temperature extremes, sun, and the use of sun-protective clothing (e.g., brim hats and sunglasses). The information should be provided before immunotherapy starts, and this is especially important for isolated pruritus.38 The daily skincare regimen should be reinforced with emphasis on gentle cleansers, skin moisturization, and sun protection – similar counseling to a patient with atopic dermatitis.37,38,47  

A prurigo nodularis 4-tier treatment ladder64 addresses neutral and immunologic mechanisms that may be applicable to ICIs-induced pruritus.64 Immunologic mechanisms are more likely to be the underlying mechanism in ICIs-induced pruritus. Patients can enter the treatment ladder at any tier and move up or down the ladder based on their clinical presentation.64  Immunologic tier 1 is more likely mechanisms can be treated with TCS, TCI, or topical calcipotriol.64  Moisturizers, including antipruritics such as pramoxine 1% or formulations with menthol or camphor, may be used as an adjunct therapy.64 

For tier 2 neural mechanisms of pruritus medications include selective nor­epinephrine reuptake inhibitors (SNRIs) (i.e., mirtazapine) and gabapentinoids (gabapentin and pregabalin), which have antipru­ritic properties.63,64  Second-generation histamine H1 receptor antagonists were shown to be effective and safe for patients receiving ICIs.64 

Additional second-line systemic therapies such as neurokinin 1 inhibitors (i.e., aprepitant) and inhibi­tors of interleukins (ILs) 4, 13 may be considered for the tier 3 immunologic mechanisms approach.64 When evaluating the need for systemic or topical treatment, possible interactions with cancer therapy are assessed together with concomitant conditions and cirAEs.9-11,15,16,23,24 37,38,47

Clinicians should take caution using systemic steroids and immunomodulators for patients receiving cancer treatment with ICIs (Table 2).9-11

Table 2: Isolated pruritus

Isolated Pruritus
Treatment recommendations First-line Second-line Third-line
Moisturization and sun avoidance
 
Antipruritic additives: pramoxine, menthol, camphor
 
Second-generation H1 antihistamine 
 
TCS, TCI, PDE4 inhibitors
 
TCA (i.e. doxepin)
 
SNRIs (i.e., mirtazapine) and gabapentinoids (gabapentin and pregabalin)
Neurokinin 1 inhibitors (i.e., aprepitant), inhibi-tors of ILs 4, 13, (i.e., dupilumab, tralokinumab)
 
Phototherapy  
 
Methotrexate Inhibitors of IL31 (i.e. nemolizumab – only in trials)
Other comments
Take caution using systemic steroids 
 
Consider biopsy if pruritus is extreme or non-responsive to treatments to rule out pre-bullous pemphigoid
 
Look for alternative diagnoses (e.g., scabies)
 
Quality of life (QoL), Selective nor¬epinephrine reuptake inhibitors (SNRIs), Interleukins (ILs), Topical corticosteroids (TCS), Topical calcineurin inhibitors (TCI), Not applicable (n.a.)

Psoriasiform Eruptions

T helper (Th) cells producing interleukin (IL)-17, IL-22, and tumor necrosis factor (TNF) drive the pathogenesis of psoriasis by orchestrating inflammation in the skin triggering proliferation of keratinocytes and endothelial cells.65 Besides Th17 cells, other immune cells that are capable of producing IL-17-associated cytokines participate in psoriatic inflammation.66-70  ICIs are thought to be driven through the TH17 pathway, and this may have implications on the development of psoriasis but also on the use of systemic psoriasis medications.64,65,71,72

Immunotherapy with anti-PD1 and anti-PDL1 can exacerbate psoriasis, although more severe flares were noted in patients treated with durvalumab.65-68,70,71

A personal or family history of psoriasis is a significant risk factor that should be identified before starting immunotherapy.65,66,68 These patients require regular skin monitoring enabling early diagnosis and treatment of psoriatic exacerbations.65,66,68 Prompt diagnosis can prevent a severe impact on QoL of this cirAE that may compromise therapeutic protocols and final cancer prognosis (Figure 4).53,65,66 A skin biopsy to confirm the diagnosis, although not required, includes parakeratosis, hypogranulosis, regular acanthosis, suprapapillary plate thinning, and neutrophils at various layers of the epidermis.68,70

CaSMO Management of Cutaneous Toxicities Associated with Immune Checkpoint Inhibitors: A Practical Primer - image
Figure 4: Psoriasiform cAEs
Photo property of MB Sauder.

Psoriasiform cirAEs can affect skin, nails, and joints and significantly impact QOL.65-71 Psoriasis can be associated with arthritis as patients have a greater-than-expected incidence of psoriatic arthritis when it occurs as a cirAE of cancer treatment.33,65,66 The choice of therapy takes many factors into account, including the extent and area of involvement, lifestyle, other health issues, and medications. First-line treatments include topical treatment with TCS, TCI, calcitriol, or a combina­tion of a super potent TCS with calcipotriol or tazarotene can be given.65-71  For recalcitrant cases, consider adjuvant narrowband ultraviolet B therapy or oral treatment such as systemic retinoids [acitretin] or apremilast.72-75  Although there is no direct evidence in ICIs treated cancer patients, methotrexate is immunosuppressive and may reduce the therapeutic effect of ICIs, similar to systemic steroids that have been shown to reduce RFS and OS.32,33,35

Acitretin or apremilast is preferred over methotrexate; however, there is an absence of data for apremilast on the effect it may have on the anticancer treatment.68-75  If the lesions persist, treatment with biologics may be required to treat psoriasiform cirAEs. IL-23 agents (gulselkumab, risankizumab, tildrakizumab) are considered most targeted and have the least direct evidence of immunosuppression (e.g. infection and TB reactivation). However, the risk/benefit needs to be considered before starting treatment with biologics (Table 3).76-86

Table 3: Psoriasiform eruption

Psoriasis
Treatment recommendations First-line Second-line Third-line

Moisturization and avoidance of sun exposure

TCS, TCI

Combination steroid products

Vitamin D analogues

Phototherapy

Systemic retinoids (acitretin)

Apremilast

Methotrexate

Biologics: consider most targeted biologics such as  IL-23 inhibitors (guselkumab, tildrakizumab, risankizumab)
Other comments

Differential diagnosis: lichen planus

Avoid cyclosporine

Consider biopsy if extensive, atypical, or non-responsive to therapy
Quality of life (QoL), Interleukin (IL), Body surface area (BSA), Topical corticosteroids (TCS), Topical calcineurin inhibitors (TCI)

Lichen Planus

Immunotherapy-induced lichen planus is a pruritic, papulosquamous disease that can affect skin, hair, and nails (Figure 5). Treatment is required, especially if pruritic when it can severely impact QoL.7-19,27,33,46,63,77,78 When examining the patient exclude guttate psoriasis, mucositis, and bullous disease. Specific investigations include a detailed medical history and medication use, liver function tests including hepatitis C status, and, if required, a skin biopsy.79

CaSMO Management of Cutaneous Toxicities Associated with Immune Checkpoint Inhibitors: A Practical Primer - image
Figure 5: Lichenoid cAEs
Photo property of MB Sauder.

Pruritus may be treated with second-generation histamine H1 receptor antagonists (e.g., blistane, rupatadine, cetirizine and fexofendadine).63,64,79 A retrospective analysis of clinical data investigating if commonly used medications might influence responses to checkpoint inhibitors. The researchers found that concurrent use of second-generation histamine H1 receptor antagonists correlated with significantly improved survival outcomes in those with melanoma or lung cancer. In addition, the antihistamine was effective for treating pruritus in cirAEs.63,64,79 The choice of therapy takes many factors into account, including the extent and area of involvement, other health issues, and medications.8-11,67-69,78,80 First-line treatment includes moisturization and sun avoidance, antihistamines, TCS, and TCI.80,81 Other second-line treatments include oral retinoids (acitretin), apremilast, sulfasalazine, metronidazole, hydroxychloroquine (especially for scalp or presentation with arthritis).80 Additionally, narrowband or broadband UVB or PUVA may be used. Treatment with cyclosporine should be avoided as it may exert effects on the immune system (Table 4).25,31,78

Table 4: Lichen planus

Lichen Planus
Treatment recommendations First-line Second-line Third-line
Moisturization and skincare
 
TCS, TCI
 
Second-generation H1 antihistamine 
 
Metronidazole, 
 
Hydroxychloroquine (especially for scalp or presentation with arthritis)
 
Systemic retinoids (acitretin)
nbUVB
 
PDE4 inhibitors 
 
Sulfasalazine, Griseofulvin, Calcipotriol ointment 
 
Low-dose corticosteroid
 
Other comments
Differential diagnosis: psoriasis, mucositis, bullous disease
 
Avoid cyclosporine
 
Consider concomitant conditions and immune-related AEs
 
Consider biopsy if extensive, atypical, or non-responsive to therapy; biopsy can help distinguish lichen planus from guttate psoriasis  
 
Psoralen and ultraviolet A (PUVA), Body surface area (BSA), Topical corticosteroids (TCS), Topical calcineurin inhibitors (TCI), Phosphodiesterase (PDE)

Eczematous Eruptions

Eczematous eruptions (EE) are associated with altered immune function, epidermal barrier dysfunction, genetic and environmental factors, and poorly understood interaction between these factors.82-85 EE presents clinically as relapsing erythematous and pruritic patches of skin with varying severity (Figure 6).9-11,82-85  Skin biopsies may be taken if the condition is extensive or not responding to therapy or if the diagnosis is in doubt.9-11 Canadian and US guidelines for topical treatment of atopic dermatitis include education and avoiding triggers, routine skincare with gentle cleansers and moisturizers, and EE management foundation regardless of disease severity and prescription treatment.82,83 Conventional moisturizers contain occlusives, humectants, and emulsions. Newer moisturizers designed to restore skin barrier defects include distinct ratios of lipids that resemble physiological compositions.36,38,47,82,83 If EE is not controlled with a moisturizer alone, TCS, TCI, or crisaborole ointment, an anti-inflammatory inhibitor of PDE4, is recommended while continuing skincare.9-11,82-86   Treatment with crisaborole ointment achieved early sustained control of atopic dermatitis flares (reduced pruritus, erythema, exudation, excoriation, induration/papulation, and lichenification).84,85  The topical PDE4 inhibitor offered a safe alternative to TCS and TCI.84,85

CaSMO Management of Cutaneous Toxicities Associated with Immune Checkpoint Inhibitors: A Practical Primer - image
Figure 6: Eczematous eruptions
Photo property of Joel Claveau

For those not adequately controlled with topical treatment, nbUVB may be considered.82,83 Dupilumab is a humanized monoclonal antibody against the shared alpha subunit of IL-4 and IL-13 receptors, blocking these cytokines commonly found in atopic dermatitis-affected skin. The use of dupilumab may be considered for extensive, moderate-to-severe EE. Two large, long-term RCTs demonstrated its efficacy and safety.9-11,82,83,86  

Another option is tralokinumab (IL-13 inhibitor) which was shown to be effective and safe for the treatment of severe atopic dermatitis.87

Treatment with the biologic is preferred to methotrexate which is an immunosuppressive and may reduce the anticancer effect.9-11 TCS, TCI, and crisaborole can be used with any systemic treatment to address an EE flare (Table 5).82-85,93

Table 5: Eczematous eruptions

Eczematous Eruptions
Treatment recommendations (grouped into 1st, 2nd, and 3rd line) First-line Second-line Third-line
Moisturization and sun avoidance
 
TCS 
 
TCI 
 
Topical PDE4 inhibitor (crisaborole)
 
Second-generation H1 antihistamine 
 
Dupilumab
 
Tralokinumab 
 
nbUVB 
 
Methotrexate
 
Systemic steroids
 
Other comments
Look for a classic presentation
 
Consider previous history or family history of atopy
 
JAK inhibitors are not recommended (pending further data)
 
Consider biopsy if extensive or non-responsive to therapy
 
 
Quality of life (QoL), Body surface area (BSA), Janus kinase (JAK), Topical corticosteroids (TCS), Topical calcineurin inhibitors (TCI), Phosphodiesterase (PDE)

Morbilliform (maculopapular) Eruptions

These cirAEs may present as pruritic, nontender, erythematous papular, and macular eruptions on various body locations.37,38,46,47 Physical examination focusing on the morphology and the distribution, starting on the trunk and then spreading to limbs, of the presented morbilliform eruptions is required to distinguish between the various cirAEs (Figure 7).37,38,46,47 Laboratory tests may be needed and include a complete blood count, electrolytes, renal and liver function, and inflammatory markers.37,38,46,47 Peripheral blood eosinophilia (≥500 eosinophils/microL) may be caused by numerous conditions, including allergic, infectious, inflammatory, and neoplastic disorders (Table 6).37,38,46,47  Mild to moderate conditions may be treated with general skin care measures, TCS, TCI, and oral histamine H1 receptor antagonists to reduce pruritus.63,64,94 Generally, this condition can be managed with aggressive topical treatment; however, for very severe and symptomatic cases, ICIs-treatment may be held, and a short course of systemic corticosteroids may be indicated.94 

CaSMO Management of Cutaneous Toxicities Associated with Immune Checkpoint Inhibitors: A Practical Primer - image
Figure 7: Morbilliform eruptions
Photo from MB Sauder.

Table 6: Morbilliform eruptions

Morbilliform (maculopapular) Eruptions
Diagnosis
Drug eruptions can range from a mild lesion involving only the skin to severe complex eruptions with systemic involvement, such as TEN. Systemic involvement should be explored even in a mild cutaneous eruption due to a drug because skin manifestation does not necessarily mirror the severity of the systemic involvement. 
 
Ensure another drug does not cause symptoms; take a drug history, looking for medications started 2-3 weeks before the eruption. 
 
Consider biopsy if the eruptions are extreme or non-responsive to treatment. 
 
Treatment recommendations (grouped into 1st, 2nd, and 3rd line) First-line Second-line Third-line
Moisturization and skincare TCS
 
If accompanied by itch: pramoxine, menthol, camphor 
 
Second-generation H1 antihistamine 
 
Systemic corticosteroids
Other comments
Topical corticosteroids (TCS)

Bullous Pemphigoid

Bullous pemphigoid (BP) is an autoimmune bullous dermatosis caused by a variety of medications. One of the more common medications to cause BP now are ICIs.88,90,91 A skin biopsy may be required to confirm BP, including direct immunofluorescence to identify the presence of antibodies in the skin, typically demonstrating linear IgG and C3. Further studies on serum can detect circulating autoantibodies (anti-BP180 or anti-BP230) with either ELISA or indirect immunofluorescence. Clinically, BP classically presents with pruritic tight tense bullae filled with serous fluid on erythematous skin on various body locations (Figure 8).88,90,91 The pre-bullous phase presents as intense pruritic urticoid plaques that can be diagnostically challenging and may be mistaken for the more common cirAE of isolated pruritus or urticaria.88,90,91 The post-bullous phase presents as hemorrhagic crusts on a background of erythema representing ruptured bullae.

CaSMO Management of Cutaneous Toxicities Associated with Immune Checkpoint Inhibitors: A Practical Primer - image
Figure 8: Bullous eruptions
Severe bullous pemphigoid associated with pembrolizumab therapy
Photo from MB Sauder

cirAE BP can occur at any time, including after discontinuation of ICI. The onset to developing BP is within days or weeks of ICI exposure or may be delayed several months after starting ICI.31 Upon diagnosis ICIs is to be held and may require permanent discontinuation.3,15,46 In milder cases, aggressive topical treatments with class I TCS may be as effective as systemic steroids.92  cirAE BP may require systemic treatment and temporary, if not permanent, cessation of immunotherapy.3,15,46 Systemic corticosteroids may be used the achieve control quickly while waiting for steroid-sparing treatments to take effect.88,90-92 Non-immunosuppressive systemic treatment includes oral tetracycline-class antibiotics combined with oral niacinamide 500 mg twice a day, omalizumab, IVIg, and dupilumab.31,88,90-92 However, steroid-sparing agents that are immunosuppressive may be required in order to control BP ongoing, including methotrexate, mycophenolate mofetil or rituximab.91 While rituximab may be immunosuppressive, it works on B-cells and may not hinder the T-cell activation from ICIs. In patients with cirAE BP, obtaining disease control as quickly as possible is ideal not only for improved QoL, and decreased morbidity but also if treatment is to be reinitiated (Table 7).93,95

Table 7: Bullous eruptions

Bullous Eruptions
Diagnostics
Prompt clinical/pathologic workup is essential (urgent referral & triage within 2-3 days), including biopsy & blood work. 
 
Biopsy can help determine if the eruption is bullous pemphigoid, bullous lichen planus, or TEN and blood work can identify pemphigus and pemphigoid antibodies 
 
Treatment recommendations (grouped into 1st, 2nd, and 3rd line) First-line Second-line Third-line
High-dose TCS
 
Tetracycline & nicotinamide
 
Systemic coticosteroids
 
Methotrexate
 
Dapsone
 
Cellcept (with caution)
 
Rituximab (some evidence); off-label, Intravenous immunoglobin,
 
Omalizumab
 
Dupilumab
 
Other comments
With mild symptoms, consider holding immunotherapy and consult a dermatologist to determine resuming treatment.  
 
In severe cases, immediately hold immunotherapy and seek urgent care from a dermatologist, oncologist, or ER.
 
Immunotherapy is usually discontinued until eruptions are under control, 
 
Avoid azathioprine & cyclosporine 
 
Cutaneous adverse events (cAEs), Quality of life (QoL), Toxic epidermal necroly¬sis (TEN), Topical corticosteroids (TCS)

Limitations

The details provided on the mode of action of ICIs were limited to what is clinically relevant for this primer on cirAEs. Clinical studies on prescription medications and skin care for cirAEs frequently are case reports, case series, or have small sample sizes. For this reason, grading the evidence resulting from the literature searches was irrelevant. Therefore, recommendations given by the CaSMO project advisers are based on information from the guidelines, algorithms, consensus papers, and systematic reviews coupled with their clinical experience and resulting from discussions. More work is required to identify the best practice evidence-based management of cirAEs.

Summary and Conclusions

Immunotherapy for cancer patients is fast-developing into the standard of care for many malignancies. Although many publications are available on cirAEs, publications on the treatment using prescription medications and skincare are scarce.

The CaSMO practical primer focuses on isolated pruritus, psoriasiform eruptions, lichenoid eruptions, eczematous eruptions, and bullous pemphigoid. The practical primer addresses the need for physician awareness, patient education on the occurrence of cirAEs, prompt diagnosis, prevention, treatment, and ongoing skincare started before ICI.

References

  1. https://cdn.cancer.ca/-/media/files/cancer-information/resources/publications/2021-canadian-cancer-statistics-special-report/0835-2976-2021-canadian-cancer-statistics-en.pdf?rev=8e016fe8c5ea4c23b05ea08bf1018ca6&hash=C5BAEC543E496EDD8460CBBF6D44A010&_ga=2.47880293.391373246.1644172159-1100777747.1644172159
  2. Canadian Cancer Society; 2021. Available at: https://www.cancer.ca/en/cancer-information/cancer-101/cancer-statistics-at-a-glance/
  3. Mittmann N, Liu N, Cheng SY, Seung SJ. Health system costs for cancer medications and radiation treatment in Ontario for the most 4 most common cancers: a retrospective cohort study. Can Med Association J Open 2020;8:E191-8. Epub 2020 March 16 doi: https://doi.org/10.9778/cmajo.20190114
  4. Dine J, Gordon R, Shames Y, et al: Immune checkpoint inhibitors: An innovation in immunotherapy for the treatment and management of patients with cancer. Asia Pac J Oncol Nurs 2017; 4:127-135.
  5. American Society of Clinical Oncology (ASCO). Understanding immunotherapy therapy. Accessed at https://www.cancer.net/ navigating-cancer-care/how-cancer-treated/immunotherapy-and-vaccines/understanding-immunotherapy on January 16, 2022.
  6. Haslam A, Gill J, Prasad V: Estimation of the percentage of US patients with cancer who are eligible for immune checkpoint inhibitor drugs. JAMA Netw Open 2020;3:e200423.
  7. Malviya N, Tattersall IW, Leventhal J, et al. Cutaneous immune-related adverse events to checkpoint inhibitors. Clin Dermatol 2020;38:660-678.
  8. Lacouture ME, Sibaud V. Toxic side effects of targeted therapies and immunotherapies affecting the skin, oral mucosa, hair, and nails. Am J Clin Dermatol. 2018 Nov;19(Suppl 1):31-39.
  9. Haanen J, Carbonnel F, Robert C, et al. management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017;28(7) (suppl_4):119-142.
  10. Schneider BJ, Naidoo J, Santomasso BD, et al. management of immunotherapy-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology (ASCO) guideline update. J Clin Oncol 2021;39(12):4073-4126. PIMD: 34724392.
  11. Thompson JA, Schneider BJ, Brahmer J et al. NCCN Guidelines insights: Management of immunotherapy-related toxicities, Version 1.2020. J Natl Compr Cancer Netw. 2020;18(3):230-241.DOI: 10.6004/jnccn.2020.0012
  12. Lacouture ME, Mitchell EP, Piperdi B, et al. Skin toxicity evaluation protocol with panitumumab (STEPP), a phase II, open-label, randomized trial evaluating the impact of a pre-emptive skin treatment regimen on skin toxicities and quality of life in patients with metastatic colorectal cancer. J Clin Oncol. 2010 March 10;28(8):1351-7.
  13. Qin Q, Patel VG, Wang B, et al. Type, timing, and patient characteristics associated with immune-related adverse event development in patients with advanced solid tumors treated with immune checkpoint inhibitors. J Clin Oncol 2020;38:e15160.
  14. Ricciuti B, Genova C, de Giglio A, et al. Impact of immune-related adverse events on survival in patients with advanced non-small cell lung cancer treated with nivolumab. J Cancer Res Clin Oncol. 2019 Feb;145(2):479-48
  15. Sibaud V. Dermatologic Reactions to Immune Checkpoint Inhibitors : Skin Toxicities and Immunotherapy. Am J Clin Dermatol. 2018 Jun;19(3):345-361.
  16. Curry JL, Tetzlaff MT, Nagarajan P, et al. Diverse types of dermatologic toxicities from immune checkpoint blockade therapy. J Cutan Pathol. 2017;44(2):158-76.
  17. Tang SQ, Tang LL, Mao YP, et al: The pattern of time to onset and resolution of immune-related adverse events caused by immune checkpoint inhibitors in cancer: A pooled analysis of 23 clinical trials and 8,436 patients. Cancer Res Treat 2021;53:339-354.
  18. Druyts E, Boye M, Agg H, et al. Immune-related adverse events and efficacy outcomes in patients treated with immunotherapy: A systematic review and meta-analysis. J Clin Oncol 2020;38.
  19. Patel AB, Pacha O: Skin reactions to immune checkpoint inhibitors. Adv Exp Med Biol 2018;995:117-129.
  20. Geisler AN, Phillips GS, Barrios DM, et al. Immune checkpoint inhibitor-related dermatologic adverse events. J Am Acad Dermatol 2020; 83:1255-1268.
  21. Kaul S, Kaffenberger BH, Choi JN, et al. Cutaneous adverse reactions of anticancer agents. Dermatol Clin 2019;37:555-568.
  22. Wang LL, Patel G, Chiesa-Fuxench ZC, et al. Timing of onset of adverse cutaneous reactions associated with programmed cell death protein 1 inhibitor therapy. JAMA Dermatol 2018;154:1057-1061.
  23. Simonsen AB, Kaae J, Ellebaek E, et al. Cutaneous adverse reactions to anti-PD-1 treatment—A systematic review. J Am Acad Dermatol 2020;83:1415-1424.
  24. Hussaini S, Chehade R, Boldt RG, et al. Association between immune-related side effects and efficacy and benefit of immune checkpoint inhibitors—A systematic review and meta-analysis. Cancer Treat Rev 2021;92:102134.
  25. Raschi E, Gatti M, Gelsomino F, et al. Lessons to be learned from real-world studies on immune-related adverse events with checkpoint inhibitors: A clinical perspective from pharmacovigilance. Target Oncol 2020;15:449-466.
  26. National Cancer Institute: Common Terminology Criteria for Adverse Events Version 5.0. NIH Publication, Bethesda, MD, 2018
  27. Schaberg KB, Novoa RA, Wakelee HA, et al. Immunohistochemical analysis of lichenoid reactions in patients treated with anti-PD-L1 and anti-PD-1 therapy. J Cutan Pathol. 2016 Apr;43(4):339-46.
  28. Voudouri D, Nikolaou V, Laschos K, et al.Anti-PD1/PDL1 induced psoriasis. Curr Probl Cancer. Nov-Dec 2017;41(6):407-12
  29. Rofe O, Bar-Sela G, Keidar Z, et al. Severe bullous pemphigoid associated with pembrolizumab therapy for metastatic melanoma with complete regression. Clin Exp Dermatol. 2017 Apr;42(3):309-12.
  30. Vivar KL, Deschaine M, Messina J, et al. Epidermal programmed cell death-ligand 1 expression in TEN associated with nivolumab therapy. J Cutan Pathol. 2017 Apr;44(4):381-84.
  31. Puzanov I, Diab A, Abdallah K et al. Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations for the Society for immunotherapy of Cancer (SITC) Toxicity Management Working group. J Immuno Therapy Cancer 2017;5:95 DOI 10.1186/s40425-017-0300-z
  32. Teulings HE, Limpens J, Jansen SN et al. Vitiligo-like depigmentation in patients with stage III-IV melanoma receiving immunotherapy and its association with survival: a systematic review and meta-analysis. J Clin Oncol. 2015 March 1;33(7):773-81.
  33. Eggermont AMM, Kicinski M, Blank CU et al. Association between immune-related adverse events and recurrence-free survival among patients with stage III melanoma randomized to receive pembrolizumab or placebo. A secondary analysis of a randomized clinical trial. JAMA Oncol. 2020;6(4):519-527. Doi: 10.1001/jamaoncol.2019.5570 PMID: 31895407
  34. Herbst RS, Baas P, Kim DW et al. Pembrolizumab versus docetaxel for previously treated, PD‐L1‐positive, advanced non‐small‐cell lung cancer (KEYNOTE‐010). Lancet 2016;387:1540–1550.
  35. Bai X, Hu J, Betof Warner A et al. Early Use of High-Dose Glucocorticoid for the management of irAE Is Associated with Poorer Survival in Patients with Advanced Melanoma Treated with Anti-PD-1 Monotherapy. Clin Cancer Res. 2021 Nov 1;27(21):5993-6000. doi: 10.1158/1078-0432.CCR-21-1283. Epub 2021 August 10. PMID: 34376536.
  36. Hua C, Boussemart L, Mateus C et al. Association of virtiligo with tumor response in patients with metastatic melanoma treated with pemobrolizumab. JAMA Dermatol. 2016;15(1)45-51. Doi: 10.1001/jamadermatol.2015.2707
  37. Sauder MB, Addona M, Andriessen A, et al. The role of skin care in oncology patients. Skin Ther Letter; 2020 S Oct(10):1-12.
  38. Sauder MB, Andriessen A, Claveau J, et al. Canadian skin management in oncology (CaSMO) algorithm for patients with oncology treatment-related skin toxicities. Skin Ther Letter; 2021 S March 21:1-10.
  39. Hijal T, Sauder MB, Andriessen A, Claveau J, Lynde CW. Canadian Skin Management in Oncology Group (CaSMO) algorithm for the prevention and management of acute radiation dermatitis. Skin Ther Letter; 2021 S.
  40. Brouwers M, Kho ME, Browman GP, et al. AGREE Next Steps Consortium. AGREE II: advancing guideline development, reporting and evaluation in healthcare. Can Med Association J 2010,182:E839-42
  41. Trevelyan EG, Robinson N. Delphi methodology in health research: how to do it? Eur J Integrative Med 2015;7(4):423-428.
  42. Smith Begolka W, Elston DM, Beutner KR. American Academy of Dermatology evidence-based guideline development process: responding to new challenges and establishing transparency. J Am Acad Dermatol.2011 Jun;64(6):e105-12. doi: 10.1016/j.jaad.2010.10.029.
  43. Spain L, Diem S, Larkin J. Management of toxicities of immune checkpoint inhibitors. Cancer Treat Rev. 2016;44:51–60.
  44. Digklia A, Duran R, Homicsko K et al. Cancer Immunotherapy: A Simple Guide for Interventional Radiologists of New Therapeutic Approaches. Cardiovasc Intervent Radiol. 2019 Sep;42(9):1221-1229. doi: 10.1007/s00270-018-2074-1.
  45. Qin Q, Patel VG, Wang B, et al. Type, timing, and patient characteristics associated with immune-related adverse event development in patients with advanced solid tumors treated with immune checkpoint inhibitors. J Clin Oncol 2020;38:e15160.
  46. Belum VR, Benhuri B, Pastow MA et al. Characterization and management of dermatologic adverse events to agents targeting the PD-1 receptor. Eur J Cancer. 2016;60(6) 12–25. doi:10.1016/j.ejca.2016.02.010.
  47. Lacouture ME, Choi J, Ho A et al. US cutaneous oncodermatology management (USCOM) a practical algorithm. J Drugs Dermatol 2021; 20(9)(Suppl):s3-19.
  48. Larkin J et al. Combined Nivolumab and Ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373(1):23–34.
  49. Anforth RM, Blumetti TCMP, Kefford RF, et al. Cutaneous manifestations of dabrafenib in patients with metastatic melanoma. Br J Dermatol. 2012 Nov;167(5):1153-60.
  50. Shi VJ, Rodic N, Gettinger S et al. Clinical and histologic features of lichenoid mucocutaneous eruptions due to anti-programmed cell dealth 1 and anti-programmed cell death ligand 1 immunotherapy. J Am Med Assoc Dermatol 2016; 152: 1128–1136.
  51. Tetzlaff MT, Nelson KC, Diab A et al. Granulomatous/sarcoid-like lesions associated with checkpoint inhibitors: a marker of therapy response in a subset of melanoma patients Am J Dermatopathol 2017; 39: 121–129.
  52. Koelzer VH, Buser T, Willi N et al. Grover’s-like drug eruption in a patient with metastatic melanoma under ipilimumab therapy. J Immunother Cancer 2016;4(8): 47. DOI: 1186/s40425-016-0151-z
  53. Boada A, Carrera C, Segura S, et al. Cutaneous toxicities of new treatments for melanoma. Clin Transl Oncol. 2018;20:1373-1384.
  54. Bayer V, Amaya B, Baniewicz D, et al. Cancer immunotherapy: An evidence-based overview and implications for practice. Clin J Oncol Nurs 2017;21:13-21.
  55. Keiser MF, Patel AB, Altan M: Cutaneous toxicities in lung cancer patients on immune checkpoint inhibitor therapy. Clin Lung Cancer 2021;22:195-e1.
  56. Ala-Leppilampi K, Baker NA, McKillop C, et al. Cancer patients’ experiences with immune checkpoint modulators: A qualitative study. Cancer Med 2020;9:3015-3022.
  57. Kumar V, Chaudhary N, Garg M, et al. Current diagnosis and management of immune-related adverse events (irAEs) induced by immune checkpoint inhibitor therapy. Front Pharmacol 2017; 8:49.
  58. Prior LM et al. Toxicities in immunotherapy: Can they predict response? J Clin Oncol. 2016;34(15 suppl):e14534-e14534.
  59. Muntyanu A, Netchiporouk E, Gerstein W, et al. Cutaneous immune-related adverse events (irAEs) to immune checkpoint inhibitors: A dermatology perspective on management. J Cutan Med Surg 2021;25:59-76.
  60. Ai L, Gao J, Zhao S et al. Nivolumab-associated DRESS in a genetic susceptible individual. J Immunother Cancer 2021;9:e002879. doi:10.1136/jitc-2021-002879
  61. Das S, Johnson DB. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J Immunother Cancer 2019;7:306
  62. Matsubara T, Uchi H, Takamori S et al. Acute Generalized Exanthematous Pustulosis Caused by the Combination of Pembrolizumab Plus Chemotherapy in a Patient With Squamous-Cell Carcinoma. Clin Lung Cancer 2020;21(2):e54-e56.
  63. Ständer S, Pereira MP, Yosipovitch G et al. IFSI-guideline on chronic prurigo including prurigo nodularis. Itch 2020;5:e42 http://dx.doi.org/10.1097/itx.0000000000000042
  64. Elmariah S, Kim BS, Yosipovitch G, et al. Practical approaches for diagnosis and management of prurigo nodularis: United States expert panel consensus. J Am Acad Dermatol 2021;(3):747-760. doi.org/10.1016/j.jaad.2020.07.025
  65. Guven DC, Kilickap S, Guner G, et al. Development of de novo psoriasis during nivolumab therapy in a patient with small-cell lung cancer. J Oncol Pharm Pract 2020;26:256-258.
  66. Voudouri D, Nikolaou V, Laschos K, et al. Anti-PD1/PDL1 induced psoriasis. Curr Probl Cancer 2017;41:407-412.
  67. Choi Y, Jang D, Byun HJ et al. Psoriasiform dermatitis related with T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains inhibitor in a patient with non-small-cell lung cancer. Ann Dermatol 2020;32(2):172-174. PMID: 33911733 | PMCID: PMC7992544
  68. Amaris N, Geisler BS, Lacouture ME et al. Immune checkpoint inhibitor-related dermatologic adverse events. J Acad Dermatol 2020;83(5):1255-1268.
  69. Phillips GS, Wu J, Hellmann MD, et al. Treatment outcomes of immune-related cutaneous adverse events. J Clin Oncol. 2019;37:2746–2758.
  70. Kato Y, Otsuka A, Miyachi Y, Kabashima K. Exacerbation of psoriasis vulgaris during nivolumab for oral mucosal melanoma. J Eur Acad Dermatol Venereol. 2016;30:e89–e91.
  71. Totonchy MB, Ezaldein HH, Ko CJ, Choi JN. Inverse psoriasiform eruption during pembrolizumab therapy for metastatic melanoma. JAMA Dermatol. 2016;152:590–592.
  72. Brahmer JR, Lacchetti C, Schneider BJ, et al. management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of clinical oncology clinical practice guideline. J Clin Oncol 2018;36:1714–68.doi:10.1200/JCO.2017.77.6385
  73. Elmets CA, Leonardi CL, Davis DMR, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with awareness and attention to comorbidities. J Am Acad Dermatol. 2019; 80: 1073–113. x
  74. Siciliano MA, Dastoli S, d’Apolito M et al. Pembrolizumab-Induced Psoriasis in Metastatic Melanoma: Activity and Safety of Apremilast, a Case Report. Front. Oncol. 2020;10:579445. doi: 10.3389/fonc.2020.579445
  75. Fattore D, Annunziata MC, Panariello L, Marasca C, Fabbrocini G. Successful treatment of psoriasis induced by immune checkpoint inhibitors with apremilast. Eur J Cancer 2019;110:107–9. doi: 10.1016/j.ejca.2019.01.010
  76. Frieder J, Kivelevitch D, Menter A: Secukinumab: a review of the anti-IL-17A biologic for the treatment of psoriasis. Ther Adv Chronic Dis. 2018;9:5-21. 1177/2040622317738910
  77. De Bock M, Hulstaert E, Kruse V, Brochez L: Psoriasis vulgaris exacerbation during treatment with a PD-1 checkpoint inhibitor: case report and literature review. Case Rep Dermatol. 2018;10:190-197. 10.1159/000491572
  78. Martinez-Domenech A, Garcia-Legaz Martinez M, Magdaleno-Tapial J, et al. Digital ulcerative lichenoid dermatitis in a patient receiving anti-PD-1 therapy. Dermatol Online J 2019;25.
  79. De la Paz Sarasola M, Taquez Delgado MA, Medina VA et al. Histamine in cancer immunology and immunotherapy. Current status and new perspectives. Pharmacol Res Perspect. 2021;9(5):e00778.
  80. Tziotzios C, Lee JYW, Brier T, et al. Lichen planus and lichenoid dermatoses. J Am Acad Dermatol 2018; 79: 789–818.
  81. Tattersall IW, Leventhal JS: Cutaneous toxicities of immune checkpoint inhibitors: The role of the dermatologist. Yale J Biol Med 2020;93:123-132.
  82. Lynde CW, Bergman J, Fiorillo L, et al. Clinical insights about topical treatment of mild-to-moderate pediatric and adult atopic dermatitis. J Cutan Med Surg. 2019 May/Jun;23(3_suppl):3S-13S.
  83. Eichenfield LF, Tom WL, Berger TG, et al. Guidelines of care for the management of atopic dermatitis: section 2. Management and treatment of atopic dermatitis with topical therapies. J Am Acad Dermatol. 2014;71:116–132.
  84. Yosipovitch G, Simpson EL, Tan H et al. Effect of crisaborole topical ointment, 2%, on atopic dermatitis–associated pruritus: an extended analysis of 2 phase 3 clinical trials. Itch 2019;4(1)p e12.
  85. Paller AS, Tom WL, Lebwohl MG, et al. Efficacy and safety of crisaborole ointment, a novel, nonsteroidal phosphodiesterase 4 (PDE4) inhibitor for the topical treatment of atopic dermatitis (AD) in children and adults. J Am Acad Dermatol. 2016 Sep;75(3):494-503.e6
  86. Simpson EL, Bieber T, Guttman-Yassky E, et al. Two phase 3 trials of Dupilumab versus placebo in atopic dermatitis. N Engl J Med. 2016;375(24):2335–2348
  87. Wollenberg A, Blauvelt A, Guttman-Yassky E et al. Tralokinumab for moderate-to-severe atopic dermatitis: results from two 52-week, randomized, double-blind, multicentre, placebo-controlled phase III trials (ECZTRA 1 and ECZTRA 2). Br J Dermatol. 2021;184(3):437-449.
  88. Fontecilla NM, Khanna T, Bayan CY, et al. Bullous pemphigoid associated with a new combination checkpoint inhibitor immunotherapy. J Drugs Dermatol 2019;18:103-104.
  89. Kubicki SL, Welborn ME, Garg N, et al. Granulomatous dermatitis associated with ipilimumab therapy (ipilimumab associated granulomatous dermatitis). J Cutan Pathol 2018;45:636-638.
  90. Siegel J, Totonchy M, Damsky W, et al: Bullous disorders associated with anti-PD-1 and anti-PD-L1 therapy: A retrospective analysis evaluating the clinical and histopathologic features, frequency, and impact on cancer therapy. J Am Acad Dermatol 2018;79:1081-1088.
  91. Lopez AT, Khanna T, Antonov N, et al. A review of bullous pemphigoid associated with PD-1 and PD-L1 inhibitors. Int J Dermatol 2018;57:664-669.
  92. Joly P, Roujeau JC, Benichou J et al. A comparison of oral and topical corticosteroids in patients with bullous pemphigoid. N Engl J Med 2002; 346:321-327
    DOI: 10.1056/NEJMoa011592
  93. EsfahaniK, Elkrief A, Calabrese C, et al. Moving towards personalized treatments of immune-related adverse events. Nat Rev Clin Oncol 2020;17:504–15. doi:10.1038/s41571-020-0352-8
  94. Fatima S, Veenstra J, Antonyan AS et al. Severe recalcitrant morbilliform eruption from dual immune checkpoint blockade. JAAD Case Rep 2018;4(6):593-595. PMID: 29955646
  95. Aizman L, Nelson K, Sparks AD, et al. The influence of supportive oncodermatology interventions on patient quality of life: A Cross-Sectional survey. J Drugs Dermatol. 2020May 1;19(5):477-82.
]]>
Tirbanibulin for the Treatment of Actinic Keratosis: A Review https://www.skintherapyletter.com/actinic-keratosis/tirbanibulin/ Mon, 15 Aug 2022 14:10:43 +0000 https://www.skintherapyletter.com/?p=13485 Austinn C. Miller, MD1; Susuana Adjei, MD1; Laurie A. Temiz, BA1,2; Stephen K. Tyring, MD, PhD, MBA1,3

1Center for Clinical Studies, Webster, TX, USA
2Meharry Medical College, Nashville, TN, USA
3Department of Dermatology, University of Texas Health Science Center, Houston, TX, USA

Conflict of interest:
Stephen Tyring received grant support, paid to the Center for Clinical Studies, from Athenex. The other authors have no conflicts of interest.

Abstract:
Actinic keratosis (AK) is a common precancerous condition found on chronically sun-damaged skin, particularly on the face, scalp, arms, and legs. Early and effective treatment of AKs is important to prevent progression to squamous cell carcinoma. Many topical treatments for AKs are often limited because of poor tolerability, prolonged treatment duration, and reduced adherence. Tirbanibulin 1% ointment, a new topical field therapy for AKs, reduces these issues. It requires a consecutive 5-day application period and is effective, demonstrating complete (100%) clearance of AK lesions in 49% of patients, partial (>75%) clearance in 72%, and a median reduction in lesion count of 87.5% while exhibiting a favorable safety profile, mild adverse events, improved tolerability, and long-term results.

Key Words:
tirbanibulin; actinic keratosis, field therapy, 5-fluorouracil, diclofenac, imiquimod, face, scalp

Introduction

Actinic keratosis (AK) is a common, recurrent precancerous condition found on chronically sun-damaged skin, particularly on the face, scalp, arms, and legs.1 Clinically AKs appear as macules, papules, or hyperkeratotic plaques with an erythematous background.2 Its prevalence steadily increases with age and prolonged sun-exposure. The American Academy of Dermatology (AAD) notes that approximately 60% of predisposed individuals over the age of 40 are diagnosed with at least one AK.3 Other risk factors include male gender, fair skin (Fitzpatrick type I-II), ultraviolet (UV) exposure, immunosuppression, previous history of AKs or skin cancer, human papillomavirus (HPV) infection, and genetic diseases.3

Cumulative UV exposure is considered a major risk factor for AK development because of the resultant modification of cellular repair mechanisms in keratinocytes.4 UVB irradiation causes the formation of thymidine dimers in DNA and mutations of the telomerase gene, whereas UVA indirectly induces DNA mutation through photo-oxidative stress.3 The clinical significance of AKs is secondary to the associated discomfort, cosmetic burden, and the possibility of progression to invasive squamous cell carcinoma (SCC).5 Rates of malignant transformation vary from 0.025% to 16%, and the risk of progression increases in patients with multiple AKs (more than five).3 While AKs are a risk factor for the development of SCC, it is impossible to predict which lesions will transform, therefore treatment of all AKs is recommended.3

Many treatments exist for AKs. Single or few discrete AKs are typically treated with cryosurgery.1 Treatment of multiple lesions and surrounding photo-damaged skin (field cancerization) includes topical agents and photodynamic therapy (Table 1).1 These treatments may be associated with local skin reactions of pain, irritation, redness, flaking, erosions, ulcerations, and irreversible skin changes of pigmentation and scarring.1 Furthermore, some treatments have to be administered over periods of weeks or months, which may reduce adherence and undermine treatment success.1

Table 1

Medication Mechanism of Action Dosing/ Application Efficacy Drawback/Adverse Effects Contraindications
Tirbanibulin 1% ointment Microtubule inhibitor and Src kinase inhibitor with potent antiproliferative activity against keratinocyte growth Applied once daily on face or scalp for 5 consecutive days; up to 25 cm2 contiguous treatment surface • 49% complete (100%) clearance of lesions

• 72% partial reduction (>75%) of lesions

• Median reduction in lesion count was 87.5%
• Local skin reactions: mostly mild-to-moderate erythema, flaking

• Scaling, crusting, swelling, vesiculation/ pustulation and erosion/ ulceration observed less commonly
None
5-fluorouracil 5% cream Antimetabolite cytotoxic agent (antipyrimidine group); interferes with DNA synthesis Applied to affected areas 1-2 times daily for 3-4 weeks • 38% of patients experienced complete clearance of lesions in 6 months8

• Overall 73% reduction in lesions8
• Local skin reactions: pain, pruritus, burning, erythema, erosion, inflammation, hyperpigmentation Pregnant women, patients with dihydropyrimidine dehydrogenase enzyme deficiency, and/or hypersensitivity to any components of the cream
Diclofenac 3% gel Inhibits the cyclooxygenase pathway, resulting in decreases in prostaglandin E2 synthesis Applied twice-daily applications for 8-12 weeks; up to 25 cm2 treatment surface • 47% of patients experienced complete clearance at 90 days of twice daily application • Local skin reactions: atopic dermatitis, cutaneous dryness, edema, pruritus, scaly rash, ulcerations, vesiculobullous rash

• Long duration

• No systemic adverse events
Patients with diclofenac, polyethylene glycol monomethyl ether 350, benzyl alcohol, and/or hyaluronate sodium hypersensitivity
Imiquimod 3.75% cream Imidazoquinoline-derivative promotes activation of innate immunity One cycle of 3 applications per week for 4 weeks, cycle can be repeated once; up to 25 cm2 contiguous treatment surface • 34% of patients experienced complete clearance

• 54% of patients experienced partial clearance
• Local skin reactions: pruritus, burning, erythema, pain, edema, dryness, crusting, erosions, ulcerations, scabbing

• Systemic reactions are rare

• Reactions occur less during a second treatment cycle
None
Table 1. Topical FDA-approved field therapies for AKs of the scalp and face.
Modified from: Dao D-PD, et al. 20214 and Dlott AH, et al. 202112

More recently, the role of the Src kinase in carcinogenesis has shed light on an alternative therapeutic option. In UV damaged skin, a cascade of events activates peroxisome proliferator activated receptor (PPAR) beta/delta which stimulates the Src oncogene expression, increases Src kinase activity and enhances the EGFR/Erk1/2 signaling pathway, resulting in increased epithelial-to-mesenchymal transition (EMT) marker expression.6 Elevated levels of Src have been linked to AKs and SCCs, and play a role in both primary tumor growth and metastases.7 Therefore, Src inhibitors were viewed as a plausible therapeutic option and many have since been developed. Tirbanibulin is a novel compound that inhibits Src kinase signaling and tubulin polymerization in rapidly dividing cells. It has shown promise as a new therapeutic agent for the treatment of AKs on the face or scalp.

Background

Mechanism of Action

The chemical name of tirbanibulin is N-benzyl-2-(5-(4-(2-morpholinoethoxy)phenyl)pyridine-2-yal) acetamide (Figure 1).4 It is a synthetic, first-in-class, anti-proliferative agent that disrupts Src kinase signaling and inhibits tubulin polymerization (Figure 2).8 Through these mechanisms, it also promotes the induction of p53, G2/M arrest of proliferating cell populations, and subsequent apoptosis.8

Chemical structure of tirbanibulin.
Figure 1. Chemical structure of tirbanibulin.
Borrowed from: Bartlett G. Line diagram of tirbanibulin [Internet]. 2020 [cited 2021 Oct 20]. Available from: https://commons.wikimedia.org/wiki/File:Tirbanibulin-line.png (Under the Creative Commons License)9
figure of two mechanisms of tirbanibulin
Figure 2. Two mechanisms of tirbanibulin. (Top) In UV-damaged skin, a cascade of events activates PPAR which stimulates Src oncogene expression and increased Src kinase activity that contributes to the pathogenesis of AKs and SCCs. (Bottom) By inhibiting dimerization of alpha and beta tubulin, tubulin polymerization is prevented, and microtubule formation is inhibited, thus halting essential cellular functions such as protein transport and cell division which leads to apoptosis and cell death.

Clinical Trials

Phase I8

The Phase I trial was an open-label, single-center study in subjects aged ≥18 years with clinically typical AKs on the forearm. Thirty participants were enrolled into 4 sequential cohorts (n=4, 10, 8, and 8 in each cohort, respectively): Cohort 1 received tirbanibulin ointment 1% 50 mg/day once daily for 3 days over 25 cm2 treatment area with 4-8 AK lesions; Cohort 2 received 200 mg/day once daily for 3 days over 100 cm2 treatment area with 8-16 AK lesions; Cohort 3 and Cohort 4 were similar to Cohort 1 and Cohort 2, respectively, but treatment was for 5 days. The follow-up period was through day 45.

To assess tirbanibulin activity, AK lesion numbers at baseline (day 1), days 10, 17, 31, and 45 were collected. Complete (100%) and partial (≥75%) AK clearance rates (defined by the reduction in AK lesions in the treatment area at day 45 compared with baseline) were evaluated for each cohort.

Twenty-nine participants completed the study with one participant withdrawing consent on day 2. Reductions in lesion counts from day 1 to 45 were observed in all cohorts. On day 45, Cohorts 1-4 demonstrated 25%, 0%, 50%, and 12.5% of complete AK clearance in the treatment area, respectively.

Dermal safety clinical studies in healthy subjects demonstrated that tirbanibulin 1% ointment did not cause contact sensitization, phototoxic skin reactions, or photoallergic skin reactions.4

Phase II8

The Phase II trial was an open-label, uncontrolled, dose-regimen- finding, multicenter study in subjects aged ≥18 years with clinically typical AKs on the face or scalp. One hundred and sixty-eight participants were enrolled into 2 sequential cohorts (n=84 in each cohort): Cohort 1 received 50 mg/day tirbanibulin ointment 1% once daily for 3 days over 25 cm2 treatment area with 4–8 AK lesions; Cohort 2 received the same treatment for 5 days.

AK lesion counts at baseline (day 1), 8, 15, 29, and 57 were collected, with a 12-month follow-up period for participants that achieved complete AK clearance to monitor for recurrence. All 168 participants completed the trial. Extensive overall AK clearance on the face or scalp was demonstrated in both cohorts. More participants had 100% clearance at day 57 in the 5-day (43% [95% Confidence Interval (CI) = 32, 54]) vs. the 3-day cohort (32% [95% CI = 22, 43]). Partial clearance rates were also slightly higher in the 5-day (56% [95% CI = 45, 67]) vs. the 3-day cohort (52% [95% CI = 41, 63]). There was a consistent decrease in lesion counts across all visits from baseline to day 57 for both cohorts.

All 63 participants who had 100% clearance at day 57 in the Phase 2 study were included in the Recurrence Follow-up Set. At 12 months post-day 57, recurrence rates for the 5-day cohort (57% [95% CI = 41, 73]) were lower than the 3-day cohort (70% [95% CI = 51, 87]). Most recurrence occurred within 6 months post-day 57.

Phase III1

The Phase III trial was a randomized, double-blind, parallel-group, vehicle-controlled, multicenter (62 US centers) trial in subjects aged >18 years with 4-8 clinically typical AKs on the face or scalp within a contiguous area measuring 25 cm2. A total of 702 participants, divided among two identical trials (n=351 at each site), were randomly assigned in a 1:1 ratio to receive either tirbanibulin 1% ointment for 5 days self-administered to a 25 cm2 contiguous area or vehicle ointment (placebo). Enrollment across patients was controlled to achieve a 2:1 ratio of facial: scalp treatment areas. The primary outcome was the percentage of patients with a complete reduction in the number of lesions in the application area at day 57. The secondary outcome was the percentage of patients with a partial reduction in the number of lesions within the application area at day 57. The incidence of recurrence was evaluated at 1 year.

Tirbanibulin demonstrated higher complete clearance levels in trial 1 (44% [95% CI = 32, 47]; P<0.001) and trial 2 (54% [95% CI = 33, 51]; P<0.001) compared to the vehicle groups (5% and 13% in trials 1 and 2, respectively). Pooled data from both trials demonstrated complete clearance in 49% ([95% CI = 35, 47]; P<0.001) of patients in the tirbanibulin groups and in 9% of the vehicle groups.

Partial clearance was significantly higher in both tirbanibulin groups at 68% (trial 1) and 76% (trial 2) than in the vehicle groups. Pooled data revealed partial clearance in 72% ([95% CI = 48, 60]; P<0.001) of patients in the tirbanibulin groups and in 18% of the vehicle groups. At 1 year, 47% of patients with complete clearance experienced recurrence of AKs.

Median reduction in AK lesion count in patients received tirbanibulin was 87.5% vs. 20% for vehicle (P<0.0001).1

Safety and Tolerability

Throughout all phases, no serious adverse events (AEs) were reported secondary to tirbanibulin.1,8 Local skin reactions (LSRs) consisted mostly of mild-moderate erythema, flaking or scaling, application-site pruritus, and application-site pain.1 Erythema (93%) and flaking (82%) were most common.1 Severe LSRs (all types) were observed in <10% of patients, with severe erythema in 6% of patients.1 Other LSRs included crusting, swelling, scaling, vesiculation or pustulation, and ulcerations or erosions.4 LSRs typically appeared on treatment day 2 and peaked on treatment day 8, before spontaneously resolving in ~2 weeks-1 month.1,8 No participants were withdrawn from the clinical trials due to AEs or LSRs.

Laboratory evaluation of blood chemistry, hematology, urine analysis, vital signs, electrocardiograms, and physical examinations in association with the phase III trials were not indicative of any systemic side effects.1 Tirbanibulin produces adverse ophthalmic reactions, and therefore, patients must be careful not to transfer the drug into the periocular area or eyes.

There has been no data on possible birth defects and/or adverse fetal/maternal outcomes during pregnancy with the use of tirbanibulin.4 However, extremely high doses were noted to cause birth defects in rats and rabbits (more than 70 and 159 times the recommended human dose, respectively).4

Regulatory Approval

The topical 1% ointment formulation of tirbanibulin was approved by the US FDA in December 2020 for the treatment of AKs on the face and scalp.7 One packet of ointment (250 mg) contains 2.5 mg (1%) of tirabanibulin.4 The medication is applied on the affected area of the face or scalp once a day for 5 consecutive days.4 Each packet will cover up to 25 cm2 on the face or scalp and is disposed of after its one-time use.4

There are no contraindications listed in the FDA-approved prescribing information.4

Discussion

Compared to other older topical treatments for AKs, tirbanibulin demonstrates several advantages. Tirbanibulin effectively and completely cleared AK lesions with a 5-day application while demonstrating a favorable safety profile and long-term results. The most common AEs for tirbanibulin were mild and included erythema, flaking, pruritus and pain at the application site. Unlike most other topical treatments for AKs, severe local
reactions, including vesiculation or pustulation and erosion or ulceration, were infrequent.1 The favorable tolerability is attributed to tirbanibulin mechanism of action in which it induces apoptosis rather than necrosis. Apoptosis is associated with little or no inflammation.10

Treatment adherence and patient satisfaction has shown to be significantly better with shorter duration topical treatments for AK.11 Current first-line topical field therapies for AKs include 5-fluorouracil (5-FU), imiquimod, and diclofenac gel, all of which have unfavorable aspects undermining compliance and thus successful treatment (Table 1).12 These therapies more frequently result in AEs, severe LSRs, and prolonged patient discomfort, at times necessitating a pause in treatment until the healthy skin has healed.12 In addition, each requires an extended period of application ranging from 3-4 weeks to 8-12 weeks with more than one application daily and/or a second cycle.12 Ingenol mebutate, a promising short duration (3-day) AK treatment, recently lost its first-line status after being discontinued in 2020 secondary to increased risk of skin malignancy.

A combination of 5% 5-FU cream plus 0.005% calcipotriol ointment has been used off-label to treat AKs with variable results.13 The evidence was based on a single center study using the combination topical treatment twice daily for 4 days applied to 25 cm2 area.13 Twenty-seven percent of patients achieved complete clearance of AK lesions on the face, and <20% on the scalp at week 8.13 Severe erythema on the face was observed in 80% of patients.13

The incidence of recurrence with conventional treatments has ranged from 20% to 96%.1 However, owing to the chronic nature of AKs, recurrence of lesions in sun-damaged areas is expected.1 With tirbanibulin, 47% of patients with complete clearance experienced a recurrence at 1 year in the phase III trials. Despite the apparent benefits of tirbanibulin, no direct comparisons have been made with other AK treatments.1 Ultimately, head-to-head studies will be needed to determine true superiority/inferiority.

Other uses of tirbanibulin are being explored. Phase I trials are underway in Taiwan to investigate its potential as a treatment for psoriasis.7 Tirbanibulin was noted to successfully eradicate periungual SCC in a patient that failed to clear with imiquimod 5% in combination with monthly cryotherapy, suggesting its potential as a nonsurgical therapeutic option for SCC.14

Conclusion

Early and effective treatment of AKs is important to prevent progression to SCC.4 Many topical treatments for AKs are often limited because of patient tolerability, treatment duration, and adherence. Tirbanibulin effectively and completely cleared AK lesions with a 5-day application period while demonstrating a favorable safety profile, mild AEs, improved tolerability, and long-term results, making it a promising field therapy for AKs.

References



  1. Blauvelt A, Kempers S, Lain E, et al. Phase 3 Trials of tirbanibulin ointment for actinic keratosis. N Engl J Med. 2021 Feb 11;384(6):512-20.

  2. Reinehr CPH, Bakos RM. Actinic keratoses: review of clinical, dermoscopic, and therapeutic aspects. An Bras Dermatol. 2019 Dec;94(6):637-57.

  3. Dianzani C, Conforti C, Giuffrida R, et al. Current therapies for actinic keratosis. Int J Dermatol. 2020 Jun;59(6):677-84.

  4. Dao DD, Sahni VN, Sahni DR, et al. 1% tirbanibulin ointment for the treatment of actinic keratoses. Ann Pharmacother. 2022 Apr;56(4):494-500.

  5. Balcere A, Rone Kupfere M, Čēma I, et al. Prevalence, discontinuation rate, and risk factors for severe local site reactions with topical field treatment options for actinic keratosis of the face and scalp. Medicina (Kaunas). 2019 Apr 4;55(4):E92.

  6. Montagner A, Delgado MB, Tallichet-Blanc C, et al. Src is activated by the nuclear receptor peroxisome proliferator-activated receptor β/δ in ultraviolet radiation-induced skin cancer. EMBO Mol Med. 2014 Jan;6(1):80-98.

  7. Markham A, Duggan S. Tirbanibulin: first approval. Drugs. 2021 Mar;81(4): 509-13.

  8. Kempers S, DuBois J, Forman S, et al. Tirbanibulin ointment 1% as a novel treatment for actinic keratosis: phase 1 and 2 results. J Drugs Dermatol. 2020 Nov 1;19(11):1093-100.

  9. Bartlett G. Line diagram of tirbanibulin [Internet]. 2020 Dec 17 [cited 2021 Oct 20]. Available from: https://commons.wikimedia.org/wiki/File:Tirbanibulinline.png.

  10. Wallach D, Kovalenko A. Keeping inflammation at bay. Elife. 2014 Mar 25;3:e02583.

  11. Grada A, Feldman SR, Bragazzi NL, et al. Patient-reported outcomes of topical therapies in actinic keratosis: a systematic review. Dermatol Ther. 2021 Mar;34(2):e14833.

  12. Dlott AH, Di Pasqua AJ, Spencer SA. Tirbanibulin: topical treatment for actinic keratosis. Clin Drug Investig. 2021 Sep;41(9):751-5.

  13. Cunningham TJ, Tabacchi M, Eliane J-P, et al. Randomized trial of calcipotriol combined with 5-fluorouracil for skin cancer precursor immunotherapy. J Clin Invest. 2017 Jan 3;127(1):106-16.

  14. Moore AY, Moore S. Topical tirbanibulin eradication of periungual squamous cell carcinoma. JAAD Case Rep. 2021 Jun 26;14:101-3.


Purchase Article PDF for $1.99

]]>
Transition of Topical Therapy Formulation in Psoriasis: Insights from a Canadian Practice Reflective https://www.skintherapyletter.com/psoriasis/transition-topical-therapy/ Fri, 01 Apr 2022 20:00:38 +0000 https://www.skintherapyletter.com/?p=13288 David N. Adam, MD, FRCPC1-3; Sonya J. Abdulla, MD, FRCPC4; Patrick Fleming, MD, FRCPC1; Melinda J. Gooderham, MD, FRCPC5; John Ashkenas, PhD6; Clinton B. McCracken, PhD7

1Division of Dermatology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
2Baywood Dermatology and CCA Medical Research, Ajax, ON, Canada
3Probity Medical Research, Waterloo, ON, Canada
4Dermatology on Bloor, Toronto ON, Canada
5Skin Centre for Dermatology, Peterborough, ON, Canada
6imc North America, Toronto, ON, Canada
7LEO Pharma Inc. Canada, Thornhill, ON, Canada

Conflict of interest:
D. Adam has been an investigator, speaker, or advisory board member for LEO Pharma, AbbVie, Amgen, Arcutis, Bausch Health, Boehringer Ingelheim, BMS, Celgene, Coherus, Dermira, Dermavant, Eli Lilly, Galderma, Incyte, Janssen, Merck, Novatrtis, Pfizer, Regeneron, Sanofi Genzyme, Sun Pharma, and UCB. S. Abdulla has been a speaker or advisory board member for LEO Pharma, AbbVie, Celgene, Eli Lilly, Galderma, Janssen, Novartis, Pfizer, Sanofi Genzyme, UCB, and Bausch/Valeant. P. Fleming has received honorarium and/or consulting and/or advisory boards and/or speaking fees for AbbVie, Altius, Aralez, Bausch Health, Cipher, Galderma, Eli Lilly, UCB, Janssen, Novartis, Pfizer, and Sanofi-Genzyme. M. Gooderham has been an investigator, speaker, or advisory board member for LEO Pharma, AbbVie, Amgen, Akros, Arcutis, Boehringer Ingelheim, BMS, Celgene, Dermira, Dermavant, Eli Lilly, Galderma, GSK, Janssen, Kyowa Kirin, Medimmune, Merck, Novartis, Pfizer, Regeneron, Sanofi Genzyme, Sun Pharma, UCB, and Bausch/Valeant. J. Ashkenas received support via imc North America (Toronto, ON) from LEO Pharma Inc. Canada for participating in the development of the practice reflective and for analyzing the findings. He has no other financial interest to declare. C. McCracken is employed by LEO Pharma Inc. Canada.

Abstract:
Patient preferences for psoriasis treatment may affect treatment adherence and disease control; changing topical formulation may improve adherence and patient acceptance of treatment. This study explored dermatologists’ reasons for transitioning psoriasis patients from an ointment or gel (Dovobet®) formulation to an aerosol foam (Enstilar®) formulation of calcipotriol and betamethasone dipropionate (Cal/BD), and to assess the success of this transition. Medical records of 81 Canadian patients from 9 dermatologists were retrospectively reviewed for symptoms affecting quality of life, reasons for transitioning treatment, and whether transition was successful. Reasons for transition included efficacy, quality of life, and patient adherence. At follow-up, median psoriasis severity and body surface area affected had decreased from baseline, and patients experienced improved quality of life. Itch and itch-related sleep loss, which were identified as burdensome in 63% of patients at baseline, had resolved in 33% and improved in 54% of patients at follow-up. Dermatologists deemed the transition successful in 85% of patients, with the most common reasons being patient-reported success, clearance of signs/symptoms, and continued prescription refills. Transition from Cal/BD ointment or gel to aerosol foam was generally deemed successful by patients and dermatologists, and was associated with improved quality of life and improved itch control.

Key Words:
psoriasis; topical treatment; foam; formulation; fixed combination

Introduction

Psoriasis is a chronic condition that commonly requires topical treatment, either as monotherapy or in combination with phototherapy or systemic treatment. Given the challenge of consistent long-term adherence to treatment for psoriasis,1-3 patient acceptance of topical treatment is an important consideration.4,5 Formulations with more appealing cosmetic features could affect patient acceptance, and therefore adherence and ultimately disease control.6-9

Calcipotriol and betamethasone dipropionate (Cal/BD) are the active ingredients in Dovobet® ointment or gel (Cal/BD ointment or gel) and Enstilar® aerosol foam (Cal/BD foam). Cal/BD aerosol foam has improved skin penetration and is reported to be better accepted by patients, relative to Cal/BD ointment or gel.10 Switching to Cal/BD foam can improve disease control. Significantly more patients using Cal/BD foam achieved a 2-grade improvement according to the physician’s global assessment of disease severity, with their psoriasis being clear or almost clear, by week 4 compared with patients using Cal/BD gel by week 8 (38.3% vs. 22.5%, respectively; P < 0.001).11,12 Given that the active ingredients are the same, comparisons between the products relate purely to formulation.

In this study, medical records of Canadian psoriasis patients who transitioned from Cal/BD ointment or gel to Cal/BD foam were examined retrospectively. The objective was to explore Canadian dermatologists’ reasons for changing topical treatment formulation in these patients and to assess reasons the transition was judged successful or unsuccessful. As patients on topical monotherapy may have different motivations for and success with treatment transition, compared with those receiving concomitant topical and non-topical therapies (i.e., systemic treatment or phototherapy), the study included patients from both populations.

Patients and Methods

In this retrospective chart review, 9 Canadian dermatologists reviewed the medical records of their patients with plaque psoriasis who were previously treated with Cal/BD ointment or gel and were switched to treatment with Cal/BD foam. The study protocol was approved by RRB (Research Review Board Inc., Waterloo, ON, Canada; approval # 2019.538). The need for informed consent was waived due to the retrospective design of the study.

The dermatologists reviewed the medical records from their 10 most recent patients meeting all inclusion criteria: ≥18 years of age, with active psoriasis, and transitioned from Cal/BD ointment or gel to Cal/BD foam at the baseline visit (defined as the visit where the change in treatment occurred). Patients could not have made any change in prescribed systemic treatment or phototherapy from 3 months before to 1 month after the switch to Cal/BD foam. Baseline findings were from the visit during which the dermatologist first prescribed Cal/BD aerosol foam. Follow-up data were from the first subsequent visit, no less than 4 weeks and no more than 32 weeks after the baseline visit.

Using the data collected from the medical records, the dermatologists completed an online questionnaire developed from a pilot study carried out by 3 authors of the current study. The pilot study sought to identify classes of data that could be reliably found in medical records for psoriasis patients seen in normal Canadian dermatology practice. Pilot study data were not included in the current report.

Demographic and clinical characteristics extracted from the baseline visit records included age, gender, symptoms affecting quality of life, the dermatologist’s reasons for transitioning the patient to Cal/BD foam, the number of clinic visits in the 12 months prior to baseline, and history of psoriasis therapies in the 24 months prior to baseline. Patients were considered to be on topical monotherapy if they were using Cal/BD ointment or gel with or without other prescription or non-prescription topical agents, but without systemic treatment or phototherapy. Conversely, patients were considered as on combination therapy if they were using Cal/BD ointment or gel concomitant with phototherapy or systemic therapy. Physicians submitted their findings for each patient electronically until they had completed 10 reports. After the seventh report of patients in one class of treatment (topical monotherapy or combination therapy), the physician received an automatic reminder that both classes should be represented among their 10 reports.

The dermatologist’s assessments of psoriasis severity, as noted in the medical records, for the patient’s lifetime worst severity, severity at baseline, and severity at follow-up, were reported in the questionnaire using a 5-point scale (0 = absent; 1 = mild; 2 = moderate; 3 = severe; 4 = very severe). Estimated percent body surface area (BSA) affected by psoriasis was extracted from the baseline and follow-up visits. The dermatologist’s assessments of improvement in quality of life, the impact of Cal/BD foam on overall psoriasis disease and itch or itch-related sleep loss, and whether the transition to Cal/BD was successful, were extracted from the records of the follow-up visit. Improvement in quality of life was rated on a 4-point scale (0 = no improvement; 4 = significant improvement). The level of impact on overall psoriasis disease and itch or itch-related sleep loss was rated as ‘Worsened’, ‘Same’, ‘Improved’, or ‘Resolved’. Dermatologists were asked to identify one or more specific signs or symptoms with the greatest impact on quality of life. If the patient’s record did not include the relevant information, dermatologists indicated ‘Unknown/Not documented’.

Descriptive statistics were analyzed using Microsoft® Excel for Mac version 16.40 and reported as mean ± standard deviation (SD), median (range), and proportions. Missing data were not imputed, and the results for each variable were reported based on the number of patients for whom data were available.

Results

Nine dermatologists completed questionnaires for 10 patients each. Of those 90 questionnaires, 9 were excluded, leaving 81 patient records for analysis. Reasons for exclusion were that insufficient data were included (n = 1), Cal/BD foam was not used between baseline and follow-up (n = 4), and the follow-up visit was not within the 4 to 32-week timeframe (n = 4).

Baseline demographic and clinical characteristics are shown in Table 1. More patients were on monotherapy than on combination therapy. Psoriasis at lifetime worst was less severe in patients on topical monotherapy vs. combination therapy. The most commonly affected body areas were the legs, arms, trunk, and scalp.

Baseline characteristic All patients
(N = 81)
Patients on monotherapy
(n = 48)
Patients on combination therapy
(n = 33)

Na


Mean ± SD or n (%)


Na


Mean ± SD or n (%)


Na


Mean ± SD or n (%)


Male 45 (55.6) 26 (54.2) 19 (57.6)
Age, years 51.2 ± 17.5 51.8 ± 17.7 50.5 ± 17.6
Duration of psoriasis, years 46 11.1 ± 8.6 25 11.2 ± 9.1 21 11.0 ± 8.1
Psoriasis severity at worst 75 2.5 ± 0.9 44 2.3 ± 0.9 31 2.9 ± 0.8
Affected body areas 73 41 32
Scalp 31 (42.5) 14 (34.2) 17 (53.1)
Face 12 (16.4) 7 (17.1) 5 (15.6)
Body 65 (89.0) 34 (82.9) 31 (96.9)
Arms 65 (89.0) 34 (82.9) 31 (96.9)
Trunk 39 (53.4) 14 (34.2) 25 (78.1)
Legs 50 (68.5) 25 (61.0) 25 (78.1)
Hands 13 (17.8) 5 (12.2) 8 (25.0)
Skin folds 1 (1.4) 0 (0.0) 1 (3.1)
Other 9 (12.3) 4 (9.8) 5 (15.6)
Table 1: Patient demographics and baseline characteristics
aNumber of patient records from which the relevant data were available if less than the total.

 

Complete data for psoriasis severity (at worst, at baseline, and at follow-up) were available for 72 patients (Figure 1); with the remainder missing data from at least one of those time points. In those patients with complete data, severity at worst was mild or moderate in 36 (50%) patients. At baseline and at follow-up, 53 (74%) patients and 62 (86%) patients, respectively, experienced mild or moderate disease.

Bar chart of number of patients per category of psoriasis severity for psoriasis at its worst, at baseline, and at follow-up in patients for whom psoriasis severity was reported all timepoints (n = 72).
Figure 1. Number of patients per category of psoriasis severity for psoriasis at its worst, at baseline, and at follow-up in patients for whom psoriasis severity was reported all timepoints (n = 72).
Severity score: 0 = absent; 1 = mild; 2 = moderate; 3 = severe; 4 = very severe.

Patients who had experienced severe, or very severe psoriasis at its worst, were more likely to be on combination therapy and had greater baseline psoriasis severity and BSA affected, relative to those with mild or moderate psoriasis at worst (Table 2). For patients with severe or very severe disease, scaling, itch or itch-related sleep loss, and redness were identified as the signs and symptoms with the greatest impact on the quality of life. Among patients with mild or moderate psoriasis at worst, itch or itch-related sleep loss had the greatest impact on the quality of life. Similarly, redness had the greatest impact on quality of life for 8 of 14 (57%) patients on combination therapy but 3 of 14 (21%) patients on monotherapy, whereas itch or itch-related sleep loss had the greatest impact in 10 of 14 (71%) patients on monotherapy but 7 of 14 (50%) patients on combination therapy.

Baseline characteristic Severity score for psoriasis at worst Patients on monotherapy Patients on combination therapy
1 or 2
(N = 37)


3 or 4
(N = 38)


(N = 48)


(N = 33)


Na mean ± SD or n (%) Na mean ± SD or n (%) Na mean ± SD or n (%) Na mean ± SD or n (%)
Psoriasis severity at baseline 1.9 ± 0.7 37 2.3 ± 0.9 45 2.1 ± 0.8 32 2.1 ± 0.8
BSA at baseline 27 4.9 ± 5.5 29 10.9 ± 11.9 36 8.0 ± 9.9 23 7.5 ± 9.2
Symptoms with greatest impact on QoL 11 16 14 14
Itch/itch-related sleep loss 8 (72.7) 9 (56.3) 10 (71.4) 7 (50.0)
Scaling 5 (45.5) 10 (62.5) 8 (57.1) 7 (50.0)
Redness 3 (27.3) 8 (50.0) 3 (21.4) 8 (57.1)
Stigma 4 (36.4) 5 (31.3) 6 (42.9) 4 (28.6)
Dryness 3 (27.3) 5 (31.3) 3 (21.4) 5 (35.7)
Poor sleep 0 (0.0) 1 (6.3) 0 (0.0) 1 (7.1)
Other 1 (9.1) 2 (12.5) 0 (0.0) 3 (21.4)
Type of therapy
Monotherapy 27 (73.0) 17 (44.7)
Combination therapy 10 (27.0) 21 (55.3)
Table 2: Correlates of psoriasis severity at its worst
aNumber of patient records from which the relevant data were available if less than the total. QoL, quality of life.

 

The most common reasons cited for transitioning to Cal/BD foam were to improve clinical efficacy (74/81; 91%), quality of life (49/81; 61%), and treatment adherence (41/81; 51%). These reasons were ranked similarly for patients on monotherapy and on combination therapy (data not shown).

In patients for whom data were available at both baseline and follow-up, median psoriasis severity score (range) was 2.0 (1.0-4.0) at baseline and 1.0 (0.0-4.0) at follow-up. Median BSA was 4.0 (1.0-45.0) at baseline and 1.5 (0.0-25.0) at follow-up. The median and overall ranges for psoriasis severity and BSA were similar for patients on mono- and combination therapy. These findings were comparable across all 9 dermatologists’ patient sets.

At follow-up, quality of life as assessed by the investigator had improved for 79% and worsened for 4% of the 80 patients for whom quality of life data were reported (Figure 2A). Improvement was also experienced by 5/10 (50%) patients for whom stigma was reported as having a major impact on quality of life. Itch or itch-related sleep loss resolved in 33% and improved in 54% of patients overall (Figure 2B). Of note, very similar improvements were observed for both quality of life and itch/itch-related sleep loss measures in subsets of patients who cited those specific respective factors as the reason for seeking to transition. Overall, disease resolved in 16%, improved in 66%, and stayed the same in 15% but worsened in 3% of patients (n = 79) at the follow-up visit.

Transition of Topical Therapy Formulation in Psoriasis: Insights from a Canadian Practice Reflective - image
Figure 2. The proportion of patients for whom (A) quality of life and (B) itch or itch-related sleep loss worsened, stayed the same, improved, or resolved after transitioning from a Cal/BD ointment or gel to a Cal/BD foam. Cal/BD, calcipotriol and betamethasone dipropionate.

In each category of psoriasis severity at baseline, at least half of patients experienced improved quality of life, improved or resolved overall disease, and improved or resolved itch or itch-related sleep loss (Table 3). When grouped by baseline severity, the proportions of patients showing improved quality of life (mild: 15/17, 88%; moderate: 33/40, 83%; severe/very severe: 12/20, 60%) and improved itch or itch-related sleep loss (mild: 11/17, 65%; moderate: 20/40, 50%; severe: 11/20, 60%) were highest for the mild severity patients but roughly similar across groups; the study was not powered to draw any conclusions with respect to improvement according to baseline severity.

Outcome Psoriasis severity at baseline
Mild
n (%)
Moderate
n (%)
Severe/Very severe
n (%)
Number of patients 17 40 20
QoL improved 15 (88.2) 33 (82.5) 12 (60.0)
Overall disease improved or resolved 14 (82.4) 65 (87.5) 13 (65.0)
Itch or itch-related sleep loss improved or resolved 11 (64.7) 20 (50.0) 11 (55.0)
Table 3: Outcomes correlated with baseline psoriasis severity
Reported for the 77 patients for whom the baseline severity data were available. QoL, quality of life.

 

Transition was considered successful in 69 of 81 (85%) patients. Of the 12 patients for whom transition was deemed unsuccessful, 7 needed systemic treatment, including biologics (3 and 4 patients on monotherapy and combination therapy, respectively), and no improvement was seen in 2 (1 on monotherapy; 1 on combination therapy). In addition, 1 patient disliked the cosmetic features of the aerosol foam; for 2 patients, no reason was cited. The most common reasons cited for considering transition successful were that the patient reported success (40/69; 58%), signs and symptoms of psoriasis had cleared at follow-up (32/69; 46%), and the patient continued to fill prescriptions (22/69; 32%).

Discussion

In this retrospective chart review, the transition from a Cal/ BD ointment or gel formulation (Dovobet®) to a Cal/BD foam formulation (Enstilar®) was rated successful in most cases. The most common reason cited was patient-reported success, and most patients experienced improvement in the specific measure (e.g., itch or itch-related sleep loss) cited as a reason for wanting to transition. These findings are supported by results of a previous survey in which Canadian respondents generally rated Cal/BD foam as more pleasant cosmetically, relative to previously used topical products,8,9 consistent with other data that foam products are well accepted relative to other formulations.6

Transition success in the current study did not appear related to whether the patient was on mono- vs. combination therapy, as similar benefits were found in both groups. These benefits included improvements in overall disease, quality of life, and itch and itch-related sleep loss.

Itch is a pervasive challenge for psoriasis management, and itch severity is broadly associated with clinical severity.13 In this population, itch or itch-related sleep loss was reported in all severity groups. Following transition to Cal/BD foam, most patients with mild disease at baseline, as well as approximately half of others, experienced improvement or resolution of itch or itch-related sleep loss. Resolution was reported at a similar rate in the overall population and in patients for whom itch control was identified as a reason for treatment change. Given the clear disease burden associated with itch, dermatologists should ask their patients about their experience of itch and itch-related sleep loss, regardless of overall disease severity, and they should consider a change of topical treatment regimen in those who are dissatisfied with their current level of itch control.

Strengths and Limitations

This was a retrospective chart review of individuals transitioning between topical formulations, namely Cal/BD ointment or gel, to Cal/BD foam. The population may not be fully representative of Canadian psoriasis patients under a dermatologist’s care, since it would be expected to disproportionately include patients who are unsatisfied with Cal/BD gel or ointment. Due to the absence of a control group, no firm conclusions can be drawn regarding changes in symptoms of psoriasis related to the use of Cal/BD foam. For similar reasons, the findings here may not be generalized to other topical formulation transitions, including other transitions to foam formulations.

Conversely, strengths of this study include the real-world setting of treatment and the fact that the questionnaire used was developed through a pilot study that helped ensure that sufficient data could be captured for most variables of interest. It is also reassuring to note that the 9 physicians reported similar levels of treatment success among their individual patient sets, suggesting that the questionnaire was reliable and transparent to the respondents.

Conclusion

The benefits of transitioning from Cal/BD ointment or gel to aerosol foam formulation in this Canadian patient population were similar to those reported in clinical studies and were seen consistently among patients with a range of treatment priorities and with differing history of psoriasis severity and treatment history. Changing the topical treatment formulation, even without a change in active ingredients, should be considered for patients who are dissatisfied with their current topical treatment, independent of their psoriasis severity.

Acknowledgements

The authors thank the dermatologists who participated in the survey, Alice Kowalczyk, PharmD (LEO Pharma Inc. Canada) and Kristel Bermejo, PhD (imc North America, integrated medhealth communication) for contributing to the study design, and Celeste Lavallee, MSc (imc North America, integrated medhealth communication) for contributing to the data analysis and writing the article.

References



  1. Albrecht L, Bourcier M, Ashkenas J, et al. Topical psoriasis therapy in the age of biologics: evidence-based treatment recommendations. J Cutan Med Surg. 2011 Nov-Dec;15(6):309-21.

  2. Ali SM, Brodell RT, Balkrishnan R, et al. Poor adherence to treatments: a fundamental principle of dermatology. Arch Dermatol. 2007 Jul;143(7):912-5.

  3. Nolan BV, Feldman SR. Adherence, the fourth dimension in the geometry of dermatological treatment. Arch Dermatol. 2009 Nov;145(11):1319-21.

  4. Canadian Psoriasis Guidelines Addendum Committee. 2016 addendum to the Canadian guidelines for the management of plaque psoriasis 2009. J Cutan Med Surg. 2016 Sep;20(5):375-431.

  5. Canadian Psoriasis Guidelines Committee. Canadian guidelines for the management of plaque psoriasis: overview. J Cutan Med Surg. 2011 Jul-Aug;15(4):210-9.

  6. Housman TS, Mellen BG, Rapp SR, et al. Patients with psoriasis prefer solution and foam vehicles: a quantitative assessment of vehicle preference. Cutis. 2002 Dec;70(6):327-32.

  7. Svendsen MT, Feldman SR, Tiedemann SN, et al. Psoriasis patient preferences for topical drugs: a systematic review. J Dermatolog Treat. 2021 Aug;32(5): 478-83.

  8. Vender R, Gooderham MJ, Guenther LC, et al. Canadian patients’ preferences in topical psoriasis care: insights from the PROPEL surveys. J Cutan Med Surg. 2018 Sep/Oct;22(5):464-71.

  9. Vender R, Gooderham MJ, Guenther LC, et al. Psoriasis patients’ preference for an aerosol foam topical formulation. J Eur Acad Dermatol Venereol. 2018 Nov;32(11):e400-e1.

  10. Lind M, Nielsen KT, Schefe LH, et al. Supersaturation of calcipotriene and betamethasone dipropionate in a novel aerosol foam formulation for topical treatment of psoriasis provides enhanced bioavailability of the active ingredients. Dermatol Ther (Heidelb). 2016 Sep;6(3):413-25.

  11. Paul C, Leonardi C, Menter A, et al. Calcipotriol plus betamethasone dipropionate aerosol foam in patients with moderate-to-severe psoriasis: sub-group analysis of the PSO-ABLE study. Am J Clin Dermatol. 2017 Jun;18(3):405-11.

  12. Paul C, Stein Gold L, Cambazard F, et al. Calcipotriol plus betamethasone dipropionate aerosol foam provides superior efficacy vs. gel in patients with psoriasis vulgaris: randomized, controlled PSO-ABLE study. J Eur Acad Dermatol Venereol. 2017 Jan;31(1):119-26.

  13. Pithadia DJ, Reynolds KA, Lee EB, et al. Psoriasis-associated itch: etiology, assessment, impact, and management. J Dermatolog Treat. 2020 Feb;31(1): 18-26.


Purchase Article PDF for $1.99

]]>
Canadian Skin Management in Oncology Group (Casmo) Algorithm for the Prevention and Management of Acute Radiation Dermatitis https://www.skintherapyletter.com/skin-cancer/casmo-algorithm-prevention-management-acute-radiation-dermatitis/ Mon, 01 Nov 2021 00:00:49 +0000 https://www.skintherapyletter.com/?p=12903 Tarek Hijal, MD, FRCPC1, Maxwell B Sauder, MD, FRCPC2, Anneke Andriessen, PhD3, Joel Claveau, MD, FRCPC4, Charles W Lynde, MD, FRCPC5

Affiliations



  1. Associate Professor, Department of Oncology, McGill University, Director, Division of Radiation Oncology, McGill University Health Centre, Montreal, QC, Canada;

  2. Diplomate, American Board of Dermatology; Fellow, Royal College of Physicians and Surgeons of Canada; Onco-dermatologist, Princess Margaret Cancer Centre, Director, Pigmented Lesion Clinic, Toronto Dermatology Centre, Toronto, ON, Canada.

  3. Radboud UMC, Nijmegen and Andriessen Consultants, Malden, The Netherlands Diplomate, American Board of Dermatology, Fellow, Royal College of Physicians and

  4. Surgeons of Canada, Associate Professor, Department of Medicine, Laval University, Director Melanoma and Skin Clinic, Le Centre Hospitalier Universitaire de Québec, Hôtel-Dieu de Québec, Quebec City, QC, Canada;

  5. Diplomate, American Board of Dermatology, Fellow, Royal College of Physicians and Surgeons of Canada, Associate Professor, Department of Medicine University of Toronto, Toronto, ON, Canada, Lynderm Research, Markham, ON, Canada


PDF Download

Acknowledgments and Disclosure: The authors disclosed receipt of the following financial support for the research, authorship, and publication of this manuscript. This work was supported by an unrestricted educational grant from La Roche-Posay Canada.

All authors contributed to the development of this work and its review and agreed with its content.


Introduction

Up to one out of two Canadians will develop cancer in their lifetime.1 It has been estimated that 225,800 Canadians were diagnosed with cancer in 2021.1

Approximately 30-40% of cancer patients receive radiotherapy.2 A study in Ontario on breast, colorectal, lung, and prostate cancer showed that in the year following the cancer diagnosis, 96,003 patients received radiation therapy.2

Radiotherapy damages cancerous cells’ by directly or indirectly damaging their DNA via ionization and free-radical formation.3-5 Radiation-induced cutaneous AEs occur in up to 95% of cancer patients who will develop some form of radiodermatitis (RD).3-5 Of those with breast cancer, 87% develop RD, and concurrent anti-cancer therapies may aggravate RD in 90% of head and neck patients.4,5

RD affects the area that received irradiation, and the severity depends on the area treated, beam energy, total dose, dose per fraction, treatment schedule, and use of concomitant systemic agents. RD varies considerably in severity, course, and prognosis and can have severe sequelae that impact the quality of life (QoL) and potentially alter cancer treatment course and outcomes.3-7

Radiation treatment’s impact on QoL may be profound. A prospective study conducted from July 2017 to June 2018 using the Skindex-16 pre-and post-treatment included eighty-three cancer patients [breast cancer (49%), head and neck cancer (45%), and anal canal cancer (6%)] receiving radiation treatment.7 All patients developed RD [Grade 1 – 59%, Grade 2 – 33%, and Grade 3 – 8%].7 The median composite Skindex-16 pre-treatment was 0, and post-treatment 34, demonstrating a marked negative impact on QoL.7

RD is categorized as acute, occurring within 1 to 4 weeks of treatment, or chronic, manifesting beyond four weeks to years after treatment.4

Strategies to prevent or mitigate RD are aimed at ensuring an optimal skin condition starting before the treatment. Skincare aims to improve comfort during radiotherapy, reducing inflammation and promoting healing of skin areas affected by RD.8-12 Patients should be educated on a daily skincare regime focusing on: hygiene, moisturization, and sun avoidance and protection measures.8-13

The Canadian Skin Management in Oncology Group (CaSMO) developed an algorithm for the reduction of severity and management of acute RD, which follows previous publications from this group that addressed general oncology-treatment related cutaneous adverse events [AEs], prevention, and skin management.8,13

Scope

The CaSMO project initiated by La-Roche Posay Canada aims to improve cancer patients’ and survivors’ quality of life by offering tools for preventing and managing cancer-treatment-related cutaneous AEs.

The CaSMO panel of clinicians who treat cancer patients with radiation treatment-related cutaneous AEs developed, discussed, and reached a consensus on an evidence-based algorithm supporting prevention, treatment, and maintenance using OTC skin care measures for acute RD. The algorithm focuses on skincare measures to prevent and treat RD using prevention measures and a skincare regime involving hygiene, moisturization, and sun protection measures and products. The algorithm aims to improve patient outcomes and determine the best approach for oncology skin care programs for all stakeholders in the Canadian health care setting. These include oncologists, family practice/internal medicine physicians, dermatologists, oncology nurses, advanced practice providers (APPs), nurse practitioners (NP), physician assistants and pharmacists.

Methods

In the algorithm’s development, the panel used a modified Delphi approach following the AGREE II instrument.14,15 The modified Delphi method is a communication technique for interactive decision-making for medical projects.15 The used process was adapted from face-to-face meetings to a virtual meeting to discuss the outcome of literature searches and reach a consensus on the algorithm based on the selected literature.15,16 The virtual discussion was followed by online follow-up replacing the use of a questionnaire.15 The process entailed preparing the project, selecting the panel, and conducting systematic literature searches followed by summarizing the literature search results, grading the literature, and drafting the algorithm. On January 23, 2021, a virtual panel meeting reviewed the systematic literature review results, discussed and adopted the algorithm using evidence coupled with the expert opinion and experience of the panel. The online process was to fine-tune the algorithm and to prepare and review the publication. (Figure 1)

The CaSMO working group’s consensus on the algorithm was established as a hundred percent agreement was obtained.

Canadian Skin Management in Oncology Group (Casmo) Algorithm for the Prevention and Management of Acute Radiation Dermatitis - image
Figure 1: Process used for the development of the algorithm

Literature Review

A literature review included guidelines, consensus papers, and publications describing current best-practice in radiation-treatment related to cutaneous AEs that benefit from topical treatment using OTC skincare, clinical, and other research studies in the English language from January 2010 to December 2020. Excluded were articles with no original data (unless a review was deemed relevant), articles not dealing with skincare for prevention and treatment of RD, and publication language other than English. A dermatologist and a physician/scientist conducted the searches on January 3 and 4, 2021, on PubMed and Google Scholar as a secondary source of the English-language literature, using the terms:

RD; skincare and topical regimes for prevention, treatment, and maintenance of RD; QoL of patients with RD; adjunctive skincare use; education of staff and patients; communication strategies; adherence; concordance; efficacy; safety; tolerability; skin irritation.

The results of the searches were evaluated independently by two reviewers, resolving discrepancies by discussion. The searches yielded one hundred and twenty-two publications. After excluding duplicates and articles deemed not relevant for the algorithm ([n = 52] other subjects, low quality, a small number, case studies), seventy papers remained. Twenty-five review articles (including one guideline, one algorithm and, eighteen systemic literature reviews), and forty-five were clinical studies (including eighteen randomized controlled trials, eighteen clinical evaluations, and cohort studies, four randomized trials, and five other).

Radiation Dermatitis, Prevention, and Treatment

The goal of radiation therapy is either curative or palliative.3,4 It can be delivered in the neoadjuvant or adjuvant setting, or can be the main treatment, with or without combined systemic treatment.3,4 The multidisciplinary team guiding the patient through the radiation treatment process comprises the radiation oncologist, the radiation oncology nurse, the radiation therapist, medical physicist, and dosimetrist.3,4

Radiation therapy can be external [i.e. 3-D conformal radiation therapy, intensity-modulated radiation treatment, image-guided, Thomotherapy, and stereotactic radiosurgery] or internal, such as brachytherapy, radioisotope therapy, radioimmunotherapy, and radioembolization (Table 1).17 RD is defined as a cutaneous inflammatory reaction induced by exposure to biologically effective levels of ionizing radiation.17 The mechanisms associated with a radiation induced AE include an inflammatory response and oxidative stress, which interact and promote each other.18 Inflammatory markers involved in acute inflammation secondary to ionizing radiation including IL-1, IL-6, TNF-alpha and TGF-Beta, can be expressed within hours after receiving the first radiation treatment.18 Radiation treatment exhibits biological effects within hours to weeks of exposure, through irreversible breaks of the nuclear DNA strands that cause extensive genetic damage and inhibit cells’ ability to divide and replicate.3,18 Further damage comprises structural tissue destruction, generation of reactive oxygen species, a decrease in functional stem cells, initiation of epidermal and dermal inflammation, and skin cell necrosis.3,18 Radiation damage is complex, and a variety of factors influence its severity.

External beam radiation therapy
2D and 3D external radiation therapy
Intensity-modulated radiation therapy
Volumetric-modulated arc therapy
Image-guided radiation therapy
Stereotactic radiosurgery
Stereotactic body radiation therapy
Proton therapy
Total body irradiation

Table 1: External beam radiation therapy

 

RD is generally limited to the irradiated area and depends on several factors, including the target area, dose and fractionation schedule, patient’s condition, and the use of other concomitant cancer treatments (e.g. platinum-based regimens, cetuximab, 5-fluorouracil).3,4-6,18-22

Intrinsic factors that influence the occurrence and severity of RD include demographic or patient-related characteristics.6 These include age, ethnic origin, skin type, smoking, obesity, breast size, hormonal status, presence of infection, and co-existing diseases, such as diabetes or cardiovascular disease.6,17-22

Extrinsic factors include treatment-related characteristics such as technique, dose, volume, fractionation, beam energy, use of bolus, immobilization devices, and the use of combined systemic anti-cancer therapies. Further factors comprise the clinical site of treatment, e.g., areas with skin folds [head and neck, breast and axilla].17-22

Certain drugs (e.g. platinum-based regimens, cetuximab, 5-fluorouracil) increase sensitivity to radiation treatment, causing increased cellular damage and delayed tissue repair.22 Conventional chemotherapy agents and cancer treatment with EGFR inhibitors increase the risk for severe RD (Box 1).22,23

RD is categorized as acute, occurring within 1 to 4 weeks of radiation treatment, or chronic, manifesting beyond four weeks to years after treatment (Table 2).3,4,6,18-23

Location of the treatment field RD may be more severe depending on the location of the treatment field i.e. sites with intertriginous skin folds such as the breast or buttocks
Chemotherapeutic agents Bleomycin, cisplatin, dacarbazine, docetaxel, doxorubicin, etoposide, 5-fluorouracil, gemcitabine, methotrexate, temozolomide.
Any radio-sensitizers or drugs that cause immune suppression
Antibiotics and anti-tuberculosis medication
Comorbidities Anemia, diabetes mellitus, cardiovascular disease, suppression of the immune system, connective tissue diseases, radiosensitive disorders, ataxia telangiectasia, xeroderma pigmentosa
Other factors Advanced age, female sex, obesity, tobacco use, chronic sun exposure, previous breast reconstruction/implants, nutrition and hydration status, mobility status

Box 1: Contributing factors to radiodermatitis (RD)


Canadian Skin Management in Oncology Group (Casmo) Algorithm for the Prevention and Management of Acute Radiation Dermatitis - image

Table 2: Acute and chronic radiation dermatitis20

*Grades of Acute Dermatitis (CTCAE V5– common terminology criteria for adverse events (National Cancer Institute)19


Cutaneous effects of radiotherapy vary considerably in severity, course, and prognosis and can have severe sequelae that impact the quality of life and disrupt cancer treatment.7,8 The two most prevalent grading systems of RD are the RTOG (Radiation Therapy Oncology Group) and CTCAE v5 (Common Terminology Criteria for Adverse Events) systems. The RTOG assessment tool classifies RD from 0 [no visible signs of RD] to 4 [ulceration, bleeding, necrosis].20,21 For the present algorithm, the CTCAE v5 grading system for acute RD from the National Cancer Institute (NCI) is used. This tool has five classes from 1 [faint erythema and dry desquamation] to 5 [death] (Table 3).19,22

CTCAE v5 – DCTD Grading RTOG Grading
Grade Definition Grade Definition
1 Faint erythema or dry desquamation 0 No visible change to skin
2
  • Moderate to brisk erythema;
  • Patchy moist desquamation mostly confined to skin folds and creases;
  • Moderate edema
1
  • Faint or dull erythema.
  • Mild tightness of skin and itching may occur
3
  • Moist desquamation in areas other than skin folds and creases;
  • Bleeding induced by minor trauma or abrasion
2
  • Bright erythema / dry desquamation.
  • Sore, pruritus, and tight skin
4
  • Life-threatening consequences;
  • Skin necrosis or ulceration of full-thickness dermis;
  • Spontaneous bleeding from the involved site;
  • Skin graft indicated
2.5
  • Patchy moist desquamation
  • Yellow/pale green exudate. Soreness with edema
5 Death 3
  • Confluent moist desquamation.
  • Yellow/pale green exudate
  • Soreness with edema
4 Ulceration, bleeding, necrosis (rarely) seen

Table 3: CTCAE and RTOG grading for acute RD

Division of Cancer Treatment & Diagnosis Dermatitis Radiation Grading (DCTD); Radiation Therapy Oncology Group (RTOG)
Grades of Acute Dermatitis (CTCAE – common terminology criteria for adverse events (National Cancer Institute)19
A-B – Grade 1 – faint erythema and dry desquamation
C-D – Grade 2 – moderate erythema and/or edema; patchy desquamation confined to folds
E-F – Grade 3 – moist desquamation in areas other than folds and bleeding from minor trauma
Grade 4 – Life-threatening consequences; full-thickness necrosis, spontaneous bleeding; skin graft indicated


 

Algorithm for the Prevention and Management of Acute Rd

The CaSMO algorithm for acute RD used the mnemonic RECUR (Reliable, Efficient, Clear instructions, Understandable, Remember easily).24

A clinical algorithm’s function is to standardize and support medical decision-making, such as regulating the selection and use of treatment regimens, thereby improving adherence to evidence-based guidelines.14 The algorithms have inputs and outputs, precisely defined specific steps, and uniquely defined results that depend on the preceding steps.24 The current algorithm for the prevention and management of acute RD focused on preventing or reducing and managing RD using OTC skincare and topical treatment (Figure 2). Detailed information on the various grades of RD is given in Figure 3.

The algorithm details measures to be taken before radiation treatment which include education, avoidance of skin irritants, preventative skincare, and sun avoidance and sunscreen use; it also describes prophylactic measures to be taken when radiation starts. Finally, it describes how treatment assessment of skin condition and grading of RD should take place during treatment while the preventative measures continue.

Each section is discussed in the order as they appear in the algorithm.

Canadian Skin Management in Oncology Group (Casmo) Algorithm for the Prevention and Management of Acute Radiation Dermatitis - image
Figure 2: CaSMO radiation dermatitis algorithm

Moist desquamation (MD); Sunscreen (SS); Topical corticosteroids (TCS); Radiation dermatitis (RD); Twice/day (BID) Grade 0: No visible RD; Grade 1: Faint erythema or dry desquamation; Grade 2: Moderate to brisk erythema; Patchy moist desquamation, mostly confined to skin folds and creases; moderate edema; Grade 3: Moist desquamation in areas other than skin folds and creases; Bleeding induced by minor trauma or abrasion; Grade 4: Life-threatening consequences; Skin necrosis or ulceration of full-thickness dermis; Spontaneous bleeding from the involved site; Skin graft indicated.
*Photograph grade 1: RD on the left breast showing erythema during radiation therapy for breast cancer.
*Photograph grade 2 no MD: RD on the neck showing erythema and dry desquamation
*Photograph grade 2 with MD: Vulva-sarcoma RD with erythema and moist desquamation
*Photograph grade 3 with MD: RD with erythema, moist desquamation, and bleeding from minor trauma

Grading Prevention, skincare and treatment
Grade 1: Faint erythema or dry desquamation, possible pain

Canadian Skin Management in Oncology Group (Casmo) Algorithm for the Prevention and Management of Acute Radiation Dermatitis - image
Erythema

Canadian Skin Management in Oncology Group (Casmo) Algorithm for the Prevention and Management of Acute Radiation Dermatitis - image
Dry desquamation
  • Check adherence to treatment.
  • Cleanse the skin and use a moisturizer.
  • Avoid sun exposure and use sunscreen.

For dry desquamation start or continue with low to mid potency TCS to decrease progression and severity of itching, burning and irritation.3

Grade 2: Moderate to brisk erythema;
Patchy moist desquamation mostly confined to skin folds and creases;
Moderate edema, pain

Canadian Skin Management in Oncology Group (Casmo) Algorithm for the Prevention and Management of Acute Radiation Dermatitis - image
Moist desquamation in the groin
  • Check adherence to treatment.
  • Cleanse the skin and use a moisturizer.
  • Avoid sun exposure and use sunscreen.
  • Use saline compresses for cooling.
  • Continue TCS in the surrounding area of moist desquamation
Grade 3: Moist desquamation in areas other than skin folds and creases;
Bleeding induced by minor trauma or abrasion, severe pain

Canadian Skin Management in Oncology Group (Casmo) Algorithm for the Prevention and Management of Acute Radiation Dermatitis - image
Moist desquamation and bleeding
  • Check adherence to treatment.
  • Cleanse the skin and use a moisturizer.
  • Avoid sun exposure and discontinue sunscreen on irradiated area until the reaction is over.
  • Use saline compresses on the areas with moist desquamation.
  • Continue TCS in the surrounding area of moist desquamation
  • Use a wound dressing for bullae and erosions, and select the type of dressing according to the wound bed condition and the exudate production.
  • Consider putting radiation treatment on hold.

Figure 3: RD Treatment according to presentation

Grading according to CTCAE v519


Education on Prevention and Treatment of RD

Education on the type of radiation treatment, on preventive and on treatment measures for RD is essential for both clinicians and patients.13,25 Before starting the treatment, a therapeutic relationship with the patient should be built, supporting the patients’ active participation in their cancer treatment.13 As outlined in two previous publications by the CaSMO group, a detailed discussion between the patient, treating physician, and nurse or other team members includes explaining the radiation treatment protocol, potential side effects, hospital visits, diagnostic tests, management of AEs, and prophylactic and preventative measures.8,13 It is recommended to provide the patients with details on who to contact in the early stages of RD to enable addressing the AEs as early as possible.8,13 It is recommended to support education given to patients verbally by printed or online information (Box 2).8,13

Education on general measures include avoiding skin irritants, products with an elevated pH (>7), scented products and temperature extremes.8,13 Further skin trauma should be avoided such as the use of adhesive bandages and tape, rubbing, scratching and massaging the skin within the treatment area. The patient should be recommended to wear loose fitting cotton clothing which may help avoid traumatic shearing and friction injuries.

Patients should also be recommended to use electric shavers for hair removal. Wax or other depilatory creams and pre-shave liquids and aftershave are discouraged, as they can irritate the irradiated skin.

Patients should be asked not to shave the axilla if it is within the treatment field but may continue to use antiperspirants or deodorants during radiation therapy on dry intact skin.

Question Website/documentation
Cancer Care Ontario: The Prevention and Management of Acute Skin Reactions Related to Radiation Therapy https://www.cancercareontario.ca/en/guidelines-advice/types-of-cancer/846
NCI: Radiation Therapy Side Effects https://www.cancer.gov/aboutcancer/treatment/types/radiation-therapy/side-effects
Questions to ask your doctor about your cancer treatment https://www.cancer.gov/about-cancer/treatment/questions
Questions on how radiation therapy works https://www.cancer.gov/about-cancer/treatment/types/radiation-therapy
Questions about How, why and what to do during radition treatment https://www.cancer.gov/about-cancer/treatment/types/radiation-therapy#RTCCSE
Information on skincare from CDA websites https://dermatology.ca
https://dermatology.ca/recognized-products/skincare/
Information on cancer treatments, cutaneous AEs, and treatments Ruth C. The Dermatology World//December 2019
Support system. www.aad.org/dw/
Glossary and review on cutaneous AEs Sauder M, et al. Skin Ther Letter 2020;(10):1-12
Algorithm on skincare for cutaneous AEs Sauder M, et al. Skin Ther Letter 2021;S(3):1-10

Box 2: Resources and information


Sun Avoidance and Sun Protection

Patients should be educated on radiation treatment-induced photosensitivity which can result in symptoms such as severe sunburn, pruritus, erythema, or edema. It should be explained that phototoxicity may occur, primarily induced by UVA exposure.8,13

Phototoxicity symptoms may appear on the exposed area within a few minutes to several hours after exposure to UV light.3-5,8,13

Patients should be educated on sun avoidance as part of a healthy lifestyle, and the use of sun-protective clothing [e.g., brim hats and sunglasses].8,13 During the course of radiation and while skin is healing the patient should keep the treatment field out of direct sunlight. 3-5,8,13 After the skin has healed, it may be more sun sensitive and therefore requires diligent protection from sunlight and tanning beds should be avoided.8,13

Most dermatologists recommend daily sunscreen of SPF 30 or higher, especially for sun-exposed areas. Special populations that are at higher risk for sun-induced toxicities and neoplasms are advised to avoid sun exposure by using para-aminobenzoic acid (PABA) free UVA and UVB protection as well as sun-protective clothing.8,13 Sunscreen should be reapplied as needed (e.g. after swimming or heavy perspiration) (Box 3).

Patients may continue to swim in chlorinated pools but should rinse afterwards with a gentle cleanser followed by immediately applying a moisturizer. If RD has progressed beyond dry desquamation (CTCAE grade 1) swimming should be avoided.

Phototoxicity and photoallergy Photosensitivity can result in symptoms such as severe sunburn, pruritus, erythema, or edema. The two types of reactions that may occur are phototoxicity and photoallergy. UVA is primarily responsible for triggering these effects and is present throughout the day and year and is readily transmitted through window glass.

Phototoxicity symptoms may appear on the exposed area within a few minutes to several hours after exposure to UV light.

Photoallergic reactions are less common and usually occur due to topically applied medications. They involve the immune system, and symptoms can appear immediately after sun exposure or may take longer to develop. These reactions may look like eczema and can spread to areas of the body not exposed to sunlight.

Sun avoidance and protection Some anticancer drugs may cause photosensitivity reactions making certain precautions necessary:

  1. Avoid unnecessary exposure to UV light including sunlight, tanning beds, and sun lamps.
  2. When outside, cover up with a long-sleeved shirt, long pants or skirt and a broad-brimmed hat made out of tightly woven, dark coloured fabric. If light shines through the fabric, then UV can also go through it.
  3. Use a broad-spectrum sunscreen (protects against both UVA and UVB) that has SPF of at least 30.
Sunscreen application
  • Apply at least 15 to 30 minutes before sun exposure to allow time for the product to diffuse into the skin.
  • Apply generously to all exposed areas, including tops of ears, under chin, and balding areas. Avoid eye area.
  • Reapply at least every 2 hours and after swimming.
  • Discontinue use if erythema or irritation occur.

Box 3: Information on photoprotection and sunscreen application


 

Assessment of Skin Condition

Upon presentation of a patient with RD, it is important to check the location, size of the affected area, erythema, dry desquamation, moist desquamation, exudate, signs of infection, and discomfort (burning, pruritus, pulling, tenderness, dryness, scaling, flaking, peeling).3-5,8,13,19 Similarly, the healthcare provider should check for the presence of fever, pain, mucosal involvement and if there are significant blood laboratory abnormalities. The clinician must establish whether the AE is possibly dangerous or life threatening such as in the case of skin necrosis, ulceration of the full thickness dermis or spontaneous bleeding,13,19 and seek urgent care via a dermatologist, or emergency room for these patients.13,19 The patient must be asked about the duration of the AE and what products were used for cleansing, moisturizing, or dressings for erosions or bullae.3-5,8,13,19

Finally, the impact of RD on the patients’ daily activities, sleeping, eating or drinking must be verified. In the context of the assessment, the date of the last radiation or chemotherapy treatment, as well as concurrent medication use (analgesic, antibiotic, antifungal) and efficacy must be verified.3-5,13,19

Skincare Using Cleansers and Moisturizers

Skincare using OTC products comprises cleansers and moisturizers (Box 4).8,13 Although evidence is lacking on the best choice of product, basic recommendations on supportive measures using OTC skincare are given in various cancer treatment guidelines and consensus papers (Table 4).3,9-13,22,23,25-33

Cleansers8,13
  • Encourage to wash the irradiated skin daily using lukewarm water.
  • Wash cloths may cause friction and are therefore discouraged.
  • Use a soft towel to pat the skin to remove water but not completely dry.
  • Use gentle cleansers such as those with a near-physiological skin pH (4.0 – 6.0).8,13
  • Avoid the use of soap and cleansers with an alkaline pH (> 7), which may excessively remove skin lipids, elevating skin surface pH, and compromise the skin barrier function further.8,13
Moisturizers8,13
  • Apply moisturizers to the face, hands, feet, neck, and back liberally and daily.
  • Moisturizers should be applied at room temperature.
  • Formulations should be safe, effective, free of additives, fragrances, perfumes or sensitizing agents and should have a physiologic skin surface pH (4.0 – 6.0).8,13
  • Skincare product choices depend on the skin condition, availability, costs, and individual preferences.
  • Moisturizer effectiveness depends on the formulation, the vehicle, frequency, and compliance of applications.
  • Choose a moisturizer vehicle based on skin condition, level of xerosis, and patient preference.

Box 4: Criteria and application of cleansers and moisturizers


 

Subject Type Reference
Management of acute RD: a review of the literature and proposal for a treatment algorithm Systematic review and algorithm Rosenthal A, et al. J Am Acad Dermatol. 2019;81(2):558–5673
The role of skincare in oncology patients. Systematic review Sauder MB et al. Skin Ther Letter; 2020 S Oct(10):1-128
Management of acute RD Systematic review and meta-analysis Chan RJ, et al. BMC Cancer 2014;14:539
Prophylaxis and management of acute RD Systematic review Salvo N, et al. Curr Oncol 2010;17:94–11210
Evidence in RD management Critical appraisal of systematic reviews Chan et al. Int J Radiat Oncol Biol Phys 2012;84:e357–6211
Topical interventions to prevent acute RD in head and neck cancer patients Systematic review Ferreira EB, et al. Support Care Cancer. 2017;25 (3):1001–1011. doi: 10.1007/s00520-016-3521-712
Canadian skin management in oncology (CaSMO) algorithm Algorithm Sauder MB et al Skin Ther Letter; 2021 S (3):1-1013
Topical treatment of RD Systematic review Iacovelli NA,et al Drugs Context. 2020 jun 12;9:2020-4-7. 10.7573/dic.2020-4-7. eCollection 2020.22
Prevention and treatment of RD in head and neck cancer patients Systematic review Iacovelli NA, et al. Future Oncol. 2018;14(3):291–30523
Clinical practice guidelines for the prevention and treatment of acute and late RD from the MASCC Skin Toxicity Study Group Guidelines Wong RK, et al. Support Care Cancer. 2013;21(10):2933–2948.25
The use of aloe vera in cancer radiation: an updated comprehensive review. Review Farrugia CE, et al. Complement Ther Clin Pract. 2019;35:126–130.26
Hyaluronic acid for treatment of RD Systematic review Cosentino D, Piro F. Eur Rev Med Pharmacol Sci. 2018;22(21):7562–7572.27
Topical management of acute radiation dermatitis in breast cancer patients: a systematic review. Systematic review and meta-analysis Haruna F, et al. Anticancer Res. 2017;37(10):5343–5353. doi: 10.21873/anticanres.11960.28
Acute RD in head and neck cancer patients treated with radiotherapy and chemotherapy or EGFR inhibitors Review and consensus Russi EG, et al. Crit Rev Oncol Hematol. 2015;96(1):167–182.29
Evidence-based skin care management in radiation therapy: clinical update. Systematic review McQuestion M. Semin Oncol Nurs. 2011;27:e1–e17.30
Patient-rated measures of RD in breast cancer radiotherapy. Systematic review Schnur JB, et al. Am J Clin Oncol. 2011;34(5):529–536.31
RD treatment overview Review Hegedus F, et al. Int J Dermatol. 2017;9:909–914.32
Wound healing after radiation therapy Review Haubner F, Ohmann E, Pohl F, Strutz J, Gassner HG. Radiat Oncol. 2012;7:162.33

Table 4: Systematic literature reviews, consensus papers, guidelines and algorithms


Rosenthal and colleagues recommended a treatment algorithm for RD that included basic hygiene measures using mild soap and lukewarm water.3 A consensus paper and skincare algorithm by the CaSMO group recommended a daily skincare regime focusing on: hygiene, moisturization, sun protection, and, if applicable, camouflage products.8,13 They recommended to encourage patients to keep their skin moisturized using gentle products to prevent pruritus and xerosis.8,13

The skincare formulations used for patients with RD should be safe, effective, free of additives, fragrances, perfumes, sensitizing agents, and should have a near physiologic stratum corneum (SC) pH.8,13 To support adherence to the skincare regime, products should be cosmetically pleasant and easy to use.8,13

Evidence levels of components of topical treatments is shown in Table 5.3,8,13,26-28,34-59 A systematic review3 of topical agents for the treatment of RD reported no benefits from formulations containing aloe vera26, chamomile3,37, ascorbic acid, pantothenic acid, and trolamine.49 However, benefits or potential benefits were shown when using formulations containing hyaluronic acid (HA)27,28, 34,54 epidermal growth factor EGF3,39,51 granulocyte-macrophage colony stimulating factor (GM-CSF), topical corticosteroids (TCS) 40-45 or statins.58

Topical Proposed mechanism of action Clinical efficacy Level of evidence
Gentle cleansers8,13 Cleansing, antimicrobial, hydrating Proven useful, reduced RD symptoms A2
Thermal water containing skincare regimen8 Prevention and treatment of RD, anti-inflammatory, supports a balanced skin microbiome Potentially useful for prevention and treatment B2
Calendula3,38,50 Anti-inflammatory, antibacterial, antifungal, and antioxidant properties; stimulates angiogenesis Potentially useful, reduced symptoms B2
Catechins Anti-inflammatory, antibacterial, and antioxidant properties Potentially useful, reduced symptoms C3
Aloe vera26 Anti-inflammatory; rich in vitamins, enzymes, minerals, sugars, and other active constituents Not proven useful A2
Chamomile3,37 Anti-inflammatory, spasmolytic, antibacterial Not proven useful B2
b-Sitosterol Herbal formulation thought to be antibacterial with analgesic, anti-inflammatory, antiedema, and antithrombotic effects Potentially useful C2
Ascorbic acid Antioxidant, free radical scavenging Not proven useful B2
Pantothenic acid Antioxidant, incorporated into CoA and
increases glutathione levels, promotes
epithelial growth
Not proven useful C2
HA27,28,,34,54 Antioxidant, involved in epidermal moisture retention Potentially useful A2
EGF3,39,51 Stimulates proliferation of human fibroblasts and keratinocytes, promotes epithelialization Potentially useful C4
GM-CSF Promotion of chemotaxis of monocytes into tissues, stimulating macrophage maturation and activation Potentially useful C3
Corticosteroids40-45 Anti-inflammatory; inhibits radiation-induced cytokines Proven useful, reduced grade of RD A1
Statins58 Anti-inflammatory, immunomodulatory, antioxidant, metabolic, and antibacterial activities Potentially useful A2
Silicone-based film or dressing35,36,46-48 Anti-inflammatory, promotes epithelialization Potentially useful A3
Trolamine49 Nonsteroidal anti-inflammatory; promotes macrophages recruitment and stimulates of
granulation tissue
Not proven useful A2
Silver sulfadiazine53 Antimicrobial Potentially useful A2
Silver nylon dressings56 Anti-inflammatory, barrier-enhancing Proven to be useful A2
Barrier cream52 Prophylactic management Not proven useful A3
Oil-in-water emulsion55 Reduction of RD symptoms Potentially useful A3

Table 5: Evidence levels of components of topical treatments


 

Topical agents that have common ingredients known as soothing may be beneficial for the reduction of symptoms such as niacinamide, panthenol, squalene, glycerin, and allantoin.8,13 A skincare kit including all the products needed may be easy to use for the patients supporting adherence to the regimen.8,13 A thermal water containing skincare regime (La Roche-Posay) comprising two types of cleansers, a moisturizer, a healing baume, and an SPF50+ sunscreen was shown to be beneficial for RD prevention and treatment.8 The regimen was evaluated in two-hundred-fifty-three women with mostly stage I (International Union Against Cancer (UICC) /American Joint Committee on Cancer (AJCC)) breast cancer undergoing radiotherapy. The frequent users who daily used the total skincare regime showed significantly (p ≤ 0.0001) less severe RD than those who used parts of the skincare regimen infrequently.8

Contrary to the advice some cancer patients may receive when undergoing radiation treatment, skincare does not interfere with or increase the radiation dose to the skin and can be used in moderation before daily radiation treatments.60 Even if applied shortly before radiation treatment, thin or moderately applied skincare may have minimal influence on skin radiation dose regardless of beam energy or beam incidence.60 Patients are frequently concerned about toxic effects on the skin.60,61 Allowing patients to apply skincare throughout their radiation treatment period will simplify patient instructions and reduce patient confusion and anxiety.60,61 Allowing patients to apply skincare daily and liberally without restrictions on application timing is likely to improve patient quality of life and adherence to the prevention and management of cutaneous AEs using skincare.60-62

Topical Pharmaceuticals

Topical corticosteroids (TCS) have anti-inflammatory effects and may be used for the prevention and treatment of RD.3,4,25,28,40-45,59 A meta-analysis confirmed that TCS, ranging from mild to potent, significantly prevented the incidence of wet desquamation and reduced the mean RD score.28 Various studies confirmed the efficacy of mild TCS decreasing moist desquamation, lowering the incidence of severe RD and delaying time to development of grade three RD.41-44 Extensive and inappropriate use especially of high potency TCS on the face, neck or genitalia can cause AEs including skin atrophy, permanent striae, hematomas and tearing of the skin, telangiectasia, hypertrichosis, local and systemic infections, and rarely adrenal suppression.8,13

Although the efficacy of TCS is established in RD such as in breast cancer28, it is currently under evaluation in head and neck cancer where the use of TCS is frequently associated with the onset of clinically relevant adverse events.22,42,59 In particular, the prolonged use of TCS leads to skin atrophy, which may be particularly contraindicated in this condition.59

Topical use of statins may be beneficial for RD as the product may display some anti-inflammatory, immunomodulatory, antioxidant, metabolic, and antibacterial properties.48

Although topical trolamine (doxepin) is extensively used for the management of RD, its efficacy for RD has not been established.49

Wound Healing Products

Wound healing products and barrier films are widely used, as well, in oncology for cracked skin, erosion, bullae and more severe RD healing by secondary intention. Depending on the wound bed condition and exudate levels, various dressings may be used, such as a foam dressing or a non-adherent wound contact layer, including silicone-coated dressings. The frequency of dressing changes depends on exudate level and are typically twice weekly. The evidence supporting the efficacy of various dressings is scarce. Silicone based agents may have anti-inflammatory properties and are available as a gel or as coated wound dressings.35,36,46-48

Prophylactic topical antibiotics should be avoided to comply with antimicrobial stewardship preventing antibiotic resistance.

The Multidisciplinary Team

Prevention and treatment of RD is a multidisciplinary effort involving radiation oncologists, dermatologists, nurses, and advanced practitioners (APPs). Collaboration and an interprofessional approach between oncology and dermatology is effective in connecting cancer patients with dermatological care from the start of their radiation treatment through to completion.8,13,63-68

Timely intervention by a radiation-oncologist or dermatologist trained in supportive dermatology for oncology patients is critical to preventing avoidable treatment interruptions.8,13,63-68 Moreover, almost equally important is an oncology specialists’ ability to improve quality of life-related to RD and may be able to preserve cancer treatment through managing RD early.8,13

Limitations

Statements used and recommendations given in the algorithm were based on a mix of data and expert opinion. While it is possible that alternatives for RD could exist, the recommendations are suggestions for best practice developed from a panel of expert clinicians that are supported by peer-reviewed literature.

A small panel of physicians developed the algorithm, representing a few centers, and did not include patients in the development. Although limited evidence was available to guide the development, the project will hopefully encourage more skincare studies to prevent, treat and maintain RD.

Conclusions

A multidisciplinary team treating and guiding the cancer patient who receives radiation treatment may improve cancer treatment tolerance. The CaSMO algorithm on acute radiation dermatitis and general preventive measures, including cleansers and moisturizers to prevent or reduce the severity of acute radiation dermatitis, may increase awareness and help improve cancer patient outcomes.

References



  1. Canadian Cancer Society; 2021. Available at: https://www.cancer.ca/en/cancer-information/cancer-101/cancer-statistics-at-a-glance/

  2. Mittmann N, Liu N, Cheng SY, et al. Health system costs for cancer medications and radiation treatment in Ontario for the 4 most common cancers: a retrospective cohort study. CMAJ Open. 2020 Mar 16;8(1):E191-E198.

  3. Rosenthal A, Irailevich R, Mov R. Management of acute radiation dermatitis: A review of the literature and proposal for treatment algorithm. J Am Acad Dermatol. 2019 Aug;81(2):558-567.

  4. Leventhal J, Young MR. Radiation Dermatitis: Recognition, Prevention, and Management. Oncology (Williston Park). 2017 Dec 15;31(12): 894-9.

  5. Biswal SG, Mehta RD. Cutaneous Adverse Reactions of Chemotherapy in Cancer Patients: A Clinicoepidemiological Study. Indian J Dermatol. Jan-Feb 2018;63(1):41-46.

  6. Singh M, Alavi A, Wong R, Akita S. Radiodermatitis: A Review of Our Current Understanding. Am J Clin Dermatol. 2016;17(3):277-92.

  7. Rzepecki A, Birnbaum M, Ohri N, et al. Characterizing the Effects of Radiation Dermatitis on Quality of Life: A Prospective Survey-Based Study. J Am Acad Dermatol. 2019 Mar 13;S0190-9622(19)30425-6.

  8. Sauder MB, Addona M, Andriessen A, et al. The role of skin care in oncology patients. Skin Ther Letter; 2020 Oct; (Suppl):1-12.

  9. Chan RJ, Webster J, Chung B, et al. Prevention and treatment of acute radiation-induced skin reactions: a systematic review and meta-analysis of randomized controlled trials. BMC Cancer. 2014 Jan 31;14:53.

  10. Salvo N, Barnes E, van Draanen J, et al. Prophylaxis and management of acute radiation-induced skin reactions: a systematic review of the literature. Curr Oncol. 2010 Aug;17(4):94–112.

  11. Chan RJ, Larsen E, Chan P. Re-examining the evidence in radiation dermatitis management literature: an overview and a critical appraisal of systematic reviews. Int J Radiat Oncol Biol Phys. 2012 Nov 1;84(3):e357-62.

  12. Ferreira EB, Vasques CI, Gadia R, et al. Topical interventions to prevent acute radiation dermatitis in head and neck cancer patients: a systematic review. Support Care Cancer. 2017 Mar;25(3):1001-1011.

  13. Sauder MB, Andriessen A, Claveau J, et al. Canadian skin management in oncology (CaSMO) algorithm for patients with oncology treatment-related skin toxicities. Skin Ther Letter. 2021 March; (Suppl):1-10.

  14. Brouwers M, Kho ME, Browman GP, et al. AGREE II: advancing guideline development, reporting and evaluation in health care. CMAJ. 2010 Dec 14;182(18):E839-42.

  15. Trevelyan EG, Robinson N. Delphi methodology in health research: how to do it? Eur J Integrative Med. 2015;7(4):423-428.

  16. Smith Begolka W, Elston DM, Beutner KR. American Academy of Dermatology evidence-based guideline development process: responding to new challenges and establishing transparency. J Am Acad Dermatol. 2011 Jun;64(6):e105-12.

  17. Canadian Cancer Society; 2021. Available at: https://www.cancer.ca/en/cancer-information/diagnosis-and-treatment/radiation-therapy/external-radiation-therapy/?region=on

  18. Wei J, Meng L, Jiang X et al. Radiation induced skin reactions: mechanism and treatment. Cancer Manag Res. 2018 Dec 21;11:167-177.

  19. United States Department of Health and Human Services, National Institutes of Health, National Cancer Institute(NCI) Common Terminology Criteria for Adverse Events (CTCAE) Bethesda, MD: nci; 2010. Ver. 5. evs.nci.nih.gov/ftp1/CTCAE/About.html;

  20. Trueman E. Managing radiotherapy induced skin reactions in the community. J Community Nurs. 2013;27(4):16-24.

  21. Bolderston A, Cashell A, McQuestion M, et al. A Canadian survey of the management of radiation-induced skin reactions. J Med Imaging Radiat Sci. 2018 Jun;49(2):164-172.

  22. Iacovelli NA, Torrente Y, Sacerdote P et al. Topical treatment of radiation-induced dermatitis: current issues and potential solutions. Drugs Context. 2020 Jun 12;9:2020-4-7.

  23. Iacovelli NA, Galaverni M, Cavallo A, et al. Prevention and treatment of radiation-induced acute dermatitis in head and neck cancer patients: a systematic review. Future Oncol. 2018 Feb;14(3):291-305.

  24. Guenther L. Presentation at Derm. Update, Fall 2018, Montreal, Canada http://www.slideshare.net/jrcampos/03-algorithm-properties

  25. Wong RK, Bensadoun RJ, Boers-Doets CB, et al. Clinical practice guidelines for the prevention and treatment of acute and late radiation reactions from the MASCC Skin Toxicity Study Group. Support Care Cancer. 2013 Oct;21(10):2933-48.

  26. Farrugia CE, Burke ES, Haley ME, et al. The use of aloe vera in cancer radiation: An updated comprehensive review. Complement Ther Clin Pract. 2019 May;35:126-130.

  27. Cosentino D, Piro F. Hyaluronic acid for treatment of the radiation therapy side effects: a systematic review. Eur Rev Med Pharmacol Sci. 2018 Nov;22(21):7562-7572.

  28. Haruna F, Lipsett A, Marignol L. Topical Management of Acute Radiation Dermatitis in Breast Cancer Patients: A Systematic Review and Meta-Analysis. Anticancer Res. 2017 Oct;37(10):5343-5353.

  29. Russi EG, Moretto F, Rampino M, et al. Acute skin toxicity management in head and neck cancer patients treated with radiotherapy and chemotherapy or EGFR inhibitors: Literature review and consensus. Crit Rev Oncol Hematol. 2015 Oct;96(1):167-82.

  30. McQuestion M. Evidence-based skin care management in radiation therapy: clinical update. Semin Oncol Nurs. 2011 May;27(2):e1-17.

  31. Schnur JB, Love B, Scheckner BL, et al. A systematic review of patient-rated measures of radiodermatitis in breast cancer radiotherapy. Am J Clin Oncol. 2011 Oct;34(5):529-36.

  32. Hegedus F, Mathew LM, Schwartz RA. Radiation dermatitis: an overview. Int J Dermatol. 2017 Sep;56(9):909-914.

  33. Haubner F, Ohmann E, Pohl F, et al. Wound healing after radiation therapy: review of the literature. Radiat Oncol. 2012 Sep 24;7:162.

  34. Presta G, Puliatti A, Bonetti L, et al. Effectiveness of hyaluronic acid gel (Jalosome soothing gel) for the treatment of radiodermatitis in a patient receiving head and neck radiotherapy associated with cetuximab: a case report and review. Int Wound J. 2019 Dec;16(6):1433-1439.

  35. Chan RJ, Blades R, Jones L, et al. A single-blind, randomised controlled trial of StrataXRT® – a silicone-based film-forming gel dressing for prophylaxis and management of radiation dermatitis in patients with head and neck cancer. Radiother Oncol. 2019 Oct;139:72-78.

  36. Wooding H, Yan J, Yuan L, et al. The effect of Mepitel Film on acute radiation-induced skin reactions in head and neck cancer patients: a feasibility study. Br J Radiol. 2018;91:20170298.

  37. Ferreira EB, Ciol MA, Vasques CI, et al. Gel of chamomile vs. urea cream to prevent acute radiation dermatitis in patients with head and neck cancer: a randomized controlled trial. J Adv Nurs. 2016 Aug;72(8):1926-34.

  38. Schneider F, Danski MT, Vayego SA. Usage of Calendula officinalis in the prevention and treatment of radiodermatitis: a randomized double-blind controlled clinical trial. Rev Esc Enferm USP. 2015 Apr;49(2):221-8.

  39. Kang HC, Ahn SD, Choi DH, et al. The safety and efficacy of EGF-based cream for the prevention of radiotherapy-induced skin injury: results from a multicenter observational study. Radiat Oncol J. 2014 Sep;32(3):156-62.

  40. Hindley A, Zain Z, Wood L, et al. Mometasone furoate cream reduces acute radiation dermatitis in patients receiving breast radiation therapy: results of a randomized trial. Int J Radiat Oncol Biol Phys. 2014 Nov 15;90(4):748-55.

  41. Ho AY, Olm-Shipman M, Zhang Z, et al. A randomized trial of mometasone furoate 0.1% to reduce high-grade acute radiation dermatitis in breast cancer patients receiving postmastectomy radiation. Int J Radiat Oncol Biol Phys. 2018 Jun 1;101(2):325-333.

  42. Zenda S, Yamaguchi T, Yokota T, et al. Topical steroid versus placebo for the prevention of radiation dermatitis in head and neck cancer patients receiving chemoradiotherapy: the study protocol of J-SUPPORT 1602 (TOPICS study), a randomized double-blinded phase 3 trial. BMC Cancer. 2018 Sep 6;18(1):873.

  43. Miller RC, Schwartz DJ, Sloan JA, et al. Mometasone furoate effect on acute skin toxicity in breast cancer patients receiving radiotherapy: a phase III double-blind, randomized trial from the North Central Cancer Treatment Group N06C4. Int J Radiat Oncol Biol Phys. 2011 Apr 1;79(5):1460-6.

  44. Ulff E, Maroti M, Serup J, Falkmer U. A potent steroid cream is superior to emollients in reducing acute radiation dermatitis in breast cancer patients treated with adjuvant radiotherapy. A randomised study of betamethasone versus two moisturizing creams. Radiother Oncol. 2013 Aug;108(2):287-92.

  45. Neben-Wittich MA, Atherton PJ, Schwartz DJ, et al. Comparison of provider-assessed and patient-reported outcome measures of acute skin toxicity during a Phase III trial of mometasone cream versus placebo during breast radiotherapy: the North Central Cancer Treatment Group (N06C4). Int J Radiat Oncol Biol Phys. 2011 Oct 1;81(2):397-402.

  46. Herst PM, Bennett NC, Sutherland AE, et al. Prophylactic use of Mepitel Film prevents radiation-induced moist desquamation in an intra-patient randomised controlled clinical trial of 78 breast cancer patients. Radiother Oncol. 2014 Jan;110(1):137-43.

  47. Zhong WH, Tang QF, Hu LY, et al. Mepilex Lite dressings for managing acute radiation dermatitis in nasopharyngeal carcinoma patients: a systematic controlled clinical trial. Med Oncol. 2013 Dec;30(4):761.

  48. Diggelmann KV, Zytkovicz AE, Tuaine JM, et al. Mepilex Lite dressings for the management of radiation-induced erythema: a systematic inpatient controlled clinical trial. Br J Radiol. 2010 Nov;83(995):971-8.

  49. Abbas H, Bensadoun RJ. Trolamine emulsion for the prevention of radiation dermatitis in patients with squamous cell carcinoma of the head and neck. Support Care Cancer. 2012 Jan;20(1):185-90.

  50. Sharp L, Finnilä K, Johansson H, et al. No differences between Calendula cream and aqueous cream in the prevention of acute radiation skin reactions–results from a randomised blinded trial. Eur J Oncol Nurs. 2013 Aug;17(4):429-35.

  51. Kong M, Hong SE. Topical use of recombinant human epidermal growth factor (EGF)-based cream to prevent radiation dermatitis in breast cancer patients: a single-blind randomized preliminary study. Asian Pac J Cancer Prev. 2013;14(8):4859–4864.

  52. Graham PH, Plant N, Graham JL, et al. A paired, double-blind, randomized comparison of a moisturizing durable barrier cream to 10% glycerine cream in the prophylactic management of postmastectomy irradiation skin care: trans Tasman Radiation Oncology Group (TROG) 04.01. Int J Radiat Oncol Biol Phys. 2013 May 1;86(1):45-50.

  53. Hemati S, Asnaashari O, Sarvizadeh M, et al. Topical silver sulfadiazine for the prevention of acute dermatitis during irradiation for breast cancer. Support Care Cancer. 2012 Aug;20(8):1613-8.

  54. Pinnix C, Perkins GH, Strom EA, et al. Topical hyaluronic acid vs. standard of care for the prevention of radiation dermatitis after adjuvant radiotherapy for breast cancer: single-blind randomized phase III clinical trial. Int J Radiat Oncol Biol Phys. 2012 Jul 15;83(4):1089-94.

  55. Jensen JM, Gau T, Schultze J, et al. Treatment of acute radiodermatitis with an oil-in-water emulsion following radiation therapy for breast cancer: a controlled, randomized trial. Strahlenther Onkol. 2011 Jun;187(6):378-84.

  56. Aquino-Parsons C, Lomas S, Smith K, et al. Phase III study of silver leaf nylon dressing vs standard care for reduction of inframammary moist desquamation in patients undergoing adjuvant whole breast radiation therapy. J Med Imaging Radiat Sci. 2010 Dec;41(4):215-221.

  57. Lewis L, Carson S, Bydder S, et al. Evaluating the effects of aluminum-containing and non-aluminum containing deodorants on axillary skin toxicity during radiation therapy for breast cancer: a 3-armed randomized controlled trial. Int J Radiat Oncol Biol Phys. 2014 Nov 15;90(4):765-71.

  58. Ghasemi A, Ghashghai Z, Akbari J, et al. Topical atorvastatin 1% for prevention of skin toxicity in patients receiving radiation therapy for breast cancer: a randomized, double-blind, placebo-controlled trial. Eur J Clin Pharmacol. 2019 Feb;75(2):171-178.

  59. Liao Y, Feng G, Dai T, et al. Randomized, self-controlled, prospective assessment of the efficacy of mometasone furoate local application in reducing acute radiation dermatitis in patients with head and neck squamous cell carcinomas. Medicine (Baltimore). 2019 Dec;98(52):e18230.

  60. Baumann BC, Verginadis II, Zeng C, et al. Assessing the validity of clinician advice that patients avoid the use of topical agents before daily radiotherapy treatments. JAMA Oncol. 2018 Dec 1;4(12):1742-1748. doi: 10.1001/jamaoncol.2018.4292.

  61. Salzmann M, Marmé F, Hassel JC. Prophylaxis and management of skin toxicities. Breast Care (Basel). 2019 Apr;14(2):72-77.

  62. Baumann BC, Zeng C, Freedman GM, et al. Avoiding antiperspirants during breast radiation therapy: myth or sound advice? Radiother Oncol. 2017 Aug;124(2):204-207.

  63. Aizman L, Nelson K, Sparks AD, Friedman AJ. The influence of supportive oncodermatology interventions on patient quality of life: a cross-sectional survey. J Drugs Dermatol. 2020 May 1;19(5):477-482.

  64. Chen ST, Molina GE, Lo JA, et al. Dermatology consultation reduces interruption of oncologic management among hospitalized patients with immune-related adverse events. J Am Acad Dermatol. Apr;82(4):994-996.

  65. Barrios DM, Phillips GS, Feites-Martinez A, et al. Outpatient dermatology consultations for oncology patients with acute dermatologic adverse events impact anticancer therapy interruption: a retrospective study. J Eur Acad Dermatol Venereol. 2020 Jun;34(6):1340-1347.

  66. Barrios DCK, Phillips G, Lucas AS, et al. Anticancer therapy interruption and diagnostic concordance between referring clinicians and dermatologists at MSKCC. J Am Acad Dermatol. 2017;76(6): AB45

  67. Lucas AS, Ciccolini K. The role of oncodermatology in care of patients receiving cancer therapy. Semin Oncol Nurs. 2017 Nov;33(4):393-401.

  68. Kwong BY. Outcomes of Embedding Dermatologic Care Within Oncology Practices for Patients With Cancer. JAMA Dermatol. 2020 Oct 1;156(10):1051-1052.

  69. Zurfley F, Mostow EN. Association between the use of a physician extender and dermatology appointment wait times in Ohio. JAMA Dermatol. 2017 Dec 1;153(12):1323-1324.

  70. Crews J. 2019 (4). https://www.healio.com/news/hematology-oncology/20190416/telemedicine-in-oncology-virtual-solution-to-a-very-real-problem


]]>