Advances in Psoriasis A Multisystemic Guide Jeffrey M. Weinberg Mark Lebwohl Editors Second Edition 123 Contents 1Introduction to and History of Psoriasis and Psoriasis Therapy���������������������������������������������������������������������������� 1 John B. Cameron and Abby S. Van Voorhees 2Pathophysiology of Psoriasis/Novel Pathways������������������������������ 9 Jeremy M. Hugh and Jeffrey M. Weinberg 3Psoriasis: Clinical Review and Update������������������������������������������ 19 Ivan Grozdev and Neil J. Korman 4Psoriasis: Epidemiology, Potential Triggers, Disease Course���������������������������������������������������������������������������������� 27 Ivan Grozdev and Neil J. Korman 5Topical Therapy I: Corticosteroids and Vitamin D Analogues ���������������������������������������������������������������������� 39 Eric J. Yang and Shari R. Lipner 6Topical Therapy II: Retinoids, Immunomodulators, and Others���������������������������������������������������������������������������������������� 51 Lyn C. Guenther 7Topical Therapy II: Retinoids, Immunomuodulators, and Others/Ultraviolet Therapy for Psoriasis ������������������������������ 71 Kristen M. Beck and John Koo 8Laser Therapy in Psoriasis�������������������������������������������������������������� 93 Quinn Thibodeaux and John Koo 9Traditional Systemic Therapy I: Methotrexate and Cyclosporine������������������������������������������������������������������������������ 103 Erin Boh, Andrew Joselow, and Brittany Stumpf 10Traditional Systemic Therapy II: Retinoids and Others ���������������������������������������������������������������������� 119 Vignesh Ramachandran, Ted Rosen, Misha Koshelev, and Fareesa Shuja Sandoval 11Apremilast���������������������������������������������������������������������������������������� 141 Jerry Bagel and Elise Nelson 12Etanercept���������������������������������������������������������������������������������������� 145 Andrew F. Alexis and Charlotte M. Clark v vi 13Adalimumab for Psoriasis �������������������������������������������������������������� 153 Cooper B. Tye and Jennifer C. Cather 14Infliximab, Golimumab, and Certolizumab Pegol������������������������ 173 Jacob A. Mojeski and Robert E. Kalb 15Ustekinumab������������������������������������������������������������������������������������ 201 George Han, Caitriona Ryan, and Craig L. Leonardi 16Guselkumab�������������������������������������������������������������������������������������� 213 Deep Joshipura, Brooke Rothstein, and David Rosmarin 17Tidrakizumab ���������������������������������������������������������������������������������� 225 George Han 18Risankizumab���������������������������������������������������������������������������������� 235 Erica B. Lee, Deeti J. Pithadia, Kelly A. Reynolds, and Jashin J. Wu 19Secukinumab for the Treatment of Inflammatory Skin and Joint Disease �������������������������������������������������������������������� 243 Jason E. Hawkes and Avishan Vishi Hawkes 20Ixekizumab �������������������������������������������������������������������������������������� 253 Caitriona Ryan and Roisin O’Connor 21Brodalumab�������������������������������������������������������������������������������������� 263 Annika S. Silfvast-Kaiser, Dario Kivelevitch, So Yeon Paek, and Alan Menter 22Biosimilars for Psoriasis������������������������������������������������������������������ 279 Sarah Lonowski, Nirali Patel, Nika Cyrus, and Paul S. Yamauchi 23Research Pipeline I: Oral Therapeutics for Psoriasis������������������ 291 D. Grand, K. Navrazhina, J. W. Frew, and J. E. Hawkes 24Research Pipeline II: Upcoming Biologic Therapies�������������������� 303 Ahuva D. Cices and Jeffrey M. Weinberg 25Pediatric Psoriasis���������������������������������������������������������������������������� 311 Starling Tolliver, Amber N. Pepper, Salma Pothiawala, and Nanette B. Silverberg 26Challenges in Psoriasis Treatment: Nail, Scalp, and Palmoplantar Involvement������������������������������������������������������ 343 Jeffrey J. Crowley 27Psoriasis and Comorbidities����������������������������������������������������������� 363 Philip M. Laws and Richard B. Warren 28Summary of Published Treatment Guidelines������������������������������ 399 Vignesh Ramachandran, Abigail Cline, and Steven R. Feldman Index���������������������������������������������������������������������������������������������������������� 415 Contents 1 Introduction to and History of Psoriasis and Psoriasis Therapy John B. Cameron and Abby S. Van Voorhees Abstract The history of psoriasis has had a long and complicated path. With initial challenges in recognizing this disease, psoriasis was misunderstood in ancient times as documented in the Bible as well as in other ancient texts. At first mistakenly thought to be leprosy or other infectious processes such as impetigo, those with psoriasis were ostracized from their communities. It was only in the mid 1800s that Camille Gibert clarified psoriasis as a papulosquamous disease. Remarkable progress in our understanding has occurred over the millennium, some a result of scientific investigation while other has come as a consequence of serendipity. Even in the just the past decade we continue to refine our understanding of the immune dysregulation that causes this condition. Given the poor characterization of this disease, those with psoriasis also suffered from a lack of effective treatments. We review the history of the treatments that have been employed. Starting with Fowler’s solution, sometimes the treatments had significant tox- J. B. Cameron Department of History, Old Dominion University, Norfolk, VA, USA A. S. Van Voorhees (*) Department of Dermatology, Eastern Virginia Medical School, Norfolk, VA, USA e-mail: vanvooas@evms.edu © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_1 icity. The use of tar and anthralin, while still in use today by some, were some of the first modalities to provide care. Early observations by Goeckerman and Ingram demonstrated the benefit of combining these topical treatments with ultraviolet B light to allow for enhanced efficacy. In the 1970s both methotrexate and PUVA photochemotherapy were developed, allowing for the outpatient care of psoriasis. Systemic and topical retinoids followed, as did narrowband UVB. It was however, the serendipitous discovery of the benefit of cyclosporine that truly allowed us to understand the importance of the immune system in this disease. Drugs first targeting T cells were then followed by those targeting TNF, and now more recently those targeting IL17 and IL23. With each new achievement in the understanding of psoriasis, the ability to more finely target the abnormalities of the immune system has improved, and with that, the efficacy of our newest agents has greatly expanded. Key Learning Objectives 1. To understand the evolution of the understanding of the pathogenesis of psoriasis 2. To understand the evolution of the treatment paradigm for psoriasis 3. To describe the evolving understanding of the diagnosis of psoriasis over time 1 J. B. Cameron and A. S. Van Voorhees 2 History of Psoriasis BL O SP ch R ild G IN od ho OD water WI old N a PH L EG Sanguine Phlegmatic choleric Melancholic N y R bille) LEellow e bil LY HO ) (yello CH wo O r SU M You M th ER COLD AU T ma UM tur k Fire ty HOT i MELA (bla NC c The Bible Book of Leviticus, chapters 13 and 14. Arikha, N (2007) Passions and Tempers. Harper Perennial, New York. Also Crissey, JT, Parish, LC, Stelley, WB (1981) The Dermatology and Syphilology of the Nineteenth Century. Praeger Scientific, New York, pp. 4–5. air R TE e g An understanding of disease overall and, skin disease in particular, has been a unique part of modern times. Previously those with psoriasis were often mislabeled, poorly regarded, and suffered as a consequence. Symptoms were often considered the disease itself, and consequent progress in treatment was limited by this lack of understanding of the disease and its cause. The history of psoriasis therefore requires an appreciation of how the understanding of disease has progressed over time and the often serendipitous findings of treatment options. Pre-scientific societies often viewed disease as resulting from a violation of the sacred order, the malignant influence of magic or the breaking of a taboo. For example the entire thirteenth chapter of the Book of Leviticus concerns how the priests may determine if an outbreak on the skin is leprosy and the fourteenth chapter concerns which animals (lambs and birds) shall be sacrificed to purify the victim.1 The first cultures that developed notions of rational science were China and Greece. Western medicine finds its roots in the Greek belief that disease results from natural causes, that in some way the balance or integrity of the body has been disrupted. Treatment, therefore, consisted in restoring that balance or integrity. Hippocrates may be the father of western medicine but the most influential founder was more likely Galen of Pergamon (130– 200 CE) To Galen the human body was a very complex organism made up not just of the four humors but also of gradations of dry and moist and hot and cool.2 Treatment of disease was not limited to bleedings and purges but also included the use of lotions designed to restore health. Galen’s system was so completely accepted that only in the nineteenth century would the humors and miasma disappear from medical belief to be replaced by the germ theory of disease. MOIST earth DRY DIAGRAM OF HUMOURS, ELEMENTS, QUALITIES, AND SEASONS I dentification of Psoriasis as a Unique Disease It is extremely difficult to tease out the history of psoriasis and its treatment in the ancient world because of confusion between psoriasis and many other diseases. Furthermore in the past the names assigned to diseases and symptoms were arbitrary and inconsistent. Identification of psoriasis in Egypt is especially difficult because of confusion between the disease and leprosy in later times.3 However, since extensive examination of Egyptians mummies would indicate that leprosy was not present in Egypt before the common era, it is possible that psoriasis was mislabeled as leprosy. We face similar problems of terminology in Greek medicine. The Corpus Hippocraticum contains precise descriptions and treatments for many recognizable diseases of the skin.4 Again, it is likely that much of what was referred to as leprosy was in fact psoriasis. The appearance of true leprosy early in the common era compounded the confusion and sometimes lead to harsh treat- 1 2 Pusey WA (1933) History of Dermatology. Charles C. Thomas, Baltimore, pp 11–17. 4 Pusey WA (1933) History of Dermatology. Charles C. Thomas, Baltimore, pp 19–25. 3 1 Introduction to and History of Psoriasis and Psoriasis Therapy ments for those with psoriasis since lepers were often isolated and forbidden to associate with non-leprosy population. The first indisputable reference to psoriasis comes from the fifth and sixth books of Aulus Cornelius Celsus, De Re Medica (circa 25 BCE-­ circa 50 CE) a Roman who compiled an extensive list of diseases and treatment for use by estate owners.5 Celsus did not use the term psoriasis but rather describes it under the heading impetigo. After the collapse of Roman culture, the practice of scientific medicine in the west disappeared and only returned as a part of the Renaissance. Geronimo Mercurialis penned a summary of what was known of skin diseases in 1572.6 Mercuralis lumped psoriasis in with other diseases as Lepra. He mentions several treatments including wolf dung rubbed in with vinegar, blood of a mountain goat as well as the rubbing of psoriasis with cantharides. Robert Willan (1757–1812) set out clear and uniform nomenclature of skin diseases in 1809. However, his terminology unfortunately perpetuated some confusion in that he called psoriasis lepra vulgaris.7 That confusion would end by mid-century when Camille Melchoir Gibert (1792–1866) dropped lepra vulgaris and used only psoriasis as the sole term for the disease and his work made clear important distinctions among papulosquamous diseases.8 Gibert’s successors improved the distinctions. Hebra fully distinguished the clinical practice of leprosy from psoriasis; Heinrich Auspitz (1835–1886) noted bleeding points after removing scales (Auspitz sign); Heinrich Köebner made an important contribution in 1872 when Pusey WA (1933) History of Dermatology. Charles C. Thomas, Baltimore, p. 28. 6 Sixteenth Century Physician and his Methods: Mercurialis on Diseases of the Skin. Translated from De Morbis Cutaneis et Omnibus Corporis Humani Excrementis Tractatus, by Sutton RL Jr (1986) The Lowel Press,Kansas City, Missouri. 7 Pusey WA (1933) History of Dermatology. Charles C. Thomas, Baltimore, pp62ff. 8 Pusey WA (1933) History of Dermatology. Charles C. Thomas, Baltimore, pp 81–82. 5 3 he delivered an address entitled, “The Etiology of Psoriasis” pointing out the tendency of prior trauma to produce psoriasis lesions. The “Koebner Phenomenon” is still viewed as an important indication of psoriasis.9 In 1898 Munro described the micro abscesses of psoriasis now called Munro’s abscesses. With the addition in the early twentieth century of Leo van Zumbusch of generalized pustular psoriasis and Waranoff’s description of the pale halo now called “woranoff ring,” accurate diagnosis of psoriasis became commonplace. History of the Treatment of Psoriasis The history of the treatment of psoriasis has been largely driven by serendipitous findings. The late 1700s and 1800s included treatments such as arsenic, chrysarobin and ammoniated mercury. Anthralin and tar came into widespread use in the first half of the twentieth century. Starting in the 1950s topical steroids were developed followed by the arrival of methotrexate, retinoids, and immunosuppressive medications in the 1970s, 1980s and 1990s respectively. An enhanced understanding of the pathogenesis of psoriasis has allowed for more targeted drug development in the twenty-first century. Our therapeutic armamentarium now includes medications known as the biologics which target various aspects of the immune system allowing for its regulation. rsenic, Ammoniated Mercury A and Chrysarobin During the eighteenth and nineteenth centuries three topical agents are known to have been used in the treatment of psoriasis. While probably first developed by the ancient Greeks, arsenic solution was first utilized in dermatology in 1786.10 Crissey JT, Parish LC, Shelley WB (1981) The dermatology and syphilology of the nineteenth century. Praiger Publishers, New York, pp 367–369. 10 Farber M, “History of the treatment of psoriasis,” J 9 J. B. Cameron and A. S. Van Voorhees 4 The first report of its use in psoriasis though was attributed to Girdlestone in 1806. He is credited with noting the efficacy of Fowler’s solution for improving the lesions of psoriasis.11 Ammoniated mercury, another topical agent was also used at this same time.12 The use of this mercury in the topical treatment of psoriasis has been attributed to Dr. Fox in 1880. It was championed as well by Duhring. The use of both of these topical agents continued until the 1950s and 1960s when concerns about their possible toxicity risks and accidental poisonings caused their prohibition. The third topical agent that was identified was chrysarobin. A serendipitous finding, it was noted to be of benefit in the treatment of psoriasis by Balmonno Squire in 1876. His patient, who was using Goa powder to treat a presumed fungal infection, was noted to have improvement of his psoriasis. Kaposi in 1878 also published his experience with this topical approach to psoriasis. acid, zinc oxide and ultraviolet light. For many decades this combination was the mainstay of psoriasis treatment in Europe. Coal tar was also pioneered in the early 1900s. In 1925 Goeckerman noted the beneficial effect of the combination of coal tar with ultraviolet light B radiation in the treatment of psoriasis.15 While the beneficial effect of sunlight on psoriasis had been long known, Goeckerman realized that this effect might be enhanced if combined with a topical photosensitizer. The success of this approach was demonstrated by its widespread use for many decades. While both the Goeckerman protocol and the Ingram protocol were often effective, the main limitation of these approaches was that they were very time-intensive, requiring patients to remain hospitalized for weeks each year to control their disease. Anthralin and Tar The development of steroids both for systemic and topical use revolutionized the treatments of many diseases including psoriasis. First discovered in 1950, it was only two years later that the potential role of this agent in a topical form was demonstrated in psoriasis.16 Known as compound F, hydrocortisone was beneficial in treating psoriasis. From this time to the present, topical steroids have continued to play a significant role in reducing the inflammation in various cutaneous conditions. Topical steroids continue to be the most frequently prescribed medication in the treatment of psoriasis today. After the first identification of the potential benefit of chrysarobin, the 1900s were a time of its further exploration. During the early part of this century scientists learned how to convert chrysarobin to anthralin. The active agent was identified as 2-methyl dithranol. During World War 1 when natural supplies were interrupted the process of synthesizing anthralin was discovered. It was then in 1916 that Unna came to understand the potential of this compound in the treatment of psoriasis.13 In 1953, Ingram further refined the treatment of psoriasis with anthralin.14 He demonstrated that treatment could be enhanced by utilizing the combination of anthralin, salicylic Amer Acad Dermaol 1992; 27: 640–5. 11 Bechet PE, “History of the use of arsenic in dermatology,” Arch Dermatol 1931; 23: 110–7. 12 Farber EM, “History of the treatment of psoriasis,” J Amer Acad Dermatol 1992; 27: 640–5. 13 Unna PG” Cignolin als Heilmittel der psoriasis,” Dermatol Wochenschr 1916; 62: 116–86. Fry L,” Psoriasis,” 1988; 119: 445–561. 14 Ingram JT, “The Approach to Psoriasis,” Br Med J 1953; 2: 591–594. Corticosteroids Methotrexate and PUVA Methotrexate was developed in the 1950s for the treatment of malignancies. As seen with treatments before, it was only a short while before its Goeckerman WH, “Treatment of Psoriasis,” Northwest Med 1925; 24: 229. 16 Sulzberger MB, Witten VH, “The Effect of topically applied coumpound F in select dermatoses,” J Invest Dermatol 1952; 19: 101. 15 1 Introduction to and History of Psoriasis and Psoriasis Therapy potential in the treatment of psoriasis was identified. In 1946 Farber developed aminopterin for the treatment of leukemia. Five years later Gubner noted its role in the treatment of psoriasis.17 While using this agent in the treatment of Rheumatoid arthritis, he noted that his patient, who concurrently had psoriasis, also had improvement of his skin. A more stable derivative of aminopterin with less toxicity, methotrexate, was introduced in 1958 for the treatment of psoriasis.18 This agent was subsequently approved by the FDA for the treatment of psoriasis in 1972 after the first guidelines for its use were published.19 It continues to be an important agent in the treatment of psoriasis today. The efficacy of PUVA in the treatment of psoriasis was also demonstrated during the 1970s. While the combination of ultraviolet light and a photosensitizing compound had been used in the treatment of vilitigo for hundreds of years in Egypt and in India,20 the demonstration of its role in psoriasis was novel. Ancient healers had those with vitiligo ingest psoralen-rich foods such as figs and limes and then exposed their skin to natural sunlight. It took however until 197421 to demonstrate the effectiveness of the combination of psoralen and artificial UVA light exposure (320–400 nm) in psoriasis. Known as PUVA, this therapeutic approach was a highly efficacious approach for many patients with chronic psoriasis. PUVA was widely utilized since it allowed patients to achieve control of their disease without incurring long inpatient hospitalizations. 17 Guber R, August S, Ginsbergerg V,” Therapeutic suppression of tissue reactivity; effect of aminopterin in Rheumatoid arthritis and psoriasis,” Am J Med Sci 1951; 221: 176–182. 18 Edmondson W, Guy WB, “Treatment of psoriasis with folic acid antagonist,” Arch Dermatol 1958; 78(2) 200–203. 19 Roenigk HH Jr, Maibach HI, Weinstein G, “Guidelines in methotrexate therapy for psoriasis,” Arch Dermatol 1972; 105: 363–365. 20 Gupta AK, Anderson TF, “Psoralen photochemotherapy,” 1987; 17: 703–34. 21 Parrish Jam Fitzpatrick TB, Tanenbaum L, Pathak M, “Photochemotherapy of psoriasis with oral methoxsalen and longwave ultraviolet light,” New Engl J Med 1974; 291: 1207–1211. 5 However, in the late 1970s the first reports surfaced of cutaneous malignancies.22 Longitudinal studies of the original cohort of patients in subsequent years confirmed this finding.23 Stern noted the increase risk of SCC, BCC and potentially melanoma skin cancer in those treated with high-­ dose exposures and long-term therapy. Concerns about the potential risk of cutaneous malignancy has limited the utilization of this modality over the past decades. arrowband UVB, Retinoids, N Vitamin D As the potential risk of non-melanoma skin cancers associated with long-term use of PUVA became increasingly apparent, discoveries continued and additional new approaches to treat psoriasis came into prominence in the 1980s and the 1990s. Parrish and Jaenicke identified what has come to be known as narrowband UVB.24 They identified that the most therapeutically effective wavelengths of UVB were those between 300–313 nm, while the remaining wavelengths of UVB light contributed primarily to the development of erythema. Subsequently, 311 nm was identified as the most efficacious wavelength for the clearance of psoriasis lesions.25 This modality therefore allowed for clearance of the skin with more limited risk of erythema; narrowband UVB has come to replace the broad-band UVB phototherapy upon which it was based. In the 1980s systemic retinoids previously developed for acne and hyperkeratosis were also Stern RS, Thibodeau LA, Kleinerman RA, Parrish JA, Fitzpatrick TB, et al, “Risk of Cutaneous Carcinoma in patients treated with oral methoxsalen photochemotherapy for psoriasis,” New Engl J Med 1979; 300: 809–813. 23 Stern RS, Lange R, et al, “Non-melanoma Skin Cancer Occurring in patients treated with PUVA Five to Ten Years after First Treatment, J Invest Dermatol 1988; 91: 120–124. 24 Parrish JA, Jacnicke KF, “Action Sprectrum for phototherapy of Psoriasis,” J Invest Dermatol 1981; 76(5): 359–62. 25 Green C, Ferguson J, Lakshmipathi T, Johnson BE,” 311 nm UVB phototherapy-an effective treatment for psoriasis,” Br J Dermatol 1988; 119: 691–6. 22 J. B. Cameron and A. S. Van Voorhees 6 explored for their possible benefit in the treatment of psoriasis. The second-generation ­retinoid etretinate, followed by the development of its metabolite acitretin, were both shown to be beneficial.26 Etretinate was eventually removed from the market given its lipophilic nature and consequent persistence in the subcutaneous fat. Acitretin however, remains a systemic treatment of psoriasis today. A third generation retinoid, tazarotene was also developed as a topical agent that continues to be utilized in the treatment of psoriasis. Vitamin D and its derivatives were investigated in the 1980s as well. Based on a chance observation of a patient’s psoriasis skin improving with the administration of systemic Vitamin D, the development of topically applied Vitamin D derivatives began. While still not fully understood, these topical agents remain important in the armamentarium of dermatologists when treating patients with psoriasis.27 Systemic Immunosuppressive Medications Understanding the importance of immunosuppression in the treatment of psoriasis was another example of gains achieved by serendipitous findings. When cyclosporine was initially developed in the 1970s the critical role of the immune system in the pathogenesis of psoriasis was not yet appreciated. When transplant patients who coincidentally had psoriasis were placed on cyclosporine to prevent graft rejection they were noted to have improvement of their skin lesions.28 The efficacy of this treatment on the psoriasis helped to identify the importance of T cells, and the immune system more generally, in this disease. Despite its demonstrated effectiveness FDA 26 Ellis CN, Voorhees JJ, “Etretinate therapy,” J Amer Acad Dermatol, 1987; 16: 267–91. 27 Kragballe K, Beck HI, Sogaard H, “Improvement of psoriasis by a topical Vitamin D3 analogue (MC 903) in a double-blind study,” Br J Dermatol 1988; 119(2): 223–230. 28 Mueller W, Hermann B, “Cyclosporin A for psoriasis,” New Engl J Med 1979; 301 (10): 555. approval was delayed until the 1990s because of concerns of possible risks associated with this medication.29 The knowledge gained in the twenty first century from cyclosporine in the understanding of psoriasis opened the door for the development of medications that more specifically targeted the immune system. A number of biologic agents were initially developed which included those targeting T cells as well as those targeting tumor necrosis factor and IL 12/23. While heralded with great promise, the T cell targeting compounds alefacept30 and efulizumab31 have subsequently been removed from the market because of potential side effects and/or lack of efficacy. However the TNF inhibitors—adalimumab,32 etanercept,33 and infliximab34 and the IL-12/23 compound ustekinumab35 have revolutionized the care of patients with psoriasis. They have become mainstays in the treatment of this disease. The initial assumption about ustekinumab was that the IL12 portion was the key blockade. Instead research has now established that the IL23 inhibition is most important in psoriasis. This has opened our eyes to an ever-greater understanding of the Griffiths CEM, DUBrtret L, Ellis CN, et al, “CIclosporin in psoriasis clinical practice: an international consensus statement,” Br J Dermatol 2004; 150 (Suppl 67): 11–23. 30 Ellis CN, Krueger GG, “Alefacept Study Group. Treatment of CHorinc Plaque Psoriasis by selective targeting of memory effector T lymphocytes,” New Engl J Med 2001; 345(4): 248–25. 31 Gordon KB, Papp KA, Hamilton TK, et al, “Efalizumab for patients with moderate to severe plaque psoriasis: a randomized controlled trial“JAMA 2003; 290 (23): 3073–80. 32 Menter A, Tyring SK, Gordon K, et al, “Adalimumab therapy for moderate to severe psoriasis: a randomized, controlled phase 3 trial,” J Amer Acad Dermatol 2007; 58: 106–15. 33 Leonardi CL, Powers JL, Matheson RT, et al, “Etanercept Psoriasis Study group: Etanercept as monotherapy in patients with psoriasis,” New Engl J Med 2003; 349 (21): 2014–22. 34 Chaudri U, Romano P, Mulcahy LD, et al,” Efficacy and safety of infliximab monotherapy for plaque-type psoriasis: a randomized trial,” Lancet 2001; 357 (9271): 1842–7. 35 Krueger GG, Langley RG, Leonardi C, et al, “A Human interleukin-12/23 monoclonal antibody for the treatment of psoriasis,” New Engl J Med 2007; 356: 580–92. 29 1 Introduction to and History of Psoriasis and Psoriasis Therapy immune system and the key role of IL17 cells in the immunologic cascade in psoriasis. With each new class of medication developed, the role of the immune system in psoriasis has become increasingly apparent. This knowledge has allowed for the development of IL17 inhibitors including secukinumab36 and Ixekizumab,37 IL17 receptor blocker brodalumab,38 and IL23 inhibitors guselkumab39 and rizukizumab.40 New agents targeting different sites of the inflammatory cascade are currently under development and may further add to both our understanding of psoriasis and our therapeutic armamentarium. In addition to understanding the pathogenesis of psoriasis we have also come to understand that psoriasis is truly a disease of systemic inflammation. While long known that psoriasis can be 36 LangleyRG, Elewski BE, Lebwohl M, et al, “Secukinumab in plaque psoriasis-results of two phase 3 trials,” NEJM 2014; 371(4): 326–338. 37 Gordon KB, Blauvelt A, Papp KA, et al, “Phase 3 trials of Ixekizumab in Moderate to Severe Psoriasis,” NEJM 2016; 375(4): 345–356. 38 Lebwohl M, Strober B, Menter A, et al, “Phase 3 studies comparing Brodalumab with Ustekinumab in Psoriasis“NEJM 2015; 373: 1318–1328. 39 Reich K, Armstrong AW, Foley P, et al, “Efficacy and Safety of guselkumab, an anti-interleukin-23 monoclonal antibody compared with adalimumab for the treatment of patients with moderate to severe psoriasis with randomized withdrawal and retreatment: results from the phase 3, double-blind, placebo- and active comparator-controlled Voyage 2 trial,” J Amer Acad Dermatol 2017; 76(3): 418–431. 40 Gordon KB, Strober B, Lebwohl M, “Efficacy and Safety of Risankizumab in Moderate to Severe plaque psoriasis (UltiMMa-1 and UltiMMa-2): results from two double-blind, randomized, placebo-controlled and ustekinumab-controlled phase 3 trials,” Lancet 2018; 39: 650–661. 7 associated with psoriatic arthritis, we have become more aware of the frequency with which psoriasis patients suffer from other concurrent illnesses such as inflammatory bowel disease, diabetes and obesity. Patients with psoriasis have also been shown to have an increased risk of cardiovascular disease including cardiovascular death, and more frequently suffer from the metabolic syndrome, sleep apnea, renal disease and others. Thus, our understanding of psoriasis as well as the knowledge of how to treat this disease has become inextricably linked over time. With each step forward therapeutically, more and more is learned so that we enhance our understanding this disease process. This new knowledge has facilitated the development of medications with which to treat psoriasis, thereby allowing patients to achieve control of their disease. The expectation for treatment response has increased to levels unimaginable only ten years ago. Each step in the understanding of the disease as well as its possible treatments has been built upon the lessons learned previously. The National Psoriasis Foundation has set target goals for those with psoriasis of having a body surface area of 1 or less while on treatment. With the vast array of medications at our disposal and their ability to allow for clearance of the skin, achieving clear or almost clear skin has become an obtainable goal. We may soon be entering an era where “the heartbreak of psoriasis” reigns no more. Disclosures John B. Cameron Ph.D.—none Abby S. Van Voorhees, MD—She has served as a consultant for the following companies: Derm Tech, WebMD, Novartis, Lilly, UCB, Celgene. —She has served as an investigator for Celgene, Lilly, AbbVie. 2 Pathophysiology of Psoriasis/ Novel Pathways Jeremy M. Hugh and Jeffrey M. Weinberg Abstract 1. Psoriasis is a systemic inflammatory disease. 2. We now have an increased understanding of the specific cytokines involved in the disease. 3. Therapies have been developed to target these cytokines. Key Learning Objectives 1. To learn the underlying pathophysiology of psoriasis 2. To learn how treatments were developed with the increased understanding of psoriatic pathways 3. To understand novel pathways elucidated for psoriasis Psoriasis is a genetically programmed pathologic interaction among skin cells, immunocytes, and numerous biologic signaling molecules that is triggered by environmental stimuli. The immune response is a cellular one; type 1 (TH1) and type 17 (TH17) T cells are activated by IL-12 J. M. Hugh Department of Dermatology, University of Colorado, Aurora, CO, USA J. M. Weinberg (*) Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA e-mail: jmw27@columbia.edu © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_2 and IL-23 secreted by antigen-presenting cells (APCs) in the skin. Through various cytokines, such as tumor necrosis factor (TNF) α, these cells cause a chronic inflammatory state and alter epidermal hyperproliferation, differentiation, apoptosis, and neoangiogenesis that produce the cutaneous findings seen in this disease. The newer biologic therapies target the immunologic signaling pathways and cytokines identified in the pathogenesis of psoriasis and provide notable clinical improvement. Further study in the pathogenesis of psoriasis can help identify targets for future therapies. The last 30 years of research and clinical practice have revolutionized our understanding of the pathogenesis of psoriasis as the dysregulation of immunity triggered by environmental and genetic stimuli. Psoriasis was originally regarded as a primary disorder of epidermal hyperproliferation. However, experimental models and clinical results from immunomodulating therapies have refined this perspective in conceptualizing psoriasis as a genetically programmed pathologic interaction among resident skin cells; infiltrating immunocytes; and a host of proinflammatory cytokines, chemokines, and growth factors produced by these immunocytes. Two populations of immunocytes and their respective signaling molecules collaborate in the pathogenesis: (1) innate immunocytes, mediated by antigen-presenting cells (APCs)(including natural killer (NK) T lymphocytes, Langerhans cells, and neutrophils), 9 10 and (2) acquired or adaptive immunocytes, mediated by mature CD4+ and CD8+ T lymphocytes in the skin. Such dysregulation of immunity and subsequent inflammation is responsible for the development and perpetuation of the clinical plaques and histological inflammatory infiltrate characteristic of psoriasis. Although psoriasis is considered to be an immune-mediated disease in which intralesional T lymphocytes and their proinflammatory signals trigger primed basal layer keratinocytes to rapidly proliferate, debate and research focus on the stimulus that incites this inflammatory process. Our current understanding considers psoriasis to be triggered by exogenous or endogenous environmental stimuli in genetically susceptible individuals. Such stimuli include group A streptococcal pharyngitis, viremia, allergic drug reactions, antimalarial drugs, lithium, beta-­blockers, IFN-α withdrawal of systemic corticosteroids, local trauma (Köebner phenomenon), and emotional stress. These stimuli correlate with the onset or flares of psoriatic lesions. Psoriasis genetics centers on susceptibility loci and corresponding candidate genes, particularly the psoriasis susceptibility (PSORS) 1 locus on the major histocompatibility complex (MHC) class I region. Current research on the pathogenesis of psoriasis examines the complex interactions among immunologic mechanisms, environmental stimuli, and genetic susceptibility. After discussing the clinical presentation and histopathologic features of psoriasis, we will review the pathophysiology of psoriasis through noteworthy developments, including serendipitous observations, reactions to therapies, clinical trials, and animal model systems that have shaped our view of the disease process. In addition to the classic skin lesions, approximately 23% of psoriasis patients develop psoriatic arthritis, with a 10-year latency after diagnosis of psoriasis [1]. Principles of Immunity The immune system, intended to protect its host from foreign invaders and unregulated cell growth, employs two main effector pathways— J. M. Hugh and J. M. Weinberg the innate and the acquired (or adaptive) immune responses—both of which contribute to the pathophysiology of psoriasis [2]. Innate immunity responses occur within minutes to hours of antigen exposure but fail to develop memory for when the antigen is encountered again. However, adaptive immunity responses take days to weeks to respond after challenged with an antigen. The adaptive immune cells have the capacity to respond to a greater range of antigens and develop immunologic memory via rearrangement of antigen receptors on B and T cells. These specialized B and T cells can then be promptly mobilized and differentiated into mature effector cells that protect the host from a foreign pathogen. Innate and adaptive immune responses are highly intertwined; they can initiate, perpetuate, and terminate the immune mechanisms responsible for inflammation. They can modify the nature of the immune response by altering the relative proportions of type 1 (TH1), type 2 (TH2), and the more recently discovered type 17 (TH17) subset of helper T cells and their respective signaling molecules. A TH1 response is essential for a cellular immunologic reaction to intracellular bacteria and viruses or cellular immunity. A TH2 response promotes IgE synthesis, eosinophilia, and mast cell maturation for extracellular parasites and helminthes as well as humoral immunity, while a TH17 response is important for cell-mediated immunity to extracellular bacteria and plays a role in autoimmunity [3]. The innate and adaptive immune responses employ common effector molecules such as chemokines and cytokines, which are essential in mediating an immune response. Implicating Dysregulation of Immunity Our present appreciation of the pathogenesis of psoriasis is based on the history of trial-and-error therapies; serendipitous discoveries; and the current immune targeting drugs used in a variety of chronic inflammatory conditions, including rheumatoid arthritis, ankylosing spondylitis, and inflammatory bowel disease. Before the mid-­ 2 Pathophysiology of Psoriasis/Novel Pathways 1980s, research focused on the hyperproliferative epidermal cells as the primary pathology because a markedly thickened epidermis was indeed demonstrated on histologic specimens. Altered cell-­cycle kinetics were thought to be the culprit behind the hyperkeratotic plaques. Thus, initial treatments centered on oncologic and antimitotic therapies used to arrest keratinocyte proliferation with agents such as arsenic, ammoniated mercury, and methotrexate [4]. However, a paradigm shift from targeting epidermal keratinocytes to immunocyte populations was recognized when a patient receiving cyclosporine to prevent transplant rejection noted clearing of psoriatic lesions in the 1980s [5]. Cyclosporine was observed to inhibit messenger RNA transcription of T-cell cytokines, thereby implicating immunologic dysregulation, specifically T-cell hyperactivity, in the pathogenesis of psoriasis [6]. However, the concentrations of oral cyclosporine reached in the epidermis exerted direct effects on keratinocyte proliferation and lymphocyte function in these patients [7]. Thus, the question was raised as to whether the keratinocytes or the lymphocytes drove the psoriatic plaques. The use of an IL-2 diphtheria toxin-­fusion protein, denileukin diftitox, specific for activated T cells with high-affinity IL-2 receptors and nonreactive with keratinocytes, distinguished which cell type was responsible. This targeted T-cell toxin provided clinical and histological clearing of psoriatic plaques. Thus, T lymphocytes rather than keratinocytes were recognized as the definitive driver behind the psoriatic plaques [8]. Additional studies have demonstrated that treatments that induce prolonged clearing of psoriatic lesions without continuous therapy, such as psoralen plus UVA irradiation, decreased the numbers of T cells in plaques by at least 90% [9]. However, treatments that require continual therapy for satisfactory clinical results, such as cyclosporine and etretinate, simply suppress T-cell activity and proliferation [10, 11]. Further evidence has linked cellular immunity with the pathogenesis of psoriasis, defining it as a TH1-type disease. Natural killer T cells were shown to be involved through the use of a severe combined immunodeficient mouse model. 11 They were injected into prepsoriatic skin grafted on immunodeficient mice, creating a psoriatic plaque with an immune response showing cytokines from TH1 cells rather than TH2 cells [12]. When psoriatic plaques were treated topically with the toll-like receptor 7 agonist imiquimod, aggravation and spreading of the plaques were noted. The exacerbation of psoriasis was accompanied by an induction of lesional TH1-type interferon produced by plasmacytoid dendritic cell (DC) precursors. Plasmacytoid DCs were observed to compose up to 16% of the total dermal infiltrate in psoriatic skin lesions based on their coexpression of BDCA2 and CD123 [13]. Additionally, cancer patients being treated with interferon alfa experienced induction of psoriasis [14]. Moreover, patients being treated for warts with intralesional interferon alfa developed psoriatic plaques in neighboring prior asymptomatic skin [15]. Patients with psoriasis who were treated with interferon gamma, a TH1 cytokine type, also developed new plaques correlating with the sites of injection [16]. Intralesional T Lymphocytes Psoriatic lesions contain a host of innate immunocytes, such as APCs, NK cells, and neutrophils, as well as adaptive T cells and an inflammatory infiltrate. These cells include CD4 and CD8 subtypes in which the CD8+ cells predominate in the epidermis, while CD4+ cells show preference for the dermis [17]. There are two groups of CD8+ cells: one group migrates to the epidermis, expressing the integrin CD103, while the other group is found in the dermis but may be headed to or from the epidermis. The CD8+ cells residing in the epidermis that express the integrin CD103 are capable of interacting with E-cadherin, which enables these cells to travel to the epidermis and bind resident cells. Immunophenotyping reveals that these mature T cells represent chiefly activated memory cells, including CD2+, CD3+, CD5+, CLA, CD28, and CD45RO+ [18]. Many of these cells express activation markers such as HLA-DR, CD25, and CD27, in addition to the T-cell receptor (TCR). 12 T-Lymphocyte Stimulation J. M. Hugh and J. M. Weinberg psoriasis [20]. Furthermore, the cytokines produced from the immunologic response, such as Both mature CD4+ and CD8+ T cells can respond tumor necrosis factor (TNF) α, IFN-γ, and IL-2, to the peptides presented by APCs. Although correspond to cytokines that are upregulated in the specific antigen that these T cells are react- psoriatic plaques [21]. ing to has not yet been elucidated, several antiIntegral components of the immunologic syngenic stimuli have been proposed, including apse complex include co-stimulatory signals, self-­ proteins, microbial pathogens, and micro- such as CD28, CD40, CD80, and CD86, and adhebial superantigens. The premise that self-reactive sion molecules such as cytotoxic T-lymphocyte T lymphocytes may contribute to the disease antigen 4 and lymphocyte function-­ associated process is derived from the molecular mimicry antigen (LFA) 1, which possess corresponding theory in which an exuberant immune response receptors on the T cell. These molecules play a to a pathogen produces cross-reactivity with self-­ key role in T-cell signaling, as their disruption antigens. Considering that infections have been has been shown to decrease T-cell responsiveassociated with the onset of psoriasis, this theory ness and associated inflammation. The B7 fammerits consideration. However, it also has been ily of molecules routinely interacts with CD28 T observed that T cells can be activated without cells to co-stimulate T-cell activation. Cytotoxic antigens or superantigens but rather with direct T-lymphocyte antigen 4 immunoglobulin, an contact with accessory cells [19]. No single the- antibody on the T-cell surface, targets B7 and ory has clearly emerged. Researchers continue to interferes with signaling between B7 and CD28. search for the inciting stimulus that triggers the In psoriatic patients, this blockade was demonT lymphocyte and attempt to determine whether strated to attenuate the T-cell response and corT cells are reacting to a self-derived or non–self-­ related with a clinical and histologic decrease in derived antigen. psoriasiform hyperplasia [22]. Biologic therapies that disrupt the LFA-1 component of the immunologic synapse also have demonstrated efficacy T-Lymphocyte Signaling in the treatment of psoriasis. Alefacept is a human LFA-3 fusion protein that binds CD2 on T cells T-cell signaling is a highly coordinated process and blocks the interaction between LFA-3 on in which T lymphocytes recognize antigens via APCs and CD2 on memory CD45RO+ T cells and presentation by mature APCs in the skin rather induces apoptosis of such T cells. Efalizumab is than the lymphoid tissues. Such APCs expose a human monoclonal antibody to the CD11 chain antigenic peptides via MHC I or II molecules for of LFA-1 that blocks the interaction between which receptors are present on the T-cell surface. LFA-1 on the T cell and intercellular adhesion The antigen recognition complex at the T-cell and molecule 1 on an APC or endothelial cell. Both APC interface, in concert with a host of antigen-­ alefacept and efalizumab, 2 formerly marketed independent co-stimulatory signals, regulates biologic therapies, demonstrated remarkable T-cell signaling and is referred to as the immu- clinical reduction of psoriatic lesions, and alefanologic synapse. The antigen presentation and cept has been shown to produce disease remisnetwork of co-stimulatory and adhesion mole- sion for up to 18 months after discontinuation of cules optimize T-cell activation, and dermal DCs therapy [23–25]. release IL-12 and IL-23 to promote a TH1 and TH17 response, respectively. The growth factors released by these helper T cells sustain neoan- NK T Cellss giogenesis, stimulate epidermal hyperproliferation, alter epidermal differentiation, and decrease Natural killer T cells represent a subset of CD3+ susceptibility to apoptosis that characterizes the T cells present in psoriatic plaques. Although NK erythematous hypertrophic scaling lesions of T cells possess a TCR, they differ from T cells 2 Pathophysiology of Psoriasis/Novel Pathways by displaying NK receptors comprised of lectin and immunoglobulin families. These cells exhibit remarkable specificity and are activated upon recognition of glycolipids presented by CD1d molecules. This process occurs in contrast to CD4+ and CD8+ T cells, which, due to their TCR diversity, respond to peptides processed by APCs and displayed on MHC molecules. Natural killer T cells can be classified into two subsets: (1) one group that expresses CD4 and preferentially produces TH1- versus TH2-type cytokines, and (2) another group that lacks CD4 and CD8 that only produces TH1-type cytokines. The innate immune system employs NK T cells early in the immune response because of their direct cytotoxicity and rapid production of cytokines such as IFN-γ, which promotes a TH1 inflammatory response, and IL-4, which promotes the development of TH2 cells. Excessive or dysfunctional NK T cells have been associated with autoimmune diseases such as multiple sclerosis and inflammatory bowel disease as well as allergic contact dermatitis [26–28]. In psoriasis, NK T cells are located in the epidermis, closely situated to epidermal keratinocytes, which suggests a role for direct antigen presentation. Furthermore, CD1d is overexpressed throughout the epidermis of psoriatic plaques, whereas normally, CD1d expression is confined to terminally differentiated keratinocytes. An in vitro study examining cytokine-­based inflammation demonstrative of psoriasis treated cultured CD1d-positive keratinocytes with IFN-γ in the presence of alpha-­ galactosylceramide of the lectin family [29]. IFN-γ was observed to enhance keratinocyte CD1d expression, and subsequently, CD1d-­positive keratinocytes were found to activate NK T cells to produce high levels IFN-γ, while levels of IL-4 remained undetectable. The preferential production of IFN-γ supports a TH1-mediated mechanism regulated by NK T cells in the immunopathogenesis of psoriasis. Dendritic Cells Dendritic cells are APCs that process antigens in the tissues in which they reside after which they 13 migrate to local lymph nodes where they present their native antigens to T cells. This process allows the T-cell response to be tailored to the appropriate antigens in the corresponding tissues. Immature DCs that capture antigens mature by migrating to the T-cell center of the lymph node where they present their antigens to either MHC molecules or the CD1 family. This presentation results in T-cell proliferation and differentiation that correlates with the required type of T-cell response. Multiple subsets of APCs, including myeloid and plasmacytoid DCs, are highly represented in the epidermis and dermis of psoriatic plaques as compared with normal skin [30]. Dermal DCs are thought to be responsible for activating both the TH1 and TH17 infiltrate by secreting IL-12 and IL-23, respectively. This mixed cellular response secretes cytokines and leads to a cascade of events involving keratinocytes, fibroblasts, endothelial cells, and neutrophils that create the cutaneous lesions seen in psoriasis [3]. Although DCs play a pivotal role in eliciting an immune response against a foreign invader, they also contribute to the establishment of tolerance. Throughout their maturation, DCs are continuously sensing their environment, which shapes their production of TH1- versus TH2-type cytokines and subsequently the nature of the T-cell response. When challenged with a virus, bacteria, or unchecked cell growth, DCs mature into APCs. However, in the absence of a strong stimulus, DCs fail to mature into APCs and present self-peptides with MHC molecules, thereby creating regulatory T cells involved in peripheral tolerance [31]. If this balance between immunogenic APCs and housekeeping T cells is upset, inflammatory conditions such as psoriasis can result. Cytokines Cytokines are low-molecular-weight glycoproteins that function as signals to produce inflammation, defense, tissue repair and remodeling, fibrosis, angiogenesis, and restriction of neoplastic growth [32]. Cytokines are produced by J. M. Hugh and J. M. Weinberg 14 immunocytes such as lymphocytes and macrophages as well as nonimmunocytes such as endothelial cells and keratinocytes. Proinflammatory cytokines include IL-1, IL-2, the IL-17 family, IFN-γ, and TNF-α, while anti-inflammatory cytokines include IL-4 and IL-10. A relative preponderance of TH1 proinflammatory cytokines or an insufficiency of TH2 anti-inflammatory cytokines induces local inflammation and recruitment of additional immunocyte populations, which produce added cytokines [33]. A vicious cycle of inflammation occurs that results in cutaneous manifestations such as a plaque. Psoriatic lesions are characterized by a relative increase of TH1-­type (eg, IL-2, IFN-γ, TNF-α, TNF-β) to TH2-­type (eg, IL-4, IL-5, IL-6, IL-9, IL-10, IL-13) cytokines and an increase in TH17type cytokines. Natural killer T cells stimulated by CD1d-­overexpressing keratinocytes increase production of proinflammatory IFN-γ without effect on the anti-inflammatory IL-4. In addition to the cytokines produced by T cells, APCs produce IL-18, IL-23, and TNF-α found in the inflammatory infiltrate of psoriatic plaques. Both IL-18 and IL-23 stimulate TH1 cells to produce IFN-γ, and IL-23 stimulates TH17 cells. Clearly, a TH1- and TH17-type pattern governs the immune effector cells and their respective cytokines present in psoriatic skin. Tumor Necrosis Factor α Although a network of cytokines is responsible for the inflammation of psoriasis, TNF-α has been implicated as a master proinflammatory cytokine of the innate immune response due to its widespread targets and sources. Tumor necrosis factor α is produced by activated T cells, keratinocytes, NK cells, macrophages, monocytes, Langerhans APCs, and endothelial cells. Psoriatic lesions demonstrate high concentrations of TNF-­α, while the synovial fluid of psoriatic arthritis patients demonstrates elevated concentrations of TNF-α, IL-1, IL-6, and IL-8 [33]. In psoriasis, TNF-α supports the expression of adhesion molecules (intercellular adhesion molecule 1 and P- and E-selectin), angiogenesis via vascular endothelial growth factor, the synthesis of proinflammatory molecules (IL-1, IL-6, IL-8, and nuclear factor κβ), and keratinocyte hyperproliferation via vasoactive intestinal peptide [34]. A role for TNF-α in psoriasis treatment was serendipitously discovered in a trial for Crohn disease in which infliximab, a mouse-human IgG1 anti–TNF-α monoclonal antibody, was observed to clear psoriatic plaques in a patient with both Crohn disease and psoriasis [35]. Immunotherapies that target TNF-α, including infliximab, etanercept, and adalimumab, demonstrate notable efficacy in the treatment of psoriasis [36–38]. Tumor necrosis factor α is regarded as the driver of the inflammatory cycle of psoriasis due to its numerous modes of production, capability to amplify other proinflammatory signals, and the efficacy and rapidity with which it produces clinical improvements in psoriasis. IL-23/TH17 Axis A new distinct population of helper T cells has been shown to play an important role in psoriasis. These cells develop with the help of IL-23 (secreted by dermal DCs) and subsequently secrete cytokines such as IL-17; they are, therefore, named TH17 cells. CD161 is considered a surface marker for these cells [39]. Strong evidence for this IL-23/TH17 axis has been shown in mouse and human models as well as in genetic studies. IL-23 is a cytokine that shares the p40 subunit with IL-12 and has been linked to autoimmune diseases in both mice and humans [3]. It is required for optimal development of TH17 cells [40] from a committed CD4+ T-cell population after exposure to transforming growth factor β1 in combination with other proinflammatory cytokines [41, 42]. IL-23 messenger RNA is produced at higher levels in inflammatory psoriatic skin lesions versus uninvolved skin [43], and intradermal IL-23 injections in mice produced lesions resembling psoriasis macroscopically and microscopically [44]. Furthermore, 2 Pathophysiology of Psoriasis/Novel Pathways several systemic therapies have been shown to modulate IL-23 levels and correlate with clinical benefit [3]. Alterations in the gene for the IL-23 receptor have been shown to be protective for psoriasis [45–47], and the gene coding for the p40 subunit is associated with psoriasis [45, 46]. Type 17 helper T cells produce a number of cytokines, such as IL-22, IL-17A, IL-17F, and IL-26; the latter 3 are considered to be specific to this lineage [41]. IL-22 acts on outer body barrier tissues, such as the skin, and has antimicrobial activity. Blocking the activity of IL-22 in mice prevented the development of skin lesions [48], and psoriasis patients have elevated levels of IL-22 in the skin and blood [49, 50]. The IL-17 cytokines induce the expression of proinflammatory cytokines, colonystimulating factors, and chemokines, and they recruit, mobilize, and activate neutrophils [51]. IL-17 messenger RNA was found in lesional psoriatic skin but not unaffected skin [52], and cells isolated from the dermis of psoriatic skin have been shown to produce IL-17 [53]. IL-17A is not elevated in the serum of psoriatic patients (unlike other autoimmune diseases) [54], and it is, therefore, thought that TH17 cells and IL-17A production are localized to the affected psoriatic skin. Consistent with this concept is the finding that treatments such as cyclosporin A and antiTNF agents decrease proinflammatory cytokines in lesional skin but not in the periphery [55–57]. These cytokines released by TH17 cells in addition to those released by TH1 cells act on keratinocytes and produce epidermal hyperproliferation, acanthosis, and hyperparakeratosis characteristic of psoriasis [3]. New therapies have been developed to target the IL-23/TH17 axis. Ustekinumab is approved for moderate to severe plaque psoriasis. This treatment’s effect may be sustained for up to 3 years, it is generally well tolerated, and it may be useful for patients refractory to anti-TNF therapy such as etanercept [58]. Briakinumab, another blocker of IL-12 and IL-23, was studied in phase 3 clinical trials, but its development was discontinued due to safety concerns [59]. Newer 15 drugs targeting the IL-23/TH17 axis include secukinumab, ixekizumab, brodalumab, guselkumab, and tildrakizumab. Genetic Basis of Psoriasis Psoriasis is a disease of overactive immunity in genetically susceptible individuals. Because patients exhibit varying skin phenotypes, extracutaneous manifestations, and disease courses, multiple genes resulting from linkage disequilibrium are believed to be involved in the pathogenesis of psoriasis. A decade of genome-wide linkage scans have established that PSORS1 is the strongest susceptibility locus demonstrable through family linkage studies; PSORS1 is responsible for up to 50% of the genetic component of psoriasis [60]. More recently, HLA-Cw6 has received the most attention as a candidate gene of the PSORS1 susceptibility locus on the MHC I region on chromosome 6p21.3 [61]. This gene may function in antigen presentation via MHC I, which aids in the activation of the overactive T cells characteristic of psoriatic inflammation. Studies involving the IL-23/TH17 axis have shown genetics to play a role. Individuals may be protected from psoriasis with a nonsynonymous nucleotide substitution in the IL23R gene [46– 48], and certain haplotypes of the IL23R gene are associated with the disease [46, 48] in addition to other autoimmune conditions. Genomic scans have shown additional susceptibility loci for psoriasis on chromosomes 1q21, 3q21, 4q32–35, 16q12, and 17q25. Two regions on chromosome 17q were recently localized via mapping, which demonstrated a 6 Mb separation, thereby indicating independent linkage factors. Genes SLC9A3R1 and NAT9 are present in the first region, while RAPTOR is demonstrated in the second region [62]. SLC9A3R1 and NAT9 are players that regulate signal transduction, the immunologic synapse, and T-cell growth. RAPTOR is involved in T-cell function and growth pathways. Using these genes as an example, we can predict that the alterations of 16 regulatory genes, even those yet undetermined, can enhance T-cell proliferation and inflammation manifested in psoriasis. Conclusion Psoriasis is a complex disease whereby multiple exogenous and endogenous stimuli incite already heightened innate immune responses in genetically predetermined individuals. The disease process is a result of a network of cell types, including T cells, DCs, and keratinocytes that, with the production of cytokines, generate a chronic inflammatory state. Our understanding of these cellular interactions and cytokines originates from developments, some meticulously planned, others serendipitous, in the fields of immunology, cell and molecular biology, and genetics. Such progress has fostered the creation of targeted immune therapy that has demonstrated remarkable efficacy in psoriasis treatment. Further study of the underlying pathophysiology of psoriasis may provide additional targets for therapy. References 1. Gottlieb A. Psoriasis. Dis Manag Clin Outcome. 1998;1:195–202. 2. Gaspari AA. Innate and adaptive immunity and the pathophysiology of psoriasis. J Am Acad Dermatol. 2006;54(3 suppl 2):S67–80. 3. Di Cesare A, Di Meglio P, Nestle F. The IL-23/Th17 axis in the immunopathogenesis of psoriasis. J Invest Dermatol. 2009;129:1339–50. 4. Barker J. The pathophysiology of psoriasis. Lancet. 1991;338:227–30. 5. Nickoloff BJ, Nestle FO. Recent insights into the immunopathogenesis of psoriasis provide new therapeutic opportunities. J Clin Invest. 2004;113:1664–75. 6. Bos J, Meinardi M, van Joost T, et al. Use of cyclosporine in psoriasis. Lancet. 1989;23:1500–5. 7. Khandke L, Krane J, Ashinoff R, et al. Cyclosporine in psoriasis treatment: inhibition of keratinocyte cell-­ cycle progression in G1 independent effects on transforming growth factor-alpha/epidermal growth factor receptor pathways. Arch Dermatol. 1991;127:1172–9. 8. Gottlieb S, Gilleaudeau P, Johnson R, et al. Response of psoriasis to a lymphocyte-selective toxin (DAB389IL-2) suggests a primary immune, but not keratinocyte, pathogenic basis. Nat Med. 1995;1:442–7. J. M. Hugh and J. M. Weinberg 9. Vallat V, Gilleaudeau P, Battat L, et al. PUVA bath therapy strongly suppresses immunological and epidermal activation in psoriasis: a possible cellular basis for remittive therapy. J Exp Med. 1994;180:283–96. 10. Gottlieb A, Grossman R, Khandke L, et al. Studies of the effect of cyclosporine in psoriasis in vivo: combined effects on activated T lymphocytes and epidermal regenerative maturation. J Invest Dermatol. 1992;98:302–9. 11. Gottlieb S, Hayes E, Gilleaudeau P, et al. Cellular actions of etretinate in psoriasis: enhanced epidermal differentiation and reduced cell-mediated inflammation are unexpected outcomes. J Cutan Pathol. 1996;23:404–18. 12. Nickoloff B, Bonish B, Huang B, et al. Characterization of a T cell line bearing natural killer receptors and capable of creating psoriasis in a SCID mouse model system. J Dermatol Sci. 2000;24:212–25. 13. Gillet M, Conrad C, Geiges M, et al. Psoriasis triggered by toll-like receptor 7 agonist imiquimod in the presence of dermal plasmacytoid dendritic cell precursors. Arch Dermatol. 2004;140:1490–5. 14. Funk J, Langeland T, Schrumpf E, et al. Psoriasis induced by interferon-alpha. Br J Dermatol. 1991;125:463–5. 15. Shiohara T, Kobayahsi M, Abe K, et al. Psoriasis occurring predominantly on warts: possible involvement of interferon alpha. Arch Dermatol. 1988;124:1816–21. 16. Fierlbeck G, Rassner G, Muller C. Psoriasis induced at the injection site of recombinant interferon gamma: results of immunohistologic investigations. Arch Dermatol. 1990;126:351–5. 17. Prinz J. The role of T cells in psoriasis. J Eur Acad Dermatol Venereol. 2003;17(suppl):1–5. 18. Bos J, de Rie M. The pathogenesis of psoriasis: immunological facts and speculations. Immunol Today. 1999;20:40–6. 19. Geginat J, Campagnaro S, Sallusto F, et al. TCR-­ independent proliferation and differentiation of human CD4+ T cell subsets induced by cytokines. Adv Exp Med Biol. 2002;512:107–12. 20. Kastelan M, Massari L, Brajac I. Apoptosis mediated by cytolytic molecules might be responsible for maintenance of psoriatic plaques. Med Hypotheses. 2006;67:336–7. 21. Austin L, Ozawa M, Kikuchi T, et al. The majority of epidermal T cells in psoriasis vulgaris lesions can produce type 1 cytokines, interferon-gamma, interleukin-­2, and tumor necrosis factor-alpha, defining TC1 (cytotoxic T lymphocyte) and TH1 effector populations: a type 1 differentiation bias is also measured in circulating blood T cells in psoriatic patients. J Invest Dermatol. 1999;113:752–9. 22. Abrams J, Kelley S, Hayes E, et al. Blockade of T lymphocyte costimulation with cytotoxic T lymphocyte-­ associated antigen 4-immunoglobulin (CTLA4Ig) reverses the cellular pathology of psoriatic plagues, including the activation of keratinocytes, dendritic cells and endothelial cells. J Exp Med. 2000;192:681–94. 3 Psoriasis: Clinical Review and Update Ivan Grozdev and Neil J. Korman Abstract Plaque psoriasis is the most common and wellrecognized disease type. It affects more than 80% of patients. Sebopsoriasis is a term used when lesions predominate on seborrhoeic areas. Guttate psoriasis is characterized by an acute generalized eruption of small, usually less than 1 cm in diameter, erythematous scaly papules, distributed as “droplets” over the body. It is common in children and young adults with a family history of psoriasis and typically follows streptococcal infection of the upper respiratory tract. Guttate psoriasis and chronic plaque psoriasis are genetically similar conditions with a strong association to the PSORS1 genetic locus. Pustular psoriasis is a group of inflammatory skin conditions characterized by infiltration of neutrophil granulocytes in the epidermis clinically manifested with sterile pustules. The three typical pustular forms are generalized pustular psoriasis, palmoplantar pustulosis, and acrodermatitis continua of Hallopeau. According to the European Rare and Severe Psoriasis Expert Network I. Grozdev (*) Department of Dermatology, Brugmann University Hospital, Brussels, Belgium N. J. Korman Department of Dermatology, University Hospitals Case Medical Center, Cleveland, OH, USA e-mail: njk2@case.edu © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_3 consensus each subtype of pustular psoriasis is subclassified on the basis of the presence/ absence of associated features. Erythrodermic psoriasis is a severe form of psoriasis characterized by generalized inflammatory erythema and scaling involving at least 75% of the body surface area. Precipitating factors may include inappropriate use of potent topical and systemic corticosteroids. Other clinical variants of psoriasis are nail and inverse (flexural) psoriasis. In pediatric population, psoriasis presents the same clinical types. However, lesions may differ in distribution and morphology. In the future, a more clearly defined clinical classification of psoriasis is needed to provide more specific management of each psoriasis type. Key Learning Objectives 1. To understand the clinical features of psoriasis 2. To understand the different variants of psoriasis 3. To understand the differential diagnosis of psoriasis Psoriasis can present in various patterns and forms. Different classifications of the disease have been proposed based on either its epidemiology, age of onset, clinical morphology, or severity [1, 2]. However, the diagnosis is typi19 I. Grozdev and N. J. Korman 20 cally made by the recognition of the classic and distinctive lesions—well-demarcated erythematous plaques with adherent silvery scales. These correlate to the inflammation, vascular dilatation, and altered epidermal proliferation and differentiation seen histopathologically. The most common sites include symmetric involvement of the elbows, knees, lower back, and buttocks, but the disease can involve any cutaneous surface. The disease varies widely in severity and extent of involvement. There are multiple types of psoriasis that have been classified based upon a combination of morphology, distribution, and pattern. This section will review the clinical forms of the disease along with some updates on its particular subtypes. Plaque Psoriasis This is the most common and well-recognized form of psoriasis, also known as psoriasis vulgaris. It affects more than 80% of patients. It is characterized by sharply defined erythematous scaly plaques, usually distributed symmetrically over the extensor surfaces of the upper and lower extremities, lower back and scalp [3]. There can be variation in the intensity of erythema and amount of scale. The size of the lesions varies from coin-sized to palm-sized and larger. The term nummular psoriasis is used if coin-sized lesions predominate [4]. Additional features of psoriatic plaques include the Auspitz sign as the presence of pinpoint bleeding when the tightly adherent scales are removed from the surface of the plaque, and Woronoff ring as the presence of a white ring around erythematous plaques undergoing topical treatment or phototherapy [5, 6]. Psoriasis is well known to develop at sites of physical trauma (scratching, sunburn or surgery), which is the isomorphic or Koebner’s phenomenon [7] and occurs in 11 to 75% of patients [8]. The term sebopsoriasis is used if lesions predominate on seborrhoeic areas, occasionally causing difficulties separating the disease from seborrheic dermatitis [9]. Lesions of plaque psoriasis are quite stable over time. The term annular or polycyclic psoriasis is used if central clearing of the lesions with an active border is present. Psoriatic leukoderma defines the post-­ inflammatory hypopigmentation as the psoriatic plaques regress. Pruritusis present is as many as 60–90% of psoriasis patients [10]. Guttate Psoriasis Guttate psoriasis is characterized by an acute generalized eruption of small, usually less than 1 cm in diameter, erythematous scaly papules, distributed as “droplets” over the whole body surface. Usually, lesions occur over the trunk as the palms and soles are spared. Guttate psoriasis is common in children and young adults with a family history of psoriasis and typically follows streptococcal infection of the upper respiratory tract or acute stressful life events [11]. It can appear either de novo or as an acute exacerbation of preexisting psoriasis. This form of psoriasis may resemble other cutaneous conditions like pityriasis rosea or secondary syphilis. The prognosis is excellent in children as spontaneous remissions are known to occur within weeks or months. The risk of developing chronic plaque psoriasis after a first episode of guttate psoriasis has been estimated to be 25–40% [12, 13]. Guttate psoriasis and chronic plaque psoriasis are genetically similar conditions with a strong association to the PSORS1 genetic locus [14]. Although guttate psoriasis is highly associated with streptococcal infections, there is little evidence-­based data to support treatment of these patients with antibiotics or tonsillectomy [15, 16]. Pustular Psoriasis Pustular psoriasis is a group of inflammatory skin conditions characterized by infiltration of neutrophil granulocytes in the epidermis clinically manifested with sterile pustules. Historically, the three typical pustular forms (generalized pustular psoriasis, palmoplantar pustulosis, acrodermatitis continua of Hallopeau) have been integrated in the large spectrum of pustuloses. (Table 3.1). 3 Psoriasis: Clinical Review and Update Table 3.1 Generalized and localized pustuloses (from Camisa C, 2004, modified) [17] Generalized variants Von Zumbusch-type Impetigo herpetiformis Acute generalized exanthematous pustulosis Localized variants One or more plaques with pustules Palmoplantar pustulosis Annular (Subcorneal pustular dermatosis of Sneddon and Wilkinson) Acrodermatitis continua of Hallopeau Keratoderma blenorrhagicum (Previously known as Reiter’s syndrome) SAPHO syndrome (synovitis, acne, pustulosis, hyperostosis, osteitis) Generalized Pustular Psoriasis This condition was described by Leopold von Zumbusch. It is a severe form of psoriasis and can be life-threatening. It may be preceded either by plaque psoriasis or arise de novo. Withdrawal of systemic steroids may trigger the disease. This form of psoriasis is characterized by sterile pustules arising from the surface of large erythematous patches of skin distributed over the trunk and extremities. The pustules eventually dry and peel. In some cases, these pustules may form confluent large lakes of pus. Oral lesions may be present with pustules or acute geographic tongue. The eruption is usually accompanied by systemic symptoms including fever, chills, diarrhea, and arthralgias. Leukocytosis and an elevated erythrocyte sedimentation rate are commonly encountered. Generalized pustular psoriasis may be associated with polyarthritis and cholestasis from neutrophilic cholangitis [18]. It has been reported that pustular psoriasis occurs in 9% of psoriasis patients without psoriatic arthritis and in 41% of patients with psoriatic arthritis [19]. Impetigo Herpetiformis Originally described in 1872 by von Herba, impetigo herpetiformis refers to a generalized pustular eruption anytime during pregnancy in 21 a woman without prior history of psoriasis [20]. Resolution occurs with delivery. Recurrences might be associated with menstruation and oral contraceptives [21]. Some cases have been found to have IL-36RN mutations, suggesting that impetigo herpetiformis and generalized pustular psoriasos are the same disease [22]. cute Generalized Exanthematous A Pustulosis AGEP is a drug-triggered severe cutaneous reaction characterized by rapid development of nonfollicular, sterile pustules on an erythematous base. A wide variety of drugs has been associated with thecondition, antibiotics being the most common cause. Typically, within 48 h of ingesting the causative medication, there is acute onset of fever and pustulosis with leukocytosis. In severe cases, there can be mucous membrane and systemic organ involvement [23]. In some AGEP cases, the same IL-36RN mutations were found as in generalized pustular psoriasis [24]. Localized Pustular Forms Palmoplantar pustulosis is the most common variant of pustular psoriasis. It is characterized by pruritic or burning erythematous patches on the palms and soles, within which multiple pustules develop. Initially, the pustules are yellow. Later they turn dark brown, dry up and form crusts that subsequently exfoliate leaving tender and diffusely eroded surfaces [25]. Those patients experience great impairment of their quality of life with difficulty in walking and using their hands. Some authors consider palmoplantar pustulosis to be a separate dermatologic disorder as it affects predominantly female patients, has a higher age of onset, is associated with cigarette smoking (up to 100% of cases at onset), and consistently responds poorly to topical therapy. Palmoplantar pustulosis does not share the association of PSORS1 gene locus with plaque psoriasis, supporting the concept that these are distinct entities. I. Grozdev and N. J. Korman 22 Acrodermatitis continua of Hallopeau is a rare, chronic, and recalcitrant pustular eruption of the fingers and toes that is widely considered to be a localized variant of pustular psoriasis. The pustules are located on the fingertips or toes and are very painful and disabling, often leading to loss of function in the affected digits and substantial morbidity. Nail dystrophy and paronychial erythema are typically seen [26]. Consensus Statement of Phenotypes of Pustular Psoriasis For clinical use, pustular psoriasis is still grouped with psoriasis vulgaris. However, accumulating data reveal that the two conditions are genetically and phenotypically different, respond differently to treatment. The European Rare and Severe Psoriasis Expert Network was founded to define consensus criteria for diagnosis and phenotypes of pustular psoriasis, to analyze the genetics and pathophysiology, to prepare for prospective clinical trials [27]. According to the consensus of this expert group, each subtype of pustular psoriasis is subclassified on the basis of the presence or absence of associated features. Generalized pustular psoriasis is defined as primary, sterile, macroscopically visible pustules on non-acral skin, excluding cases where pustules are restricted to psoriatic plaques. It can occur with or without systemic inflammation, with or without psoriasis vulgaris, and can be either a relapsing (> 1 episode) or persistent (> 3 months) condition. Palmoplantar pustulosis is defined as primary, persistent (> 3 months), sterile, macroscopically visible pustules on the palms and/or soles and can occur with or without psoriasis vulgaris. Acrodermatitis continua of Hallopeau is defined as primary, persistent (> 3 months), macroscopically visible pustules affecting the nail apparatus, with or without psoriasis vulgaris. ErythrodermicPsoriasis Erythrodermic psoriasis is a severe form of psoriasis characterized by generalized inflammatory erythema and widespread scaling involving at least 75% of the body surface area [28]. It may develop gradually or acutely during the course of chronic psoriasis, but it may also be the initial manifestation of psoriasis. Important precipitating factors for erythrodermic psoriasis may include inappropriate use of potent topical and systemic corticosteroids. Psoriatic erythroderma is not substantially clinically different from erythroderma caused by eczematous dermatitis, seborrhoeic dermatitis, pityriasisrubra pilaris, drug induced erythroderma, lymphoma, or leukemia. Associated findings may include lymphadenopathy, hypothermia, tachycardia, peripheral edema, elevated erythrocyte sedimentation rate, hypoalbuminemia, anemia, leukocytosis or leucopenia, elevations of lactate dehydrogenase, liver transaminases, uric acid, and calcium [29]. Severe medical complications can develop due to dehydration from extensive fluid and electrolyte disturbances, protein losses, high-output cardiac failure, and infection. Manifestations of Psoriasis in Specific Locations Scalp Psoriasis The scalp is a very common location for psoriasis. Plaques typically form on the scalp and along the hair margin. Many patients discover they have psoriasis because of a dandruff-like desquamation. When the scalp is the only location of psoriasis, it may be difficult to distinguish from seborrheic dermatitis. The term sebopsoriasis is used in such cases [9]. The scales can firmly be attached to the scalp hair. This particular variety is called pseudotinea amiantacea and is more frequent in children [30]. Scarring alopecia may develop in some patients [31]. Nail Psoriasis The nails are commonly affected in patients with psoriasis. In cases where skin lesions are absent or equivocal, nail changes may be of assistance in making the diagnosis of psoriasis. Two patterns 3 Psoriasis: Clinical Review and Update of nail disorders have been shown to be caused by psoriasis [32]. Nail matrix involvement can result in features such as leukonychia, pitting, red spots in the lunula and crumbling. Onycholysis, salmon or oil-drop patches, subungual hyperkeratosis and splinter hemorrhages are classic features of psoriasis nailbed involvement. Nail psoriasis causes aesthetic and functional impairment, and is considered as an indicative of psoriasis joint involvement. 23 cum can develop psoriasiform skin lesions 1–2 months after the onset of arthritis. with involvement of the palms, soles, and the scalp [37]. The psoriasiform patch has distinctive circular scaly borders that develop from fusion of papulovesicular plaques with thickened yellow scale. Pediatric Psoriasis Approximately 30–50% of adults with psoriasis developed psoriasis before the age of 20. Inverse (Flexural, Intertiginous) The prevalence of childhood psoriasis has been Psoriasis reported to be up to 2% [38]. Although children present with the same cliniInverse psoriasis affects the skin fold areas cal types of psoriasis seen in adults, lesions may including the axillae, submammary regions, glu- differ in distribution and morphology. Plaque teal cleft, retroauricular, and inguinal folds [33]. psoriasis is the most common type of psoriasis in Obese patients often have this form of psoriasis children. Typical erythematous plaques with involving their excess skin folds [34]. Thin well-­ overlying white scales are often thinner and demarcated erythematous patches without des- smaller. Lesions tend to develop more often on quamation are the typical skin lesions. Usually the scalp, face, and flexural areas. Young children the patches are superficially eroded with fissur- usually present with a diaper rash that is irresponing in the body folds. Inverse psoriasis may occur sive to irritant diaper dermatitis treatment [39]. alone but is more frequently accompanied by Guttate psoriasis is the second most common plaque psoriasis elsewhere. In cases where it is type of psoriasis in childhood. It could be trigthe only location of psoriasis, inverse psoriasis gered by a beta-hemolytic streptococcal or viral may be confused with bacterial, fungal, or can- infection, and tends to resolve spontaneously didal intertrigo. Scrapings or cultures are then within 3–4 months of onset. It has been reported needed to exclude infection. that a proportion of children with guttate psoriasis eventually develop plaque psoriasis [40]. Pustular psoriasis is seen in 1.0–5.4% of children with psoriasis. Other less common types of Rare Forms and Some Specific Locations psoriasis in children are inverse psoriasis, palmoplantar psoriasis, isolated facial psoriasis, linear Geographic tongue also known as benign migra- psoriasis, erythrodermic psoriasis. Nail changes tory glossitis may be seen in psoriatic patients, have been reported in up to 40% of children with especially in patients with generalized pustular psoriasis. Approximately 7% of all patients with psoriasis. It is characterized by erythematous juvenile idiopathic arthritis appear to have juvepatches surrounded by a white line and loss of nile psoriatic arthritis [41]. Children suffering from psoriasis have higher filiform papillae on the dorsum of the tongue [35]. The lips can be affected in psoriasis but prevalence of comorbidities, including obesity, this should be differentiated from discoid lupus diabetes, hypertension, rheumatoid arthritis, erythematosus and desquamative cheilitis [36]. Crohn’s disease and psychiatric disorders, comIf psoriasis only affects the glans, the most com- pared to children without psoriasis [42]. Overall, 13% to 27% of children have modermon site of involvement is the proximal part [30]. It should be differentiated from erythroplasia of ate to severe disease, with a 10% body surface Queyrat. Patients with Keratoderma blenorrhagi- area affected, and may warrant the use of systemic I. Grozdev and N. J. Korman 24 Table 3.2 Differential diagnosis of variants of psoriasis (modified from Lisi P 2007) [44] References Clinical variant Guttate psoriasis 1. Griffiths CEM, Christophers E, Barker JNWN, et al. A classification of psoriasis vulgaris according to phenotype. Br J Dermatol. 2007;156:258–62. 2. Dogra S, Mahajan R. Psoriasis: epidemiology, clinical features, co-morbidities, and clinical scoring. Indian Dermatol Online J. 2016;7(6):471–80. 3. Lebwohl M. Psoriasis Lancet. 2003;361:1197–204. 4. Van de Kerkhof PCM. Clinical features. In: de Kerkhof PCM V, editor. Psoriasis. 2nd ed. London: Blackwell; 2003. p. 3–30. 5. Bernhard JD. Clinical pearl: Auspitz sign in psoriasis scale. J Am Acad Dermatol. 1997;36(4):621. 6. Varma S, Finlay AY. The Woronoff ring in psoriasis. Br J Dermatol. 2003;148910:170. 7. Diani M, Cozzi C, Altomare G. Heinrich Koebner and his phenomenon. JAMA Dermatol. 2016; 152(8):919. 8. Boyd AS, Neldner KH. The isomorphic response of Koebner. Int J Dermatol. 1990;29(6):401–10. 9. Naldi L, Gambini D. The clinical spectrum of psoriasis. Clin Dermatol. 2007;25:510–8. 10. Szepietowski JC, Reich A. Pruritus in psoriasis: an update. Eur J Pain. 2016;20(1):41–6. 11. Naldi L, Peli L, Parazzini F, Carrel CF, Psoriasis Study Group of the Italian Grouo for Epidemiological Research in Dermatology. Family history of psoriasis, stressful life events, and recent infectious disease are risk factors for a first episode of acute guttate psoriasis: results of a case-control study. J Am Acad Dermatol. 2001;44:433–8. 12. Martin BA, Chalmers RJ, Telfer NR. How great is the risk of further psoriasis following a single episode of acute guttate psoriasis? Arch Dermatol. 1996;132:717–8. 13. Pfingstler LF, Maroon M, Mowad C. Guttate psoriasis outcomes. Cutis. 2016;97(2):140–4. 14. Asumalahti K, Ameen M, Suomela S, et al. Genetic analysis of PSORS1 distinguishes guttate psoriasis and palmplantar pustulosis. J Invest Dermatol. 2003;120:627–32. 15. Owen CM, Chalmers RJ, O’Sullivan T, Griffiths CE. A systematic review of antistreptococcal interventions for guttate and chronic plaque psoriasis. Br J Dermatol. 2001;145(6):886–90. 16. Dupire G, Droitcourt C, Hughes C, et al. Antistreptococcal interventions for guttate and chronic plaque psoriasis. Cochrane Database Syst Rev. 2019;3:CD011571. 17. Camisa C. The clinical variants of psoriasis. In: Camisa C, editor. Handbook of psoriasis. 2nd ed. New York: Blackwell; 2004. p. 7–36. 18. Viguier M, Allez M, Zagdanski AM, et al. High frequency of cholestasis in generalized pustular psoriasis. Evidence for neutrophilic involvement of the biliary tract. Hepatology. 2004;40:452–8. 19. Aslanian FMNP, Lisboa FFCB, Iwamoto A, Carneirot SCS. Clinical and epidemiological evaluation of pso- Palmoplantarplaques psoriasis Palmoplantar pustulosis Generalized pustular psoriasis Erythrodermic psoriasis Nail psoriasis Inverse psoriasis Scalp psoriasis Differential diagnosis Pityriasis versicolor, Pityriasis rosea, Secondary syphilis Hand eczema, Contact dermatitis, Dermatophytosis Pompholyx, Dermatophytosis Acute generalized exanthematous pustulosis Atopic dermatitis, Drug reactions, Cutaneous T-cell lymphoma, Ichthyoses Dermatophytosis, Drug reactions, Phototoxic reactions Candida infection, Erythrasma, Seborrhoeic dermatitis, Contact allergic dermatitis, Benign familial pemphigus (Hailey-Hailey disease) Seborrheic dermatitis, Contact allergy, Ringworm therapy. Along with the conventional treatment options, biologics such as tumour necrosis factor (TNF)-alpha inhibitor etanercept and the p40 inhibitor ustekinumab are approved in the United States for severe plaque psoriasis in children. In most European countries, adalimumab, another TNF-alpha inhibitor, is also approved [43]. Differential Diagnosis of Psoriasis The clinical presentation of psoriasis is usually sufficient to make the diagnosis. Its differential diagnosis includes other common skin diseases (Table 3.2): Conclusion The continuous genetic and other experimental research on psoriasis, along with the numerous therapeutic studies in large psoriasis populations, should come out with even more clearly defined types of psoriasis. This would help for better managing the disease and its course. 3 Psoriasis: Clinical Review and Update riasis: clinical variants and articular manifestations. J Eur Acad Dermatol Venereol. 2005;19:141–2. 20. Oumeish OY, Parish LJ. Impetigo herpetiformis. Clin Dermatol. 2006;24:101–4. 21. Chaidemenos G, Lefaki I, Tsakiri A, Mourellou O. Impetigo herpetiformis: menstrual exacerbations for 7 years postpartum. J Eur Acad Dermatol Venereol. 2005;19:466–9. 22. Sigura K, Oiso N, Iinuma S, et al. IL-36RN mutations underlie impetigo herpetiformis. J Invest Dermatol. 2014;134:2472–4. 23. Szatkowski J, Schwartz RA. Acute generalized exanthemateous pustulosis (AGEP): a review and update. J Am Acad Dermatol. 2015;73(5):843–8. 24. Navarini AA, Simpson MA, Borradori L, et al. Homozygous missense mutation in IL-36RN in general pustular dermatosis with intral oral involvement compatible with both AGEP and general pustule psoriasis. JAMA Dermatol. 2015;151:452–3. 25. Misiak-Galazka M, Wolska H, Rudnicka L. What do we know about palmoplantar pustulosis? J Eur Acad Dermatol Venereol. 2017;31(1):38–44. 26. Seghal VN, Verma P, Sharma S, et al. Acrodermatitis continua of Hallopeau: evolution of treatment options. Int J Dermatol. 2011;50(10):1195–211. 27. Navarini AA, Burden AD, Capon F, et al. European consensus statement on phenotypes of pustular psoriasis. J Eur Acad Dermatol Venereol. 2017;31(11):1792–9. 28. Singh RK, Lee KM, Ucmak D, et al. Erythrodermic psoriasis: pathophysiology and current treatment perspectives. Psoriasis (Auckl). 2016;6:93–104. 29. Boyd AS, Menter A. Erythrodermic psoriasis: precipitating factors, course, and prognosis in 50 patients. J Am Acad Dermatol. 1989;21(Part 1):985–91. 30. Ayala F. Clinical presentation of psoriasis. Reumatismo. 2007;59(1):40–5. 31. van de Kerkhof PCM, Chang A. Scarring alopecia and psoriasis. Br J Dermatol. 1992;126:524–5. 32. Schons KR, Knob CF, Murussi N, et al. Nail psoriasis: a review of the literature. An Bras Dermatol. 2014;89(2):312–7. 25 33. Khosravi H, Siegel MP, Van Voorhens AS, et al. Treatment of inverse/intertriginous psoriasis: updated guidelines from the Medical Board of the National Psoriasis Foundation. J Drugs Dermatol. 2017;16(8):760–6. 34. Wang G, Li C, Cao T, Liu Y. Clinical analysis of 48 cases of inverse psoriasis: a hospital-based study. Eur J Dermatol. 2005;15:176–8. 35. Meier M, Sheth PB. Clinical spectrum and severity of psoriasis. Curr Probl Dermatol. 2009;38:1–20. 36. Zhu JF, Kaminski MJ, Pulitzer DR, Hu J, Thomas HF. Psoriasis: pathophysiology and oral manifestations. Oral Dis. 1996;2:135–44. 37. Suong J, Lenwohl M. Psoriasis – clinical presentation. In: Psoriasis and psoriatic arthritis. An integrated approach. Gordon KB, Ruderman EM (eds.), Springer Berlin, 2004: 67-72. 38. Burden-Teh E, Thomas KS, Ratib S, et al. The epidemiology of childhood psoriasis: a scoping review. Br J Dermatol. 2016;174(6):1242–57. 39. Bronckers IM, Paller AS, van Geel MJ, et al. Psoriasis in children and adolescents: diagnosis, management and comorbidities. Pediatr Drugs. 2015;17(5):373–84. 40. Mercy K, Kwasny M, Cordoro KM, et al. Clinical manifestations of pediatric psoriasis: results of a multicenter study in the United States. Pediatr Dermatol. 2013;30(4):424–8. 41. Schwartz G, Paller AS. Targeted therapies for pediatric psoriasis. Semin Cutan Med Surg. 2018;37(3):167–72. 42. Mahe E. Childhood psoriasis. Eur J Dermatol. 2016;26(6):537–48. 43. Thaci D, Papp K, Marcoux D, et al. Sustained long-­ term efficacy and safety of adalimumab in pediatric patients with severe chronic plaque psoriasis from a randomized, double-blind, phase III study. Br J Dermatol. 2019 Apr 24;181(6):1177–89. https://doi. org/10.1111/bjd.18029. 44. Lisi P. Differential diagnosis of psoriasis. Reumatismo. 2007;59(Suppl 1):56–60. 4 Psoriasis: Epidemiology, Potential Triggers, Disease Course Ivan Grozdev and Neil J. Korman Abstract Worldwide prevalence rates of psoriasis range from 0.6 to 4.8%. The disease tends to have a bimodal distribution of onset with the major peak occurring at age of 20–30, and a later smaller peak occurring at age of 50–60. While there are many potential triggers of psoriasis, infections are an important trigger and up to half of children with psoriasis have an exacerbation within 2 weeks following an upper respiratory infection. Psychological distress is a causative or maintaining factor in disease expression for many patients with psoriasis. Other well-documented triggers for flares include trauma, alcohol and smoking, as well as obesity. Plaque psoriasis is usually chronic with intermittent remissions. Plaques may persist for months to years at the same locations; however, periods of complete remission may occur. I. Grozdev (*) Department of Dermatology, Sofia Medical Faculty, Sofia, Bulgaria N. J. Korman Department of Dermatology, University Hospitals Case Medical Center, Cleveland, OH, USA e-mail: njk2@case.edu © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_4 Key Learning Objectives 1. To understand the epidemiology of psoriasis 2. To examine the triggers of psoriasis 3. To understand the clinical course of the disease Epidemiology Worldwide prevalence rates of psoriasis range from 0.6 to 4.8% [1, 2]. Women and men are equally affected. The prevalence of the disease varies depending on the climate and ethnicity, although these relationships are complicated. The Caucasian population of Europe and the US are affected equally by psoriasis [3, 4]. The disease is uncommon in the Mongoloid race. Asian Americans have a prevalence of between 0.4% [5] and 0.7% [6]. African-Americans have a prevalence of 1.3% [7], while American-Indians have prevalence of 0.2% or less [6]. Psoriasis is absent in certain populations such as South American Indians and Australian Aborigines [8], while in the Arctic Kasah’ye its prevalence is 11.8% [9]. A positive correlation between latitude and psoriasis prevalence would be expected given the efficacy of ultraviolet light as a treatment [10]. However, Jacobson et al. identified 22 population-­based surveys, case-control studies, 27 I. Grozdev and N. J. Korman 28 and reviews on psoriasis prevalence rates from numerous regions around the globe and found no correlation between absolute latitude and psoriasis prevalence [11]. These findings suggest that other factors or a combination of factors may play a role in the frequency of psoriasis rather than latitude alone. Differences in the prevalence rates of psoriasis between two areas of the same continent (West Africa prevalence rates of 0.05– 0.9% compared to South Africa prevalence rates of 2.8–3.5% [12]) suggest that genetic factors play an important role [13]. Although new onset psoriasis occurs in all age groups from newborns to age 108 [14], the disease tends to have a bimodal distribution of onset with the major peak occurring at age of 20–30, and a later smaller peak occurring at age of 50–60. Patients with early age onset of psoriasis are more likely to have a family history of psoriasis, the course of the disease tends to be more unstable with frequent remissions and relapses, the disease tends to be more resistant to treatment and more severe disease tends to be more common. Late age onset psoriasis tends to have a more stable chronic clinical course, and is more likely to be associated with psoriatic arthritis, nail involvement and palmoplantar pustular involvement [15, 16]. The mean age of onset of psoriasis varies from study to study, but nearly 75% of patients with psoriasis have an onset before the age of 40, and 12% of patients have the onset of psoriasis at age of 50–60 [17]. More recent studies have indicated an increasing prevalence of childhood psoriasis [18]. The known familial concentration of psoriasis indicates an important role of hereditary factors; however a 67%-concordance in monozygotic twins suggests that environmental factors may also be an important component of psoriasis [19]. Potential Triggers Trauma Trauma can trigger the exacerbation of psoriatic lesions or the development of new lesions (this is known as the Koebner phenomenon, originally described by Heinrich Koebner in 1872). Trauma as a trigger is more commonly seen in patients who develop psoriasis at an early age and those who require multiple therapies to control their disease [20]. In clinical studies the prevalence of the Koebner phenomenon may range from 24–51% of patients while in experimental studies among selected patients with severe psoriasis and a history of the Koebner phenomenon, it was observed in up to 92% of patients [21]. Factors that may lead to the Koebner phenomenon include acupuncture, vaccinations, scratches, removal of adhesive bandages, insect and animal bites, burns (thermal, chemical, electrosurgical), radiation, incisions, cuttings, abrasions, tattoos, irritant and allergic contact dermatitis, phototoxic dermatitis, as well as skin diseases including acne, furuncles, herpes zoster, and lichen planus. The “reverse” Koebner phenomenon which occurs when trauma within a psoriatic lesion causes clearing of that psoriatic lesion has also been observed [22]. Some treatment modalities for psoriasis are based on the reverse Koebner phenomenon such as electrodissecation, dermabrasion, cryotherapy, and CO2-laser therapy. Infections Infections have long been recognized as important triggers for psoriasis exacerbations. Up to half of children with psoriasis have an exacerbation of their disease within 2 weeks following an upper respiratory infection [23, 24]. Infection with streptococcus pyogenes has a well-known association with guttate psoriasis [25]. Up to 85% of patients with an episode of acute guttate psoriasis show evidence of a preceding streptococcal infection as demonstrated by positive anti-­streptolysin-­O titers [24]. In another study, 84% of patients with guttate psoriasis had a history of infection prior to occurrence of skin lesions, and the majority of these patients (63%) had a verified streptococcal pharyngitis [26]. Although pharyngeal origin of the infection is most common, skin infection with streptococcus pyogenes can also lead to guttate psoriasis. Streptococcal infections can elicit exacerbations 4 Psoriasis: Epidemiology, Potential Triggers, Disease Course of other types of psoriasis and psoriatic arthritis as well. Patients with psoriasis develop sore throats much more frequently than non-psoriatic individuals and it is well documented that streptococcal throat infections can trigger the onset of psoriasis, and such infections cause exacerbation of chronic psoriasis [27]. The study of 111 patients isolated streptococcus pyogenes in 13% of patients with a guttate flare of chronic plaque psoriasis, in 14% of patients with chronic plaque psoriasis, and in 26% of patients with acute guttate psoriasis, while 7% of the patients in the control group had streptococcus pyogenes isolated [28]. Although many reports suggest a possible role for antibiotics or tonsillectomy in the treatment of guttate psoriasis [27, 29–33], the data is controversial about the beneficial effect of either intervention [34]. Superantigens such as streptococcal pyogenic exotoxin [35–38] as well as peptidoglycan derived from various different bacterial sources [39, 40] can lead to the development of psoriasis due to an abnormal response of the innate immune system towards the super antigen. Development and use of experimental vaccines to treat psoriasis are based on this hypothesis [41, 42]. Other microorganisms reported to be potential triggers for psoriasis include Staphylococci, Candida, H pylori and Malassezia spp [25, 43] while infections with Yersinia spp have been reported to induce psoriatic arthritis [25]. Another important potential triggering factor for psoriasis is infection with the human immunodeficiency virus (HIV). The link between HIV infection and psoriasis onset seems paradoxical as the immunosuppression should lead to psoriasis improvement [44]. It is suggested that either HIV could function as a super antigen or other microorganisms, including opportunistic ones, could develop in the host because of the immune dysregulation [44]. The prevalence of psoriasis in patients with HIV infection is nearly 5%, about twice that seen in the general population. The clinical manifestations of psoriasis in HIV-­infected patients are similar to those in non-HIV-­infected patients. However, lesions of more than one subset of psoriasis are often found in the same HIV patient [45]. For example, a patient with chronic 29 plaque psoriasis may go on to develop guttate or pustular lesions. Psoriasis may occur at any time in the course of an HIV infection and exacerbations tend to be longer and more frequent than those in otherwise healthy psoriasis patients [46]. There is no observed relationship between psoriasis and the CD4 count. There is one report of psoriasis remission in the terminal stages of the acquired immunodeficiency syndrome [47] and another report on complete resolution of erythrodermic psoriasis in an HIV and HCV patient, unresponsive to anti-psoriatic treatments after highly active antiretroviral therapy [48]. Rapid onset of acute eruptive psoriasis, frequent exacerbations or resistance to conventional and biologic treatments should raise the possibility of underlying HIV disease [48, 49]. It has been suggested that inflammation within psoriatic lesions develops against unknown antigens and super antigens of viral origin such as human papilloma virus 5 (HPV5), human endogenous retroviruses, Coxsackie adenoviruses, Arboviruses and others [43, 50, 51]. Stress Psychological distress is a causative or maintaining factor in disease expression for many patients with psoriasis. In one study, over 60% of psoriasis patients believed that stress was the principal factor in the cause of their psoriasis [52]. Farber and colleagues surveyed over 5000 patients with psoriasis and 40% reported that their psoriasis occurred at times of worry and 37% experienced worsening of psoriasis with worry [5]. In a more recent study of 400 patients with newly developed psoriasis, 46% of the patients with plaque psoriasis and 12% of the patients with guttate psoriasis linked the onset of their disease with a life crisis, including divorce, severe or life-­threatening disease of the patient or a family member, death in the family, financial burden, dismissal, or harassment in school [53]. In another study among 50 psoriasis patients, stressful life events were seen in 26% of the patients within 1 year preceding onset or exacerbation of psoriasis, suggesting the potential value of relaxation therapies and 30 stress management programs in the management of patients with psoriasis [54]. Stress-induced relapse rates of up to 90% have been reported in children [23]. In an epidemiological study of 784 Greek psoriasis patients, stress was self-reported as the main cause for psoriasis exacerbations by 60% of patients [55]. It was demonstrated that “low level worriers” achieved clearing of their skin with PUVA (psoralen plus ultraviolet-A) a median of 19 days earlier than “high level worriers” undergoing the same treatment [56]. Other studies reveal that cognitive-behavioral therapy in conjunction with medical therapy can lead to a significantly greater reduction in the severity of psoriasis than medical therapy alone [57, 58]. The role that acute psychosocial stressors play in altering hypothalamic-­pituitary-­adrenal (HPA) responses in patients with psoriasis is an area of controversy. Some data shows that stress-exacerbated psoriasis flares lead to decreased levels of cortisol [59], while other fails to show such a correlation [60, 61]. The epidemiologic data linking psychological stress and onset or exacerbation of psoriasis is also controversial with some studies supporting this association [62], while others do not [63, 64]. Medications Various medications used for concomitant diseases may influence psoriasis in terms of precipitating or worsening it. Drug-induced psoriasis is defined as the development of psoriasis after treatment with a medication that remits when that medication is withdrawn, while drug-triggered or drug-exacerbated psoriasis is defined as the development of psoriasis after treatment with a medication whose withdrawal does not influence the clinical course [65]. The time between the start of the drug intake and the outbreak of ­psoriatic eruption may vary and depends on the drug and is classified as follows: short (<4 weeks between the start of the drug and the onset of psoriatic eruption), medium (>4 and <12 weeks), and long (>12 weeks) [66]. Several medications may trigger or worsen psoriasis. The most common medications that have been reported to trigger I. Grozdev and N. J. Korman psoriasis include lithium, beta-blockers, non-steroidal anti-inflammatory drugs, tetracyclines, and antimalarials [65, 67]. Several other medications that have reported to worsen psoriasis include angiotensin converting enzyme (ACE) inhibitors, terbinafine, clonidine, iodine, amiodarone, penicillin, digoxin, interferon-alpha, and interleukin-2. The abrupt discontinuation of systemic or superpotent topical corticosteroids can serve as triggers of psoriasis although the frequency of this association has not been studied. While these observations suggest the possibility that certain medications may trigger psoriasis worsening, no controlled trials have proven an association. Newly developed psoriasis has been reported in patients taking TNF-alpha blockers for indications other than psoriasis, including Crohn’s disease and rheumatoid arthritis [68, 69]. Interferons play an important role in the pathogenesis of psoriasis. Their use as medications such as IFN (α, β, γ) and imiquimod may induce or worsen psoriasis [43, 70]. Additionally, withdrawal of interferon use in hepatitis C commonly leads to improvement in psoriasis [71]. Alcohol and Smoking Alcohol and smoking have both been implicated as triggering factors for psoriasis exacerbations. It is well documented that the prevalence of psoriasis is increased among patients who abuse alcohol [72]. However, conflicting evidence exists as to whether increased alcohol intake in psoriasis patients is a factor in the pathogenesis or whether having a chronic disorder like psoriasis leads to greater intake of alcohol in an attempt to self-medicate. A study of 144 Finnish patients with psoriasis demonstrated that alcohol consumption in the previous 12 months was linked to the onset of psoriasis. This study suggests that psoriasis may lead to sustained alcohol abuse and that this alcohol intake may perpetuate the disease [73]. Qureshi et al. prospectively evaluated the association between total alcohol consumption and risk of incident psoriasis in a cohort of 82,869 nurses [74]. Compared with women who did not drink alcohol, the multivariate relative 4 Psoriasis: Epidemiology, Potential Triggers, Disease Course risk of psoriasis was significantly higher for an alcohol consumption of 2.3 drinks per week or more. Moreover, examining the type of alcoholic beverage, non-light beer intake was associated with an increased risk of developing psoriasis among women, while other alcoholic beverages did not increase this risk. Recently, a meta-­ analysis of case-control studies showed that the overall odds ratio of psoriasis for drinking persons compared to those with non-drinking habits was 1.531 (P = 0.002), suggesting that alcohol consumption is associated with an increased risk of psoriasis [75]. Further support of increasing alcohol abuse as a post-diagnosis condition was seen in a case-control study of 60 Australian twins who were discordant for psoriasis [76]. In this study, no difference in alcohol consumption between discordant twins, either monozygotic or dizygotic, was discovered. The influence of increasing alcohol consumption on the severity of psoriasis has also been investigated and there appears to be a tendency for heavy drinkers to develop more extensive and severe psoriasis that lighter drinkers [77]. Mortality related to alcohol use in psoriasis has also been evaluated. A population-based study of over 5000 patients followed for 22 years demonstrated that psoriatic patients have an increased mortality rate when compared to a control group [78]. However, this study did not account for previous hepatotoxic psoriasis therapies or other medical conditions, and used the most severe psoriasis patients (those who required hospital admission for their psoriasis), suggesting that the elevated mortality rates attributed to these patients may have been overstated. In summary, alcohol consumption is more prevalent in psoriasis patients, and it may also increase the severity of psoriasis. The association of alcohol with the pathogenesis and exacerbation of p­ soriasis is less clear. Prolonged alcohol abuse may lead to alcoholic liver disease, and in that way may decrease treatment responsiveness and options, which may prolong exacerbations. Patients with psoriasis are more likely than those without psoriasis to smoke. In a large cross-­sectional study from Utah, 37% of psoriatic patients acknowledged they were smokers com- 31 pared to 25% smokers in the general population [79]. Although the Finnish study of 144 psoriasis patients, mentioned above, found no association between smoking and the onset of psoriasis [73], another study of 55 women demonstrated an increased smoking rate of psoriasis patients compared to controls [80]. Naldi et al. examined 560 patients and showed that the risk for developing psoriasis was the greater in former smokers and current smokers than in those who had never smoked [81]. A hospital-based Chinese study evaluated 178 psoriasis patients and 178 controls and found a graded positive association between the risk of psoriasis and the intensity or duration of smoking [82]. Moreover, they showed that the risk of psoriasis in smokers with the HLA-Cw6 haplotype was increased by 11-fold over non-­ smokers without the HLA-Cw6 haplotype demonstrating an additive effect of genetics on that of smoking in inducing psoriasis. Gene-smoking interaction was found also by Yin et al. [83]. In a larger Italian study of 818 patients, those that smoked greater than 20 cigarettes per day were at a twofold increased risk for more severe psoriasis than those who smoked less than 10 cigarettes per day [84]. The largest and most definitive study investigating the association of smoking with psoriasis was the Nurses’ Health Study II which prospectively followed a cohort of over 78,000 US nurses over a 14 year time period. They demonstrated a “dose-response” relationship for smoking and the risk of developing incident psoriasis [85] and that the risk of incident psoriasis decreases nearly back to that of never smokers 20 years after stopping smoking. In addition, they also found that prenatal and childhood exposure to passive smoke was associated with an increased risk of psoriasis. Obesity Numerous studies demonstrate an increased prevalence of obesity, defined as a body mass index (BMI) ≥30 kg/m2, among patients with psoriasis [79, 86–90]. The findings of a systematic review and meta-analysis of multiple observational studies support an increased prevalence of obesity 32 in patients with psoriasis [90]. The pooled odds ratio [OR] for obesity for patients with psoriasis compared with a control group without psoriasis was 1.66 (95% CI 1.46–1.89). Further support for this link between psoriasis and obesity derives from a review of over 10,000 patients with moderate to severe psoriasis enrolled in clinical trials of biologic therapies [86] where the average BMI for all patients in the trials was 30.6 kg/ m2. There is also a correlation between obesity and the severity of psoriasis [79, 87, 91]. In the systematic review and meta-­analysis described above, the risk for obesity was more pronounced in patients with severe psoriasis (pooled OR 2.23, 95% CI 1.63–3.05) than patients with mild psoriasis (pooled OR 1.46, 95% CI 1.17–1.82) [91]. One study of 4065 individuals with psoriasis and 40,650 controls that was included in the meta-analysis illustrated a progressive relationship between disease severity and obesity. Among patients with mild (≤2% body surface area [BSA]), moderate (3–10% BSA), and severe psoriasis (>10% BSA), the prevalence of obesity compared to controls increased by 14, 34, and 66%, respectively [91]. Children with psoriasis also have an increased risk for obesity. In an international cross-­sectional study of 409 children with psoriasis, children with psoriasis were significantly more likely to be obese (BMI ≥95th percentile) than controls (OR 4.29, 95% CI 1.96–9.39) [92]. Similar to the general population of patients with psoriasis, a correlation between disease severity and obesity was observed. Children with severe psoriasis had a greater increase in risk for obesity than children with mild disease (OR 4.92, 95% CI 2.20–10.99 versus 3.60, 95% CI 1.56–8.30). More than one factor may contribute to the association between obesity and psoriasis. Although the negative psychosocial impact of psoriasis was initially considered the sole reason for excess weight in patients with psoriasis [79], more recent data suggest that obesity may increase risk for psoriasis. In an analysis of data collected from almost 80,000 women in the Nurses’ Health Study II, increased adiposity and weight gain were identified as strong risk factors for psoriasis [93]. Compared to women with a I. Grozdev and N. J. Korman BMI of 21.0–22.9 kg/m2 at age 18, the multivariate relative risk for the development of psoriasis for subjects with a BMI ≥30 kg/m2 at the same age was 1.73 (95% CI, 1.24–2.41), and only 0.76 (95% CI, 0.65–0.90) for women with a BMI <21 kg/m2. Data on the effects of weight loss on disease severity in psoriasis are limited. The first randomized trial designed to explore the effect of weight loss on the severity of psoriasis found a statistically non-significant trend towards greater improvement in psoriasis (as assessed by Psoriasis Area Severity Index [PASI] score) in overweight or obese psoriasis patients who were placed on a low-energy diet compared with a similar group of patients who continued to eat ordinary healthy foods [94]. The median baseline PASI score for all patients (5.4) correlated with mild to moderate psoriasis, and during the 16-week trial, patients allocated to the low-­ energy diet lost a mean of 15.8 kg compared with a mean loss of 0.4 kg in the control group. By study end, PASI scores were reduced by a mean of 2.3 in the low-energy diet group compared with only 0.3 in the control group. In addition, improvement in the Dermatology Quality of Life Index (a secondary outcome measure aimed at assessing the change in the impact of psoriasis on patient quality of life) was significantly greater in the low-energy diet group. Improvement in psoriasis following gastric bypass surgery has been previously documented [95–97]. The mechanism of this is unclear, but may be related to alterations in the production of pro-inflammatory and anti-inflammatory adipokines, or weight-loss related changes in cutaneous microflora that result in the elimination of an antigenic stimulant [98]. However, worsening of psoriasis has also occurred after weight loss and weight loss surgery [99–101]. Further study is therefore necessary to better understand the effect of weight loss on psoriasis. Obesity may impact the efficacy of some psoriasis treatments [102]. A cohort study of approximately 2400 patients receiving systemic therapy for psoriasis (including conventional and biologic agents) found that compared to individuals with a BMI of 20–24 kg/m2, obese 4 Psoriasis: Epidemiology, Potential Triggers, Disease Course subjects (BMI ≥30 kg/m2) were less likely to achieve 75% improvement in disease severity (odds ratio 0.62, 95% CI 0.49–0.79 at 16 weeks) regardless of the type of therapy [103]. In addition, in a randomized trial of 61 obese patients with moderate to severe psoriasis, weight loss improved the response to cyclosporine (2.5 mg/ kg per day) [104]. The weight-based dosing regimen for ustekinumab, a newer treatment option for psoriasis, is used to mitigate the reduction in drug efficacy observed when the standard dose is utilized in obese patients [102]. Estrogen Elevated estrogen levels may serve as a trigger for psoriasis in some patients. Reports of new onset psoriasis at puberty, psoriasis worsened by estrogen therapy, and psoriasis that may be cyclical and related to menses, all suggest an etiologic role for elevated estrogen levels [105]. However, there have also been reports of psoriasis occurring or being exacerbated at the onset of menopause, which supports the opposite interpretation. These findings demonstrate that the potential role of estrogen as a triggering factor for psoriasis is not entirely clear. Additionally, while some patients report a worsening of psoriasis during pregnancy, nearly twice as many report improvement of their psoriasis during pregnancy [106–108]. Relapse during the early postpartum period is common. The mechanism by which psoriasis tends to improve during pregnancy is not well understood. However, there is now data to suggest that the up regulation of Th2 cytokines during pregnancy counteracts the effects of proinflammatory Th1 cytokines which are key players in the pathogenesis of psoriasis [77]. Disease Course Psoriasis encompasses a spectrum of cutaneous manifestations that varies from patient to patient and even in the same patient over time. While the majority of patients have chronic plaque psoriasis throughout the typically lifelong course of their 33 disease, some patients may develop other variants including guttate, pustular, inverse or erythrodermic variants. Plaque psoriasis is usually chronic with intermittent remissions. Plaques may persist for months to years at the same locations; however, periods of complete remission do occur. Psoriatic plaques usually develop slowly over time. During exacerbations, however, plaques tend to enlarge more rapidly with an active peripheral edge of increasingly intense erythema and scale along with increases in plaque thickness. New psoriatic papules may arise in areas of normal skin surrounding the established plaques and coalesce with these to form increasingly larger plaques. Resolution of a plaque typically begins at its center. The end result of plaque clearance may be post-inflammatory hypo- or hyper-pigmentation that gradually fades giving way to normal-­appearing skin. Complete remission of psoriasis for several years followed by reoccurrence of disease can occur. Traditionally, chronic plaque psoriasis has been considered a single entity; however, recent evidence demonstrates that patients with thin and thick plaque psoriasis have differing clinical features [109]. In addition to cutaneous involvement, plaque psoriasis may be associated with internal involvement, including joints and extra-­articular sites such as the eyes. Concomitant psoriatic arthritis occurs in up to 30% of patients with cutaneous psoriasis [110]. In a minority of patients, the symptoms of psoriatic arthritis appear before skin involvement. The prevalence of ophthalmic involvement in patients with cutaneous disease is not known; however, it is thought to occur in approximately 10% of patients [111]. Psoriasis may affect almost any part of the eye, leading to blepharitis, peripheral keratopathy, acute anterior uveitis, posterior synechiae, conjunctivitis, and cataract formation. Guttate psoriasis may clear spontaneously over weeks to months. There is a tendency toward younger age of onset with elevated anti-­ streptolysin O (ASO) titer in patients with involuting course. Guttate psoriasis may become chronic and progress to plaque psoriasis, particularly in patients with a family history of psoriasis [112]. 34 Palmoplantar pustulosis, which is now considered a single entity distinct from pustular psoriasis, is a chronic disease which tends to remain localized to the palms and soles. It is significantly aggravated by extrinsic factors, such as stress, smoking, and infections. It is less responsive to standard treatment and is commonly associated with sterile inflammatory bone lesions, such as the SAPHO syndrome (synovitis, acne, pustulosis, hyperostosis, and osteitis). Localized palmoplantar psoriasis presents with sterile pustules found only on the palms and soles that may be also seen with or without evidence of classic plaque type disease. Generalized pustular psoriasis (von Zumbusch) is a severe acute form of psoriasisin which small, monomorphic sterile pustules develop in painful inflamed skin which is triggered by pregnancy, rapid withdrawal of corticosteroids, infections, and hypocalcemia. It is complicated by systemic symptoms of fever, chills, and fatigue, as well as electrolyte derangements and liver abnormalities. This variant requires aggressive treatment with systemic immunosuppressive therapy. The mortality rate of generalized pustular psoriasis due to sepsis is high without appropriately aggressive treatment [77]. References 1. Naldi L. Epidemiology of psoriasis. Curr Drug Targets Inflamm Allergy. 2004;3:121–8. 2. Lebwohl M. Psoriasis. Lancet. 2003;361:1197–204. 3. Toloza SM, Valle-Onate R, Espinoza LR. Psoriatic arthritis in South and Central America. Curr Rheumatol Rep. 2011;13(4):360–8. 4. Chandran V, Raychaudhuri SP. Geoepidemiology and environmental factors of psoriasis and psoriatic arthritis. J Autoimmun. 2010;34(3):J314–21. 5. Farber EM, Nall ML. The natural history of psoriasis in 5600 patients. Dermatologica. 1974;148:1–18. 6. Koo J. Population-based epidemiologic study of psoriasis with emphasis on quality of life assessment. Dermatol Clin. 1996;14:485–96. 7. Gelfand JM, Stern RS, Nijsten T, Feldman SR, Thomas J, Kist J, Rolstad T, Margolis DJ. The prevalence of psoriasis in African Americans: results from a population-based study. J Am Acad Dermatol. 2005; 52:23–6. 8. Shaefer T. Epidemiology of psoriasis. Review and the German perspective Dermatology. 2006;212:327–37. I. Grozdev and N. J. Korman 9. Eckes L, Ananthakrishnan R, Walter H. The geographic distribution of psoriasis [in German]. Hautarzt. 1975;26:563–7. 10. Okada S, Weatherhead E, Targoff IN, Wesley R, Miller FW. International Myositis Collaborative Study Group. Global surface ultraviolet radiation intensity may modulate the clinical and immunological expression of autoimmune muscle disease. Arthritis Rheum. 2003;48(8):2285–93. 11. Jacobson CC, Kumar S, Kimball A. Latitude and psoriasis prevalence (research letter). J Am Acad Dermatol. 2011;65(4):870–3. 12. Ouedraogo DD, Meyer O. Psoriatic arthritis in Sub-­ Saharan Africa. Joint Bone Spine. 2012;79(1):17–9. 13. Griffiths CE, Christophers E, Barker JN, Chalmers RJ, Chimenti S, Krueger GG, Leonarid C, Menter A, Ortonne JP, Fry L. A classification of psoriasis vulgaris according to phenotype. Br J Dermatol. 2007;156:258–62. 14. Buntin D, Skinner R, Rosenberg E. Onset of psoriasis at age 108. J Am Acad Dermatol. 1983;9:276–7. 15. Henseler T, Christophers E. Psoriasis of early and late onset: characterization of two types of psoriasis vulgaris. J Am Acad Dermatol. 1985;13(3):450–6. 16. Ferrandiz C, Pujol RM, Garcia-Patos V, Bordas X, Smandia JA. Psoriasis of early and late onset: a clinical and epidemiologic study from Spain. J Am Acad Dermatol. 2002;46:867–73. 17. Holgate MC. The age-of-onset of psoriasis and the relationship to parental psoriasis. Br J Dermatol. 1975;92:443–8. 18. Tollefson MM, Crowson CS, McEvoy MT, Maradit Kremers H. Incidence of psoriasis in children: a population-based study. J Am Acad Dermatol. 2010;62(6):979–87. 19. Farber EM, Nall ML. Genetics of psoriasis: twin study. In: Farber EM, Cox AJ, editors. Proceedings of the international symposium on psoriasis. Stanford: Stanford University Press; 1971. p. 7–13. 20. Melski JW, Bernhard JD, Stern RS. The Koebner (isomorphic) response in psoriasis: associations with early age of onset and multiple previous therapies. Arch Dermatol. 1983;19:655–9. 21. Weiss G, Shemer A, Trau H. The Koebner phenomenon: review of the literature. J Eur Acad Dermatol Venereol. 2002;16(3):241–8. 22. Eyre RW, Krueger GG. Response to injury of skin involved and uninvolved with psoriasis, and its relation to disease activity: Koebner and ‘reverse’ Koebner reactions. Br J Dermatol. 1982;106(2):153–9. 23. Nyfors A, Lemholt K. Psoriasis in children: a short review and a survey of 245 cases. Br J Dermatol. 1975;92:437–42. 24. Whyte HJ, Baughman RD. Acute guttate psoriasis and streptococcal infection. Arch Dermatol. 1964;89:350–6. 25. Norrlind R. The significance of infections in the origination of psoriasis. Acta Rheumatol Scand. 1955;1(2):135–44. 5 Topical Therapy I: Corticosteroids and Vitamin D Analogues Eric J. Yang and Shari R. Lipner Abstract Key Learning Objectives Psoriasis is a common disease affecting 1. To understand the mechanism of action approximately 2% of the population worldof topical steroids and vitamin D wide. Topical therapies play an integral role in analogues the treatment of patients with mild-to-­ 2. To understand the appropriate use and moderate disease. The mainstays of treatment efficacy of topical steroids and vitamin are topical corticosteroids, vitamin D anaD analogues logues, or a combination of the two. This 3. To understand the safety issues of topichapter will discuss the pharmacokinetics and cal steroids and vitamin D analogues mechanism of action of topical corticosteroids and vitamin D analogues for the treatment of psoriasis. Additionally, long-term adverse effects of these medications, vehicle selection, Topical Corticosteroids potency, and combination use with other treatment modalities will also be discussed. Topical corticosteroids (TCS) are a mainstay in treatment of a wide range of inflammatory dermatoses. There are seven classes of topical steroids ranging from superpotent (class 1) to very low-potency topical steroids (class 7). These classes have been developed based on vasoconstrictor assays [1]. These vasoconstrictor assays involve preparing the test corticosteroid in 95% alcohol and then applying it to the volar surface of a normal volunteer’s forearm. The alcohol is left to evaporate and then the test area is covered with an occlusive dressing for 16 h. Afterwards, E. J. Yang the area is washed off and vasoconstriction is Chicago Medical School, Rosalind Franklin University, North Chicago, IL, USA assessed. The vasoconstrictive assay is reproducible and correlates well with the clinical efficacy S. R. Lipner (*) Department of Dermatology, Weill Cornell Medical of TCS (Fig. 5.1). College, New York, NY, USA e-mail: shl9032@med.cornell.edu © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_5 39 E. J. Yang and S. R. Lipner 40 Fig. 5.1 Topical corticosteroid CH2OH 21 18 12 HO O 20 17 OH 11 13 19 1 16 9 14 2 8 15 3 7 5 O 4 Pharmacokinetics/Mechanism of Action Three factors determine the pharmacokinetics and potency of a topical corticosteroid: the structure of the corticosteroid molecule, the vehicle, and the skin onto which the corticosteroid is applied [2]. Hydrocortisone is the central structure of most topical corticosteroids. Variations are formed by placing hydroxyl groups onto the 11-β, 17-α, and 21 positions. Additionally, ketone groups at the 3 and 20 positions and a double bond into the 4 position of the glucocorticoid nucleus distinguish between classes. Adding or altering functional groups such as hydroxyl, hydrocarbon, ester, fluoro, chloro, acetonide or ketone groups at certain positions can vastly impact the molecule’s pharmacokinetics [2]. The alteration of hydroxyl groups modifies the molecule’s lipophilicity, solubility, percutaneous absorption and glucocorticoid receptor binding ability [2]. Glucocorticoid potency is increased by adding a double-bond at position one, or with additional fluorination or chlorination [2]. Additionally, halogenation at the 6-α or 9-α position increases glucocorticoid receptor binding activity [2]. Decreased mineralocorticoid activity, as in 6 d­examethasone, betamethasone and triamcinolone, is accomplished by the addition of a 16-α methyl, 16-β methyl, or 16-α hydroxyl group. Finally, epidermal enzymes cause de-­ esterification of topical corticosteroids into inactive metabolites. Increased potency can be accomplished by inhibiting de-esterification through halogenation at the 21 position. Vehicle The vehicle of a topical corticosteroid can influence percutaneous absorption and therapeutic efficacy. When choosing a topical steroid, one must first decide on the desired potency based on the severity and the location of the skin disease. Then, one must decide on the vehicle based on the type of lesion to be treated, need for hydration or drying effect, location and potential for irritation by components of the vehicle, and patient preference. Lotions are often preferred for the face, ointments work well for dry lesions, and gels are more useful in hairy areas or for a drying effect for wet lesions. Patients typically prefer vehicles that are quickly absorbed, non-greasy, and easy to apply [3], such as lotions and foams, but patient preferences may vary greatly [4]. 5 Topical Therapy I: Corticosteroids and Vitamin D Analogues Potent and superpotent topical steroids should be avoided on the thin skin of the face and intertriginous areas due to an increased risk of skin atrophy. The vehicle may alter the pharmacokinetics of a topical steroid molecule, thereby affecting its potency. A novel formulation for halobetasol propionate 0.01% lotion has demonstrated similar efficacy for psoriasis as compared to halobetasol 0.05% cream despite a lower drug concentration [5], due to increased drug delivery of the active ingredient by the vehicle [6]. Propylene glycol and alcohol, which are common solvents, can affect percutaneous absorption by altering the topical corticosteroid molecule’s solubility in the vehicle. Propylene glycol enhances potency through increasing penetration through the stratum corneum (Fig. 5.2). For some agents, brand-name preparations are not always equivalent to generics and may have higher or lower potency. For example, Valisone 0.1% cream (Schering) and Kenalog 0.1% cream (Westwood-Squibb) have both demonstrated increased vasoconstriction over generics [2]. In addition, Synalar 0.025% cream is also more potent than generic fluocinolone acetonide 0.025% cream (Fougera and Company) [2]. However, Aristocort 0.025% cream and Aristocort 0.05% cream (Lederle Laboratories) are significantly less potent than generic triamcinolone 0.025 and 0.05% cream (Fougera and Company) [2]. In general, generic vs. brand-name ointments tend to be closer in vasoconstrictive assays than creams. Additionally, there are differences between different generic preparations as well as different brand-name preparations of the same topical corticosteroids [7]. Bioavailability and penetration of the topical corticosteroid increase with inflamed or diseased skin, as well as with increased hydration of the stratum corneum. The thickness of the stratum corneum is inversely proportional to the degree of penetration of the topical corticosteroid. Very occlusive agents, such as ointments, increase the absorption of topical corticosteroids through increased hydration of the stratum corneum [8]. 41 Immunologic Mechanisms Topical corticosteroids are closely involved with all aspects of inflammation in the body, affecting both adaptive and innate immunity. TCS have been shown to decrease the number and function of Langerhans’ cells, which are antigen presenting cells found in the skin important in initiating immune responses. Neutrophils are decreased, less adherent to vascular endothelium and have decreased phagocytic function with corticosteroid use [9–11]. Similarly, leukocytes show decreased antibody-dependent cellular toxicity and natural killer cell function [12, 13]. In addition, the production of many cytokines is decreased including interleukin (IL)-1, IL-2, interferon (IFN)-γ, tumor necrosis factor and granulocyte-monocyte-stimulating factor [2]. Topical steroids decrease the mitotic rate of the epidermis, thereby causing thinning of the stratum corneum and granulosum and flattening of the basal layer [14]. TCS also cause atrophy of the dermis through inhibition of fibroblast proliferation, migration, chemotaxis and protein synthesis. They have also been shown to cause inhibition of fibroblast synthesis of both glycosaminoglycans and collagen [15–17]. Use in Psoriasis The antiproliferative and atrophogenic characteristics of TCS are useful in treating psoriasis. Topical corticosteroids are the mainstay of treatment and often first-line for the management of mild to moderate psoriasis, as well as for intertriginous areas and genitalia, as these areas can become irritated with the use of other topical agents [18]. In general, for the treatment of localized plaque-type psoriasis, high potency or superpotent TCS are prescribed twice daily. Optimal improvement with high potency TCS is often achieved after 2 weeks. Superpotent TCS are recommended for the treatment of nail psoriasis [19], whereas low-to-mid-potency TCS are typically used for intertriginous and genital psoriasis [18]. E. J. Yang and S. R. Lipner 42 Fig. 5.2 Diagram of steroid molecule. (Top) Clacitriol chemical structure, (Bottom) Calcipotrience chemical structure Calcitriol chemical structure 22 21 20 18 12 24 23 27 OH 25 26 17 11 16 13 9 8 15 14 7 6 5 19 4 10 3 HO 1 2 OH Calcipotriene chemical structure OH H H HO OH •H2O 5 Topical Therapy I: Corticosteroids and Vitamin D Analogues Katz and colleagues in several studies indicated the efficacy of clobetasol ointment or betamethasone dipropionate ointment in clearing plaque type psoriasis and found that remission could be maintained by applying 3.5 g three times a week [20, 21].In a placebo-controlled trial, Katz et al. demonstrated that with maintenance therapy consisting of 12 weeks of weekend-­only use of betamethasone dipropionate ointment, 74% of patients remained in remission as compared to 21% of the patients receiving placebo [22]. Occlusion can greatly increase penetration and efficacy of TCS. Studies have demonstrated that triamcinolone acetonide 0.1% ointment under occlusion is more effective than clobetasol propionate 0.05% cream twice daily or triamcinolone acetonide 0.1% ointment alone [23, 24]. Flurandrenolide (Cordran) tape is frequently prescribed due to its occlusive nature and has been shown to be superior to twice-daily diflorasone diacetate ointment in a randomized bilateral comparison study of plaque-type psoriasis [25]. Clobetasol propionate lotion applied under occlusion with a hydrocolloid dressing (Duoderm ET) once weekly also showed faster remission of psoriasis than unoccluded clobetasol propionate ointment applied twice daily [26, 27]. Foams have been found to have increased efficacy over lotions of the same class of TCS when treating the scalp [28, 29]. Combination with Other Therapies In patients with psoriasis, vitamin D analogues are frequently added at the onset, as there is a synergistic effect with TCS. Topical corticosteroids work synergistically with light therapy as well as many systemic agents. Psoriasis clears faster when using psoralen plus ultraviolet A (PUVA) with TCS versus PUVA alone. The addition of topical corticosteroids to cyclosporine therapy also leads to more rapid clearance of psoriasis [30]. Topical steroids may also be combined with other topical agents, such as tazarotene [31, 32], salicylic acid [33, 34], or anthralin [35], providing increased efficacy due to increased penetration. 43 Adverse Effects Systemic adverse effects from topical corticosteroids are uncommon and are increased with young age, liver disease, renal disease, the potency of the drug, amount of skin surface involvement, the use of occlusion, frequency of application and the duration of treatment [2]. The liver metabolizes corticosteroids and the kidneys excrete both metabolized and unmetabolized corticosteroid [36]. Infants and young children are particularly predisposed to systemic adverse effects from TCS due to a higher skin surface-to-­ body ratio, increased cutaneous permeability, and immature renal function [37]. Catch-up growth is expected when topical corticosteroids are discontinued in this population. Cushing’s syndrome and hypothalamic-pituitary-adrenal (HPA) axis suppression has been noted in patients applying high quantities of topical corticosteroids for prolonged periods of time [38–40]. Screening for HPA axis suppression is done using the 8 AM plasma cortisol level and definitive diagnosis requires the cosyntropin test. Local adverse effects are also rare but occur more frequently than systemic adverse effects. Cutaneous atrophy is the most commonly observed side effect, and is characterized by telangiectasias, striae, hypopigmented, wrinkled or shiny skin [41]. Striae are typically seen after many weeks to months of topical steroid use; risk factors include the potency of corticosteroid, location of application, occlusion, and use in infancy/childhood. “Corticosteroid phobia” is an exaggerated and often irrational fear of using topical steroids, and is common amongst patients [42], often resulting in treatment nonadherence [43]. Patients’ primary concerns often stem from TCS “thinning the skin,” but a 2011 study by Hong et al. demonstrated that appropriate long-­ term use of topical corticosteroids in children with dermatitis does not cause skin atrophy [44]. Application of TCS may also result in perioral dermatitis, characterized by erythematous papules in a periorificial distribution. Treatment for perioral dermatitis consists of an oral tetracycline in addition to a long taper with a non-fluorinated topical corticosteroid, such as hydrocortisone acetate cream. E. J. Yang and S. R. Lipner 44 Prolonged use of topical glucocorticoids on the eyelids can lead to glaucoma and cataracts, and thus is not recommended [45]. These complications may also occur in patients who apply TCS peripherally on their body, if inadvertent eye contact occurs. Allergic contact dermatitis to topical steroids may occur and can be suspected when a patient fails to respond to topical steroid therapy or flares with topical steroid therapy [46, 47]. This allergy may be to the vehicle or the actual corticosteroid molecule, which can be confirmed with patch testing. Topical corticosteroids often have a delayed reaction and persist for at least 96 h, thus requiring a delayed check [48]. Loss of clinical effect may occur with repeated application of topical corticosteroids—known as tachyphylaxis. This phenomenon occurs more commonly with higher strength topical corticosteroids, but often subsides after a rest period of a few days. There is no established regimen to prevent tachyphylaxis. A commonly recommended regimen is twice daily application of TCS for 2 weeks followed by a 1 week rest period or weekend-only application [49]. Vitamin d Analogues Structure, Biosynthesis and Mechanism of Action Vitamin D as a treatment for psoriasis was first discovered after a patient receiving oral vitamin D for osteoporosis was cured of psoriasis [50]. Calcitriol, the active form of vitamin D3, was found to inhibit the keratinocyte proliferation and modulate the keratinocyte differentiation [51]. However, the therapeutic doses of oral vitamin D3 for osteoporosis produce hypercalcemia and hypercalciuria, thus limiting its practicality for use in dermatology. As a result, topical vitamin D analogues were developed to minimize the risk of hypercalcemia, while maintaining the other beneficial cellular effects of vitamin D. There are currently four vitamin D3 analogues out in the market: calcipotriene, calcitriol, tacalcitol and maxacalcitol. The skin both synthesizes vitamin D (where 7-dehydrocholesterol is converted to vitamin D3 in the presence of ultraviolet (UV) radiation) and is a target organ for vitamin D activity. Vitamin D receptors transduce the effects of 1, 25-dihydroxyvitamin D3 and have been identified in keratinocytes, Langerhans’ cells, melanocytes, fibroblasts and endothelial cells [52]. The vitamin D receptor (VDR) is activated by binding to its ligand (1,25-dihydroxyvitamin D3) or a synthetic analogue such as calcipotriene or calcitriol. This vitamin D receptor complex, in association with the retinoid X receptor-α (RXR-α), then binds to specific DNA binding sites called Vitamin D Response Elements (VDREs), resulting in induction or repression of their target gene. In addition to inhibiting the proliferation of keratinocytes and promoting epidermal differentiation, vitamin D promotes the formation of the cornified envelope by increasing gene expression and thereby increasing levels of involucrin and transglutaminase [53]. Vitamin D also possesses anti-inflammatory benefits. It has been shown to increase levels of anti-inflammatory cytokine interleukin (IL)-10 and decrease levels of pro-inflammatory cytokine IL-8in psoriatic plaques [54]. In addition, it has been shown to inhibit the production IL-2 and IL-6 by T cells, blocks transcription of interferon (IFN)-γ and inhibits cytotoxic T cell and natural killer cell activity [55]. Calcitriol Calcitriol is the natural active form of vitamin D3. Calcium metabolism is affected by calcitriol through release of calcium from bone, decreasing parathyroid hormone, increasing tubular resorption of calcium in the kidney, and stimulating calcium transport in the intestines. Thus, if applied excessively, calcitriol may result in hypercalcemia and hypercalciuria. Calcitriol is available in an ointment form as Vectical (USA) and Silkis (Europe). 5 Topical Therapy I: Corticosteroids and Vitamin D Analogues Calcipotriene (Calcipotriol) 45 Maxacalcitol (1α,25-dihydroxy-22-oxacalcitriol) is available as Oxarol in Japan and has been shown to be 10 times more potent than calcitriol and tacalcitol in inhibiting keratinocyte ­proliferation and 60 times less calcemic than calcipotriene [56]. It has shown benefit in the treatment of psoriasis and has not posed a significant risk of hypercalcemia. roids in the treatment of psoriasis [57–59]. Calcipotriene applied twice daily has been shown to be more effective than once daily applications, without increased skin irritation [60, 61]. Calcipotriene is associated with slightly more skin irritation than topical steroids, which rarely leads to withdrawal of therapy [59]. In a study of 114 patients by Bruce et al., calcipotriene ointment was superior to fluocinonide ointment in the treatment of plaque psoriasis through 6 weeks [62]. A 2003 study of 258 psoriasis patients by Camarasa et al. in 2003 found that though betamethasone dipropionate 0.05% ointment was associated with slightly higher global improvement than calcitriol ointment, a statistically significantly higher proportion of patients remained in remission following calcitriol therapy (48%) than betamethasone therapy (25%) [63]. Calcipotriene may be used for intertriginous psoriasis, though burning and irritation are commonly encountered [18, 60]. Less frequent application and alternation with low-potency topical steroids in these areas may be less irritating. Calcipotriene is an effective and well-tolerated modality for treating scalp psoriasis, and in combination with other topical agents, may lead to improved response to treatment. In long-term studies, calcipotriene has been shown to be a safe and effective therapy for the chronic management of psoriasis. Sustained disease improvement has been documented with twice daily use for 1 year without elevation of serum calcium levels [64]. Vitamin D analogues have been shown to be effective and well-tolerated in children. In an uncontrolled pilot study of 12 children under the age of 15 with long-term follow-up of 106 weeks by Park et al., patients showed significant improvement in PASI scores compared to baseline, with no detected serious adverse effects or hypercalcemia [65]. Indication for Psoriasis Use with Other Treatment Modalities Vitamin D analogues perform as well as midpotency steroids, but less well than superpotent ste- Topical steroids are commonly used in conjunction with vitamin D analogues, as these therapies Calcipotriene is a synthetic form of calcitriol. It was the only vitamin D analogue that was available in the U.S. for many years. Its molecular structure differs slightly from calcitriol, as it contains a double bond and ring structure in its side chain which enables it to be metabolized much more rapidly. As a result, it is less likely to cause hypercalcemia than calcitriol. It is available under ointment, cream, solution, and foam forms under the trade names Dovonex (USA), Sorilux (USA), Daivonex (Europe, Asia), Psorcutan (Europe) and Dermocal (South America). Tacalcitol Tacalcitol’s (1,24(OH)2D3) structure is slightly different from calcitriol, as it contains a hydroxyl group at the 24-position rather than the 25-­position. Nevertheless, it has a similar affinity for vitamin D receptors and similar therapeutic effects. Tacalcitol is less selective than calcipotriene in its effect on calcium metabolism and has been shown to induce hypercalcemia at equivalent doses to calcitriol. It is available under ointment, cream, lotion and solution forms in Japan and under lotion and ointment forms in Europe as Curatoderm. Maxacalcitol E. J. Yang and S. R. Lipner 46 have a synergistic effect when used in combination. Combination therapy of TCS with vitamin D analogues improves the clinical response rate and minimizes the side effects of both treatments [60, 66, 67]. Topical steroids reduce or eliminate the irritation associated with calcipotriene use. Additionally, a study by Lebwohl demonstrated that patients using superpotent topical steroids on weekends and calcipotriene during the week maintained a longer remission than patients using superpotent topical steroids alone [68]. Formulations of a combination of calcipotriene and betamethasone dipropionate ointment have demonstrated greater efficacy and a more rapid onset of action compared to either medication alone [69]. This combination is highly effective for scalp psoriasis and is associated with significantly fewer side effects than with calcipotriol alone [70, 71]. The foam formulation has demonstrated superior efficacy for psoriasis as compared to the gel formulation in previous studies [72]. The calcipotriol/betamethasone propionate combination is available in the ointment (Daivobet, Taclonex), topical suspension (Taclonex), and foam (Enstilar) forms. Combining vitamin D analogues and phototherapy has also been shown to clear lesions more rapidly than either entity alone and produces a greater reduction in Psoriasis Area and Severity Index (PASI) [73, 74]. Studies combining PUVA with calcipotriene have also demonstrated increased efficacy than when using PUVA alone [75]. Total cumulative UVA exposure required for clearance of psoriasis with vitamin D analogues is reduced, thus decreasing the risk of developing skin cancer. Vitamin D analogues should be applied following phototherapy, as the application prior to UV radiation leads to ­degradation of vitamin D analogues and can alter the transmission of UV light [76]. Calcipotriene has been shown to enhance psoriasis patients’ response to both acitretin [77] and cyclosporine [78, 79], while allowing for lower dosing and less toxicity. Calcipotriene paired with methotrexate has also allowed for decreased dosing of methotrexate and increased time to relapse following the discontinuation of methotrexate [80]. Additionally, a study by Kircik d­emonstrated that the combination betamethasone dipropionate 0.064% with calcipotriene 0.005% maintains the efficacy of etanercept after a step down dose to 50 mg weekly from 50 mg twice weekly [81]. Patients with inadequate response to etanercept also demonstrated improved efficacy with combination calcipotriol and etanercept treatment [82]. Adverse Effects Vitamin D analogues are typically well tolerated, with the main side effects being application-site burning and irritation. These symptoms are more common on the thin skin of the face and in the intertriginous areas, with irritation developing in about 20% of patients treating those areas [83]. Irritation is self-limited and resolves quickly once the drug is discontinued. Patients are recommended to use less than 100 g of topical calcipotriene weekly [84], as topical calcipotriene demonstrates a dose-­ dependent suppression of serum parathyroid hormone production and 1,25 dihydroxyvitamin D3 levels [65, 85]. Patients undergoing long-term treatment exceeding 100 g of topical vitamin D analogues weekly should have their serum parathyroid hormone and vitamin D levels checked. Patients with renal disease are at increased risk of developing hypercalcemia with topical vitamin D analogue therapy, and thus may benefit from regular monitoring, even when applying less than 100 g per week. References 1. Cornell RC, Stoughton RB. Correlation of the vasoconstriction assay and clinical activity in psoriasis. Arch Dermatol. 1985;121:63–7. 2. Wolverton SE. Comprehensive dermatologic drug therapy. Philadelphia: Saunders Elsevier; 2007. p. 595–624. 3. Eastman WJ, Malahias S, Delconte J, DiBenedetti D. Assessing attributes of topical vehicles for the treatment of acne, atopic dermatitis, and plaque psoriasis. Cutis. 2014;94:46–53. 4. Hong CH, Papp KA, Lophaven KW, Skallerup P, Philipp S. Patients with psoriasis have different 6 Topical Therapy II: Retinoids, Immunomodulators, and Others Lyn C. Guenther Abstract Tazarotene is the only topical retinoid approved for treatment of psoriasis. Although it is used primarily for plaque psoriasis, it may be beneficial for palmoplantar and nail psoriasis. Use in combination with a mid- to superpotency topical steroid enhances efficacy and reduces irritancy and the atrophogenic potential of steroids. Thrice a week use with a twice weekly superpotent topical steroid may maintain improvement long-term. The once daily fixed combination lotion containing the superpotent steroid halobetasol propionate and tazarotene 0.045% (Duobrii™) is convenient, providing synergistic efficacy and unrestricted duration of use until clearance occurs. Use of tazarotene with broad band and narrow band UVB can enhance efficacy and decrease the cumulative dose of UV. Addition of tazarotene to broadband or narrowband UVB, or PUVA phototherapy enhances efficacy and decreases the total dose of ultraviolet radiation. The calcineurin inhibitors pimecrolimus and tacrolimus and phosphodiesterase-4 inhibitor crisaborole are not approved for treatment of psoriasis, although they are efficacious and L. C. Guenther (*) Division of Dermatology, Western University, The Guenther Dermatology Research Centre, London, ON, Canada e-mail: dgue@guenthermpc.com © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_6 well tolerated in the treatment of intertriginous, facial and genital psoriasis. The use of tar and anthralin has declined with the development of cosmetically elegant, efficacious treatments that do not stain the skin or clothing. Although coal tar contains many known carcinogens, use in psoriasis has not been associated with an increase in skin cancer. Addition of tar to erythemogenic UVB does not enhance efficacy. Key Learning Objectives 1. To understand the mechanism of action of topical retinoids, immunomodulators, and other topicals. 2. To understand the appropriate use and efficacy of topical retinoids, immunomodulators, and other topicals. 3. To understand the safety issues of topical retinoids, immunomodulators, and other topicals. In addition to the commonly used topical corticosteroids and vitamin D analogues, there are several other topical therapies which are used in the management of psoriasis. Tazarotene, a receptor-­selective topical retinoid, is the first and only topical retinoid to be approved to treat psoriasis. Topical tretinoin and isotreti51 L. C. Guenther 52 noin, two other ­retinoids, have been abandoned due to the variable efficacy and irritancy of tretinoin [1–5], and failure to show superior efficacy of isotretinoin compared to placebo [6]. The calcineurin inhibitors pimecrolimus and tacrolimus are commonly used off-label for intertriginous, facial, and genital psoriasis [7]. Coal tar and anthralin were once in common use, however their use has declined with the development of more cosmetically elegant, efficacious agents [8]. Tazarotene Tazarotene is available as a 0.05 and 0.1% gel and cream [9] (Fig. 6.1). Figure 6.1 also known as; Tazorac, Zorac, Avage, 118292-40-3, tazaroteno, tazarotenum, Suretin, Tazoral, AGN-190168. It is also available in a fixed combination lotion with halobetasol (Duobrii). Tazarotene (marketed as Tazorac, Avage and Zorac) is a prescription topical retinoid sold as a cream or gel. Halobetasol propionate/tazarotene lotion is a topical prescription superpotent steroid/retinoid combination. Tazarotene is approved for treatment of psoriasis, acne and sun damaged skin, while halobetasol/tazarotene is approved for treatment of plaque psoriasis in adults. Unlike other products that contain topical steroids, this combination product does not have a limitation on how long the product can be used. S C C N O O Mechanism of Action Tazarotene is a synthetic acetylenic retinoid which is a prodrug. Its free-acid active metabolite tazarotenic acid, binds to the nuclear retinoic acid receptor (RAR) β and γ, weakly to RARα, but not to retinoid X receptors (RXRs) [10]. RARγ is the predominant subtype in the epidermis [11]. RARs affect gene transcription after forming heterodimers with RXRs [12]. Tazarotenic acid cannot be converted to other retinoids since it does not contain isomerizable double bonds [10]. Tazarotene decreases inflammation and normalizes the abnormal keratinocyte hyperproliferation and differentiation seen in psoriasis [10]. The lymphocytic infiltrate in the dermis, number of HLA-DR and intracellular adhesion molecule (ICAM-1) positive cells in the epidermis and dermis, expression of epidermal growth-factor receptor (EGFR), the hyperproliferative keratins K16 and K6, skin-derived antileukoproteinase (SKALP) and macrophage migration inhibitory factor-related-protein-8 (MRP-8), keratinocyte transglutaminase type 1 (TGase K), and involucrin, are reduced, and filaggrin expression in the upper stratum spinosum and stratum granulosum increased [10, 13]. SKALP is an elastase inhibitor [14] in the suprabasal layers of psoriatic epidermis, which is not present in normal epidermis [15]. The enzyme TGase K and protein involucrin are involved in formation of the cross-linked envelope and are prematurely expressed in psoriasis [16]. Tazarotene can also induce expression of tazarotene-induced genes (TIG). TIG1 is a cell adhesion molecule which promotes cell to-cell contact and reduces keratinocyte proliferation [17]. TIG2 is not anti-proliferative, but is involved in keratinocyte differentiation [18, 19]. TIG3 regulates keratinocyte terminal differentiation and cornified envelope formation through the activation of type 1transglutaminase (TG1) [20]. Pharmacokinetics Fig. 6.1 Also known as; Tazorac, Zorac, Avage, 118292-­ 40-­ 3, tazaroteno, tazarotenum, Suretin, Tazoral, AGN-­ The half life of tazarotene is 2–18 min [10]. 190168 (Molecular Formula: C12H21NO2S. Molecular Weight: 351.46194) Tazarotene is converted via esterase metabolism 6 Topical Therapy II: Retinoids, Immunomodulators, and Others to its active metabolite tazarotenic acid which has linear pharmacokinetics and a 1–2 h elimination half-life [10]. Tazarotenic acid is then metabolized into inactive sulphoxide and sulphone metabolites and more polar conjugate metabolites [10]. Fecal elimination peaks approximately 2.5 days after dosing and is for all intensive purposes complete by 1 week [21]. Urinary excretion is virtually complete by 2–3 days [21]. Tazarotene and tazarotenic acid do not accumulate in tissues [10]. In a study of 6 patients with psoriasis, 4.54% of a 2 mg dose administered at a concentration of 2.5 μg/cm2 was absorbed into the stratum corneum, 1.38% into the epidermis, and 0.97% into the dermis; 0.43% was recovered in the feces and 0.33% in the urine [21]. The systemic absorption after an occluded 10 h 2 mg application (2.5 μg/cm2) of tazarotene 0.1% gel to the backs of 6 healthy males was 5.3% and drug half life in blood and urine, 17–18 h [22]. Application of the 0.1% gel to 20% body surface area (BSA) of healthy volunteers for 7 days resulted in a mean Cmax +/− SD of tazarotenic acid of 0.72 ± 0.58 ng/ml [23]. Low plasma concentrations of tazarotene (<0.15 ng/ml) and tazarotenic acid (0.05–6.1 ng/ml) were noted in 2.8% (2/72) and 47.2% (34/72) of patients treated with 0.05% or 0.1% gel [24, 25]. In a similar study, after 12 weeks of therapy only 1 psoriasis patient had a low concentration (0.069 ng/ml) of tazarotene, while 69.4% had detectable tazarotenic acid [26]. In two phase 3 cream studies, tazarotenic acid was found in approximately ½ of the samples, with a highest concentration of 0.874 ng/ml [27]. Toxicology In contrast to tretinoin, tazarotene and tazarotenic acid are not cytotoxic to Chinese hamster ovary cells [28]. In addition, tazarotene is not mutagenic [10]. In a 21-month mouse study, it was not carcinogenic, however, in the hairless mouse photocarcinogenicity study, similar to other retinoids, it enhanced the photocarcinogenicity associated with ultraviolet irradiation [21]. 53 Tazarotene 0.05% and 0.1% gels did not exhibit phototoxic or photoallergic potential in healthy adult Caucasians [29]. In common with other retinoids, systemic tazarotene is teratogenic [10]. Topical tazarotene is contraindicated in pregnancy [30], although in clinical trials, healthy babies were reported in all eight women who inadvertently became pregnant [31]. Clinical Studies in Plaque Psoriasis Clinical trials of tazarotene and other topicals are summarized in Table 6.1. In the two phase 2 trials, 0.01% tazarotene aqueous gel was not found to be efficacious, while 0.05 and 0.1% tazarotene gels once and twice daily showed similar efficacy with significant improvement in elevation, scaling, erythema and overall clinical severity of plaques as early as 1 week [32]. Treatment-­ related adverse effects (primarily burning, pruritus, stinging, and erythema) occurred in 30% in the first trial and 22.2% in the second trial. In the second trial, up to 50% of plaques had erythema of the surrounding skin. In a phase 3, placebo-controlled study, once daily tazarotene 0.05 and 0.1% gel for 12 weeks had similar efficacy and were superior to vehicle (p < 0.05) in all efficacy measures [24]. At the end of treatment, 59% on 0.05% tazarotene gel and 70% on the 0.1% gel had at least 50% improvement. Twelve weeks after treatment discontinuation, 52% in the 0.05% group and 41% in the 0.1% group continued to have at least 50% improvement. Treatment related adverse effects (AEs) consisted primarily of mild to moderate local irritation. A small uncontrolled study (n = 43) suggested that these AEs could be minimized without compromising efficacy, by short-­ contact application for 20 min followed by washing with water [33]. Two phase 3 placebo-­ controlled cream studies involving 1303 patients showed that tazarotene 0.05 and 0.1% creams were significantly better than vehicle with regards to overall assessment, global response to treatment, and reduction in plaque elevation and scaling [27]. One of the studies included a 12-week L. C. Guenther 54 Table 6.1 Clinical trials of tazarotene in psoriasis Study Krueger et al. (1998) [32] # Patients 45 (with 90 bilateral symmetrical plaques) Treatment 2 of: 0.01 or 0.05% tazarotene (taz) gel, or vehicle gel BID × 6 weeks Krueger et al. (1998) [32] 108 (with 216 bilateral symmetrical plaques) [31] 2 of the following: 0.05% taz gel OD or BID, 0.1% taz gel OD or BID × 8 weeks then 8 weeks follow-up Weinstein (1997) [24] 324 with 318 evaluable [24] Taz 0.05, 0.1% gel or vehicle gel OD × 12 weeks then 12 weeks follow-up Taz 0.05 and 0.01% similar and better than placebo in all efficacy measures (p < 0.05).59% on 0.05 and 70% on 0.1% had ≥50% improvement. At follow-up, 52 and 41% respectively maintained improvement. Weinstein et al. (2003) [27] 1303 in 2 studies Taz 0.05 or 0.1% cream (cr) or vehicle × 12 weeks (with 12 weeks follow-up in 1 study) Taz 0.05 and 0.1% more efficacious than vehicle. 12 weeks pooled data: 25.3% on vehicle, 41.1% on taz 0.05 and 44.9% on 0.1% taz had an overall lesional assessment ≤mild. Lebwohl et al. (1998) [26] 348 with 340 evaluable for efficacy Taz 0.05, or 0.1% gel OD OR fluocinonide 0.05% cr BID × 12 weeks with 12 weeks follow-up period. At week 12, no significant difference between taz &fluocinonide. More rapid relapse with fluocinonide after treatment discontinuation Tzung et al. (2005) [34] 23, but 19 evaluable with 44 lesion pairs Comparable efficacy at week 12 Taz: Greater maintenance of improvement Kaur et al. (2008) [35] 20 Taz 0.1% gel OD + petrolatum, or, calcipotriene 0.005% ung BID × 12 weeks with 4 weeks follow-up Left side: Taz 0.05% or 0.1% gel OD × 8 weeks right side: Calcipotriene 0.005% BID Kumar et al. (2010) [36] 30, with 27 evaluable for per protocol Taz 0.1% gel OD right side 5% crude coal tar (CCT) ung OD left side × 12 weeks. 8 weeks follow-up Efficacy 0.01% taz: Minimal efficacy. 45% on 0.05% gel had ≥75% improvement vs. 13% on vehicle. (p < 0.05) No significant differences in ≥75% improvement with Rx (range: 48% with 0.05% taz OD to 63% with 0.05% taz BID) or 8 weeks follow-up Comparable efficacy of OD taz 0.1% & BID calcipotriene. Greater efficacy of calcipotriene than OD taz 0.05% No significant difference between 2 sides (74.15% ESI reduction with taz; 77.37% with CCT, p > 0.05) Safety 33% of plaques had Rx related adverse effects (AEs), especially erythema and pruritus Rx-related AEs in 22.2% (burning, pruritus, stinging, erythema; 13% with 0.05% OD vs. 30% with 0.1% BID). Perilesional erythema in ½. Rx withdrawal due to AEs in 5.1%. Mild to moderate irritation: Pruritus (8% on vehicle, 17% on 0.05% taz, 23% on 0.1% taz), burning (6, 15, 19% respectively), erythema (1, 7, 8% respectively). Withdrawal due to AEs: 3, 10 and 12% respectively In the 2 studies, pruritus on vehicle: 12.2 and 8.9%, vs. 16.1 and 7.1% on taz 0.05%, and 29.4 and 15.6% on taz 0.1%. Taz: More burning, stinging, desquamation, skin irritation, erythema Taz: Mild to moderate pruritus, burning, erythema. Fluocinonide: Minimal irritation. Withdrawal due to AEs: 12% on taz 0.05,18% on 0.1% taz, 2% on steroid. Irritation in 35% on taz and 0% on calcipotriene No discontinuation due to AEs. No statistically significant difference in AEs between the 2 sides AEs in 48.1% on taz, but none on CCT 6 Topical Therapy II: Retinoids, Immunomodulators, and Others 55 Table 6.1 (continued) Study Lebwohl et al. (1998) [37] # Patients 300 with 284 evaluable for efficacy and 299 for safety Dubertret et al. (1998) [39] 398 Dhawan et al. (2005) [41] Green and Sadoff (2002) [42] 10 Koo and Martin (2001) [43] 73 Taz 0.1% gel OD + mometasone furoate 0.1% cr OD, or, mometasone furoate 0.1% cr BID × up to 12 weeks With 12 weeks follow-up if clear by week 4 or ≥ 50% better by week 12 Guenther et al. (2000) [44] 120 Taz 0.1% + 0.1% mometasone furoate OD or, calcipotriene 0.005% BID × 8 weeks +12 weeks follow-up if clear at week 2 or 4, or week 8 ≥ 50% better 259 with 229 evaluable Treatment Taz 0.1% gel OD + (fluocinolone acetonide 0.01% cr or mometasone furoate 0.1% cr or fluocinonide 0.05% cr or placebo [Glaxal® base]) × 12 weeks +4 weeks follow-up Taz 0.1% gel alternate evenings with: 1% hydrocortisone, 0.05% aclometasone dipropionate, betamethasone valerate 0.1%, or placebo Taz 0.1% cr OD + 0.12% betamethasone valerate foam OD × 12 weeks (open label) Taz 0.1% gel hs +/− am steroid (fluocinonide 0.05% ung, 0.1% mometasone furoate ung, 0.05% diflorasone diacetate ung, 0.05% betamethasone dipropionate cr, 0.005% fluticasone propionate ung, or 0.05% diflorasone diacetate cr) × 12 weeks Efficacy Taz + mometasone furoate 0.1% or fluocinonide 0.05% was superior to taz 0.1% + placebo after 2, 8 and 12 weeks. Similar efficacy of fluocinolone & placebo groups Safety Burning peaked at week 4 and was seen in 19% in the taz + placebo group compared to 7–15% in the taz + steroid groups Greater reduction in elevation, scaling and erythema with taz + betamethasone valerate. Median time to 50% improvement was 2 weeks vs. 4 weeks in the other groups 2 clear at week 4. No AEs 4 clear at week 8.1 patient did not have any improvement The greatest efficacy was seen with betamethasone dipropionate cr (50% mean reduction vs. 20% with monotherapy, p ≤ 0.001), followed by mometasone furoate ung (41% reduction, p ≤ 0.05) and diflorasone Diacetate ung (38% reduction, p ≤ 0.05). Maximal improvement at 8 weeks Greater, more rapid global improvement, plaque elevation and scaling with taz + steroid vs. steroid monotherapy (p ≤ 0.05 by week 4 or 8) Fewer treatment-related AEs with steroids (36% with hydrocortisone, 32% with aclometasone, 31% with betamethasone) vs. 42% with placebo At week 2, ≥75% improvement in 45% on taz + steroid vs. 26% on calcipotriene (p ≤ 0.05). Also greater reduction in BSA, elevation, scaling and erythema No AEs The mometasone furoate ung regime was best tolerated (17% incidence of drug-related AEs vs. 40% with taz monotherapy). There were no treatment-­ related withdrawals due to AEs in the mometasone furoate ung regimen vs. 18% on taz monotherapy 1 dermatitis on mometasone. Taz + steroid: 19% Rx related AEs at week 4, 17% at week 8 and 0% at week 12. Burning 11%, pruritus 11%, irritation 9%, eruption 6%, new psoriasis or exacerbation 6% Greater AEs with taz + steroid. [42% vs. 8% burning, 32% vs. 13% pruritus, 28% vs. 12% irritation, 25% vs. 7% erythema (p ≤ 0.05) for each AE] (continued) L. C. Guenther 56 Table 6.1 (continued) Study Tanghetti et al. (2000) [45] # Patients 1393 Treatment Taz 0.05% or 0.1% gel OD for up to 12 weeks either as monotherapy or in combination with other topicals (open label) Efficacy Increased efficacy with adjunctive emollient and/or corticosteroid Adjunctive mid- or high potency steroid is at least as efficacious and often superior to super potent steroid Marked reduction in scaling, elevation and overall lesional severity on both sides (p < 0.0001) with no difference between the 2 sides. More improvement of erythema with clobetasol (p < 0.01) At week 8, 52.5%, 33.3%, 18.6% and 9.7% respectively had treatment success (IGA clear/almost clear + at least a 2-grade improvement from baseline) Bowman et al. (2002) [48] 15 with 28 lesion pairs Taz 0.1% gel OD + calcipotriene 0.005% gel BID, or, clobetasol ung BID × 2 weeks, then 4 weeks follow-up (open label) Sugarman JL et al. (2017) [50] 212 Phase 2. Halobetasol propionate (HP) 0.01%/taz 0.045% lotion vs. HP vs. taz vs. vehicle OD x 8 weeks Stein Gold L et al. (2018) [51] 418 2 phase 3 studies HP/taz lotion or vehicle od X 8 weeks Lebowohl MG et al. (2019) [52] 555 HP/taz lotion daily x 8 weeks, then as needed in 4 week intervals up to 24 continuous weeks. Koo (2000) [53] 54 patients with 108 target lesions Broad band UVB 3×/week ½ body with +2/3: No topical, vehicle, or taz 0.1% gel OD × 2 weeks pre-UV, then 3×/week Time to 50% improvement reduced by ½ (25 days vs. 53 days) cumulative UVB reduced by 76% Behrens et al. (2000) [54] 10 ½ body hs taz 0.05% gel or emollient +311 nb UVB 5×/week After 4 weeks, 64% PASI reduction taz vs. 48% At week 8, 35.8% (study 1) and 45.3% (study 2) had treatment success (IGA clear/almost clear + at least a 2-grade improvement from baseline) At week 24, 20.9% discontinued treatment due to lack of efficacy Safety Increased tolerability with adjunctive steroid more AEs with monotherapy (22% at week 4) vs. 13% with mid- or high potency steroid or 12% with super-potent steroid No Rx withdrawal due to AEs no Rx-related AEs with Clobetasol Taz/calcipotriene: Asymptomatic erythema in 53%, peeling in 33%, pruritus in 7 and irritation in 7% Application site pain, pruritus, and erythema most frequent with taz (22.4%) and 0.2%HP/taz (10.2%) Comparable number of patients on HP/taz (3.4%) and vehicle (3.2%) reporting ≥1AE leading to discontinuation vs. 12.2% on taz monotherapy Contact dermatitis (6.3%), pruritus (2.2%) and pain at the application site (2.6%) were the most common treatment-related AEs 7.5% discontinued due to treatment-emergent AEs: Dermatitis (7 people), pruritus (7 people) and pain (6 people), being the most common Taz + UVB: No photosensitivity or phototoxicity 16.7% had Rx-related AEs (irritation, burning and pruritus) Mild irritation with taz, but no phototoxicity 6 Topical Therapy II: Retinoids, Immunomodulators, and Others 57 Table 6.1 (continued) Study Stege et al. (1998) [55] Dayal et al. (2018) [56] # Patients 20 Behrens et al. (1999) [58] 12 ½ body taz 0.05% gel or vehicle + bath PUVA 4×/ week Tzaneva et al. (2002) [59] 31 Oral PUVA 4 × weeks +0.1% taz gel (0.05% if not tolerated) 1 lesion 1 side, tacalcitol ung 1 lesion opposite side 30 Treatment 0.1% taz gel or 5% salicylic acid 1 week before & during 3 weeks narrow band (nb) UVB 2 target plaques on right and left sides of body. Right side treated with taz 0.05% gel + nbUVB BIW; left side with nb UVB BIW follow-up phase; treatment response was generally maintained after the drug was discontinued. In this study, an overall lesional score of mild or better was noted at the end of the 12 weeks treatment period and 12 weeks follow-up period in 24.4 and 21.8% respectively on vehicle, 41.7 and 33.4% respectively on 0.05% tazarotene, and 39.4 and 30.3% respectively on tazarotene 0.1% cream. The skin-associated treatment-related AEs including pruritus, burning, erythema, skin irritation, stinging and desquamation were more common in the tazarotene arms, particularly the 0.1% arm. In a steroid comparison study, after 12 weeks of therapy, tazarotene 0.05 and 0.1% gels had comparable efficacy to fluocinonide 0.05% cream [26]. However, the psoriasis returned faster in the steroid group. In those patients who achieved an overall lesional score of mild or better at the end of treatment, relapse to a score of moderate or worse was noted at the end of the 12 weeks follow up period in 18% in the 0.1% arm, 37% in the 0.05% tazarotene arm, and 55% in the fluocinonide arm (p < 0.05% between Efficacy Significantly faster and greater efficacy in ½ body treated with taz Safety At week 12, target plaque score reduced by 99.64% and 84.92% respectively Complete clearance in 96.7% and 6.7% respectively 16.4 +/− 2.8 and 23.33 +/− 1.2 sessions respectively for clearance Faster and greater efficacy with taz. At 3 weeks, 76.5% median PASI reduction vs. 58.5%. (p < 0.05) Similar efficacy with taz and tacalcitol. Compared to PUVA mono-therapy, the cumulative UVA was less with taz or tacalcitol (p < 0.01) Lesional irritation 6.67%, burning 3.33%, pruritus 6.67% with taz No statistically significant difference between the 2 sides (p = 0.352) No photo-toxicity. With taz, mild irritation (transient burning and erythema) Tacalcitol: 1 mild irritant dermatitis, 1 hypertrichosis Taz: 7 had AEs (dryness, irritant dermatitis, pruritus, burning); resolved after changed to 0.05% tazarotene 0.1% gel and fluocinonide). A small right/left study showed similar efficacy of tazarotene 0.1% gel once daily + emollient once daily and calcipotriene BID, and however tazarotene was more irritating, but had a better maintenance effect after treatment discontinuation (overall severity p = 0.007, erythema p = 0.01, scaling p < 0.001, elevation p < 0.001) [34]. In a small open label right/left pilot study of 20 patients, twice daily calcipotriene was also found to be comparable to once daily 0.1% tazarotene, but was more efficacious than once daily 0.05% tazarotene [35]. A small (n = 30) open-label right/ left study showed that tazarotene and 5% crude coal tar ointment had similar efficacy as measured by erythema, scaling and induration (ESI) [36]. Addition of a mid-potency corticosteroid (mometasone furoate 0.1% cream) or high potency cream (fluocinonide 0.05% cream) improved efficacy and reduced adverse effects [37]. At least 50% improvement was noted in 91% and 95% in the mid- and high-potency ste- 58 L. C. Guenther roid arms respectively compared to 80% in the severity [46]. In another similar subset study placebo arm. The cumulative rates of burning involving 246 patients switched from calcipotriwere only 61% as frequent in the mid-potency ene + steroid at baseline to tazarotene + steroid, steroid arm (14%), and 52% as frequent in the 75% achieved at least 50% global improvement high-potency steroid arm (12%) compared to the at the final visit (up to 12 weeks) [47]. A small placebo arm (23% rate). Addition of a low-­ open-label right/left comparison pilot study potency steroid (fluocinolone acetonide 0.01%) (n = 15) showed similar efficacy of tazarotene + had minimal additive benefit. Tazarotene can also calcipotriene ointment, and the superpotent stereduce the development of steroid induced epi- roid clobetasol ointment [48]. dermal atrophy. In a 4-week long study of healthy Tazarotene can be used to maintain improvevolunteers, epidermal thickness was reduced by ment. After a 6 weeks open-label treatment phase 43% with diflorasone diacetate ointment mono- with tazarotene 0.1% gel + clobetasol propionate therapy vs. 28% when used in combination with 0.05% ointment, a double-blind 5 months maintazarotene 0.1% gel (p ≤ 0.003) [38]. Another tenance phase showed that those on tazarotene study showed that combination therapy with taz- Monday, Wednesday, Friday + clobetasol arotene 0.1% gel used alternate evenings with Tuesday, Thursday maintained 75% global betamethasone valerate 0.1% enhanced efficacy improvement (p ≤ 0.001 vs. vehicle group, and tolerance [39, 40]. The foam formulation of p ≤ 0.05% vs. tazarotene/vehicle), those on tazbetamethasone valerate (0.12%) was studied in a arotene Monday, Wednesday, Friday + vehicle small open-label study involving 10 patients; all Tuesday, Thursday 50% improvement, and those but 1 had improvement and 4 were clear at week on vehicle Monday, Wednesday, Friday + white 8 [41]. In an effort to determine the optimal ste- petrolatum Tuesday, Thursday, 25% improveroid to use with tazarotene, tazarotene monother- ment [49]. apy was compared to combination therapy with 3 A fixed combination of halobetasol propiodifferent high-potency and 3 different mid-to-­ nate (HP) 0.01% and tazarotene (taz) 0.045% high-potency steroids [42]. Greatest efficacy was lotion had greater treatment success (i.e. seen when tazarotene 0.1% gel was used in com- Investigator Global Assesssment (IGA) of clear/ bination with betamethasone dipropionate 0.05% almost clear with at least a 2-grade improvement cream, followed by mometasone furoate 0.1% from baseline) at week 8 (52.5%) vs. HP (33.3%, ung and diflorasone diacetate 0.05% ung. The p = 0.033), taz (18.6%, p < 0.001), and Vehicle best tolerated regimen was the tazarotene + (9.7%, p < 0.001) in a phase 2, multicenter, mometasone furoate 0.1% ung, making this regi- double-­blind study [50]. In two phase 3 trials, by men the one with the optimal balance of efficacy week 8, treatment success occurred in 35.8% and tolerability. Tazarotene 0.1% gel in combina- (study 1) and 45.3% (study 2) compared with tion with mometasone furoate 0.1% cream once 7.0% and 12.5% on Vehicle (p < 0.001) [51]. A daily was shown to be more efficacious than rapid onset of action was noted in both studies; twice daily mometasone furoate cream [43] and by 2 weeks, HP/Taz lotion was statistically sigtwice daily calcipotriene 0.005% ointment [44]. nificantly superior to vehicle [50, 51]. In a 1-year A large (n = 1393) open-label effectiveness open label study (n = 555) with a focus on safety, study also showed that mid- to high potency ste- HP/taz lotion was applied daily for 8 weeks, then roids were optimal potencies to be used in combi- as needed [52]. Those without an IgA of clear/ nation with tazarotene, and that superpotent almost clear at week 8 were treated for additional steroids were not superior [45]. A subset of 166 4 weeks. At week 12, those with no improvement patients were switched from calcipotriene +/− a in IGA from baseline were discontinued (4.7%). steroid to tazarotene + a steroid. There was a sub- For the remainder of the study, patients were stantial improvement in efficacy and patient sat- treated for 4 weeks with HP/taz if they had an isfaction of these patients with 71% having at IGA of 2 or more at a visit, to a maximum of least a 1 grade improvement in overall psoriasis 24 weeks continuous treatment. At week 24, 6 Topical Therapy II: Retinoids, Immunomodulators, and Others 20.9% discontinued treatment due to lack of efficacy. The rate of adverse events was similar to that in the pivotal trials, peaked at day 60 and remained stable from day 90 until the end of the study. Treatment-emergent AEs resulted in discontinuation of 7.5% of study participants. Addition of tazarotene to phototherapy can enhance efficacy and decrease he total dose of ultraviolet (UV) radiation. Daily pre-treatment with 0.1% tazarotene gel for 2 weeks, then three times a week immediately after broad band UVB treatment, increased the rapidity of improvement and overall efficacy [53]. The time to reach 50% improvement decreased from 53 days to 25 days with an associated 76% reduction in median cumulative UVB exposure (390 vs. 1644 mJ/ cm2). The time to reach 75% improvement was 28 days earlier. By day 81, 75% improvement or better occurred in 50% on UVB monotherapy versus 82% on UVB + tazarotene 0.1% gel. No treatment related photosensitivity or phototoxicity were noted. Similar results were seen with narrow-band (nb) UVB. In a small study (n = 10), after 4 weeks of 5×/week nb UVB, the psoriasis area and severity index (PASI) score decreased from 18.3 to 6.5 (95% confidence interval (CI) 5.29–7.91) with the addition of tazarotene 0.05% gel hs, compared to 9.5 (95% CI 7.70–11.70) with emollient (p < 0.05) [54]. In a half body study involving 20 patients, tazarotene in combination with nb UVB was more efficacious than 5% salicylic acid with nb UVB [55]. In an open-­ label trial (n = 30), a target plaque was picked from similar sites on each side of the body [56]. Both sides were treated with nb UVB, but the right side was also treated with topical tazarotene 0.05% gel. After 12 weeks of therapy, the mean target plaque was reduced by 99.64% in the taz + nb UVB side vs. 84.92 on the nb UVB monotherapy side. Complete clearance was achieved at the end of 12 weeks in 96.67% vs. 6.67%. Fewer treatment sessions were required for clearance on the tazarotene side (16.40 =/−2.799 vs. 23. 33 =/−1.212. Tazarotene nb UVB combination therapy was found to be similar to calcipotriene nb UVB combination therapy in a study of 10 patients [57]. 59 Psoralen ultraviolet A (PUVA) studies have also shown added benefit with topical tazarotene. In a ½ body bath PUVA study (n = 12), the side treated with tazarotene improved faster and to a greater extent [58]. After 3 weeks, the median PASI reduction was 76.5% (95% confidence interval (CI) 65–86) compared to 58.5% (95% CI 50–69). No phototoxic effects were seen. In an oral PUVA study (n = 31) addition of tazarotene gel or tacalcitol ointment resulted in faster clearing and 14 rather than 16 PUVA exposures [59]. linical Studies in Other Types C of Psoriasis (Palmoplantar, Nail) Tazarotene is efficacious in the treatment of palmoplantar and nail psoriasis. In a 12-week randomized trial with 30 patients with palmoplantar psoriasis randomized to once daily 0.1% tazarotene cream or 0.05% clobetasol propionate cream, there was no significant difference in the reduction in the erythema scaling fissures and induration (ESFI) score between the two groups (83.2 and 89.1% respectively) and complete clearance (52.9 and 61.5% respectively) [60]. A double-blind, vehicle-controlled 24-week study (n = 31) showed greater reduction in onycholysis (p ≤ 0.05% weeks 4 and 12) and pitting (p ≤ 0.05 at week 24) in occluded and nonoccluded tazarotene 0.1% gel treated nails [61]. An open-label study (n = 35) showed fingernail improvement after only 4 weeks with nonoccluded tazarotene 0.1% gel applied to the nail plates, nail folds and periungual skin [62]. Hyperkeratosis and oil drop changes responded faster; pitting was the most persistent. After 12 weeks of treatment of fingernails and toenails, the mean visual assessment score for onycholysis decreased from 26 to 2, hyperkeratosis 25 to 2, oil spots 18 to 3, and pitting 13 to 1 (p < 0.0001 for each change). A case report in a 6-year-old child showed that 8 weeks treatment with nonoccluded 0.05% tazarotene gel improved nail psoriasis, especially hyperkeratosis [63]. Occluded tazarotene 0.1% cream and clobetasol propionate 0.05% cream had similar efficacy in a double-­ L. C. Guenther 60 blind study of 46 patients with nail psoriasis [64]. Both treatments showed significant improvement in onycholysis, hyperkeratosis, salmon patches and pitting. Application Tips Since tazarotene is photostable, it can be applied at any time of day [65]. When used in combination with a topical steroid, both compounds can be used at the same time of day without adversely affecting each other’s stability [66]. The gel and cream formulations rub in well and do not stain. Only a small quantity is needed; larger amounts can increase the risk of irritation [67]. A cotton-­ tipped applicator to apply tazarotene and application of moisturizer around psoriatic lesions can minimize perilesional irritation from inadvertent application to unaffected skin. The gel and cream should be dry before clothes are worn to minimize spread onto unaffected skin. The 0.05% formulation should be considered for individuals with sensitive skin [67]. If irritation should occur, use of the 0.05% cream formulation rather than the gel, alternate day treatment, and short contact treatment for as little as 5 min followed by a corticosteroid or emollient immediately after the tazarotene has been washed off, should be considered [67]. opical Calcineurin Inhibitors T (Immunomodulators) Pimecrolimus and Tacrolimus Topical pimecrolimus is available as a 1% cream and tacrolimus as a 0.03 and 0.1% ointment. They were approved by the FDA in 2001 and 2000 respectively [68]. Pimecrolimus is a derivative of the ascomycin macrolactam while tacrolimus was isolated from the bacterial strain Streptomyces tsukubaensis [68]. They are indicated to treat atopic dermatitis. Although they are not FDA approved for the treatment of psoriasis, they are efficacious in the treatment of intertriginous, facial and genital psoriasis [69]. (Fig. 6.2). A macrolide isolated from the culture broth of a strain of Streptomyces tsukabaensis that has strong immunosuppressive activity in vivo and prevents the activation of T-lymphocytes in response to antigenic or mitogenic stimulation in vitro. Mechanism of Action After binding to macrophilin-12, topical calcineurin inhibitors inhibit the calcium dependent phosphatase calcineurin, which in turn results in inhibition of translocation of nuclear factor of activated T cells (NFAT) and down regulation of cytokine synthesis [7]. Toxicity Application site burning is the most frequent adverse drug reaction [7]. The FDA issued a controversial lymphoma “black box” warning in March 2006 [68]. This warning noted that, although a causal relationship has not been ­established, rare cases of malignancy (e.g., skin and lymphoma) have been reported in patients treated with topical calcineurin inhibitors. A study of 293,253 patients with atopic dermatitis, did not find an increased risk of lymphoma associated with use of calcineurin inhibitors [70]. The main factor associated with an increase risk of lymphoma was disease severity [70]. A 2015 Cochrane review of tacrolimus found no evidence to support a possible increased risk of cancer [71]. In contrast to topical steroids, calcineurin inhibitors are not atrophogenic [72]. Tacrolimus has been shown to increase collagen synthesis [73] and pimecrolimus to reverse skin atrophy [74]. Clinical Studies in Psoriasis In the treatment of plaque psoriasis, non-occluded tacrolimus was effective in two small studies 6 Topical Therapy II: Retinoids, Immunomodulators, and Others Fig. 6.2 Also known as: TACROLIMUS MONOHYDRATE, Protopic (TN), 109581-93-3, FK-506 monohydrate, Tacrolimus (USAN/ INN), Tacrolimus hydrate (JP16), F4679_ SIGMA (Molecular Formula: C44H71NO13; Molecular Weight: 822.03344) 61 H H O H O O H H O O H H O O H N O O O O O H O (one monotherapy [75] and one with concurrent 6% salicylic acid gel [76]) but not in another [77], while occluded tacrolimus [75, 78] and pimecrolimus [79] were. Studies in inverse psoriasis are summarized in Table 6.2. In one study (n = 57), after 2 weeks of 1% pimecrolimus, 71.4% were clear/almost clear vs. 20.7% on vehicle (p < 0.0001) [80]. Only 1 patient on pimecrolimus had a treatment-related AE (paresthesia). However a 4-week study did not show superiority of pimecrolimus over vehicle [81]. In a small study (n = 15), the mean total score for erythema, scaling and thickness on the face, genitalia and intertriginous area decreased from 6.88 at baseline to 0.37 after 60 days of tacrolimus 0.1% oint- ment. Two patients experienced a warm sensation in facial lesions after application. In an 8-week open label study of 21 patients with intertriginous and/or facial psoriasis treated with 0.1% tacrolimus, all patients had at least 75% improvement and 81% were clear [82]. In a study of 167 patients with intertriginous and/or facial psoriasis, by day eight 24.8% treated with 0.1% tacrolimus vs. 6% on vehicle were at least 90% better (p = 0.004); by 8 weeks, the numbers had risen to 66.7 and 36.8% respectively (p < 0.0001) [83]. The face and intertriginous areas showed similar improvement (42% with facial and 48% with intertriginous lesions were clear) [84]. Similar efficacy (47.6% clear) was seen in an open label L. C. Guenther 62 Table 6.2 Studies of calcineurin inhibitors in inverse psoriasis Study Gribetz et al. (2004) [80] # Patients 57 Treatment Pimecrolimus (P) 1% cream or vehicle (V) BID × 8 weeks Kreuter et al. (2006) [81] 80 P 1% cream or calcipotriene (C) 0.005% or 0.1% betamethasone valerate (B) or vehicle (V) × 4 weeks Matyin Rzquerra et al. (2006) [75] 15 0.1% tac ung × 60 days (open label) Freeman et al. (2003) [82] Lebwohl et al. (2004, 2005) [83, 84] 21 intertriginous and/or facial psoriasis (2/21 face only) 104 intertriginous 167 with intertriginous and/or facial psoriasis 0.1% tac (tac) × 8 weeks (open label) Steele et al. (2005) [86] 13 children 12 with 0.1% tac 1 with 0.03% tac (retrospective review) Brune et al. (2007) [87] 8 children with intertriginous/11 with face and/or intertriginous psoriasis 0.1% tac BID × 180 days (open label) 0.1% tac ung or V BID × 8 weeks facial study (n = 21), although 5/10 with complete clearing had recurrences during 1 month of follow-up [85]. Adverse effects of tacrolimus. were not significantly different than in the placebo arm [83]. Studies in children have also shown efficacy of tacrolimus in facial and intertriginous psoriasis [86, 87]. In one open label study, marked improvement was noted in all 10 patients with long-standing genital and facial psoriasis after 1 week of 0.1% tacrolimus ointment [88], and in another one (n = 12), male gen- Efficacy Clear/almost clear: Day 3: 14.3% on P, 0% on V, p = 0.0477 Week 8: 71.4% on P, 20.7% on V, p < 0.0001 Mean M-PASI score reduction: B: 86.4%, C: 62.4%, P: 39.7%, V: 21.1%. No significant difference between B and C, P and C, and P and V. B better than P, p < 0.05 and V, p < 0.01. C better than V, p < 0.01. Erythema decreased from 2.9 at baseline to 0.19, infiltration from 2.1 to 0.11 and desquamation from 1.8 to 0.07. (for each, p < 0.001) N.B. includes face + genital psoriasis Complete clearing in 81% and 75–99% clearing in 19% (N.B. includes 2 pts. with only facial lesions) 48% with intertriginous psoriasis on tac were clear vs. 14% on V (p < 0.001) 12/12 on 0.1% tac had complete clearance within 2 weeks No improvement in 1 patient on 0.03% but difficulty with adherence Reduction in overall severity incl. Face (1.63 to 0.71, p < 0.0001) in the 8 who completed Safety 1 mild application site paresthesia with P 1 moderate tenderness with V. no withdrawal due to AEs P: 5/20 mild itching and burning C: 2/20 increased erythema, warmth, irritation B: No AEs V: 1/20 herpes genitalis 2 had a transient warm sensation 2 had itching and a feeling of warmth Burning in 8% on tac, 7.3% on V, hyperesthesia in 4.5% on tac and 0% on V, itching in 7.1% on tac and 1.8% on V. all NS 1 on 0.03% had burning and irritation Pruritus in 1 ital PASI decreased from 15.8 to 1.2 (p < 0.001) after 8 weeks of twice daily tacrolimus 0.1% ointment [89]. Success in male genital psoriasis has also been reported with pimecrolimus [90] Crisaborole 2% Ointment Crisaborole 2% ointment is a phosphodiesterase­4 inhibitor approved for the treatment of mild-to-­ moderate atopic dermatitis. It is not approved for 6 Topical Therapy II: Retinoids, Immunomodulators, and Others treatment of psoriasis. In a double-blind vehicle-­ controlled study involving 21 individuals with intertriginous, anogenital, or facial psoriasis, after 4 weeks of therapy, the crisaborole arm had 66% improvement in the Target Lesion Severity Scale (TLSS) compared to 9% improvement with vehicle [91]. There were no application site reactions, atrophy or telangiectasia [91]. Tar Although the German Guidelines [7] do not recommend tar for psoriasis treatment and describe it as “obsolete,” tar is still widely used in many parts of the world [92]. Tar has been used for more than 2000 years and became standard treatment with ultraviolet B phototherapy after Goeckerman’s report in 1925 [92]. There are 3 types of tar, coal tar, wood tar (pine, beech, birch, juniper), and shale (ichthammols, bituminous tars) [93]. Coal tar comes from coal distillation in the manufacture of coke and contains approximately 10,000 compounds including polycyclic aromatic hydrocarbons such as benzenes, naphthalenes, creosoles and phenols [94]. The temperature of the distillation and type of coal used affect the final composition [95]. Liquor Carbonis Detergens (LCD) is an alcoholic extract of coal tar that can be mixed in cream, ointment or lotions bases, usually in a concentration of 5–15% [94]. Mechanism of Action Tar inhibits DNA synthesis which in turn results in decreased epidermal proliferation [92]. It is also said to be vasoconstrictive, antifungal, antiparasitic, and antipruritic [92]. Toxicity Coal tar is malodorous, can stain hair and fabric and unlike wood and shale tars, is photosensitizing at wavelengths of 330–550 nm [892. It can also cause burning, stinging (“tar smarts”), fol- 63 liculitis, acneiform eruptions, irritation, allergic contact dermatitis, erythroderma, tar keratoses and keratoacanthomas [92]. Acute tar intoxication may occur if tar is ingested, or if large amounts of tar are absorbed after topical application; erythrodermic psoriasis patients and young children are at a higher risk [92]. Carcinogenicity concerns arose after Sir Percival Pott, a London surgeon, linked the increased risk of scrotal cancer in chimney sweeps to soot [96], however there was no increase in the expected rate of skin cancer a study of 719 patients with psoriasis [97] and a 25-year follow-up study of 280 patients treated with crude coal tar and UVB [98]. Clinical Studies in Psoriasis Tar extract in oil was superior to the oil base in 5 subjects [99], however coal tar was significantly less effective than betamethasone valerate (38% mean PASI reduction vs. 69%) in another study [100]. Concentrations of 1 and 6% crude coal tar have similar efficacy when used with UVB [101]. Daily UVB and tar ointment three times a day for an average of 20 days yielded good to excellent improvement in 95% of 123 patients with remission rates of 2 months to 8 years (average 1.7 years) [102]. In a similar study, 60% of patients were still in remission 2 years after treatment [103]. With erythemogenic doses of UVB, a number of studies failed to show increased efficacy with tar compared to petrolatum [104–108]. Studies with suberythemogenic UVB showed less UVB energy and fewer side effects with tar oil vs. emollients in one study [109], faster improvement with 1% crude coal tar in petrolatum vs. petrolatum (22%/week vs. 14.7%/week, p < 0.0005) and 5% tar extract in oil vs. oil base (19.6%/week vs. 11.4%/week, p < 0.0005) in another [107], and no benefit in another [110]. Menkes et al. showed similar efficacy of suberythemogenic UVB with tar oil, and maximally erythemogenic UVB with emollients in a study of 49 patients with psoriasis, however the total UVB dose to clearing was 44% less in the suberythemogenic UVB with tar group [111]. L. C. Guenther 64 Anthralin Anthralin (1,8-dihydroxyanthrone) was first synthesized in 1916 by Galewsky in Germany, after chysarobine, a related compound from Goa powder produced from the araroba tree in Brazil, was noted to be an effective treatment for psoriasis [112]. In the 1930s Ingram used anthralin in Lassar’s paste (petrolatum, salicylic acid, zinc oxide, starch) with tar baths and UVB to treat psoriasis [112]. The Ingram regimen was in common use for several decades, but is now rarely used. Anthralin has also been formulated as an ointment [112], gel [113] and in a cream base which washes off easily and has minimal staining [114]. Commercial formulations are no longer available in Canada due to the development of efficacious, convenient, more cosmetically acceptable alternatives. Mechanism of Action In vitro, anthralin has been shown to inhibit DNA replication and DNA repair synthesis [115], interfere with mitochondria [116], decrease keratinocyte transforming growth factor-α expression and epidermal growth factor (EGF) receptor binding [117], inhibit leukotriene production by neutrophils [118], and inhibit monocyte secretion of interleukin (IL)-6, IL-8 and tumour necrosis factor (TNF)-α [119]. Toxicity Adverse effects include staining of skin, nails and clothing due to oxidation of anthralin, burning, and irritant and allergic contact dermatitis [112]. In order to minimize irritation, care should be taken to avoid application to uninvolved skin and intertriginous areas should not be treated. Clinical Studies The benefits of the Ingram regime in 2120 patients were reported by Maclennan and Hellier in 1961; 95% were clear in a mean time of 15.2 days as inpatients and 19.5 days as outpatients [120]. Average clearance times in other inpatient series have varied from 11.0 to 20 days [112]. Short contact anthralin ointment with a 10 min to 1 h contact time was tried in an effort to minimize staining and irritation [121]. It was as effective as conventional treatment with anthralin ointment [122], difluorosone acetate [122], and twice daily calcipotriene [123]. If anthralin is considered in the outpatient setting, short contact application (20–30 min) with an initial 1% concentration, increasing the concentration as tolerated, has been recommended [69]. Moisturizers and Keratolytics Moisturizers and keratolytics such as salicylic acid and urea are considered ‘Basic Therapy’ of psoriasis [7], although there are no placebo-­ controlled studies supporting their use. Moisturizers decrease scaling, limit painful fissuring and are anti-pruritic. Salicylic acid should be not used to large areas since systemic toxicity might occur, especially if applied to >20% of the body surface, or to patients with impaired renal or hepatic f­ unction [69]. Conclusions Although tazarotene is efficacious as monotherapy, it is more commonly used in combination with a topical steroid to enhance tazarotene’s tolerance, increase efficacy and decrease steroids’ atrophogenic potential [50, 124]. The once daily lotion containing the superpotent steroid halobetasol propionate 0.01% and tazarotene 0.045% (Duobrii™) is convenient with greater efficacy and fewer AEs than tazarotene monotherapy. There is no duration limitation in the U.S. labeling, although treatment should be stopped when the skin is clear. Use of tazarotene with broad band and narrow band UVB can enhance efficacy and decrease the cumulative dose of UV. Although phototoxicity was not noted in clinical studies, if tazarotene is added to ongoing phototherapy, it 6 Topical Therapy II: Retinoids, Immunomodulators, and Others might be prudent to reduce the UVB dose by 30–50% or UVA dose by 2 J/cm2 once scaling and induration are reduced, since one study showed that application of tazarotene for 2 weeks prior to phototesting significantly decreased the average UVB MED from 56.25 to 42.5 mJ/cm2 (p < 0.01) [125]. Palmoplantar and nail disease, particularly nail bed disease, also respond to tazarotene. The topical calcineurin inhibitors particularly tacrolimus, and phosphodiesterase-4 inhibitor crisaborole do not cause atrophy and are efficacious and well-tolerated in the treatment of inverse, facial and genital psoriasis. The use of tar and anthralin has declined with the introduction of more cosmetically elegant, efficacious products. Several studies have questioned the efficacy of tar; there does not appear to be any benefit of adding tar to erythemogenic UVB. References 1. Frost PF, Weinstein GD. Topical administration of vitamin a acid for ichthyosiform dermatoses and psoriasis. JAMA. 1969;206:1863–8. 2. Fredriksson T. Antipsoriatic activity of retinoic acid (vitamin a acid). Dermatologica. 1971;142:133–6. 3. MacDonald A, McMinn RM, Fry L. Effect of retinoic acid in psoriasis. II. Long-term study. Br J Dermatol. 1972;87:256–60. 4. Orfanos CE, Schmidt HW, Mahrle G, et al. Retinoic acid in psoriasis. Its value for topical therapy with and without corticosteroid. Clinical, histological, and electron microscopical studies on forty-four hospitalized patients with extensive psoriasis. Br J Dermatol. 1973;88:167–82. 5. Günther S. The therapeutic value of retinoic acid in chronic, acute, guttate, and erythrodermic psoriasis. Clinical observations on twenty-five patients. Br J Dermatol. 1973;89:515–7. 6. Bischoff R, De Jong EM, Rulo HF, et al. Topical application of 13-cis-retinoic acid in the treatment of chronic plaque psoriasis. Clin Exp Dermatol. 1992;17:9–12. 7. Nast A, Kopp I, Augustin M, et al. German evidence-­ based guidelines for the treatment of Psoriasis vulgaris (short version). Arch Dermatol Res. 2007;299:111–38. 8. Murphy G, Reich K. In touch with psoriasis: topical treatments and current guidelines. J Eur Acad Dermatol Venereol. 2011;25(Suppl 4):3–8. 65 9. Guenther LC. Tazarotene, a receptor-selective topical retinoid, in the treatment of psoriasis. Today’s Ther Trends. 1999;17(2):133–45. 10. Chandraratna RA. Tazarotene—first of a new generation of receptor-selective retinoids. Br J Dermatol. 1996;135(Suppl 49):18–25. 11. Elder JT, Fisher GJ, Zhang Q-Y, et al. Retinoic acid receptor gene expression in human skin. J Invest Dermatol. 1991;96:425–33. 12. Pfahl M. Nuclear retinoid receptors as central regulators of retinoid response pathways. In: Burgdorf WHC, Katz SI, editors. Dermatology progress and perspectives. New York: Parthenon; 1993. p. 155–9. 13. Esgleyes-Ribot T, Chandraratna RA, Lew-Kaya D, et al. Response of psoriasis to a new topical retinoid AGN 190168. J Am Acad Dermatol. 1994;30:581–90. 14. Molhuizen HO, Alkemade HA, Zeeuwen PL, et al. SKALP/elafin: an elastase inhibitor from cultured human keratinocytes. J Biol Chem. 1993;268:1228–32. 15. Schalkwijk J, van Vlijmen IM, Alkemade JA, de Jongh GJ. Immunohistochemical localization of SKALP/elafin in psoriatic epidermis. J Invest Dermatol. 1993;100:390–3. 16. Bernard BA, Asselineau D, Schaffar-Deshayes L, Darmon MY. Abnormal sequence of expression of differentiation markers in psoriatic epidermis: inversion of two steps in the differentiation program? J Invest Dermatol. 1988;90:801–5. 17. Nagpal S, Patel S, Asano AT, et al. Tazarotene-­ induced gene 1 (TIG1), a novel retinoic acid receptorresponsive gene in skin. J Invest Dermatol. 1996;106:269–74. 18. Nagpal S, Patel S, Jacobe H, et al. Tazarotene-­ induced gene 2 (TIG2), a novel retinoid-responsive gene in skin. J Invest Dermatol. 1997;109:91–5. 19. Zheng Y, Luo SJ, Zeng WH, Peng ZH, Tan SS, Xi YP. Alteration of tazarotene induced gene-2 expression in psoriasis vulgaris. Nan Fang Yi Ke Da Xue Xue Bao. 2008;28(10):1792–4. 20. Eckert RL, Sturniolo MT, Rans R, et al. TIG3: a regulator of type 1 transglutaminase activity in epidermis. Amino Acids. 2009;36(4):739–46. 21. Marks R. Pharmacokinetics and safety review of tazarotene. J Am Acad Dermatol. 1998;39(4):S134–8. 22. Franz TJ, Lehman PA, Franz S, et al. Percutaneous absorption of AGN 190168, a new synthetic retinoid, through human skin in-vivo (Abstr.). J Invest Dermatol. 1992;98:650. 23. Tang-Liu DD, Matsumoto RM, Usansky JI. Clinical pharmacokinetics and drug metabolism of tazarotene. Clin Pharmacokinet. 1999;37:273–87. 24. Weinstein GD, Krueger GG, Lowe NJ, et al. Tazarotene gel, a new retinoid, for topical therapy of psoriasis: vehicle-controlled study of safety, ­efficacy, and duration of therapeutic effect. J Am Acad Dermatol. 1997;37:85–92. 25. Weinstein GD. Safety, efficacy and duration of therapeutic effect of tazarotene used in the treatment 7 Topical Therapy II: Retinoids, Immunomuodulators, and Others/ Ultraviolet Therapy for Psoriasis Kristen M. Beck and John Koo Abstract Total-body ultraviolet therapy (UV) for moderate-­to-severe psoriasis consists of narrowband and broadband-UVB, psoralen plus UVA (PUVA—where psoralen can be ingested orally or applied topically), inpatient phototherapy (i.e. Goeckerman Therapy, Ingram therapy), non-office-based phototherapy (i.e. use of commercial sunlamps/sunbeds or home UVB for psoriasis treatment, heliotherapy, climatotherapy), and combined UVB/PUVA with retinoid or biologic agents. For each type of UV therapy discussed in this chapter, essential information regarding dosage and administration, efficacy (including comparator data if available), short-term side effects, and long-­ term photocarcinogenic risks are discussed. A well-balanced understanding of the advantages and drawbacks of each photo-­therapeutic option can help phototherapy practitioners optimize clinical outcomes as well as enhance the quality of life for patients affected by this chronic skin condition. K. M. Beck (*) · J. Koo Department of Dermatology, University of California, San Francisco, CA, USA © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_7 Key Learning Objectives 1. To understand the mechanism of action and types of phototherapy for psoriasis. 2. To understand the appropriate use and efficacy of phototherapy for psoriasis. 3. To understand the safety concerns of utilizing phototherapy for psoriasis. UVB Phototherapy UVB phototherapy has been used worldwide since the early 1900s. It consists of narrowband UVB (NB-UVB) and broadband UVB (BB-UVB), which are still the most commonly chosen phototherapeutic options for patients with psoriasis. Dosage and Administration The calculation of the initial UVB dose can be done after assessment of the MED (the minimal erythema dose that induces barely perceptible erythema on non-involved skin) or the Fitzpatrick skin type of each patient [1]. Skin testing to determine MED can add significant time to the first phototherapy visit. Therefore, dosimetry is often based on estimation of Fitzpatrick skin types. Subsequent dosing depends on response of psoriasis 71 72 and phototoxic reactions to previous doses. For example, if a patient experiences mild-­to-­moderate pruritus or discomfort (but no skin burns), he or she should be treated with the same light dose until these reactions resolve [1]. If skin burns or intense skin inflammation (i.e. “beefy red” erythema, severe pruritus, etc.) develops, light treatment should be withheld from patients until the erythema completely resolves. In the latter case, phototherapists might consider the use of “cooling procedures” to calm intense skin inflammation, for example, with very potent topical steroids, before cautiously re-instituting phototherapy. Skin that recently experienced phototoxicity reactions is often extra-sensitive to re-exposure to UVB. Please see Tables 7.1 and 7.2 [2]. The optimal number of UVB exposures per week is three, since less than three treatments per week may result in lower efficacy, and more than three treatments per week have not been consistently shown to be more effective [3]. Once Table 7.1 Dosing guidelines for broadband ultraviolet B According to skin type Initial UVB UVB increase after dose, mJ/cm2 each treatment, mJ/ cm2 Skin type I 20 5 II 25 10 III 30 15 IV 40 20 V 50 25 VI 60 30 According to MED Initial UVB 50% of MED Treatments Increase by 25% 1–10 of initial MED Treatments Increase by 10% 11–20 of initial MED Treatments As ordered by ≥21 physician If subsequent treatments are missed for 4–7 day Keep dose same 1–2 week Decrease dose by 50% 2–3 week Decrease dose by 75% 3–4 week Start over MED Minimal erythema dose, UV ultraviolet. Administered 3–5×/week. Reproduced with permission from Menter et al. [2] K. M. Beck and J. Koo Table 7.2 Dosing guidelines for narrowband ultraviolet B According to skin type Initial UVB UVB dose, mJ/cm2 increase after each Maximum treatment, dose, mJ/ mJ/cm2 cm2 Skin type I 130 15 2000 II 220 25 2000 III 260 40 3000 IV 330 45 3000 V 350 60 5000 VI 400 65 5000 According to MED Initial 50% of UVB MED Treatments Increase by 1–20 10% of initial MED Treatments Increase as ≥21 ordered by physician If subsequent treatments are missed for 4–7 day Keep dose same 1–2 week Decrease dose by 25% 2–3 week Decrease dose by 50% or start over 3–4 week Start over Maintenance therapy for NB-UVB after >95% clearance 1 ×/ week NB-UVB for Keep dose 4 week same 1 ×/ 2 week NB-UVB for Decrease 4 week dose by 25% 1 ×/ 4 week NB-UVB 50% of highest dose MED Minimal erythema dose, NB narrowband, UV ultraviolet. Administered 3–5 ×/ week. Because there is broad range of MED for NB-UVB by skin type, MED testing is generally recommended. It is critically important to meter UVB machine once weekly. UVB lamps steadily lose power. If UV output is not periodically measured and actual output calibrated into machine, clinician may have false impression that patient can be treated with higher doses when machine is actually delivering much lower dose than number entered. Minimum frequency of phototherapy sessions required per week for successful maintenance as well as length of maintenance period varies tremendously between individuals. Above table represents most ideal situation where patient can taper off phototherapy. In reality, many patients require 1 ×/ week. NB-UVB phototherapy indefinitely for successful long-­ term maintenance. Reproduced with permission from Menter et al. [2] 7 Topical Therapy II: Retinoids, Immunomuodulators, and Others/Ultraviolet Therapy for Psoriasis patients achieve marked improvement in their psoriasis, then the therapeutic regimen can be tapered slowly to once weekly. Since this maintenance therapy is less intensive and well tolerated, it may be continued for as long as possible in order to prevent recurrence of psoriasis [1, 3]. Figures 7.1, 7.2, and 7.3 are intended to illustrate the office-based practice of UVB ­ phototherapy. Efficacy Rare comparator studies have documented a much larger difference in efficacy between broadband and narrowband UVB than what is typically observed in practice. In a study by Green et al., 52 patients with plaque psoriasis treated with NB-UVB achieved clearance in 6.6 weeks, whereas 25 patients treated with BB-UVB achieved clearance in 22 weeks [4]. At 1-year 73 follow-up, 38% of the NB-UVB cohort retained clearance, compared with 5% of the BB-UVB cohort. Such magnitude of efficacy difference has not yet been replicated by other studies or in the usual practice settings. More moderate results are be found in another study involving 22 patients with psoriasis who received half-body exposure to BB- or NB-UVB. After 3 weeks of treatment, clinical resolution occurred in 86% of NB-UVB-treated plaques and 73% of the BB-UVB-treated plaques [5]. Long-term efficacy of NB-UVB was reported by Karawaka et al., who reported that 56% of their 52-patient cohort retained PASI 50 response for at least 1 year after a 4-week course of NB-UVB phototherapy [6]. The difference in efficacy between BB-UVB and NB-UVB—where the latter is, to some extent, superior as a treatment for psoriasis—deserves explanation. Some of the UV rays emitted by BB-UVB lamps (i.e. 254, 280, or 294 nm rays) are Fig. 7.1 Nurse stations and light boxes for office-based UVB phototherapy K. M. Beck and J. Koo 74 less effective than UV rays with longer wavelengths (i.e. 311 or 312 nm rays) at clearing psoriasis [7]. The 311 nm rays emitted by NB-UVB lamps are capable of clearing psoriatic plaques with a little as 0.4 times the MED. Furthermore, NB-UVB therapy confers the benefit of causing fewer phototoxic reactions for patients than BB-UVB therapy at the same doses. Side Effects and Safety Side effects of UVB phototherapy include visible erythema, burning, blistering, discomfort, and post-inflammatory hyperpigmentation [1, 5, 8]. In addition, “over-exposure” to UVB has been associated with precipitation of erythrodermic psoriasis in two reported cases [9]. Do not perform UVB if the patient is erythrodermic or near-erythrodermic. Fig. 7.2 Eye protection for whole-body UVB irradiation Fig. 7.3 Whole-body UVB irradiation using a stool to increase lower-body exposure Photocarcinogenicity of UVB Phototherapy Studies with mice have often not mirrored human exposure to UVB. They should not be extrapolated at face value to determine the carcinogenic risk of UVB phototherapy in humans. For instance, UVB irradiation of 30 hairless, lightly pigmented mice who are not cancer-prone for 30 weeks was linked with an 83% rate of SCC development [10]. This study lacked a control mice group. Moreover, the mice received UVB 5 days per week, while humans with psoriasis are usually treated three times weekly. In another study using hairless, lightly pigmented mice, all mice developed skin tumors, while mice receiving 311 nm UVB developed skin tumors earlier than mice treated with BB-UVB [11]. Of note, some of the mice had total-body exposure to UVB at doses that were multitudes of the MED (also known as “supra-­ erythemogenic” doses). In humans, total body use of supraerythemogenic doses not done since intense UVB irradiation of non-involved skin is likely to result in frequent, severe skin burns, which can increase the risk of skin cancer. 7 Topical Therapy II: Retinoids, Immunomuodulators, and Others/Ultraviolet Therapy for Psoriasis Meanwhile, review of the worldwide literature on human experience revealed no convincing data of an increased photocarcinogenic risk of longterm UVB phototherapy for psoriasis. In a publication by Lee et al., no increased carcinogenic risk associated with UVB phototherapy was appreciated in 10 out of the 11 studies that were reviewed in the medical literature [12]. One exception is the report featuring a 30-case/137-­control sub-cohort from a large-scale Finnish study in psoriasis patients, where the relative risk ratio of developing SCC in patients with a history of UVB treatments was 1.6. This risk is slightly elevated but turned out to be not statistically significant. In addition, there was no increased risk of skin cancer in 484 Northern Irish patients who had at least 18 NB-UVB exposures individually [13]. A larger retrospective study involving 3886 patients treated with UVB at a Scottish hospital found 27 BCC, 7 SCC, and 6 melanomas, which were comparable to expected incidences for the 3 types of skin cancer, respectively, in the matched populations [14]. In this study, only patients who received both NB-UVB and PUVA had a higher incidence of BCC [15] than the expected incidence [14] in the general Scottish population, but not among those who received UVB alone [14]. In a follow-up study of 195 patients who received either BB- or NB-UVB treatment for psoriasis, only one patient developed melanoma in situ. This patient’s tumor was diagnosed in the same year phototherapy was instituted; hence, its development may have been unrelated to the limited UVB exposure [16]. Another study followed 1908 Scottish patients who had long-term UVB treatment for an average of 4 years. Remarkably, the study found no incidence of melanoma or squamous cell carcinoma (SCC) [17]. Ten patients developed basal cell carcinoma (BCC) mostly on the face compared with the expected incidence of 4.7 in the matched population [17]. The predilection of BCC for the face attested to the possible increase in solar exposure as an uncontrolled variable, since patients with psoriasis often engage in this practice to augment office-based phototherapy. In contrast, the face’s patient is usually covered during whole-body UV irradiation. 75 PUVA (Psoralen Plus PUVA) Dosage and Administration PUVA is the combination of the plant-originated compound psoralen and UVA (320–400 nm) irradiation for the treatment of psoriasis. Available as Oxsoralen Ultra® 10 mg capsules in the US, psoralen can be taken orally or diluted in a bath solution for topical administration within the hour before UVA irradiation. In the beginning of their treatment course, patients are encouraged to undergo PUVA treatment thrice weekly. For the purpose of maintaining marked improvement or clearance of psoriasis, the number of weekly office visits for PUVA can be tapered slowly to once weekly, once every other week, or even a less frequent schedule. Dosimetry of PUVA is described in Tables 7.3 and 7.4 [2]. Information about dosage following missed PUVA exposures can be found in Table 7.5. Please refer to Fig. 7.4 for an illustration of “hands” PUVA (PUVA just for the hands) and to Figs. 7.5, 7.6, 7.7, and 7.8 for an illustration of bath PUVA. Efficacy Following activation by UVA irradiation, psoralen cross-links with DNA of various cell types in psoriatic skin (i.e. keratinocytes, T cells, etc.) to inhibit inflammation and cell proliferation. A randomized, double-blind, placebo-controlled trial involving 40 psoriasis patients revealed an 86% PASI 75 response rate in the PUVA arm versus 0% PASI 75 response rate in the UVA arm after 12 weeks of therapy [18]. In another study, Table 7.3 Dosing of 8-methoxypsoralen for oral psoralen plus ultraviolet A Patient weight lb <66 66–143 144–200 > 200 kg <30 30–65 66–91 >91 Drug dose, mg 10 20 30 40 Reproduced with permission from Menter et al. [2] K. M. Beck and J. Koo 76 Table 7.4 Dosing of ultraviolet A radiation for oral psoralen plus ultraviolet A Skin type I II III IV V VI Initial dose, J/cm2 0.5 1.0 1.5 2.0 2.5 3.0 Increments, J/ cm2 0.5 0.5 1.0 1.0 1.5 1.5 Maximum dose, J/cm2 8 8 12 12 20 20 Reproduced with permission from Menter et al. [2] Table 7.5 Subsequent dosing protocol for PUVA Number of missed days 3–5 6–14 15–21 22–28 >28 Subsequent dosing Continue routine dosing increase Hold dose Decreased dose by 25% Decreased dose by 50% Re-institute phototherapy and dosimetry Fig. 7.5 Oxsoralen Ultra® 10 mg capsules for bath PUVA Adapted from UCSF Psoriasis Center with permission of the author (JK) Fig. 7.6 Boiled water to dissolve Oxsoralen Ultra*** 10 mg capsules study, both PUVA and NB-UVB irradiation were done thrice weekly for a maximum of 30 expoFig. 7.4 PUVA for the hands/feet psoriasis or eczema sures or clearance, whichever came first. In another study of 28 patients with psoriasis, 54% 88.8% of 3175 patients with psoriasis achieved of the PUVA-treated cohort versus 78% of the 95–100% clearance after an average of 20 PUVA NB-UVB-treated cohort achieved clearance after exposures [19]. a maximum of 30 exposures [21]. Interestingly, When compared against NB-UVB photother- in this study PUVA was administered twice apy, PUVA demonstrates inferior efficacy in weekly, which is not the optimal regimen to small-scale studies but superior efficacy in large-­ achieve rapid clearance of psoriasis. scale studies. A comparator trial involving only In terms of a larger “head to head” comparison 17 patients reported a PASI score reduction of study, Gordon et al. treated 100 patients thrice 45% in the PUVA-treated cohort compared with weekly with either PUVA or NB-UVB photo77% in the NB-UVB-treated cohort [20]. In this therapy. Eighty-four percent of the PUVA-treated 7 Topical Therapy II: Retinoids, Immunomuodulators, and Others/Ultraviolet Therapy for Psoriasis 77 et al. involving 93 patients showed an 84% clearance rate in the PUVA arm after 17.0 exposures compared with 65% in the NB-UVB arm after 28.5 exposures [23]. Sixty-eight percent of the PUVA-treated subjects versus thirty-five percent of the NB-UVB-treated subjects remained in remission for 6 months after treatments had ended. Evidence from these large, randomized trials seem to support superior efficacy of PUVA over NB-UVB, especially with regard to duration of therapeutic effects. Side Effects Fig. 7.7 Concentrated solution made by dissolving Oxsoralen Ultra 10 mg capsules in hot water Fig. 7.8 Measuring water level for bath PUVA. The water level corresponding to 100 L is indicated on the ruler Dose-dependent effects of PUVA include skin irritation, skin burns, and tanning. In addition, ingestion of psoralen has been associated with nausea, headaches, dizziness, and extremely rare instances of psychiatric disturbance (i.e. insomnia, depression) [8]. To minimize nausea, patients can try decreasing the dose of psoralen and/or ingesting psoralen with food later in the day [8]. After decades of use, the association of PUVA with an increased risk of cataracts has not been substantiated in an evidence-based fashion [24]. The author (JK) feels that an ophthalmic examination is mainly useful for documenting the presence or absence of pre-existing cataracts. Therefore, for the author (JK), even the presence of pre-PUVA cataracts does not necessarily constitute an absolute contraindication for initiating PUVA therapy. There is no convincing human data after decades of worldwide use to prove that PUVA increases the risk of cataracts, provided that the patient is compliant with UVA eye protection measures for 24 hours after psoralen ingestion. Long-term exposure to PUVA is associated with photoaging marked by premature cutaneous degeneration and pigmentation [25]. cohort achieved clearance of psoriasis after 16.7 exposures compared with sixty-three percent of NB-UVB-treated cohort after 25.3 exposures Photocarcinogenicity of PUVA [22]. Moreover, the number of PUVA-treated patients who retained clearance 6 months after Long-term UVA irradiation exposure has been therapy was almost double that of NB-UVB-­ associated with melanocytic atypia (i.e. large, treated patients [22]. A similar study by Yones angular hyperchromatic nuclei and binucleated K. M. Beck and J. Koo 78 melanocytes) called “PUVA lentigines”, suggesting aberrant cellular kinetics [26]. A list of 25 PUVA follow-up studies conducted worldwide can be found in Table 7.6. Out of 25 published studies, 24 did not show an elevated risk of melanoma associated with long-term PUVA exposure. However, one study published by Stern et al. recorded an increased risk of cutaneous melanoma associated with PUVA. In this study, 1380 patients across the USA were monitored for up to 21 years during and after photochemotherapy. During the first 15 years of follow-up, the relative Table 7.6 Risk of melanoma associated with PUVA treatment Authors (Year) Honigsmann et al. (1980) [27] Lobel et al. (1981) [28] Lassus et al. (1981) [29] Lindskov (1983) [30] Ros et al. (1983) [31] Reshad et al. (1984) [32] Eskelinen et al. (1985) [33] Tanew et al. (1986) [34] Henseler et al. (1987) [35] Barth et al. (1987) [36] Cox et al. (1987) [37] Torinuki & Tagami (1988) [38] Abdullah & Keczkes (1989) [39] Forman et al. (1989) [40] Bryunzeel et al. (1991) [41] Chuang et al. (1992) [42] Lever & Farr (1994) [43] Gritiyarangsan et al. (1995) [44] Maier et al. (1996) [45] McKenna et al. (1996) [46] Hannuksela et al. (1996) [47]b Cockayne & August (1997) [48] Follow-up periods (years) 1–5 Mean or median follow-up Relative risk of invasive melanoma a a Country Austria Number of Number of melanoma cases subjects 418 0 Australia 489 0 a a a Finland 525 0 1–3.6 2.2 a Denmark Sweden United Kingdom Finland 198 250 216 0 0 0 1–6 0–7 0–7 3.5 4.1 4.8 a 1047 0 0–8 a a Austria 297 0 3.4–8.0 5.3 a Europe 1643 1 invasive 0–11 8.0 a Germany 6820 0 0–10 a a United Kingdom Japan 95 0 0–8 a a 151 0 0–10 a a United Kingdom United States Netherlands 198 1 in situ 0–10 a a 551 1 invasive 0–10 4.8 a 260 0 0–12.8 8.7 a United States United Kingdom Thailand 492 0 0–14 5.4 a 54 0 6–15 11.2 a 113 0 2–14 6.2 a Austria 496 0 0.4–17.2 6.2 a Northern Ireland Finland 245 0 2–15 9.5 a 527 0 0–16 11 a 150 0 5–17 a a United Kingdom a a 7 Topical Therapy II: Retinoids, Immunomuodulators, and Others/Ultraviolet Therapy for Psoriasis 79 Table 7.6 (continued) Number of Number of melanoma cases subjects 1380 18 invasive 7 in situ 1 ocular Follow-up periods (years) 0–24 Mean or median follow-up 19 [49] 0–21 16 0–18 14.7 Authors (Year) Stern et al. (1997) [49] & Stern (2001) [50] Country United States Lindelof et al. (1999) [51] Sweden 4799 Hannuksela-Svahn et al. (1999) [52]b Sweden, Finland 944 0 Relative risk of invasive melanoma 1975–1990: 1.0 1991–1996: 4.7 (1.4–16.1) 1996–1999: 7.4 (2.2–25.1) M: 1.1 (0.4–2.3) F: 1.0 (0.5–2.2) M: 0.7 (0.0–3.8) F: 1.3 (0.0–7.2) Information not provided or not able to be calculated from data Bath PUVA only a b incidence of melanoma was not different than expected in the general US population, based on cancer statistics from the SEER (Surveillance, Epidemiology, and End Results) Program [49]. In the next 5 years of follow-up, seven new cases of melanoma were diagnosed, giving a relative risk ratio of 5.4 [49]. The authors thus hypothesized that a period of latency exists before the risk of melanoma related to long-term PUVA treatment becomes recognizable. Comprehensive knowledge of how SEER derives its cancer statistics can influence how one interprets Stern et al.’s results and the ­implications of their study. SEER approximates melanoma prevalence in the general US population based on chart reviews of hospitals and “search of private laboratory records” in designated regions, which, as of 1990, comprised 9.6% of the total US population. Unlike systemic cancers for which SEER database was primarily intended, many cases of melanoma in situ and early-stage invasive melanoma are excised on an outpatient basis by practicing dermatologists without being registered into any of the above databases [15, 53]. As a result, data generated by SEER may underestimate the actual prevalence of melanoma in the general US population. This could inflate the melanoma risk for PUVA that was calculated in Stern et al.’s study. Additionally, the study lacked a matched control group to account for confound- ing variables such as a history of sunburns, family history of skin neoplasm, other UV light treatments, or medications that possess carcinogenic risk, etc. An extension of the original study by five more years followed the same cohort of 1380 PUVA-treated patients. The relative risk ratio within this latest five-year period is 7.4, once again based on cancer statistics from SEER [50]. In contrast, the largest PUVA study that has been published to date was conducted in Sweden by Lindelof et al., who examined a cohort of 4799 PUVA-treated psoriasis patients. The data published by Lindelof et al. revealed no increased risk of melanoma compared to the general Swedish population during an average 15–16 years of follow-up [51]. There was not even an increased risk of melanoma in a sub-­ cohort of 1867 patients who were followed up for longer than 15 years [51]—this is where Stern et al. claimed to have observed an increased carcinogenic risk of PUVA. The prevalence of melanoma used to calculate relative risk ratio of melanoma in Lindelof et al.’ study comes from the Swedish Cancer Registry. It is likely to reflect the actual prevalence in the Swedish population, since reporting of melanoma diagnoses in Sweden is compulsory, unlike the situation in the USA where no melanoma-reporting requirement exists. 80 Concerning the risk of NMSC, the results of Lindelof et al.’s study supported an increased risk of SCC associated with long-term PUVA use: relative risk ratios were 3.6 for women and 5.6 for men [51]. Furthermore, analysis of the 1380-patient US PUVA cohort by Nijsten and Stern revealed higher than expected incidences of SCC and BCC compared to the general US population (based on SEER statistics). This NMSC risk is particularly notable in patients with more than 200 lifetime PUVA treatments [54]. Conversely, a follow-up study in 944 Swedish and Finnish psoriasis patients treated only with bath PUVA, not systemic PUVA, did not show any increased risk of SCC or malignant melanoma after a mean follow-up period of 14.7 years [52]. Whether different cellular mechanisms are involved or the total UVA dose is lower, bath PUVA appears to have less photocarcinogenic risk than oral PUVA. It is important to recognize that available data about long-term safety of PUVA therapy mostly come from studies involving Caucasians. Murase et al. conducted the only review of PUVA-related skin cancer incidence in non-Caucasian patients with various photosensitive dermatoses. Their analysis included 4294 long-term PUVA patients who had at least 5 years of follow-up in studies conducted in Japan, Korea, Thailand, Egypt, and Tunisia. The relative risk of developing skin neoplasm in these patients compared to the general dermatology oupatient population in each respective country where the PUVA follow up data originated was 0.86 (with a confidence interval of 0.36–1.35). This means that the increased risk of skin cancer related to long-term PUVA in Asian and North African patients has yet to be demonstrated [55]. Further investigation into the protective nature of darker skin phototypes is needed. Inpatient Photothearpy Goeckerman Therapy Formulated in 1925, Goeckerman’s eponymous therapy is one of the oldest forms of phototherapy for the treatment of psoriasis [56] and K. M. Beck and J. Koo enjoyed widespread use in the USA until the Diagnostically Related Groups (DRG) made prolonged hospitalization for dermatologic conditions impossible. It is still considered by many dermatologists as the gold standard treatment based on high efficacy, safety of not requiring any internal agents, and induction of prolonged remission times. Furthermore, Goeckerman therapy can be used to treat patients who have failed a number of therapeutic modalities beyond topical steroids, such as PUVA, UVB, and biologics [57]. Dosage and Administration Goeckerman therapy is an intensive therapy requiring all-day, everyday continuous participation for typically 4–6 weeks in a hospital unit or psoriasis day care center (Figs. 7.9 and 7.10) [58]. The treatment regimen begins each day with UVB irradiation, since UV light will not penetrate after application of crude coal tar (CCT) [59]. The second step is to lather the patient’s skin with CCT in petroleum (Fig. 7.11), which comes in 2%, 5%, and 10% preparations, and to soak the patient’s scalp in 20% liquor carbonis detergens (LCD) in Neutraderm lotion (Fig. 7.12) [58]. Twenty percent LCD in Aquaphor ointment is applied to the total body of each patient before he/she leaves the facility. It is also sent home with the patient to be done once more at bedtime. If the patient’s condition is so severe that tar plus UVB do not provide adequate control, then compounded anthralin and even topical PUVA can be added [1]. Infrequently, truly recalcitrant psoriasis might necessitate the concomitant use of retinoids and topical vitamin D derivatives in order to achieve clearance [1]. Efficacy Goeckerman therapy has high efficacy for recalcitrant psoriasis, even when single or multiple biological agents have failed to show improvements [57]. In a prospective study, 95% and 100% of 25 psoriasis patients who were treated with Goeckerman therapy reached PASI 75 by week 8 and week 12, respectively [60]. In another study, clearance of psoriasis has been documented in 90% of 300 patients after only 18 days 7 Topical Therapy II: Retinoids, Immunomuodulators, and Others/Ultraviolet Therapy for Psoriasis 81 Fig. 7.9 The common area for Goeckerman therapy patients at a psoriasis day care center, which often turns into a therapeutic milieu (i.e. where the ambient supportive atmosphere is experienced by the patients as therapeutic) Fig. 7.10 Physicians performing rounds on a patient undergoing Goeckerman therapy 82 K. M. Beck and J. Koo Fig. 7.11 Nursing staff applying crude coal tar with occlusion to psoriatic skin of treatment. Eight-month remission was recorded in 90% of these subjects [58]. It should be noted that the second study featured a more intensive treatment regimen than the commonly encountered Goeckerman therapy: UVB and tar therapy were administered twice daily, and patients attended the program six instead of 5 days a week [58]. This may explain their significantly rapid clearance rate. In 1979 DesGroseilliers et al. implemented a modified design to render Goeckerman therapy less time-intensive and more cost-effective. 200 patients were treated with coal tar application at home plus office-based UVB irradiation the following day 5 days a week. After 1 month of treatment, 86% of patients in this cohort achieved clearance of their psoriasis [61]. The mean r­ emission duration of 5.1 months was calculated based on 185 patients who experienced relapse during the observation period [61]. However, there were 15 patients in the study who did not relapse during the entire observation period. Had these 15 patients been included, the calculated mean remission duration would have been longer than 5.1 months. ide Effects and Long-term Safety S Side effects of Goeckerman therapy include folliculitis and phototoxic reactions, which are can be managed by dose adjustment of tar and UVB. The pustules associated with Goeckerman therapy can develop on non-involved skin and are therefore distinct from pustular psoriatic lesions [62]. Rarely, precipitation of erythrodermic 7 Topical Therapy II: Retinoids, Immunomuodulators, and Others/Ultraviolet Therapy for Psoriasis 83 consists of a succession of tar bath, light treatment, and application of anthralin paste or ointment to involved skin. Adding crude coal tar to anthralin has been shown to reduce skin irritation without compromising antipsoriatic efficacy [66]. In a retrospective study by De Bersaques, 275 psoriasis patients who were treated in a hospital with Ingram therapy achieved clearance after an average of 25 days [65]. Patients did not need another treatment or hospitalization for 8 to 11 months after therapy [65]. However, being a single center study, these results might have been influenced by loss to follow-up of patients who sought care from other practitioners due to early relapse. Non-office-based Phototherapy Commercial Tanning Therapy Fig. 7.12 Nursing staff applying 20% liquor carbonis detergens in Neutraderm lotion to a psoriasis patient’s scalp p­ soriasis has been documented in patients allergic to crude coal tar [63]. To investigate longterm safety, a 25-year follow-up study was conducted in 280 psoriasis patients treated with Goeckerman therapy over a 5-year period. The results consisted of 1 melanoma, 22 BCC, 7 SCC, and 3 tumors of unknown type [64]. Based on cancer statistics from a matched population, there was no increase in the risk of skin cancer in these patients [64]. Ingram Therapy Ingram therapy is the inpatient treatment of psoriasis with UV irradiation and anthralin, which was introduced to dermatology in 1916 [65]. It In commercial tanning facilities, phototherapy for psoriasis can be conducted using “tanning beds” that emit high-intensity UVA rays mixed with small quantities of contaminant UVB. This is different from office-based, outpatient UVB phototherapy, where healthcare professionals frequently perform clinical assessments and light dose adjustments. For 16 out of 20 psoriasis patients treated with tanning beds, the initial mean PASI score of 7.96 (+/− 1.77) was diminished to 5.04 (+/− 2.5) after 6 weeks of treatment [67]. Even those who did not complete the entire treatment course had a 23.5% improvement of their PASI score [67]. Furthermore, most patients reported significant improvements in health-­ related quality of life, while mild burning and transient pruritus were infrequent and well tolerated [67]. There is controversy surrounding the existence of an association between exposure to commercial UV tanning devices (i.e. sunlamps, sunbeds) and an increased risk of melanoma. In 2006 the International Agency for Research on Cancer issued a detailed report on the photocarcinogenic risk of artificial UV exposure, which addressed 23 case-control studies related to indoor tanning and melanoma. The risks for K. M. Beck and J. Koo 84 Table 7.7 Meta-analysis of all studies included Exposure Ever use of indoor tanning facility First exposure in youth Exposure distant in time Exposure recent in time Number of studies 19 7 5 5 Heterogeneity Summary relative risk P-value (95% CI) χ2 H 1.15 0.013 1.37 (1.00–1.31) a 1.75 (1.35–2.26) 0.55 1.49 (0.93–2.38) 0.018 1.10 (0.76–1.60) 0.81 Table 7.8 Meta-analysis of the cohort and population-­ based case–control studies included Exposure Ever use of indoor tanning facility First exposure in youth Exposure distant in time Exposure recent in time 0.91 1.65 0.67 Reproduced with permission from the IARC (2005) [68] a The degrees of freedom for the Chi-square are given by the number of databases included minus one, not by the number of studies ­ elanoma associated with “ever use” (any expom sure to sunlamps/sunbeds in one’s lifetime), “first use in youth” (exposure to sunlamps/sunbeds starting at 35 years old or younger), “distant use” (5 years or more from the time of the interview), and “recent use” (less than 5 years from the time of the interview) of indoor tanning devices were compared and contrasted using reported relative risk ratios. Meta-analysis of 19 of these 23 studies (Table 7.7) revealed a calculated summative relative risk of 1.15 (95% confidence interval: 1.00–1.31) for “ever use” of indoor tanning facility [68]. The same relative risk becomes slightly elevated to 1.17 (0.96–1.42) when data only from cohort and population-based case-control studies were employed for calculation (Table 7.8) [68]. In both meta-analyses the summative relative risk is higher for first exposure in youth than for “ever use” of indoor tanning facility and also higher for “distant” exposure versus “recent” exposure [68]. In a systematic review of case-control studies, 6 out of 19 studies reported a dose-response relationship between tanning lamp/bed exposure and melanoma risk [69]. Another review of 10 case-­ control studies reported that the odds ratios spanned anywhere from 0.7 (0.5–1.0) to 2.9 (1.3– 6.4) for “ever use” versus “never use” (no exposure at all during one’s lifetime) of sunlamps/ sunbeds [70]. Of note, the dose-dependent risk for invasive melanoma is likely to be more Number of studies 10 5 2 2 Heterogeneitya Summary relative risk P-value (95% CI) χ2 H 0.011 1.540 1.17 (0.96– 1.42) 1.71 (1.25– 2.33) 1.58 (0.25– 9.98)a 1.24 (0.52– 2.94) 0.435 0.973 0.502 0.830 0.762 0.521 Reproduced with permission from the IARC (2005) [68] The confidence interval is very wide because this analysis includes only 2 studies, one of which has two estimates a p­ rofound in Caucasian female users with more than 10 lifetime tanning sessions compared with those having less than 10 sessions [71]. The results of this study do not apply to Caucasian male users, possibly due to the fact that they visit tanning facilities less frequently than their female counterparts [71]. In addition, this cut-off of 10 tanning sessions is an arbitrary proposal based on the results of one study. More convincing data is needed to establish a photocarcinogenic threshold for UV exposure through tanning devices. Home UVB Therapy Most devices employed in home phototherapy emit UVB. This form of phototherapy is tailored to patients with stable psoriasis and logistical difficulty, such as lack of time to do office phototherapy or no access to transportation to dermatology practices with UVB capacity. In a randomized trial involving 196 psoriasis patients, 70% of patients receiving home UVB therapy reached PASI 50 compared to 73% in the outpatient group [72]. Short-term safety profiles were not notably different between the groups [72]. Moreover, the burden of therapy (a function of method and time commitment) was significantly lower for home phototherapy patients than for office-based outpatients [73]. 7 Topical Therapy II: Retinoids, Immunomuodulators, and Others/Ultraviolet Therapy for Psoriasis 85 Heliotherapy UVB and Retinoid Therapy Heliotherapy is the therapeutic use of sunlight to treat psoriasis and other photosensitive dermatoses. There are not many large, randomized, controlled studies to determine the efficacy of heliotherapy, let alone comparator studies involving other forms of phototherapy. In a study with 373 subjects, the rate of clearance of psoriasis was 22% and that of PASI 75 response was 84% after 1 month of treatment [74]. The median remission time was 2.6 months (80 days) [74]. Short-term side effects are similar to other phototherapeutic modalities: skin burns, irritation, pruritus, erythema, heat, etc. Skin cancer risks associated with long-term heliotherapy in patients with psoriasis have not been studied. Acitretin, an oral retinoid agent, has been shown to enhance the efficacy of UVB phototherapy. In an 8-week, randomized, comparator study involving 82 psoriasis patients, 60% of the combined BB-UVB and acitretin cohort (re-UVB) versus 24% of the BB-UVB cohort reached PASI 75 [78]. Phototherapy was administered 3–5 times weekly, and acitretin was available as 25 mg tablets taken once daily [78]. Treatment with acitretin 25 mg plus thrice weekly NB-UVB resulted in 75% improvement of psoriasis in 30 out of 40 patients from another study [79]. The phenomenon “delayed retinoid burn” was described in 2011 after a psoriasis patient experienced a severe phototoxic reaction as a result of adding acitretin 25 mg daily to a previously tolerated UVB dose [80]. Since acitretin induces cell differentiation and leads to sloughing of keratin layers from psoriatic plaques, the introduction of acitretin in the middle of already ongoing phototherapy can lead to too much light penetration causing phototoxicity to previously tolerated dosimetry. Therefore, phototherapy providers should consider reducing UV dose when starting concomitant retinoid therapy. The authors recommended cutting UVB dose by 50% as soon as acitretin is added, and gradually going back toward the pre-acitretin UVB dose level if no phototoxicity occurs [80]. How this is conducted in each phototherapy provider’s office is a judgment call. Climatotherapy at the Dead Sea Climatotherapy is the exposure of skin to a special kind of climate for the treatment of psoriasis. Its efficacy was established in a study involving 64 patients: PASI 75 response after 1 month of climatotherapy at the Dead Sea was 75.9%, and median duration of remission was 5.8 months (23.1 weeks) [75]. Analysis of skin cancer data from a cohort of 1738 Danish patients who were followed up for an average of 6.1 years revealed no association between climatotherapy and an increased risk of melanoma [76]. There were, however, 8 SCC and 28 BCC compared with 0.8 expected SCC and 6.6 expected BCC [76]. It was later discovered that the subjects in this follow­up study had received more than three times the necessary amount of UVB to achieve therapeutic results [77]. This important information might partially explain the significant difference between observed and expected incidence of non-melanoma skin cancer. Combination Therapy For patients with severe psoriasis based on large BSA coverage and/or resistance to traditional single therapies, combinations of therapies are available as treatment options. PUVA and Retinoid Therapy Similarly, rapid improvement of recalcitrant psoriasis might be achieved by adding oral retinoid agents to PUVA. In a study comparing the ­efficacies of combined PUVA plus acitretin one mg/kg body weight/day (re-PUVA) versus PUVA alone, clearance was achieved in 96% of the combination cohort and 80% of the PUVA cohort after 11 weeks of treatment [81]. Had the 48 subjects of this study undergone PUVA treatment three instead of four times weekly, the clearance rates in both arms might not have been as high and their difference easier to appreciate. 86 Furthermore, another head-to-head comparison study between re-PUVA and re-UVB showed 100% clearance rate in the re-PUVA cohort compared with 93% in the re-UVB cohort after a 6-week treatment course [82]. Etretinate one mg/ kg body weight/day was used instead of acitretin. Duration of therapeutic effects was longer for re-­ PUVA patients versus re-UVB patients [82]. However, this study was limited in its small size (each cohort consisted of 15 patients) and by the fact that PUVA or UVB was done twice weekly for each combination therapy. When photo(chemo)therapy was instituted at the optimal treatment regimen (thrice weekly) in 60 psoriasis patients, clearance was achieved in 63.3% of the re-PUVA cohort versus 56.6% of the re-UVB cohort [83]. The reported clearance rates were lower in this study than in the previous studies because acitretin was dosed at 0.3–0.5 mg instead of 1 mg/kg body weight/day. In spite of the dosing differences between these comparison studies, all consistently proved that re-PUVA is more effective at reducing clinical severity of psoriasis than re-UVB combination therapy. UVB and Biologics UVB therapy can be added to biological agents to expedite clearance of moderate-to-severe psoriasis. A six-week study involving 14 patients showed that combined UVB and etanercept helped patients achieve a 64 +/− 28.8% reduction of their modified PASI scores compared with the 53.7 +/− 36.9% PASI score reduction in patients treated with etanercept alone [84]. UVB was given thrice weekly, and etanercept was dosed at 25 mg twice weekly. De Simone et al. performed a single-arm, open label study in which 33 patients with moderate-to-severe psoriasis were treated for 8 weeks with etanercept 50 mg once weekly plus NB-UVB thrice weekly, after which only etanercept was continued for another 4 weeks. At Week 4, 8, and 12 of treatment, PASI 75 was reached in 24.2%, 66.7%, and 81.8% of the study participants [85]. The results of a single-arm, single-center, open-label study involving 20 patients with K. M. Beck and J. Koo severe psoriasis were promising: after 12 weeks of combined adalimumab 40 mg every other week and NB-UVB three times weekly, 95% and 85% of the patients reached PASI 75 and clearance, respectively [86]. Moreover, 65% of patients retained PASI 75 response 12 weeks following the end of treatment [86]. A similar but larger trial (known by the acronym “UNITE”) with NB-UVB thrice weekly plus etanercept 50 mg twice weekly also showcased high efficacy of combination therapy: at week 12, 84.9% of 86 psoriasis patients reached PASI 75 [87]. There was also an equally important and significant improvement in health-related quality of life in almost all the UNITE study subjects [87]. Photocarcinogenicity of Combination Therapy Oral retinoid agents can lower the skin cancer risk associated with phototherapy through promotion of keratinocyte differentiation. There is a case report of one patient on once daily acitretin 25 mg whose number of squamous cell carcinomas decreased while on acitretin and increased when taken off acitretin [88]. In addition, the results of a 15-year follow-up study in 135 psoriasis patients who received PUVA and acitretin/ etretinate for more than 6 months seemed convincing: the numbers of SCC diagnosed during the time of using and not using acitretin/etretinate were 196 and 302, respectively [89]. The adjusted incidence rate ratio of SCC was calculated to be 0.79, which implicates the potential of oral retinoid agents to decrease non-melanoma skin cancer risk in phototherapy patients [89]. Whether or not these agents have any effect on melanoma risk is a question that has not yet been addressed in the literature. The carcinogenicity of combined phototherapy plus biological agents is an on-going concern. In 2011, Gamblicher et al. compared psoriatic plaques that were treated with either BB-UVB at two MED or combined BB-UVB at two MED plus etanercept 50 mg one time in 11 study subjects. Punch biopsies of the treated areas were taken at 1, 24, and 72 hours after 7 Topical Therapy II: Retinoids, Immunomuodulators, and Others/Ultraviolet Therapy for Psoriasis 87 Fig. 7.13 Excimer laser therapy requires the most powerful excimer laser machine to adequately treat moderate-to-severe psoriasis BB-UVB treatment [90]. Immunohistochemical analysis revealed increased expression of survivin (a tumor marker) and decreased activity of cyclin D and p53 (regulators of tumor development) in BB-UVB-plus-etanercept-treated sites compared with BB-UVB-monotherapy-treated sites [90]. Large randomized controlled trials are needed to determine if an increased risk of melanoma or NMSC exists in psoriasis patients treated with combined photo(chemo)therapy and biologics. therapy can confer some skin cancer chemo-­ protective effects limited to the periods of retinoid use [92]. In terms of efficacy, both forms of UVB therapy are not as effective as Goeckerman or PUVA but fare better than heliotherapy [91, 92, 94]. The combination of UVB and oral retinoid or biological agents can lead to more successful therapy for patients than any of these modalities alone. Short-term side effects of UVB phototherapy are well tolerated, and no studies so far have convincingly demonstrated any relationships between long-term UVB treatment and an Conclusion increased risk of skin cancer. If office UVB is not feasible, climatotherapy, commercial and home All day, every day Goeckerman therapy is one of UV therapies should still be considered as useful the most effective regimens for moderate-to-­ alternatives in the care of patients with psoriasis. severe psoriasis, and it still maintains an appealFinally, excimer laser therapy, to which ing safety profile [91, 92]. PUVA has been shown another chapter of this book is dedicated, has to result in high clearance rates, but the associa- been conducted worldwide for more than 10 years tion of PUVA with an increased risk of NMSC in to target recalcitrant plaque psoriasis (Fig. 7.13). fair-skinned Caucasian patients has rendered it Current data on its efficacy revealed that fewer less popular nowadays than narrowband UVB exposures to light are needed to induce remission [92]. Until this date whether or not PUVA causes [95–98]. No signal regarding an increased risk of an increase in melanoma risk is a controversial skin cancer from therapeutic use of excimer laser topic due to contradictory results between a has been reported. Furthermore, excimer laser United States 16-center study and numerous spares non-involved skin, so there is a possibility, other studies primarily from Europe [49, 93]. in terms of cutaneous malignancy safety, that this Fortunately, adding oral retinoid agents to PUVA targeted phototherapy may even be better than 88 traditional total-body irradiation exposure with UVB or PUVA. Authorship Responsibilities and Attributions This manuscript represents valid work. Neither this manuscript nor one with substantially similar content under my authorship has been published or is being considered for publication elsewhere. Dr. John Koo and I (Kristen Beck) both have significant contribution to the manuscript. He has agreed to designate me as the primary correspondent with the editors to review the edited typescript and proof, and to make decisions regarding release of information in the manuscript to the media, federal agencies, or both. Financial Disclosure Both authors have no relevant financial interest in this manuscript. References 1. Gattu S, Pugashetti R, Koo JY. The art and practice of UVB phototherapy and laser for the treatment of moderate-to-severe psoriasis. In: JYM K, Lee CS, Lebwohl MG, Weinstein GD, Gottlieb A, editors. Moderate-to-severe psoriasis. 3rd ed. New York: Informa Healthcare; 2009. p. 75–113. 2. Menter A, Korman NJ, Elmets CA, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis: Section 5: Guidelines of care for the treatment of psoriasis with phototherapy and photochemotherapy. J Am Acad Dermatol. 2010;62(1):114–35. 3. Do AN, Koo JYM. Initiating narrow-band UVB for the treatment of psoriasis. Psoriasis Forum. 2004;10(1):1–6. 4. Green C, Ferguson J, Lakshmipathi T, Johnson BE. 311 nm UVB phototherapy--an effective treatment for psoriasis. Br J Dermatol. 1988;119(6):691–6. 5. Coven TR, Burack LH, Gilleaudeau P, Keogh M, Ozawa M, Krueger JG. Narrowband UV-B produces superior clinical and histopathological resolution of moderate-to-severe psoriasis in patients compared with broadband UV-B2. Arch Dermatol. 1997;133(12):1514–22. 6. Karakawa M, Komine M, Takekoshi T, et al. Duration of remission period of narrowband ultraviolet B therapy on psoriasis vulgaris. J Dermatol. 2011;38(7):655–60. 7. Parrish JA, Jaenicke KF. Action spectrum for phototherapy of psoriasis. J Invest Dermatol. 1981;76(5):359–62. 8. Lee ES, Heller MM, Kamangar F, Park K, Koo JYM. Current treatment options: phototherapy. Fut Med. 2011:1–13. https://doi.org/10.2217/ebo.11.18. K. M. Beck and J. Koo 9. Boyd AS, Menter A. Erythrodermic psoriasis. Precipitating factors, course, and prognosis in 50 patients. J Am Acad Dermatol. 1989;21(5):985–91. 10. Flindt-Hansen H, McFadden N, Eeg-Larsen T, Thune P. Effect of a new narrow-band UVB lamp on photocarcinogenesis in mice. Acta Derm Venereol. 1991;71(3):245–8. 11. Wulf HC, Hansen AB, Bech-Thomsen N. Differences in narrow-band ultraviolet B and broad-spectrum ultraviolet photocarcinogenesis in lightly pigmented hairless mice. Photodermatol Photoimmunol Photomed. 1994;10(5):192–7. 12. Lee E, Koo J, Berger T. UVB phototherapy and skin cancer risk: a review of the literature. Int J Dermatol. 2005;44(5):355–60. 13. Black RJ, Gavin AT. Photocarcinogenic risk of narrowband ultraviolet B (TL-01) phototherapy: early follow-up data. Br J Dermatol. 2006;154(3):566–7. 14. Hearn RMR, Kerr AC, Rahim KF, Ferguson J, Dawe RS. Incidence of skin cancers in 3867 patients treated with narrow-band ultraviolet B phototherapy. Br J Dermatol. 2008;159(4):931–5. 15. Whitmore SE, Morison WL. Melanoma after PUVA therapy for psoriasis. N Engl J Med. 1997;337(7):502–3. 16. Weischer M, Blum A, Eberhard F, Rocken M, Berneburg M. No evidence for increased skin cancer risk in psoriasis patients treated with broadband or narrowband UVB phototherapy: a first retrospective study. Acta Derm Venereol. 2004;84(5):370–4. 17. Man I, Crombie IK, Dawe RS, Ibbotson SH, Ferguson J. The photocarcinogenic risk of narrowband UVB (TL-01) phototherapy: early follow-up data. Br J Dermatol. 2005;152(4):755–7. 18. Sivanesan SP, Gattu S, Hong J, et al. Randomized, double-blind, placebo-controlled evaluation of the efficacy of oral psoralen plus ultraviolet A for the treatment of plaque-type psoriasis using the Psoriasis Area Severity Index score (improvement of 75% or greater) at 12 weeks. J Am Acad Dermatol. 2009;61(5):793–8. 19. Henseler T, Wolff K, Honigsmann H, Christophers E. Oral 8-methoxypsoralen photochemotherapy of psoriasis. The European PUVA study: a cooperative study among 18 European centres. Lancet. 1981;1(8225):853–7. 20. Snellman E, Klimenko T, Rantanen T. Randomized half-side comparison of narrowband UVB and ­trimethylpsoralen bath plus UVA treatments for psoriasis. Acta Derm Venereol. 2004;84(2):132–7. 21. Dawe RS, Cameron H, Yule S, et al. A randomized controlled trial of narrowband ultraviolet B vs bath-­ psoralen plus ultraviolet A photochemotherapy for psoriasis. Br J Dermatol. 2003;148(6):1194–204. 22. Gordon PM, Diffey BL, Matthews JN, Farr PM. A randomized comparison of narrow-band TL-01 phototherapy and PUVA photochemotherapy for psoriasis. J Am Acad Dermatol. 1999;41(5):728–32. 23. Yones SS, Palmer RA, Garibaldinos TT, Hawk JLM. Randomized double-blind trial of the treatment of chronic plaque psoriasis: efficacy of Psoralen-­ 8 Laser Therapy in Psoriasis Quinn Thibodeaux and John Koo Abstract Treatment options for psoriasis are numerous and span a wide array of modalities. For mild-­ to-­moderate disease, topical therapy may be adequate for complete control; however, moderate-­to-severe disease often requires systemic agents or phototherapy. Laser therapy is also a helpful tool in the armamentarium against psoriasis. For localized disease that is not adequately controlled with topicals or for lesions in difficult-to-treat areas, laser therapy may be an ideal treatment option. Lasers enable the delivery of therapeutic radiation to lesional skin only, thus sparing non-lesional skin from any possible adverse events. Over the past few decades, various lasing devices have been demonstrated to be safe and effective treatment options for psoriasis. The most commonly used devices include the 308-nm excimer laser and the 585 or 595-nm pulsed dye laser. Despite recent advances in targeted biologic therapy, lasers continue to play an important role in the management of psoriatic disease. Q. Thibodeaux (*) · J. Koo Department of Dermatology, University of California San Francisco, San Francisco, CA, USA e-mail: John.koo2@ucsf.edu © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_8 Key Learning Objectives 1. To understand the mechanism of action and types of laser therapy for psoriasis 2. To understand the appropriate use and efficacy of laser therapy for psoriasis 3. To understand the safety concerns of utilizing laser therapy for psoriasis Introduction Psoriasis is a chronic inflammatory disorder of the skin and nails that affects over 3% of adults in the United States [1]. The most common form of the disease is plaque psoriasis, which presents as scaly, erythematous lesions frequently involving the scalp, elbows, knees, umbilicus, and lower back. First line therapies for mild to moderate psoriasis include topical agents such as corticosteroids, vitamin D analogues, coal tar, and topical retinoids. For more severe disease, oral systemic agents, injectable biologics, or phototherapy may be required for adequate control. For localized disease or when limited plaques are recalcitrant to other therapies, laser therapy may be beneficial. When compared to non-­ lesional skin, psoriatic plaques are able to withstand greater doses of radiation without burning or blistering. Treatment with lasers allows the targeting of lesional skin with supraerythemo93 Q. Thibodeaux and J. Koo 94 genic doses (i.e. beyond the minimal erythema dose) of radiation leading to increased efficacy. Sparing of non-lesional skin helps to reduced treatment-related adverse effects such as burning, blistering, and hyperpigmentation. Laser therapy is also ideal for difficult-to-treat areas such as the scalp, lower legs, palms, soles, and intertriginous regions. Multiple lasers have been evaluated for their efficacy and safety in the treatment of psoriasis. In the following chapter, the literature regarding excimer, pulsed dye, Nd:YAG, and carbon dioxide lasers will be reviewed. Excimer Laser Background The excimer laser is the most widely-used targeted phototherapy device for psoriasis and is also used in the treatment of other dermatologic conditions including vitiligo, alopecia areata, and cutaneous T-cell lymphoma. The device was invented in 1970 and produces a monochromatic wavelength upon the dissociation of “excited dimers”, which gives the technology its name. A xenon dimer was used initially; however, further development resulted in a xenon-chloride (XeCl) dimer that produced a 308 nm wavelength within the ultraviolet B (UVB) spectrum (290–320 nm). In 1997, Bónis et al. were the first to report the use of excimer laser in the treatment of psoriasis. They found that in comparison to treatment with narrowband UVB (311 nm), the excimer laser was able to clear plaques of psoriasis in less time and with lower cumulative doses of radiation [2]. Mechanism of Action The mechanism of action of excimer laser in psoriasis is thought to be multifactorial and is drawn from studies analyzing other forms of UVB phototherapy. Numerous mechanistic theories have been proposed and include shifting of lesional cytokine profiles away from the Th1/Th17 axis, inducing apoptosis in epidermal and dermal kera- tinocytes and T lymphocytes, promoting local immunosuppression, and inducing cell-cycle arrest [3]. p53, a promotor of cell-cycle arrest and apoptosis has been found to be elevated in lesional skin following irradiation with excimer laser [4]. Efficacy A pair of early efficacy studies evaluated the differences between medium and high-dose treatment regimens. In the medium-dose study, 20 subjects received thrice weekly treatments for up to 8 weeks. The treatment regimen allowed possible dose increments of 25–30% per treatment. Of the 15 subjects who completed the protocol, >95% clearance was seen in an average of 10.6 treatments with a mean remission time (<25% disease recurrence) of 3.5 months [5]. The high-­ dose study treated 32 plaques in 16 patients with doses of 8 and 16 times their MED (minimal erythema dose). Following this single high-dose treatment, mean modified PASI scores decreased from 6.31 at baseline to 3.875 at follow-up 2 months later (P = 0.002) with partial clearing noted in 11 of the 16 after only 1 month [6]. The earliest large-scale study evaluating excimer laser in plaque psoriasis recruited 124 patients from multiple centers. Subjects were initially treated with an average of 3 MED and then twice weekly with dose adjustments in response to treatment. Seventy-two percent of patients who completed the protocol experienced at least 75% improvement of their lesions in an average of 6.2 treatments. In all, 84% of patients achieved 75% improvement in less than 10 treatments, with 50% reaching clearance of 90% or higher [7]. In another large, open-label study, 120 patients received an initial 3 MED dose followed by increases of 1 MED per session. Treatments were administered twice weekly for 3 weeks and then weekly until clearance was attained. Within 10 treatment sessions, 65.7% of subjects had achieved 90% improvement, with 85.3% reaching PASI90 after 13 sessions. The average length of treatment was 7.2 weeks [8]. 8 Laser Therapy in Psoriasis A smaller study evaluated 26 patients with macular-type and 14 patients with chronic plaque psoriasis. After an average of 13.7 treatment sessions, PASI improvements of 90.12 +/− 10.12% and 77.34 +/− 17.04% were noted in the macular and plaque groups, respectively [9]. Safety Judicious use of the excimer laser has proven to be a safe treatment option for psoriasis with no serious or long-term side effects reported. Common adverse events are similar to those of more generalized phototherapy but are limited to the areas receiving radiation. These common reactions include pain, burning, erythema, blisters, erosions, and hyperpigmentation and are typically mild and self-resolving [7, 8]. Although no studies exist on the relationship between the treatment of psoriasis with excimer laser and skin carcinogenesis, a retrospective cohort study of over 5000 Korean patients with vitiligo found no increased risk of actinic keratosis, non-melanoma skin cancers, or melanoma [10]. This relationship is likely to hold true for the treatment of psoriasis as well, as no reports have surfaced of an increased risk of skin cancer, even anecdotally, despite over 20 years of use. Dosing/Protocols To the authors’ knowledge, little data exists comparing the efficacies of various treatment ­regimens. In general, initial dosages are based on either a predetermined MED or by the induration of individual plaques. The MED is determined by applying increasing dosages of radiation to healthy, non-psoriatic skin and observing the first dose that creates a well-demarcated, minimally erythematous macule. In the earliest large, open-­ label study, initial dosages were one to three times the MED, regardless of plaque characteristics [7]. Other studies have utilized the induration component of the mPASI score to determine the initial dose for specific plaques independent of the patient’s skin type. Subsequent doses are then 95 altered in response to changes in induration [11]. In general, doses are increased or decreased based on response to therapy or the development of adverse events. Treatments should be separated by at least 48 h, and generally occur two to three times per week in most protocols. While the above protocols are most commonly used, other, more aggressive treatment algorithms have also been implemented successfully. Reports include a patient who achieved PASI 79 after only two treatments using a plaque-based sub-blistering dosimetry [12] and a cohort of 13 patients whose global PASI scores decreased by 42% 8 weeks after a single “TURBO” dose of 10 times their MED [13]. Higher fluences were noted to be more efficacious early in the development of the excimer laser, and Asawanonda et al. were the first to report quicker clearance and longer remission of psoriasis with high dose (8 and 16 times MED) versus medium (2, 3, 4, and 6 times MED) and low (0.5 and 1 times MED) dose treatments [14]. Few maintenance strategies have been evaluated. One protocol involved reducing the frequency of treatments after achieving PASI 75 to once per week for 4 weeks, then once every other week for 4 weeks, and then to one final treatment 4 weeks later. With this regimen, none of the five patients treated experienced >50% PASI worsening during the 3-month taper [15]. In general, remission times of 3–4 months have been reported following discontinuation of therapy [5, 6, 14]. ther Morphologies and Special O Populations In addition to chronic plaque psoriasis, treatment with excimer laser has also shown efficacy in other forms of psoriasis as well, including palmoplantar pustulosis [16, 17], TNF-alpha inhibitor-­ induced palmoplantar pustular psoriasis [18], inverse psoriasis [19, 20], scalp psoriasis [21, 22], and palmoplantar psoriasis [21, 23–25]. A side-by-side study with 10 patients compared the efficacy of excimer laser to topical PUVA for the treatment of palmoplantar psoriasis and found Q. Thibodeaux and J. Koo 96 near equivalent reductions in PASI after 5 weeks (−63.57% for excimer; −64.64% for topical PUVA) [26]. The safety and efficacy of excimer laser has also been demonstrated in a pediatric population [27]. Pulsed Dye Laser Background The pulsed dye laser (PDL) is a lasing device that is comprised of various combinations of organic dyes mixed in solvents. These combinations are “pumped” by an external high-energy source that Adjuvant Therapies excites the dye molecules, leading to the pulsed emission of radiation at a wavelength of 585 or The safety and efficacy of excimer laser has been 595 nm. At this wavelength, oxyhemoglobin acts shown to increase with the concomitant use of as the main target within the skin. The pulsed dye various topical medications. One right-versus-­ laser is mainly used in the treatment of vascular left study compared twice weekly excimer treat- lesions such as port wine stains, hemangiomas, ment to twice weekly excimer plus twice daily and telangiectasias; however, its efficacy in the calcipotriene ointment. By week 6 the average treatment of psoriasis has also been evaluated. PASI of the combination group (1.82 +/− 0.74) was significantly lower than that of the monotherapy group (5.08 +/− 1.65). In addition to bet- Mechanism of Action ter efficacy, the combination group also required significantly less cumulative radiation [28]. The pulsed dye laser exerts its biological effects Another study comparing excimer laser with based on selective photothermolysis, whereby a concomitant flumetasone/salicylic acid oint- specific targeted tissue is preferentially destroyed ment to excimer laser alone had similar results, leaving the surrounding tissues undamaged [32]. with the laser plus topical group experiencing In the case of PDL, the absorption of energy by greater improvement in PASI scores with lower oxyhemoglobin leads to the destruction of the average cumulative doses of phototherapy [29]. microvasculature. Because one of the defining Combinations of various topicals in addition to histologic features of psoriasis is dilated and torexcimer have also been evaluated. One study tuous capillaries in the dermal papillae, PDL is treated patients for 12 weeks with twice weekly able to treat psoriatic disease by ablating the vesexcimer laser in addition to alternating months of sels that supply blood, oxygen, and nutrients to twice daily clobetasol spray and calcitriol oint- skin lesions. ment. Of the 30 patients enrolled, 83% achieved Reductions in plaque severity scores have PASI75 by week 12 [30]. been correlated to decreases in plaque microvesExcimer laser has also been evaluated as an sel area following PDL treatment, with more adjuvant therapy to PUVA treatment. One study clinically significant improvement seen in evaluated 272 patients who were treated with plaques with larger pre-to-post treatment area either PUVA four times weekly monotherapy or reductions [33, 34]. One early histologic study PUVA four times weekly plus up to four suggested that a more tortuous or horizontal vasexcimer treatments within the first 2 weeks. cular pattern was associated with poorer response Overall efficacy between the two treatments to treatment in comparison to more vertically oriwas equivalent, with 67.3% of the monotherapy ented vessels [35]. Another immunohistochemigroup and 63.6% of the PUVA plus excimer cal evaluation found that the thermolytic effect of group achieving PASI 90; however, the PUVA PDL treatment was limited to the superficial capplus excimer group achieved PASI 90 in almost illary bed and did not affect the deeper vessels in half the treatment time (15 +/− 6 vs. 27 +/− the upper reticular dermis. In a similar pattern, a 7 days) and with significantly less cumulative reduction of CD4+ and CD8+ T cells was seen in UVA exposure [31]. the papillary dermis but not in the epidermis or 8 Laser Therapy in Psoriasis 97 upper reticular dermis [36]. Although thermolysis of superficial capillaries occurs as a result of plaque treatment with PDL, overall blood flow continues to be significantly elevated when compared to uninvolved skin, suggesting that deeper vessels are also involved in the pathogenesis of psoriasis [37]. Another study compared the efficacy of PDL, NB-UVB, and the combination of PDL plus NB-UVB versus no treatment. It found that while both modalities led to significant improvement by week 13, there was no significant difference between the two and the benefits of both modalities were not synergistic [44]. Efficacy Safety Hacker and Rasmussen were the first to report the efficacy of PDL in chronic plaque psoriasis. They treated a single plaque in 20 patients with fluences of 5.0, 7.0, and 9.0 J/cm2, leaving one quadrant as an untreated control. Eleven (57%) of 19 patients experienced positive effects after a single dose of the 9.0 J/cm2 fluence, with negligible improvement in quadrants treated with the 5.0 and 7.0 J/cm2 doses and no improvement in the control quadrant [38]. Following this proof-­ of-­concept, Katugampola et al. recruited eight patients with symmetric chronic plaque psoriasis and treated one plaque with PDL every 2 weeks for three sessions, leaving the corresponding control plaque untreated. At week 16, five of the eight patients showed improvement of >/= 50% of the treated plaque. One patient remained clear after 10 months of follow-up [39]. Another early study by Ros et al. found noticeable improvement in 6 of 10 treated patients after 1–3 sessions [40]. Zelickson et al. treated 36 patients across two treatment groups and found that PDL significantly improved plaque psoriasis in two to five sessions. No significant difference was noted between treatment with short (450 μs) and long (1500 μs) pulse-widths [35]. Two studies have compared PDL to topical active comparators. Each found that PDL significantly improved the targeted psoriatic plaques; however, one study found the active comparator to be superior (clobetasol propionate + salicylic acid) while the other found the active comparator to be inferior (calcipotriol/betamethasone) [41, 42]. One case series compared PDL to dermabrasion. Of the 11 patients treated with PDL, three experienced complete remission in comparison to five of six treated with dermabrasion [43]. Side effects of PDL are similar to those of other light-based therapies; however, because the modality directly targets and destroys the vasculature, more specific effects from the extravasation of red blood cells have been noted. The most common adverse events seen with PDL include petechia, purpura, pain, erosions, hyperpigmentation, hypopigmentation, and atrophic scarring [39, 40, 44]. Aside from rarely reported events of scarring, side effects are typically mild and short-lived. Other Morphologies de Leeuw et al. demonstrated that PDL may be effective for palmoplantar psoriasis. Their study involving 41 patients treated once every 4–6 weeks found that 76% of participants achieved >70% improvement in their lesions after an average of 4.2 treatments with over 50% experiencing greater than 90% improvement [45]. Much of the research into PDL and psoriasis has focused on the treatment of nail disease. A study comparing the efficacy of PDL and PDT (methyl-aminnolaevulinic acid occlusion for 3 hours followed by PDL as a light source) in the treatment of nail psoriasis found significant improvement in both nail matrix and nail bed disease for both modalities, with no significant difference noted between the two [46]. Another small study of 5 patients treated with monthly PDL for 3 months found improvements in both nail matrix (mean NAPSI from 7.0 +/− 1.4 to 2.2 +/− 0.7) and nail bed (mean NAPSI from 14.6 +/− 2.5 to 0.5 +/− 0.5) lesions [47]. A larger study involving 20 patients and 79 nails also showed Q. Thibodeaux and J. Koo 98 significant improvement in both nail bed and nail matrix NAPSI scores with no significant difference noted between longer (6 ms) and shorter (0.45 ms) pulse durations [48]. In a randomized, single-blind study involving 20 patients and 192 diseased nails, treatment with monthly PDL for 6 months resulted in significant improvement in both nail matrix and nail bed disease. In the treatment group, mean NAPSI scores decreased from 25.45 +/− 5.38 at baseline to 4.95 +/− 4.03 1 month after the last treatment session [49]. Nd:YAG Laser Background The Nd:YAG laser is a solid-state device that utilizes a crystal of neodymium-doped yttrium aluminum garnet as a lasing medium. The laser emits infrared radiation at a wavelength of 1064 nm and is used as an ablative device in multiple specialties including ophthalmology, oncology, urology, and dermatology; however, little evidence exists for its use in the treatment of psoriasis. Because the laser is able to penetrate up to 7 mm into the skin, it was postulated that improvement in psoriatic lesions could result from the destruction of deeper vasculature structures. Efficacy Ruiz-Esparza was the first to report the use of Nd:YAG laser in psoriasis in a 1999 case series. Using a 1320 nm Nd:YAG laser, he treated three patients with four sessions each and noted improvement in all irradiated lesions. No quantitative measurements or validated scales were used in this report [50]. van Lingen et al. compared monthly treatment with 1064 nm Nd:YAG to daily calcipotriol/betamethasone dipropionate (CBD) ointment for 3 months in four patients using an intrapatient left-to-right study design. Application of CBD ointment was found to significantly improve plaques at weeks 4, 8, and 12 of the study. Nd:YAG laser treatment using two different spot sizes significantly improved lesions at week 4, but these improvements were not maintained through the remainder of the study. They concluded that the laser was not “of additional value in the array of treatment modalities for chronic localized plaque psoriasis.” [51] Nd:YAG laser has also been evaluated for the treatment of psoriatic nail disease. Kartal et al. treated 16 patients with three monthly sessions of Nd:YAG and assessed for changes in NAPSI scores. Patients’ mean NAPSI score decreased from 26 +/− 7.2 at baseline to 22 +/− 6.6, 13 +/− 6, and 5.7 +/− 4.3 after treatment sessions one, two, and three, respectively. NAPSI scores at all timepoints were significantly improved from baseline [52]. Carbon Dioxide Laser Background The carbon dioxide (CO2) laser is a high-­powered lasing device that operates in the infrared spectrum with wavelengths generally ranging from 9.4 to 10.6 micrometers. At this wavelength, water absorbs and converts the energy to heat, leading to local tissue destruction. The device is used in soft-tissue surgery in multiple medical specialties and has rarely been utilized for psoriasis due to the high likelihood of adverse events and plaque recurrence. Efficacy Békássy and Astedt were the first to report the use of CO2 laser in the treatment of psoriasis. Their case series included three patients who underwent vaporization to approximately 1 mm of multiple plaques after the administration of local anesthesia. The lesions were allowed to heal over 4–6 weeks, and remission times of 3–5 years were reported; however, their recovery was complicated by two instances of bacterial superinfection that required systemic antibiotics [53]. Alora et al. compared the effects of varying the depth of ablation by using curettage, debridement, and 8 Laser Therapy in Psoriasis increasing numbers of CO2 laser passes in separate quadrants of individual plaques. In one cohort, five of six patients noted recurrence of their entire plaque within 8 weeks, while the final patient noted recurrence after 12 weeks. In the second cohort, four of six patients experienced relapse by 8 weeks, but two subjects experienced no recurrence 4 months after treatment [54]. Another study compared the effects of CO2 laser and electrodesiccation and curettage within a single plaque. Asawanonda et al. found that treatment with CO2 laser and ED&C both significantly improved psoriatic lesions over controls at 4 months of follow up; however, this improvement was then lost 2 months later. No significant differences were noted between treatment with CO2 laser versus ED&C at any timepoint [55]. Safety Adverse events have been frequently reported with CO2 laser therapy and include secondary wound infection, depigmentation, and hypertrophic scarring [53, 55]. Comparative Studies One recent single-blinded left-to-right study compared excimer laser to pulsed dye laser in the treatment of nail psoriasis. Forty-two patients underwent twice weekly excimer laser treatment to right hand nails and pulsed dye laser (PDL) once every 4 weeks to left hand nails. Mean NAPSI scores in the excimer group improved from 29.8 at baseline to 16.3 at week 24, while mean NAPSI scores in the PDL group improved from 29.5 at baseline to 3.2 at week 24. NAPSI improvement in the PDL group was significantly greater than in the excimer group with minimal side effects [56]. Another study involving 22 patients with plaque psoriasis compared twice weekly excimer laser versus PDL with salicylic acid pretreatment every 4 weeks and found that mean PASI improvement was higher in the excimer-treated plaques (4.7 SD 2.1) than in the PDL-treated plaques (2.7 SD 2.4) [57]. An intra- 99 patient left-to-right study comparing PDL and Nd:YAG laser for the treatment of nail psoriasis found no statistical difference between reductions in NAPSI scores between the two modalities when each was combined with calcipotriol/ betamethasone gel [58]. References 1. Rachakonda TD, Schupp CW, Armstrong AW. Psoriasis prevalence among adults in the United States. J Am Acad Dermatol. 2014;70(3):512–6. 2. Bonis B, Kemeny L, Dobozy A, Bor Z, Szabo G, Ignacz F. 308 nm uvb excimer laser for psoriasis. Lancet. 1997;350(9090):1522. 3. Wong T, Hsu L, Liao W. Phototherapy in psoriasis: a review of mechanisms of action. J Cutan Med Surg. 2013;17(1):6–12. 4. Bianchi B, Campolmi P, Mavilia L, Danesi A, Rossi R, Cappugi P. Monochromatic excimer light (308 nm): an immunohistochemical study of cutaneous t cells and apoptosis-related molecules in psoriasis. J Eur Acad Dermatol Venereol. 2003;17(4): 408–13. 5. Trehan M, Taylor CR. Medium-dose 308-nm excimer laser for the treatment of psoriasis. J Am Acad Dermatol. 2002;47(5):701–8. 6. Trehan M, Taylor CR. High-dose 308-nm excimer laser for the treatment of psoriasis. J Am Acad Dermatol. 2002;46(5):732–7. 7. Feldman SR, Mellen BG, Housman TS, Fitzpatrick RE, Geronemus RG, Friedman PM, Vasily DB, Morison WL. Efficacy of the 308-nm excimer laser for treatment of psoriasis: results of a multicenter study. J Am Acad Dermatol. 2002;46(6):900–6. 8. Gerber W, Arheilger B, Ha TA, Hermann J, Ockenfels HM. Ultraviolet b 308-nm excimer laser treatment of psoriasis: a new phototherapeutic approach. Br J Dermatol. 2003;149(6):1250–8. 9. He YL, Zhang XY, Dong J, Xu JZ, Wang J. Clinical efficacy of a 308 nm excimer laser for treatment of psoriasis vulgaris. Photodermatol Photoimmunol Photomed. 2007;23(6):238–41. 10. Bae JM, Eun SH, Oh SH, Shin JH, Kang HY, Kim KH, Lee SC, Choi CW. The 308-nm excimer laser treatment does not increase the risk of skin cancer in patients with vitiligo: a population-based retrospective cohort study. Pigment Cell Melanoma Res. 2019; 11. Taneja A, Trehan M, Taylor CR. 308-nm excimer laser for the treatment of psoriasis: induration-based dosimetry. Arch Dermatol. 2003;139(6):759–64. 12. Debbaneh MG, Levin E, Sanchez Rodriguez R, Leon A, Koo J, Rosenblum MD. Plaque-based sub-­blistering dosimetry: reaching pasi-75 after two treatments with 308-nm excimer laser in a generalized psoriasis patient. J Dermatolog Treat. 2015;26(1):45–8. 9 Traditional Systemic Therapy I: Methotrexate and Cyclosporine Erin Boh, Andrew Joselow, and Brittany Stumpf Abstract This chapter will review the traditional systemic drugs, methotrexate and cyclosporine, both of which are used in treating patients with moderate to severe or recalcitrant psoriasis. The pharmacology and use of these drugs in the treatment of patients with psoriasis will be covered as well as the parameters to screen patients, monitoring guidelines and adverse effects associated with each. Key Learning Objectives 1. To understand the mechanism of action of methotrexate and cyclosporine. 2. To understand the appropriate use and efficacy of methotrexate and cyclosporine. 3. To understand the safety concerns and appropriate monitoring for methotrexate and cyclosporine. E. Boh (*) · A. Joselow · B. Stumpf Department Dermatology, Tulane University School of Medicine, New Orleans, LA, USA e-mail: eboh@tulane.edu; ajoselow@tulane.edu; boswald@tulane.edu © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_9 Introduction Psoriasis is a chronic inflammatory disease affecting over seven million individuals. It can be debilitating and associated with systemic co-­ morbidities such as arthritis, hypertension, hyperlipidemia and insulin resistance [1–3]. Recently, it has been suggested that psoriasis may be associated with or part of metabolic syndrome [4–6]. In addition to topical therapy and phototherapy, systemic agents, including a newly developed class referred to as biologics, have been used for its treatment. Systemic therapies are often reserved for patients with moderate to severe psoriasis. The goals of treatment are to quickly clear or reduce psoriatic lesions, to make the patient comfortable by alleviating symptoms and to provide long-term maintenance with minimal toxicity for the patient. In the past, systemic therapy has been used for those individuals with moderate to severe psoriasis as defined by greater than 10% body surface area or debilitating disease. With the development of biologic agents, the treatment algorithm has changed tremendously, giving us additional therapeutic options that are potentially less toxic to the liver, kidneys, and bone marrow and are not teratogenic. There still remains a place for traditional agents for some patients with moderate to severe disease. While the biologics have a very good safety and efficacy profile, they have not totally replaced traditional agents for the 103 E. Boh et al. 104 treatment of psoriasis. Traditional systemic therapies continue to play an important role in the treatment of psoriasis with their oral route of administration and low cost (compared with biologics) making them an important treatment option in the appropriate patient. Contraindications due to comorbid conditions, insurance restrictions as well as cost may be limiting factors for the use of these biologic agents. This chapter will review the traditional systemic drugs, methotrexate and cyclosporine, and how they can be used as monotherapy or even adjunctive therapy (combination therapy) in treating patients with moderate to severe or recalcitrant psoriasis. Methotrexate Introduction Methotrexate is the most widely used systemic treatment for moderate to severe psoriasis. The National Psoriasis Foundation and American Academy of Dermatology recently published detailed dosing and monitoring guidelines [7, 8]. Methotrexate is a synthetic analogue of folic acid, which exhibits immunosuppressive as well as anti-inflammatory properties. It has been used successfully to treat psoriasis for over 50 years. While Gubner et al. in 1951 [9] first reported clearance of psoriasis in cancer patients Fig. 9.1 Structure of methotrexate (Methotrex ate|C20H22N8O5 https:// pubchem.ncbi.nlm.nih. gov/compound/ Methotrexate. Accessed June 14, 2019. Source: U.S. National Library of Medicine) being treated with aminopterin, a precursor to methotrexate, Edmunson and Guy first reported methotrexate’s efficacy in treating psoriasis in 1958 [10]. The FDA approved methotrexate in 1972 for the indication of psoriasis [11]. Interestingly, it was not until the early 1980s that methotrexate was approved for rheumatoid arthritis. Since these early years, methotrexate has become the “gold standard” for treating moderate to severe psoriasis and psoriatic arthritis [12]. Over the years, dosing has been modified to minimize the toxicity associated with methotrexate [7, 8]. Mechanism of Action Methotrexate has anti-proliferative, anti-­ inflammatory and immunosuppressive actions. As a synthetic analogue of folic acid, methotrexate competitively inhibits dihydrofolate reductase, an enzyme responsible for conversion of folic acid to reduced folate (tetrahydrofolate) (Fig. 9.1). Tetrahydrofolate is required for the transfer of 1-carbon units in the thymidylate and purine synthesis pathway [13]. This inhibition blocks synthesis of deoxythymidylic acid, which is required for DNA synthesis. As a consequence, methotrexate causes the arrest of cell division in the S phase. Studies have shown that the anti-proliferative effects of high-dose methotrexate can be reversed H H N N N N N N H N O H N H O O O O H H 9 Traditional Systemic Therapy I: Methotrexate and Cyclosporine 105 by high doses of folic acid, thus supporting the standing, methotrexate appears to have multiple mechanism of inhibition of DNA synthesis [13, mechanisms of action at play. Methotrexate has 14]. Low-dose methotrexate may exhibit other been shown to help restore a typical immune balmechanisms of action, including anti-­inflammatory ance of T-cells by lowering the level of Th1 and and immunomodulatory effects. This observation Th17 cells and increasing Th2 and T-reg cells – is supported by the widespread practice by derma- essentially shifting from a pro-inflammatory to tologists and rheumatologists of administering low an anti-inflammatory T-cell phenotype [19]. doses of folic acid (i.e. 1 mg per day) to patients on methotrexate to minimize adverse effects without compromising efficacy [15]. Absorption and Bioavailability While high-dose methotrexate inhibits DNA synthesis, primarily through the inhibition of Methotrexate may be given orally, intramuscudihydrofolate reductase, lower doses as used in larly, subcutaneously or intravenously. Food treatment of psoriasis have more anti-­intake does not influence absorption in adults but inflammatory and immunomodulatory effects. may in children [14]. Once absorbed, methotrexThese anti-inflammatory effects seem to involve ate is actively transported into cells where it is the formation of intracellular metabolites of metabolized into polyglutamate imidazole prodmethotrexate [13, 16]. ucts, which appear to be the more active drug. Once absorbed, methotrexate is metabolized These products accumulate, resulting in increased intracellularly to polyglutamate derivatives, adenosine contributing both to toxicity and effipotent inhibitors of a number of folate-dependent cacy [16]. Current data suggests that methotrexenzymes leading to the accumulation of intracel- ate is metabolized intracellularly, including in the lular imidazole products. These inhibit adenosine liver, to form polyglutamate metabolites that deaminase, ultimately resulting in accumulation exert effects on various tissues. of adenosine [16]. Adenosine suppresses the Peak levels are attained fairly rapidly, approxisecretion of inflammatory cytokines by macro- mately one hour after oral administration, slower phages and neutrophils, as well as diminishes the relative to other modes of administration. expression of adhesion molecules. Additionally, Absorption after ingestion may be affected by adenosine, through binding to the A2 receptor, dosing: higher doses having incomplete or variexhibits potent anti-inflammatory effects by able absorption. Steady-state levels of methotrexinhibiting neutrophil leukotriene synthesis [13]. ate may be more reliable in smaller doses over the Activated T cells play a pivotal role in psoriasis, course of 12–24 h one day per week. Plasma levand it appears that methotrexate not only inhibits els have a triphasic response. Once the drug is DNA replication of these T-cells but also dimin- absorbed, plasma levels decrease rapidly within ishes antigen-stimulated T-cell proliferation. By the first hour, reflecting distribution throughout inhibiting mitogen-activated T-cells, these cells body tissues. The second phase of reduction become more susceptible to apoptosis [17]. reflects renal excretion over the next 2–4 h. Johnston et al. recently refuted this notion of Importantly, renal function such as glomerular filincreased susceptibility to apoptosis [18]. These tration and tubular secretion can significantly authors propose that methotrexate exerts its affect blood levels. The final phase occurs through effects through both folate-dependent (decreased slow release from the tissues over 10–27 h after synthesis of antigen stimulated T-cells) and ingestion reflecting its half-life (T ½). folate-independent mechanisms (altered adheApproximately 50% of methotrexate is transsion molecule expression through alterations in ported bound to plasma proteins. The free or adenosine). Most likely, the benefits seen involve unbound portion is the active form of the drug. both processes, and methotrexate should be Plasma levels can be affected by the binding of viewed as an immunosuppressing and immuno- other drugs to these plasma proteins either modulating agent. At this point in our under- ­displacing or blocking methotrexate, resulting in E. Boh et al. 106 elevated blood levels. Drug interactions will be discussed below. Orally administered MTX has been found to have a saturable intestinal absorption and nonlinear pharmacokinetics, with significant consequences on drug bioavailability and clinical efficacy. The current evidence shows that parenterally administered MTX results in rapid and complete absorption, higher serum levels, and less variable exposure than oral dosing. The use of parenteral MTX, particularly when administered as a subcutaneous (SC) injection, has recently raised great interest in order to overcome the limitations of oral MTX. Various clinical experiences have suggested that SC MTX is more effective than oral MTX and may provide significant benefit even in patients in whom oral MTX proved to be inadequate. The increased efficacy of SC MTX resulting from higher drug exposure compared with oral MTX has been associated with a similar safety profile and in various reports even with a lower frequency of gastrointestinal complaints [20]. In the future, novel colloidal drug delivery systems may provide advantages over prior formulations with improved skin permeability and targeted delivery systems may improve efficacy, and decrease toxicity in psoriasis patients [21]. psoriasis including specific guidance on dosing, route of administration, risk and monitoring of liver toxicity [8, 24]. There are several important parameters to consider when choosing methotrexate as a form of therapy. These include the severity of the disease, the patient’s co-morbidities such as associated arthritis, socioeconomic factors, and the ability of the patient to participate in treatment. As a general rule, methotrexate is suggested for patients with at least 10% body surface area (BSA) affected, who have failed or can no longer perform topical therapy, or who have failed or have contraindications to phototherapy. Absolute and relative contraindications to methotrexate therapy, listed in Table 9.1, should be reviewed carefully and discussed with the patient prior to initiation of the drug. Considerations for patient selection should be based on patient history, laboratory findings and co-morbidities. Absolute contraindications mandate an alternative form of therapy. Methotrexate is a teratogen, and women of childbearing potential should use birth control. It is contraindicated in pregnancy as well as during breastfeeding. Men should also be counseled on not impregnating a woman until 3 months off therapy. Relative contraindications include Type Table 9.1 Contraindications to methotrexate therapy Use in Psoriasis Methotrexate is FDA-approved for the treatment of moderate and moderate-to-severe psoriasis in adults and psoriatic arthritis [13]. The first treatment guidelines on the use of MTX in psoriasis were published by Roenigk et al. in 1973 [22]; and, since that time, many newer treatment guidelines for the use of methotrexate and other systemic agents in psoriasis have been published [7, 8, 14, 23]. The most recent guidelines for the treatment of psoriasis with methotrexate were published by the National Psoriasis Foundation and by the American Academy of Dermatology in 2019 [7, 8]. These guidelines, as well as a systematic review of the literature, has led to evidence-based recommendations on the use of methotrexate in Absolute contraindications Excessive alcohol consumption resulting in liver damage Alcoholic liver disease or other chronic liver diseases, including hepatitis B or C Bone marrow abnormalities: Anemia, thrombocytopenia, leukopenia Immunodeficiency Nursing mothers Pregnancy Methotrexate hypersensitivity Active infection Relative contraindications Renal insufficiency Advanced age Alcohol consumption: History of or current alcohol consumption Peptic ulcer disease Concomitant use of hepatotoxic drugs Family history of inheritable liver disease Diabetes mellitus Hyperlipidemia Morbid obesity Active infection 9 Traditional Systemic Therapy I: Methotrexate and Cyclosporine 2 diabetes, insulin resistance, alcohol consumption, obesity and hypertriglyceridemia. As a general rule, these patients should be evaluated for other treatments. Patients with relative contraindications can use methotrexate with appropriate precautions. For instance, patients with peptic ulcer disease may be treated with concomitant proton pump inhibitor therapy. The decision to treat should be based on the patient’s individual situation. While used to treat psoriatic arthritis for years, the SEAM-PsA trial is only one of a few studies that suggest methotrexate (20 mg/week) is beneficial as a monotherapy in treatment-naïve psoriatic arthritis patients or in combination with a biologic [25]. Similarly, Appani et al., found methotrexate initiation (>15 mg/week) resulted in significant improvement in dactylitis along with other psoriatic arthritis symptoms [26]. Dosing Methotrexate is generally given as a single weekly oral dose administered over a 12 h period. It is available as a 2.5 mg, 7.5 mg, 10 mg, 15 mg tablets and an injectable 25 mg/ml solution. Intramuscular or subcutaneous administration is helpful when there is gastrointestinal intolerance to oral dosing or if there are concerns regarding patient compliance. Subcutaneous injection is equally effective and can be self-administered at home. Dosing is equivalent whether the oral or injectable form is used. Many practitioners give a single test dose of 2.5 or 5 mg to evaluate for significant bone marrow suppression or acute hepatic injury in susceptible patients. Although there are no established maximum or minimum dosages of methotrexate, weekly dosages usually range from 7.5 to 25 mg. After the test dose of 5 mg of methotrexate, blood tests (complete blood count with platelets and renal and liver function tests) are obtained 5–6 days later. If blood work is within normal limits, treatment doses are started at 10–15 mg once weekly. Methotrexate can be increased by 107 2.5–5 mg every 2–4 weeks until a response is seen, up to approximately 25 mg weekly, the maximum recommended dose. There are published reports suggesting that folic acid protects against gastric upset, nausea, oral ulcerations and may even impact bone marrow toxicity, especially in rheumatoid arthritis patients [15]. A recent study found drug-survival, the time period of treatment with a specific drug before its cessation, was significantly higher in those patients whose methotrexate therapy was supplemented with folic acid compared to those who received methotrexate alone (27.6 months vs 18.5 months, respectively) [27]. Therefore, a 1 mg per day folic acid supplement is recommended based on expert evaluation [15]. Some practitioners recommend not taking folic acid on the day patients dose with methotrexate, but the significance of this has not been studied. All dosing schedules should be adjusted to the individual patient to obtain or maintain adequate disease control with minimal side effects. In order to ensure some degree of safety, monitoring should be done on a continuous basis. Vena et al. reviewed the current literature on use of subcutaneous vs oral methotrexate for patients with inflammatory arthritis and psoriasis [20] .These authors concluded that current evidence indicates that SQ administration of MTX is characterized by linear and predictable pharmacokinetics resulting in higher bioavailability as compared with oral methotrexate, especially with weekly doses of 15 mg. Subcutaneous MTX is useful for those with GI intolerability or for those individuals who have not gotten maximal response from orally dosed levels of MTX. Monitoring Guidelines Prior to initiation of methotrexate treatment, the patient should have pretreatment blood tests, including liver function tests, creatinine, hepatitis B and C screening, tuberculosis screening, a complete medical history with emphasis on medications and co-morbidities, and a complete 108 Table 9.2 Monitoring methotrexate guidelines Baseline History and physical Careful review of drug history and evaluation of low risk versus high-risk patients Laboratory Complete blood count with differential and platelet counts (CBC) Comprehensive metabolic panel- renal and hepatic (CMP) Hepatitis B and C screening Tuberculosis screening with PPD or blood test Serum or urine pregnancy screening On-going monitoring Periodic review of drug history Physical exam Laboratory CBC with differential and platelets q month for 3 months then every 3 months CMP q month for 3 months, then every 3 months Tuberculosis screening every year physical examination performed. Patients should be counseled on methotrexate and its potential side effects. Documentation should include the type of psoriasis, past treatments, patient’s quality of life measures, the body surface affected, risk factors for hepatotoxicity and other potential toxicities. Patients taking methotrexate should be monitored regularly for potential organ toxicity. Baseline and ongoing monitoring guidelines are listed in Table 9.2. Monitoring of the blood parameters should be done approximately 2–4 weeks after adjusting the dose. It is important to remain at the minimal effective dose and to record the total cumulative dose of methotrexate while maintaining disease control and medication tolerance. It is also important to consider a liver biopsy in those patients on long-term therapy, or those with significant risk for hepatotoxicity. While practitioners have their individual styles of practice, it is advisable to perform a periodic history and physical exam on patients on methotrexate therapy to ensure the highest quality of care and to minimize potential adverse events. If laboratory abnormalities occur, blood tests may be repeated and more frequent monitoring may be necessary. Liver function tests E. Boh et al. Table 9.3 High risk factors for hepatic toxicity from methotrexate Risk factors History of or current alcohol consumption Persistent abnormal liver chemistry studies History of liver disease, including chronic hepatitis B or C Family history of inheritable liver disease Diabetes mellitus Obesity History of significant exposure to hepatotoxic drugs or chemicals Potential significant drug interactions Lack of folate supplementation Hyperlipidemia should be drawn at a 5-day interval from the last dose since values may be elevated 1 to 2 days after ingestion of methotrexate. If a significant persistent abnormality in liver chemistry develops, methotrexate therapy should be withheld for 1 to 2 weeks and repeat blood work should be performed. Discuss with the patient if any new medications were prescribed or other situations have developed. Liver Biopsy Guidelines for liver biopsy in psoriasis patients were published in 1998 and updated in the American Academy of Dermatology (AAD) guidelines for treatment of psoriasis in 2009 [23, 24, 28]. Initially, guidelines suggested liver biopsy at the start of therapy and after every 1.5 gram cumulative dose thereafter [28]. The AAD and National Psoriasis Foundation guidelines for monitoring have been updated to differentiate patients at low risk for hepatotoxicity from those at high risk [7, 8]. Table 9.3 outlines those patients at high risk for hepatotoxicity. For low-risk patients, methotrexate can be initiated without performing a baseline liver biopsy. A liver biopsy should be performed after approximately a 3.5– 4.0 gram cumulative dose. High-risk patients should obtain a liver biopsy after approximately a 300–600 mg cumulative dose to ensure a response to therapy before subjecting the patient to the risks of a biopsy. Table 9.4 summarizes the grading system proposed by Roenigk et al. for interpretation of liver biopsy histologic results [28]. 9 Traditional Systemic Therapy I: Methotrexate and Cyclosporine Table 9.4 Evaluation of liver biopsy findings [25] Grade I II IIIA Findings Fatty infiltration; mild; nuclear variability; mild; portal inflammation Moderate to severe fatty infiltration; moderate to severe nuclear variability; portal tract expansion; portal tract inflammation and necrosis Mild fibrosis; slight enlargement of portal tracts without disruption IIIB Moderate to severe fibrosis; cirrhosis IV Cirrhosis Disposition Normal Continue methotrexate Continue methotrexate Fibrosis Discontinue methotrexate Repeat biopsy sooner Fibrosis Discontinue methotrexate Cirrhosis Discontinue methotrexate As a general rule, patients with grade I and II can continue methotrexate. Those with grade IIIA may cautiously continue therapy with more stringent monitoring if better alternatives are not available. Only those patients without other options should continue methotrexate. Patients with grade IIIB and IV should discontinue the drug regardless. It is important to closely work with a hepatologist who is also up to date with the current published guidelines. Although noninvasive tests for monitoring MTX-induced hepatic fibrosis have been studied and shown to be useful in reducing the number of liver biopsies, no single test has emerged as a replacement for liver biopsy. A family of related noninvasive tests for hepatic fibrosis is available; these tests consist of panels of serum biomarkers and patient biometrics that are put into patented mathematical models that generate scores predictive of hepatic fibrosis. The nonalcoholic steatohepatitis (NASH) FibroSure test (LabCorp) is similar to the FibroTest used for assessing fibrosis in Hepatitis C infection but was specifically developed for patients with NAFLD. With a computational algorithm to provide a quantitative surrogate marker of liver fibrosis, steatosis, and nonalcoholic steatohepatitis (NASH), the NASH 109 FibroSure test uses 10 biochemical markers in combination with age, sex, height, and weight. The NASH FibroSure test is reported to have a sensitivity of 83% and a specificity of 78% in detecting risk of significant fibrosis and a sensitivity of 71% and a specificity of 72% in identifying risk of significant steatosis in patients with NAFLD [29]. Bauer et al. postulate that a liver biopsy cannot be entirely replaced by the NASH FibroSure test; they do, however, support the idea that the number of liver biopsies can be significantly reduced by the use of noninvasive tests such as the NASH FibroSure [30]. Adverse Effects There are a number of common and uncommon adverse effects reported with methotrexate usage. Common minor complaints by patients include fatigue, nausea, vomiting, headache and stomatitis. Many of these adverse effects are eliminated by administration of folic acid. If gastrointestinal symptoms persist, methotrexate can be administered by injection, subcutaneously or intramuscularly. Dosing may also be administered in divided doses over a 24-h period. The three primary areas of potential major toxicity include hepatotoxicity, myelopsuppression and pulmonary fibrosis. Hepatotoxicity There are both acute and chronic hepatotoxic adverse effects. Acute hepatocellular damage, as manifested by elevated liver enzymes, may result from high blood levels of methotrexate. High blood levels occur when the dose exceeds 25 mg per week or when levels are increased by a significant drug interaction or displacement of bound methotrexate by another medication. Acute hepatocellular damage almost always results in abnormal liver enzyme studies. However, chronic damage to the liver may occur even when liver blood studies remain normal. Chronic hepatotoxicity results from the cumulative effects of methotrexate on the portal system resulting in fibrosis. The histologic damage and subsequent fibrosis can only be assessed by liver E. Boh et al. 110 biopsy. As discussed above, biopsy of the liver can be delayed in those patients who are at low risk of hepatotoxicity but should be done at an earlier interval if the patient is at high risk. The histopathologic features of methotrexate-induced liver toxicity resemble nonalcoholic steatohepatitis (NASH), a similar pattern observed in patients who are obese, hyperlipidemic or diabetic. Methotrexate likely aggravates preexisting steatohepatitis. As stated, the risk of methotrexate-associated liver damage is increased in patients with preexisting risk factors such as obesity and hypercholesterolemia. While older studies estimate the risk of elevated transaminases and elevations twice the upper limit of normal as nearly 50% and 17% respectively, recent studies suggest, with appropriate monitoring and dosing, the risk may be significantly lower [31]. In their recent review, Conway et al. [31] noted a more accurate level of risk for elevated transaminases and those elevated beyond twice the upper limit of normal is likely closer to 20% and 1% respectively. Serum assays for assessing liver fibrosis, such as the measurement of the amino-terminal peptide of procollagen III, have been used in Europe as an alternative to liver biopsy but are not currently available in the United States [32]. Currently, there are no blood assays or diagnostic tests adequate to monitor for chronic liver toxicity other than biopsy or FibroSure testing. methotrexate therapy as well as monitor while on therapy to minimize these risks. If anemia, thrombocytopenia or leucopenia occurs acutely, it can be reversed with folic acid administration or in severe cases, folinic acid (leucovorin) rescue. High dose folic or folinic acid can be given by mouth or intravenously at doses of 15 mg every 6 h for 1 to 2 days or until the methotrexate levels approach zero. Administration of daily folic acid while on methotrexate may minimize gastrointestinal and liver toxicity, but its impact on the bone marrow toxicity remains controversial [15]. Pulmonary Fibrosis Pulmonary fibrosis and interstitial pneumonitis may uncommonly occur in patients with psoriasis being treated with methotrexate, but is more common in patients being treated for rheumatoid arthritis. Pulmonary fibrosis is more commonly associated with high dose methotrexate therapy. Patients should be monitored for signs and symptoms such as dry cough, dyspnea at rest and low-­ grade fever. Other very uncommon pulmonary complications include cryptogenic organizing pneumonia (COP), pleuritis and pleural effusions. Drug Interactions Methotrexate has many reported and presumptive drug interactions that may result in decreased or increased levels of methotrexate and potentiate end-organ toxicity. Therefore, it is very important Myelosuppression Myelosuppression is potentially a very signifi- to take a complete medication history as part of cant toxicity associated with methotrexate admin- pre-methotrexate screening. It is also important istration. Methotrexate can cause leukopenia, to counsel patients regarding possible drug interthrombocytopenia and anemia. It is usually dose-­ actions, especially with regards to over-the-­ dependent and due to direct toxic action on the counter and commonly prescribed drugs. bone marrow. Rarely, there is an idiosyncratic Table 9.5 has some of the common potential drug myelosuppression, which occurs early in treatment. Idiosyncratic reactions may be more likely Table 9.5 Common drug Interactions with methotrexate in patients with advanced age, renal insufficiency, Trimethoprim Phenothiazines underlying bone marrow disease, hypoalbumin- Sulfonamides Salicylates emia, concomitant medications or folate defi- Chloramphenicol NSAIDs Tetracyclines Systemic Retinoids ciency. For this reason, physicians administer the Dapsone Probenacid test dose of methotrexate. It is imperative to Phenytoin Triamterene screen patients appropriately before starting 9 Traditional Systemic Therapy I: Methotrexate and Cyclosporine interactions. There are numerous medications that increase or decrease blood methotrexate levels either by displacing it from protein binding sties, blocking its binding, and altering the metabolism of it through the cytochrome P-450 system. Other medications may also have an additive effect on end-organ damage. and originally used as an immunosuppressive agent in organ transplantation, it was first shown to be effective for psoriasis in 1979 [33]. Cyclosporine primarily acts by inhibiting T-cell function and interleukin IL-2 via binding to cyclophyllin. The original formulation (Sandimmune) had considerable variations in absorption and bioavailability. Newer microemulsion formulations (Neoral/ cyclosporine modified) have better and more predictable bioavailability and are more cost effective. Neoral was FDA approved in 1997 for psoriasis and rheumatoid arthritis. Cyclosporine is considered safe and effective for psoriasis. However, there is still reticence about its use in psoriasis. It has a particularly useful role in managing psoriatic crises due to its rapid onset and its efficacy in treating psoriasis unre- Cyclosporine Introduction Cyclosporine, a cyclic peptide of 11 amino acids, was isolated from the soil fungus Tolypocladium inflatum Gams in 1970 (Fig. 9.2). CSA is a highly effective and rapidly acting systemic agent for the treatment of psoriasis. Discovered in 1970 Fig. 9.2 Structure of cyclosporine (Cyclospor ine|C62H111N11O12 https:// pubchem.ncbi.nlm.nih. gov/compound/5280754. Accessed June 14, 2019. Source: U.S. National Library of Medicine) 111 O O H N N H O O N N H O N N O O N N O N N H N O O O H H H O E. Boh et al. 112 sponsive to other modalities, bridging to other therapies, and treating psoriasis within a rotational scheme of other medications [34]. Appropriate patient selection and monitoring will significantly decrease the risks of side effects. Despite published guidelines for use in psoriasis, there still remains controversy and disagreement over treatment strategy and monitoring [34]. In contrast to other traditional agents such as methotrexate and hydroxyurea, cyclosporine is not remarkably cytotoxic, does not suppress the bone marrow and is not teratogenic. It does have potential significant renal side effects, including hypertension. Numerous clinical trials have demonstrated the efficacy of CSA in psoriasis. The original studies evaluating CSA’s efficacy in psoriasis provided evidence that psoriasis is a T-cell driven immunologic disorder rather than a disorder of altered keratinocyte growth and differentiation. CSA given at 2.5 to 5 mg/kg/d for 12 to 16 weeks leads to rapid and dramatic improvement in psoriasis in up to 80% to 90% of patients [35]. When dosed at 3 mg/kg/d, CSA leads to a PASI 75 response in 50% to 70% of patients and a PASI 90 response in 30% to 50% of patients [36]. A large, well-conducted systematic review published in 2016 found no significant difference in efficacy between CSA and MTX in PASI or PGA in the treatment of psoriasis [37]. transcription of proinflammatory genes, most notably IL-2, and the up-regulation of the IL-2 receptor [38]. As a consequence, T-cell activation and production of inflammatory cytokines is impaired. It is now widely accepted that psoriasis is mediated by these activated T cells and their cytokine products. Cyclosporine appears to also dampen expression of intercellular adhesion molecule (ICAM)-1 on keratinocytes and endothelial cells. Cyclosporine also decreases tumor necrosis factor, a key cytokine in psoriasis pathogenesis, and suppresses the Th17 genes, IL-17, IL-22 and the IL-23p19 subunit, all of which are overexpressed in psoriatic skin [39]. Absorption and Bioavailability Cyclosporine is lipophilic and exhibits very poor solubility in water. As a consequence, suspension and microemulsion forms of the drug have been developed for oral administration and for injection. The first formulation, Sandimmune, was produced in 1983. It had variable and unpredictable bioavailability among patients secondary to its high dependence on bile solubility. This was improved by the introduction of a microemulsion formulation, Neoral, in July 1995. Several other cyclosporine modified formulations have subsequently been brought to market. The bioavailability of generic cyclosporine is approximately 30% that may be slightly increased with some branded versions. Cyclosporine capsules and liquid oral Mechanism of Action formulations are considered bioequivalent. There Cyclosporine primarily acts by inhibiting T-cell are few published reports comparing bioavailfunction and interleukin (IL-2) [38]. After an ability and efficacy among different generic forantigen-presenting cell binds to a T cell, intracy- mulations although the perception that differences toplasmic levels of calcium increase leading to in these agents exists is prevalent [40]. Since calmodulin activation of calcineurin phospha- cyclosporine has a narrow therapeutic window, tase. Calcineurin phosphatase dephosphorylates careful monitoring and consistency in generic cytoplasmic nuclear factor of activated T cells formulations is required. Cyclosporine is absorbed in the small intesallowing translocation into the nucleus and enabling transcription of proinflammatory genes, tine with peak concentrations occurring from including IL-2, IL-4, interferon gamma and 1–8 h. The absorption is dependent on bile salts, increased with fatty foods. Metabolism is affected transforming growth factor beta. Calcineurin inhibition by a cyclosporine-­ with concurrent grapefruit juice ingestion [41]. cyclophilin complex prevents activation of calci- Cyclosporine distributes through multiple organ neurin phosphatase, thus preventing downstream systems, including the skin, and can be found in 9 Traditional Systemic Therapy I: Methotrexate and Cyclosporine breast milk. It crosses the placenta but not the blood brain barrier. Cyclosporine is metabolized by the hepatic cytochrome P-450 3A4 (CYP3A4) enzyme system, and almost completely excreted in bile through the feces. The dose may need to be reduced in patients with hepatic insufficiency but not in renal insufficiency or with hemodialysis. The elimination half-life of cyclosporine is about 19 h, and its metabolism is age-dependent with a two-fold increase in half-life in adults compared to children. Use in Psoriasis Cyclosporine is FDA approved for severe psoriasis, resistant/recalcitrant psoriasis and disabling psoriasis. It has several advantages over other traditional oral agents- it has rapid onset of action and it is weight based dosing. These features allow for rapid control of flaring psoriasis. Numerous studies have been published on cyclosporine evaluating its efficacy, its comparison to methotrexate and etretinate, and its long-term maintenance/remission data. In a randomized study comparing methotrexate and cyclosporine after 16 weeks, cyclosporine and methotrexate appeared to be equally effective, with PASI 75 improvement being 71% and 60%, respectively [37, 42]. In a study comparing the oral retinoid etretinate with cyclosporine, PASI 75 ­improvement was seen in only 47% of patients on etretinate compared to 71% on cyclosporine [43]. Cyclosporine rapidly controls psoriasis, but rapid withdrawal may result in rebound. Open label studies evaluating discontinuation of cyclosporine after one year of therapy with a gradual (1 mg/kg/ wk) taper or abrupt cessation were published in 1999 and 2001 [35, 44]. No significant rebound was noted in either group, and the median time to relapse was not significantly different. Since more than 50% of patients will relapse after 4 months, new treatments should be added at the time of taper or once psoriasis returns. According to recently published recommendations by the National Psoriasis Foundation, cyclosporine should be used with caution and for 113 the more severe cases [34]. For reasons not totally understood, dermatologists do not use cyclosporine often for recalcitrant psoriasis and view it only as a rescue drug treatment. The drug is very safe to use if proper patient selection is done, with knowledge of the patient’s co-morbidities and medications and with proper clinical monitoring. Since the drug is very effective with rapid onset of action, offering dramatic clearance in as little as 2–4 weeks, it can be used to clear patients quickly allowing time to plan and transition to a long-term maintenance regimen. It has also been used successfully for erythrodermic and pustular psoriasis. While cyclosporine’s beneficial effects on psoriasis were first observed during trials for rheumatoid arthritis, it is not as useful for arthritis as for psoriasis lesions. While not approved for children or pregnant women, cyclosporine has been used with good results in these patient populations. It is considered category C and should be used with caution in pregnancy and during lactation [45]. Cyclosporine has been associated with low birth weight (<2500 gm) and prematurity (< 37 weeks) when used in renal transplant patients. Limited information is known about the effect of cyclosporine in pregnant women with psoriasis, who typically are prescribed lower doses. Cyclosporine does not appear to be a teratogen and has been used in pregnant women with successful outcomes. Overall, it is the authors’ opinion that cyclosporine is safe to use in high risk psoriasis patients during pregnancy if the benefits far outweigh the risks in severely debilitating disease. However, at this point in time, there are biologics which appear to be safe to use in pregnancy [46]. If cyclosporine is used in a pregnant patient, the mother and fetus should be carefully followed by high-risk obstetrician. Cyclosporine has been used in children and appears to be safe and efficacious [47]. Doses are similar to that used in the adult population, 2.5–3.0 mg/kg per day in divided doses. Treatment courses should be limited to 6-month intervals [34, 44]. Risks of malignancy and lymphoproliferative disorders seem to be minimal in children treated for skin diseases due to limited courses of therapy and dosages that are below 5 mg/kg/day. 114 Cyclosporine is contraindicated in patients with uncontrolled or difficult to control hypertension and in those individuals with significant renal disease or frequent infections. Careful attention should be paid to individuals with a personal history of cutaneous malignancies as cyclosporine can increase the numbers of skin cancers over time. Particular caution should be given with patients who have had extensive PUVA therapy (greater than 200 treatments) as the risk of nonmelanoma skin cancers, particularly squamous cell carcinoma, and melanoma may be increased [48]. As with methotrexate, the decision to use cyclosporine should be made based on a patient’s individual situation. Dosing Generally, patients are treated with 2.5–3 mg/kg/ day, usually in divided doses. The package insert suggests dosing at ideal body weight; but, in the authors’ experience, dosing is subtherapeutic at ideal weights in significantly obese patients. Dosing may need to be adjusted, especially in the first few weeks as bioavailability is variable and may vary with different generics. Cyclosporine is best absorbed on an empty stomach in children. However, adult patients can take it with food but should be consistent with dosing regimens, same time and amount of food ingested. Absorption may even be improved slightly with food in adults. Levels may be decreased with grapefruit juice [41] in both populations. Unlike transplantation patients, peak and trough levels do not need to be routinely done to ensure adequate blood levels in psoriasis patients. Levels may be checked if there is a question of compliance or issues with absorption. Cyclosporine therapy is generally used on a short-term basis to control severe flares, rarely more than 6–12 months. Monitoring Guidelines Prior to initiating cyclosporine therapy, the patient should have pretreatment blood tests, including creatinine, urinalysis, magnesium, E. Boh et al. tuberculosis screening, a complete medical history with emphasis on medications and co-­ morbidities, complete physical exam and baseline blood pressure measurements. Patients should be counseled on adverse effects and the need for careful monitoring. Baseline and ongoing monitoring guidelines are listed in Table 9.6. Frequent monitoring, especially for iatrogenic hypertension and signs of renal insufficiency, is required [34]. Usually office visits with repeated counseling and education parallel the blood evaluation schedule. If warranted, patients can also monitor blood pressure at home. The package insert recommends stopping cyclosporine if the blood pressure remains elevated after an attempt to lower the cyclosporine dose on several occasions. Many practitioners initiate low dose amlodipine or other calcium channel blocker in normotensive adult patients for renal protection [49]. Because of possible permanent damage to the kidney and loss of renal function in patients on long-term therapy, cyclosporine is a drug that requires careful patient selection and subsequent monitoring to be used safely. Therefore, a careful assessment of psoriasis disease severity is critical when assessing the risk-benefit ratio of treatment Table 9.6 Monitoring guidelines for cyclosporine Baseline History and physical Careful review of drug history Blood pressure evaluation Laboratory screening Complete blood count with differential and platelet counts (CBC) Comprehensive metabolic panel- renal and hepatic (CMP) Hepatitis B and C screening Tuberculosis screening with PPD or blood test Serum or urine pregnancy screening On going monitoring at q 2 weeks x 1 then q 4-weeks x 3 months then q 2 months Periodic review of drug history Physical exam BP at every visit and home monitoring if warranted Laboratory CBC with differential and platelets at 2 weeks then q month for 3 months then every 2 months CMP at 2 weeks then q month for 3 months then every 2 months Tuberculosis screening every year 9 Traditional Systemic Therapy I: Methotrexate and Cyclosporine with cyclosporine. With the wide availability of other useful agents for psoriasis, it is reasonable to consider an alternative if the blood pressure or kidney function remains elevated after two readings. Patients on cyclosporine are also immunosuppressed and are more susceptible to infections, bacterial, viral and fungal. Patients should be examined and screened for these types of infection before as well as during treatment. Adverse Effects While the frequency and severity of side effects is reduced when used at doses for psoriasis, the major toxicities associated with cyclosporine remain renal insufficiency and hypertension Table 9.7. The most common side effects associated with cyclosporine administration include headaches (15%), hypertrichosis (6%), and gingival hyperplasia, which is seen more frequently in transplant patients but can uncommonly occur in psoriasis patients. Other side effects include hirsurtism, tremor, diarrhea, hypertriglyceridemia, hypomagnesemia, nausea/vomiting, paresthesias and influenza-like symptoms. Blood abnormalities include hypomagnesemia, hyperlipidemia and hyperuricemia. As a consequence of these potential abnormalities, magnesium and uric acid l­evels should be obtained regularly. Lipid monitoring is less frequently needed but should be considered in patients on longer treatment periods. Rare side effects include neuroTable 9.7 Side effects of cyclosporine Increased risk of malignancies Increased risk of infections Renal impairment Hypertension Malignancies Headache, tremor, paresthesia Cutaneous Hypertrichosis Gingival hyperplasia Hyperbilirubinemia Worsening acne Increased risk of infection Nausea/vomiting/ diarrhea Flu-like symptoms Myalgias Lethargy Hypertriglyceridemia Hypomagnesemia Hyperkalemia 115 logic complaints, including lowering the seizure threshold, transient gastrointestinal complaints and respiratory complaints of cough and rhinitis. Nephrotoxicity Nephrotoxicity is the most common and clinically significant adverse effect and can present as acute azotemia or as a chronic, slowly progressive renal insufficiency or failure. Although reversible changes in the kidney may be related to the vascular effect, long-term therapy may lead to permanent scarring with loss of renal function. Therefore, it is very important to routinely monitor the renal function with a serum creatinine and a urinalysis as well as the blood pressure in patients on cyclosporine. It is important to remember that elderly patients may have a decrease in the glomerular filtration rate (GFR) without a decrease in creatinine. Patients who are on cyclosporine for a year are suggested to get an annual GFR. This is not routinely done, since patients do not usually remain on cyclosporine for greater than a year at a time. Patients with elevations of serum creatinine greater than 25% from baseline on two occasions (separated by 2 weeks) should have a 25% to 50% decrease in their dosage. If the creatinine level remains elevated, the cyclosporine dose should once again be decreased by 25% to 50%. If after these changes, the creatinine does not return close to baseline, cyclosporine should be discontinued. Hypertension Hypertension is caused by renal vasoconstriction and sodium retention, and usually presents early in the course of treatment. Dose reduction or addition of an anti-hypertensive such as amlodipine can ameliorate this adverse effect. Caution the patient that they may experience dependent edema, a side effect common to both cyclosporine and amlodipine. Since amlodipine has been reported to be renal protective in the transplant population on cyclosporine, the addition of amlodipine at this juncture is reasonable. In fact, many clinicians familiar with using the drug will start amlodipine at low doses such as 5 mg q day even in normotensive individuals as a prophylactic measure at onset of therapy [49]. E. Boh et al. 116 Table 9.8 Potential cyclosporine (CyA) drug-drug interactions Drug class Antiarythmics CCBa Diuretics Antifungals Antibiotics Anti-HIV Anti-malarials SSRIb Foods Anti-neoplastic Steroids Increases CyA Amiodarone Diltiazem;verapamil Thiazodes;furosemide Azoles Quinolones;cephalosporins doxycycline Protease inhibitors Hydroxychloroquine Fluoxetine;sertraline Grapefruit Imatinib; Dexamethasone;methylpredniso lone Anti-convulsants Others Retinoids Decreases CyA Drug levels increased by CyA Verapamil;diltiazem Griseofulvin Beta-lactams; nafcillin;rifampin Efivarenz Phenytoin;phenobarbitol; valproic acid OCPc Statins Bexarotene Calcium channel blockers; bSelective Serotonin Reuptake Inhibitors; coral contraceptive pills a Drug Interactions Since cyclosporine is metabolized by the hepatic CYP3A4 system, a variety of drug interactions can occur. Table 9.8 has a list of some common drug interactions seen with cyclosporine administration. Concomitant use of medications metabolized by the CYP3A4 system compete as substrates and may increase serum levels and potentiate toxicity. Induction or inhibition of the enzyme may decrease or increase serum levels, respectively. Foods usually do not affect cyclosporine levels but grapefruit juice increases levels of cyclosporine by CYP3A4 inhibition. Interactions may also occur with over the counter nutraceuticals, herbal and vitamin preparations. For instance, St John’s wort may decrease cyclosporine concentrations. In patients who have severe liver disease, metabolism may be decreased, leading to higher drug levels. Although heavy alcohol intake increase cyclosporine levels, mild to moderate alcohol consumption has little effect. Cyclosporine is an inhibitor of CYP3A4 affecting other drugs such as calcium channel blockers, erectile dysfunction drugs, and statins. Reports of serious rhabdomyolysis occurring in patients who are concurrently treated with a statin have been described [50]. Medications that may potentiate renal toxicity such as aminoglycosides or nonsteroidal anti-inflammatory drugs as well as medications that may elevate potassium levels should be restricted. Given the long list of possible drug interactions, a thorough medication history must be obtained for all patients before initiating treatment, and patients should be educated regarding the introduction of new drugs with continued therapy. As a general rule, drugs that alter the cytochrome P450 system should be introduced cautiously and potentially nephrotoxic drugs, such as nonsteroidal anti-inflammatory drugs, aminoglycosides, ciprofloxacin, clotrimazole, and fibrates that can impair renal function during cyclosporine treatment should be avoided if possible. Summary Both methotrexate and cyclosporine improve psoriasis by decreasing inflammation and suppression of T-cell mediated production of inflammatory cytokines that are known to be pivotal in the pathogenesis of psoriasis. While methotrexate and cyclosporine are safe and reliable treat- 9 Traditional Systemic Therapy I: Methotrexate and Cyclosporine ments for psoriasis with proper patient selection and monitoring, the risk of serious potential complications remains. Patients and physicians should be aware of these side effects and discuss risks and benefits prior to starting therapy. From the physician’s perspective, careful selection of the patient is paramount to eliminate potential complications. From the patient’s perspective, the importance of follow-up appointments and appropriate, timely blood monitoring needs to be understood. These agents may serve a useful role in bridging patients onto biologics, in those patients with a recent history of malignancy or in those patients where biologics are contraindicated or not available (Table 9.8). References 1. Gelfand JM, Neimann AL, Shin DB, et al. Risk of myocardial infarction in patients with psoriasis. JAMA. 2006;296:1735–41. 2. Takahashi H, Iizuka H. Psoriasis and metabolic syndrome. J Dermatol. 2012;39:212–8. 3. Myers WA, Gottliep AB, Maase P. Psoriasis and psoriatic arthritis: clinical features and disease mechanism. Clin Dermatol. 2006;24:438–47. 4. Henseler T, Christophers E. Disease concomitance in psoriasis. J Am Acad Dermatol. 1995;32:982–6. 5. Neimann AL, Shin DB, Wang X, et al. Prevalence of cardiovascular risk factors in patients with psoriasis. J Am Acad Dermatol. 2006;55:829–35. 6. Mallbis L, Ritchen CT, Stahl M. Metabolic disorders in patients with psoriasis and psoriatis arthritis. Curr Rheumatol Rep. 2006;8:355–63. 7. Menter A, Strober BE, Kaplan DH, et al. Joint AAD-­ NPF guidelines of care for the management and treatment of psoriasis with biologics. J Am Acad Dermatol. 2019 Apr;80(4):1029–72. 8. 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 Apr;80(4):1073–113. 9. Gubner R, Augsut S, Ginsberg V. Therapeutic suppression of tissue reactivity: effects of aminopterin in rheumatoid arthritis and psoriasis. Amer J Med Sci. 1951;221:176–82. 10. Edmunson WF, Guy WB. Treatment of psoriasis with folic acid antagonists. AMA Arch Derm. 1958;78:200–3. 11. Weinstein GD, Frost P. Methotrexate for psoriasis. A new therapeutic schedule. Arch Dermatol. 1971;103:33–8. 117 12. Weinstein GD. Methotrexate. Ann Intern Med. 1977;86:199–204. 13. Warren RB, Griffiths EM. Systemic therapies for psoriasis: methotrexate, retinoids, and cyclosporine. Clin Dermatol. 2008;26:438–47. 14. Carrerero G, Puig L, Dehesa L, Carrascosa J, et al. Guidelines on the use of methotrexate in psoriasis. Actas Dermosifiliogr. 2010;101:600–13. 15. Strober B, Menon B. Folate supplemantation during methotrexate therapy for patients with psoriasis. J Amer Acad Dermatol. 2005;53:652–9. 16. Cronstein BN, Naime D, Ostad E. The anti-­ inflammatory mechanism of methotrexate. Increased adenosine release at inflamed sites diminishes leukocyte accumulation in an vivo and in vitro model of inflammation. J Clin Invest. 1993;92:2675–82. 17. Genestier L, Paillot R, Fournel S, Ferraro C, Miossec P, Revillard JP. Immunosuppressive properties of methotrexate: apoptosis and clonal deletion of activated peripheral T cells. J Clin Invest. 1998;102:322–8. 18. Johnston A, Gudjonsson JF, Sigmundsdottir H, et al. The anti-inflammatory action of methotrexate is not mediated by lymphocyte apoptosis, but by the suppression of activation and adhesion molecules. Clin Immunol. 2005;114:154–63. 19. Priyadarssini M, Chandrashekar L, Rajappa M. Effect of methotrexate monotherapy on T-cell subsets in the peripheral circulation in psoriasis. Clin Exp Dermatol. 2019 Jul;44(5):491–7. 20. Vena GN, Cassano N, Iannone F. Update on subcutaneous methotrexate for inflammatory arthritis and psoriasis. Ther Risk Manag. 2018;14:105–16. 21. Rajitha P, Biswas R, Sabitha M, Jayakumar R. Methotrexate in the treatment of psoriasis and rheumatoid arthritis: mechanistic insights, current issues and novel delivery approaches. Curr Pharm Des. 2017;23:3550–66. 22. Roenigk HH Jr, Maibach HI, Weinstein GP. Methotrexate therapy for psoriasis. Guideline revisions. Arch Dermatol. 1973;108:35. 23. Gottlieb A, Korman N, Gordon K, Feldman S, et al. Guidelines of care for the management of psoriaisis and psoriatic arthritis, section 2: psoriatic arthritis; overview and guidlelines of care for treatment with an emphasis on the biologics. J Am Acad Dermatol. 2008;58:851–64. 24. Montaudie H, Sbidian E, Paul C, Maza A, et al. Methotrexate in psoriasis: a systematic review of treatment modalities, incidence, risk factors and monitoring of liver toxicity. J Euro Acad Dermatol Venereol. 2011;25(Suppl. 2):12–8. 25. Mease P, Gladman D, Collier D, et al. Etanercept and methotrexate as Monotherapy or in combination for psoriatic arthritis: primary results from a randomized, controlled phase III trial. Arthritis Rheumatol. 2019;71:1–13. 26. Appani SK, Devarasetti PK, Rajendra, et al. Methotrexate achieves major cDAPSA response, and improvement in dactylitis and functional sta- Traditional Systemic Therapy II: Retinoids and Others 10 Vignesh Ramachandran, Ted Rosen, Misha Koshelev, and Fareesa Shuja Sandoval Abstract Some patients with psoriasis either do not respond to or develop significant toxicities fromwell-recognized first-line systemic therapies. However, a number of second-line systemic agents may be considered. For example, acitretin is particularly effective in erythrodermic psoriasis, palmoplantar pustulosis, and generalized pustular psoriasis. Since acitretin does not induce immunocompromise, it can be used to manage psoriasis associated with HIV infection. Concomitant ultraviolet B (UVB) or psoralen plus ultraviolet A (PUVA) is synergistic with acitretin. Although not FDA-approved to treat psoriasis, hydroxyurea has demonstrated efficacy for chronic plaque psoriasis and, in short bursts, for flares of pustular psoriasis. Mycophenolate mofetil is used to treat moderate-to-severe chronic plaque psoriasis, most often as a maintenance therapy. 6-­thioguanine is used to treat plaque psoriasis and palmoplantar pustulosis which is resistant to other systemic agents, but is not recomV. Ramachandran · T. Rosen (*) Department of Dermatology, Baylor College of Medicine, Houston, TX, USA M. Koshelev University of Texas, McGovern Medical School at Houston, Houston, TX, USA mended for long-term maintenance due to possible hepatotoxicity. Systemic tacrolimus is best used to treat severe, recalcitrant plaque psoriasis; nephrotoxicity limits long-term use. Though usually used for rheumatoid arthritis, leflunomide can also be considered in treatment-­resistant chronic plaque psoriasis or psoriatic arthritis. Penicillin V or erythromycin are options for guttate psoriasis when the latter is associated bacterial infection (usually of the upper respiratory system). Apremilast, a newly approved treatment for psoriasis and psoriatic arthritis, does not induce immunosuppression, has a good safety profile, does not require laboratory monitoring, and is available in a convenient oral formulation. Nevertheless, its lower efficacy may limit utility. Key Learning Objectives 1. To understand the mechanism of action of retinoids and other oral agents used in the treatment of psoriasis. 2. To understand the appropriate use and efficacy of retinoids and other oral agents used in the treatment of psoriasis. 3. To understand the safety issues and appropriate monitoring for retinoids and other oral agents used in the treatment of psoriasis. F. S. Sandoval Westlake Dermatology, Austin, TX, USA © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_10 119 V. Ramachandran et al. 120 Psoriasis affects 1 in 50 individuals with manifestations appearing in the skin, nails, and joints [1]. Patients with psoriasis are most commonly treated with topical agents, which is effective in 70–80% of patients [2]. Systemic treatments tend to be reserved for patients with more than 10% body surface area (BSA) involvement or severe psoriasis of the scalp, palms and soles, genitalia, or intertriginous sites. Even in this era of biologics, traditional systemic therapies remain both useful and appropriate.These drugs offer comparatively inexpensive, oral alternatives with wellknown short and long-term risks [3]. Although methotrexate and cyclosporine are recognized first-line treatments for psoriasis, patients who do not respond or who develop toxicities are candidates for alternative second-line systemic agents. In this chapter, we provide a clinically-­oriented summary of several second-line systemic agents used in the treatment of psoriasis. Specifically, the medications discussed herein are: acitretin, hydroxyurea, mycophenolate mofetil (MMF), thioguanine (6TG), systemic tacrolimus, leflunomide, penicillin V, erythromycin, and apremilast (Fig. 10.1, 10.2, and 10.3). In doing so, we hope to aid clinicians navigate the myriad of alternative tradiational systemic treatment options within the patients’ clinical contexts with an evidence-based understanding of best use practices, side effects, monitoring, and efficacy. factors within cells. However, beyond this, thehe exact mechanism of action of acitretin in psoriasis is unknown though it is likely related to its ability to decrease epidermal proliferation and induce differentiation [4]. When Best to Use Acitretin is one of the suggestedtreatments for relatively stable generalized pustular psoriasis and is an effective treatment for less acute patients with exfoliative erythrodermic psoriasis [11]. It is also a suggested traditional systemic for psoriasis in the setting of human immunodeficiency virus (HIV) infection due to its lack of immunosuppression [12]. In palmoplantar pustulosis, acitretin is commonly used to ameliorate hyperkeratosis and decreases pustulation. In acitretinresponsive patients with chronic plaque psoriasis, it can be an effective maintenance ­therapy [11]. Acitretin can safely be combined with phototherapy to potentiate its efficacy. Additionally, it may be used in combination with biologic therapies, including etanercept (resulting in reduced etanercept dosing) and adalimumab. Randomized, controlled trial evidence is lacking for its use in combination with other biologics [13]. Although seemingly contraindicated by the acitretin prescribing information which warns of the risk of hepatotoxicity, acitretin and methoAcitretin trexate have been combined when monotherapy was inadequate, particularly in cases of generalized pustular psoriasis. It is crucial to monitor FDA-Approved Indication(s) liver function closely with this combination [4, Acitretin is FDA-approved for the treatment of 14]. Acitretin has been used with cyclosporine severe psoriasis in adults [4]. In women of child-­ for short-term treatment albeit with frequent bearing potential, acitretin is only advised for non- monitoring of lipids. Acitretin has also been pregnant individuals who do not respond to other combined with hydroxyurea to treat recalcitrant psoriasis medications or have c­ ontraindications palmoplantar pustulosis. to their use. The FDA recommends its prescription by physicians knowledgeable in the use of systemic retinoids [4]. Dosage Mechanism of Action Acitretin interacts with with cytosolic proteins and nuclear factors, which act as transcription The general acitretin dose ranges from 10 to 50 mg daily, administered with meals [4, 15]. Generalized pustular psoriasis typically requires an acitretin dose of 25–50 mg [8]. After clinical response, the dose is tapered to 10–25 mg daily. 10 Traditional Systemic Therapy II: Retinoids and Others 121 O OH O Acitretin S O H N N HO N H NH2 N H H2N Hydroxyurea N 6-Thioguanine HO O O OH O N O O O O O HO O Tacrolimus Fig. 10.1 Chemical structures of acitretin, hydroxyurea, 6-thioguanine, and tacrolimus; adapted from the package inserts for the corresponding medications [4–7] V. Ramachandran et al. 122 O S O NH H N O + – O NH K N O F O O F F Leflunomide Penicillin V Potassium OH HO HO O O O O OH O O O N O Erythromycin OH Fig. 10.2 Chemical structures of leflunomide, penicillin V potassium, and erythromycin; adapted from the package inserts for the corresponding medications [7–10] Exfoliative erythrodermic psoriasis requires an acitretin dose of 25–50 mg daily [15]. Clinical trials have employed varying acitretin doses to treat chronic plaque psoriasis. A daily dose of 25 mg or less minimizes adverse events. Initiation of therapy should begin with a low dose with progressive uptitration to avoid an initial disease flare and to improve tolerability [4, 15]. Maximal response is usually seen after 3–6 months of treatment [15]. Two randomized, controlled trials (8-week and 16-week) assessed the efficacy acitretin (10 mg, 25 mg, 50 mg, 75 mg) compared to ­placebo in the treatment of moderate-to-severe plaque psoriasis [13]. Low-dose treatment was defined as approximately 25 mg/day, whereas high-dose treatment 10 Traditional Systemic Therapy II: Retinoids and Others Table 10.1 Frequency of selected adverse events reported in clinical trials of acitretin [4, 15] O O O N O NH Percent of patients reporting event(s) More than 75% 50–75% O S O 123 25–50% O 10–25% Fig. 10.3 Chemical structure of apremilast (OTEZLA®) was defined as approximately 50 mg/day. The studies showed that, overall, low-dose acitretin is not only safer than high-dose acitretin, but may also be more efficacious in treating psoriasis. Adverse Events Acitretin is associated with a number of adverse effects (Table 10.1). Many of the common events are related to increased epidermis turnover, drying of the skin, and their related sequelae. Recommended Monitoring Obtain a history and physical examination, CBC with platelets, BUN/Cr, LFTs, lipid profile, and a pregnancy test if indicated before starting acitretin [14, 15]. The clinician should perform a physical examination and obtain a CBC with platelets, BUN/Cr, LFTs, and lipid profile monthly for the first 3–6 months, then every 3 months [14]. BUN/ Cr and CBC with platelets may be ordered every other monitoring period if desired. Pregnancy tests may be done monthly, if indicated. Perils and Pitfalls Acitretin is contraindicated in patients with hypersensitivity to retinoids, chronically elevated lipids, severely impaired kidney or liver function, nursing mothers, and pregnant women [4]. Less than 10% Adverse event(s) reported Cheilitis Alopecia Skin peeling Rhinitis Xerosis Nail disorder Pruritus Rigors Xerophthalmia Xerostomia Epistaxis Arthralgia Spinal hyperostosis Erythematous rash Hyperesthesia Paresthesia Paronychia Skin atrophy Sticky skin Nausea Abdominal pain Decreased night vision Headache Myalgia The average half-life of acitretin is 49 hours and that of its isomer cis-acitretin is 63 hours; however, in the presence of alcohol, etretinate, whose average half-life is 120 days, can form. Etretinate has been detected 2.1–2.9 years after stopping therapy, most likely due to storage in fatty tissue. This is why acitretin is contraindicated in women who plan to become pregnant within 3 years of stopping the medication and is given with extreme caution (if at all) in women of child-­bearing potential. Patients are also advised against donating blood from the initiation of treatment to 3 years after discontinuation. Fetal abnormalities reported with retinoid use include meningomyelocele, meningoencephalocele, decreased cranial volume, and cardiovascular malformations [4, 15]. Acitretin may interact with a number of medications. It interferes with the contraceptive effects of the microdose progestin minipill [4, 15]. When given with tetracyclines, the combination raises the risk of increased intracranial pressure and 124 manifest pseudotumor cerebri [4]. Acitretin can potentiate the glucose-lowering effects of glibenclamide and may reduce phenytoin protein binding. Acitretin should not be given with other oral retinoids nor with excessive vitamin A supplementation to avoid hypervitaminosis A [4, 15]. Hepatobiliary abnormalities such as elevated serum bilirubin and transaminases, toxic hepatitis, acute reversible hepatic injury, and cirrhosis have been associated with acitretin [1]. In clinical trials, 66% of patients treated with acitretin had triglyceride elevations; of note, these patients were likely to have increased alcohol intake, diabetes mellitus, obesity, pre-existing disturbances of lipid metabolism, or a family history of these conditions. Acitretin-induced serum triglyceride levels above 800 mg/dL have been associated with fatal fulminant pancreatitis. Acitretin-­ induced pancreatitis without an increase in serum triglycerides has also been reported. Forty percent of patients treated with acitretin in clinical trials had decreased HDL levels and one-third had serum cholesterol elevations. Changes in lipid levels resolved after stopping acitretin. Thromboembolic events, acute myocardial infarction, and pseudotumor cerebri have been reported, as have depression, thoughts of self-­harm, aggressive feelings, and other psychiatric symptoms. Adults receiving acitretin have rarely experienced abnormal ossification. V. Ramachandran et al. after 1 month depending on clinical response; the mean dose was 0.4 mg/kg/day. After 4 months, the mean PASI reduction was 59.4% [18]. There are no head-to-head trials comparing the effectiveness of acitretin to methotrexate or cyclosporine in the treatment of chronic plaque psoriasis, but it is the American Academy of Dermatology’s general consensus acitretin is a less effective monotherapy [15]. However, a head-to-head comparison between cyclosporine and etretinate, the pro-drug of acitretin which is unavailable in the United States, demonstrated cyclosporine was more efficacious in the treatment of severe plaque psoriasis [19]. Acitretin has been studied in the treatment of palmoplantar pustulosis [11, 20]. A randomized, open-label study of67 patients with palmoplantar pustulosis assessed the efficacy of acitretin. Patients received 10 mg/day for 4 weeks followed by dosage adjustment based on clinical response for 8 weeks. After the 12 weeks, patients had an average of 3.9 pustules compared to their average of 57.8 at baseline [20]. Acitretin is more efficacious when combined with phototherapy [15]. A randomized, controlledstudy of patients with moderate-tosevere psoriasis (20–80% BSA) assessed the efficacy of broadband-UVB (bb-UVB) with acitretin or placebo. Overall, combinational therapy led to 74% reduction in psoriasis severity scores after 12 weeks comparedto 42% reduction in patients treatedwith acitretin alone and 35% reduction with UVB alone [21]. Another Strength of Evidence multicenter, randomized, controlled trial of 82 patients with severe psoriasis (chronic plaque or Acitretin can be an effective short and long- examthematic-­type pustular variant) investigated term therapy for chronic plaque psoriasis [16]. A the efficacy of acitretin plus UVB compared to ­multicenter trial of 37 adults treated with acitre- placebo plus UVB. Treatment-group patients tin for psoriasis was conducted for 12 months took 35 mg acitretin daily for 4 weeks followed with 10 mg dose adjustments (up or down, range by 25 mg acitretin daily plus BB-UVB [22].]. 10–70 mg daily) occurring monthly. Seventy-­ PASI decreased on average by 79% with acitretin nine percent of patients achieved PASI 75 [17]. and UVB compared to 35% in the control group A prospective study enrolled 17 patients with (placebo and UVB). Median cumulative UVB plaque psoriasis who had failed treatment with dose needed to reach 75% improvement was methotrexate, cyclosporine, or PUVA, or had at 41% lower for the acitretin and UVB group than least 10% BSA involvement to assess the efficacy for the placebo and UVB group [22]. It is imporof acitretin [18]. Acitretin was started at 0.3 mg/ tant to note these older studies utilize BB-UVB kg/day and could be increased to 0.5 mg/kg/day in lieu of narrow-band (nb-UVB), which is the 10 Traditional Systemic Therapy II: Retinoids and Others more readily available and commonly used UVB regimen in most practices. Combination therapy with biologics are limited. However, a randomized, controlled, double-­ blinded study investigated acitretin alone versus etanercept alone versus acitretin with etanercept for patients with moderate-to-severe plaque psoriasis. Over six months, twenty patients received 0.4 mg/kg oral acitretin daily, 22 patients received 25 mg etanercept subcutaneously twice perweek, and 18 patients received 0.4 mg/kg oral acitretin daily with etanercept 25 mg subcutaneously once a week. Thirty percent of patients in the acitretin group achieved PASI 75, compared to 45% of patients treated with etanercept alone and 44% of patients treated with both etanercept and acitretin. Overall, it appears acitretin is less effective than etanercept. Adding acitretin to etanercept achieves an efficacy rate similar to that of etanercept alone. The results, however, may bedrug-­ dose-­dependent [23]. There are limited other clinical trial studies assessing acitretin in psoriasis. A multicenter, randomize, controlled, double-blinded trial of 108 moderate-to-severe plaque psoriasis patients assessed the efficacy of acitretin alone (with placebo) or in combination with “total glucosides of paeony” (TGP), a Chinese traditional herbal medication. After 12 weeks, the treatment group treated with added TPG achieved PASI 50 in 90% of patients compared to 70.5% in the control group (P < 0.05 [24]. Similarly, in another randomized, controlled, double-blinded trial of 15 patients with moderate-to-severe plaque psoriasis were treated with acitretin with or without curcumin (main active constituent of the rhizome of turmeric with various anti-inflammatory, ­ anti-­ proliferative, and anti-angiogenic effects) nanoparticles. At the end of the study period (12 weeks), higher percentages of patients treated with the adjuctive curcumin nanoparticles achieved PASI 50 (93% versus 66%), PASI 75 (43% versus 20%), and PASI 90 (36% versus 13%) compared to the control arm (all P < 0.001) [25]. These results must be reproduced with larger cohorts and assessed for possible unaccounted confounding factors to establish the strength of this data. 125 Hydroxyurea FDA-Approved Indication(s) Hydroxyurea (HYDREA®) is FDA-approved as a treatment for resistant chronic myeloid leukemia and locally advanced squamous cell carcinomas of the head and neck (excluding lip) in combination with concurrent chemoradiation [5]. Another formulation of hydroxyurea (DROXIA®) is FDA-­ approved for reduce the frequency of painful crises and to reduce the need for blood transfusions in adult patients with sickle cell [26]. Mechanism of Action The precise mechanism of action of hydroxyurea in psoriasis is unknown [5, 15, 27, 28]. Hydroxyurea inhibits ribonucleotide reductase and suppresses DNA synthesis, which is thought to contribute to an anti-proliferative role in psoriasis. When Best to Use Hydroxyurea has been given to patients with moderate-to-severe chronic plaque psoriasis who have received the recommended cumulative dose of methotrexate or who cannot tolerate adverse events related to methotrexate [29]. Hydroxyurea has also been used with low-dose cyclosporine in short-course therapy to treat recalcitrant, severe psoriasis and with acitretin to treat recalcitrant palmoplantar pustulosis [30]. Hydroxyurea may also be beneficial in generalized pustular ­psoriasis [27]. Dosage Hydroxyurea is initiated at a dose of 500 mg twice daily, and increased to 3 g daily (total) as tolerated. Dosing at 3–4.5 g weekly has also been used [15, 27, 29]. The senior author has utilized hydroxyurea as a short-term “rescue” drug during flares of von Zumbusch pustular psoriasis, V. Ramachandran et al. 126 starting at 3 g as a daily dose and decreasing by 500 mg daily, for a 1 week total course. Adverse Events Hydroxyurea is associated with a number of adverse events, most of which are related to cell turnover/production of highly proliferative cell types including the bone marrow and gastrointestinal tract (Table 10.2). Recommended Monitoring Before starting hydroxyurea, the clinician should obtain a good history,perform a general physical examination, and obtain laboratories (CBC and pregnancy test) (if indicated) [9]. Weekly CBCs are obtained until a stable dose is achieved. Thereafter CBCs are obtainedmonthly as long as treatment with this agent is continued. Repeat physical examination, focusing on skin cancer and checking for enlarged lymph nodes, should be done biannually. Periodic pregnancy testing is performed if indicated. Table 10.2 Selected adverse events reported with the use of hydroxyurea; events reported in psoriasis patients are marked with an asterisk [9, 19, 20, 22, 23] Myelosuppression, including anemia*, leukopenia*, and thrombocytopenia* Gastrointestinal symptoms, including diarrhea*, aphthous ulcers*, nausea, vomiting, constipation, anorexia, and stomatitis Dermatological symptoms, including nail pigmentation*, alopecia*, pruritus*, dermatomyositis-­ like skin changes, rash, and ulceration Systemic symptoms, including edema*, asthenia, chills, malaise, and fever Neurologic symptoms, including hallucinations, dizziness, headache, disorientation, and convulsions Serum creatinine, BUN, and uric acid elevations with temporary impairment of renal tubular function LFT elevations Rare pulmonary fibrosis Rare dysuria Nonfatal and fatal pancreatitis and hepatotoxicity, severe peripheral neuropathy in HIV patients given hydroxyurea with other antiretrovirals Perils and Pitfalls Hydroxyurea is contraindicated in patients with known hypersensitivity to hydroxyurea and in patients with leukopenia, severe anemia, or thrombocytopenia. Additionally, it is also contraindicated during pregnancy and in nursing mothers [5, 15]. Hydroxyurea may raise uric acid levels and require changes in the doses of uricosuric medications to prevent gout flares [5]. Severe peripheral neuropathy, fatal and nonfatal pancreatitis, hepatotoxicity and fatal hepatic failure have been described in HIV patients given hydroxyurea and didanosine with or without stavudine [31]. These adverse events are rare in normal hosts. Patients given long-term hydroxyurea have reportedly developed both lymphoma and nonmelanoma skin cancer. In patients treated with hydroxyurea for myeloproliferative disorders, secondary leukemia has developed. These patients have also developed gangrene and vasculitic ulcerations, but most were simultaneously receiving interferon. Self-limiting megaloblastic erythropoiesis may occur early in the course of hydroxyurea therapy. Myelosuppression may occur in patients treated with hydroxyurea. Leukopenia generally occurs first, followed by anemia or thrombocytopenia. Bone marrow recovery is rapid when treatment is stopped. Increased myelosuppression may be seen when hydroxyurea is used concurrently with radiotherapy or myelosuppressive drugs [15]. Strength of Evidence A comparative study of methotrexate to hydroxyurea in 30 patients (15 in each treatment group) assessed the efficacy of these treatments for moderate-­ to-severe plaque psoriasis [29]. Patients had ≥20% BSA involvement and a PASI of ≥10. In the methotrexate group, patients given 15 mg of the medication weekly for 4 weeks; the dose was then increased up to 20 mg weekly in patients with less than 25% PASI reduction. Patients receiving hydroxyurea were given 10 Traditional Systemic Therapy II: Retinoids and Others 500 mg twice a day on two consecutive days for 1 week, then 500 mg three times a day on two consecutive days for 3 weeks. After 4 weeks, patients with ≤25% PASI reduction were given 500 mg three times a day on three consecutive days. Overall, 10/15 patients (66.7%) treated with methotrexate and 2/15 patients (13.3%) treated with hydroxyurea achieved PASI 75 at 12 weeks (P < 0.05). Though most patients treated with hydroxyurea did not achieve PASI 75, they did have a mean PASI reduction of 48.47% while experiencing fewer adverse events than the methotrexate group [29]. However, a short-term (8-week) comparative study of methotrexate (control) and hydroxyurea treatment in 24 patients with moderate-to-severe plaque psoriasis [28]. The results of this study demonstrated that although the PASI scores before and after treatment in each group significantly decreased (P < 0.05), there was no difference in mean percentage change of PASI scores between the groups (P > 0.05) [32]. A prospective study of 34 patients with moderate-­to-severe plaque psoriasis, erythrodermic, or generalized pustular psoriasis refractory to topical treatments and methotrexate underwent a regimen of hydroxyurea to assess its efficacy in treating their condition [27]. Patients were initiated on 500 mg hydroxyurea twice daily. If hydroxyurea was tolerated and PASI reduction was less than 25% after 2 weeks, it was increased to 1.0 and 1.5 g on alternate days, and then to 1.5 g daily as tolerated. If greater than 95% clearance was achieved, treatment was tapered over 4–8 weeks. Mean PASI reduction was 76% at 10–12 weeks. Three patients developed leukopenia which recovered after discontinuing treatment. Five patients relapsed within 12 months [27] A prospective, non-randomized sries of 31 patients (including 26 with history of systemic psoriasis therapy) were given hydroxyurea 1.0– 1.5 g per day for mean follow-up of 36.1 weeks (+/− 13.8). At or before 8 weeks, nearly 75% of the cohort demonstrated adequate response (defined ≥35% PASI reduction), and over50% of patients showed >70% PASI reduction [33]. 127 Mycophenolate Mofetil (MMF) FDA-Approved Indication(s) MMF is FDA-approved in combination with corticosteroids and cyclosporine to prevent organ rejection in liver, heart, and kidney transplant patients [34]. Mechanism of Action MMF is hydrolyzed to its active metabolite, mycophenolic acid, which inhibits de novo guanosine nucleotide synthesis and selectively suppresses lymphocyte growth and division [34]. As a result, psoriasis, a T-cell mediated inflammatory disorder, is thought to be responsie to MMF for this reason. When Best to Use MMF is utilized to treat moderate-to-severe chronic plaque psoriasis in patients who cannot tolerate any of the various first-line agents [35]. MMF can be given with cyclosporine, making it useful as both a bridge and maintenance therapy for patients needing cyclosporine for acute disease control [36]. Dosage MMF can be given as 1.0–2.0 g twice daily [35]. Adverse Events Psoriasis patients given MMF reported abdominal cramping, diarrhea, nausea, elevated LFTs, severe hyperbilirubinemia, severe hypertension, life-threatening hyperuricemia, life-threatening hypokalemia, periorbital edema, urticaria, furunculosis, and pruritus [35, 37, 38]. Transplant patients given MMF reported vomiting, genitourinary urgency, genitourinary frequency, V. Ramachandran et al. 128 dysuria, sterile pyuria, headaches, insomnia, peripheral edema, hypercholesterolemia, hypophosphatemia, and hyperkalemia [15, 34]. Recommended Monitoring The clinician should obtain a history andphysical examination. Baseline labs should includeCBC with differential, serum chemistry panel, LFTs, and pregnancy test (if indicated) before starting MMF [15, 34]. After initiation of therapy, complete blood and platelet counts should be checked weekly for 1 month, then every 2 weeks for 2 months, then monthly thereafter. It is wise to also obtain serum chemistries and LFTs monthly, a physical examination focusing on skin cancer and lymph nodes biannually, and ongoing pregnancy tests (if indicated) [39]. disease [34]. Adults given prolonged courses of MMF haveincreased risk of skin cancer, especially squamous cell carcinomas Fatal cases of progressive multifocal leukoencephalopathy (PML) with the use of MMF were reported in patients with risk factors such as pre-existing immune function impairment and treatment with other immunosuppressants [39]. Strength of Evidence A randomized, open-label clinical trial evaluated the efficacy and safety of MMF versus methotrexate in patients with chronic plaque psoriasis (PASI score of at least 10) and a history of inadequate response to topical therapies [35]. Seventeen patients received MMF 1 g twice daily for 12 weeks. Fifteen patients received methotrexate 7.5 mg/week for 1 week, then 15 mg/week for 3 weeks, and then 20 mg/week for 8 weeks. Ten Perils and Pitfalls of 17 patients (58.8%) in the MMF group and 11 of 15 patients (73.3%) in the methotrexate group MMF is contraindicated in patients with hyper- achieved PASI 75 (P > 0.05) [35]. sensitivity to MMF or mycophenolic acid, during Another randomized open-label clinical trial pregnancy, and in nursing mothers [34]. MMF evaluated the efficacy of MMF versus cyclospocan cause first trimester spontaneous abortions rine in chronic plaque psoriasis patients with a and fetal malformations [25]. It can interact PASI score of at least 10 [28]. Sixteen patients with acyclovir, ganciclovir, valganciclovir, pro- received MMF 1 g twice daily for 6 weeks. Then benecid, xanthine bronchodilators, high-dose they received 500 mg MMF twice daily if they salicylates, cholestyramine, phenytoin, antibiot- had 60% or greater PASI reduction, 1 g MMF ics, calcium, iron, and aluminum or magnesium-­ twice daily if they had 25–60% PASI reduccontaining antacids [15]. Since the effects of tion, and 1.5 g MMF twice daily if they had these other medications on MMF are varied, it 25% or less PASI reduction. Twenty-one patients is recommended to consult the FDA-approval received 1.25 mg/kg cyclosporine twice daily for documentation to assess impact on bioavailabil- the first 6 weeks, and then either 1.25 mg/kg once ity and subsequent efficacy [34]. daily, 1.25 mg/kg twice daily, or 2.5 mg/kg twice Patients given MMF have developed severe daily based on PASI reduction thresholds identineutropenia. Two percent of kidney and heart cal to those in the MMF group. After 12 weeks, transplant patients and 5% of liver transplant mean PASI decreased from 22.4 to 10.6 in the patients given MMF in clinical trials developed MMF group and from 24.6 to 6.6 in the cyclofatal infection/sepsis. Live attenuated vaccines sporine group [37]. should not be given to patients taking MMF [15, Use of concomitant MMF and cyclosporine to 34]. Pure red cell aplasia, anemia, leukopenia, treat severe recalcitrant psoriasis was described and thrombocytopenia have also been reported. in nine patients who failed to clear on cyclospoBetween 0.4% and 1% of transplant patients rine alone or could not tolerate higher cyclogiven MMF with other immunosuppressive drugs sporine doses [36]. Three patients showed good developed lymphoma or lymphoproliferative clinical improvement and four patients showed 10 Traditional Systemic Therapy II: Retinoids and Others 129 moderate disease control after a follow-up period When Best to Use of 3–11 months. No additional toxicity was seen after starting MMF in patients already tak- 6TG can be used to treat plaque psoriasis and ing cyclosporine. Notably, this study included a palmoplantar pustulosis resistant to more compatient with erythrodermic psoriasis and another monly utilized systemic agents or when these with generalized pustular psoriasis [36]. other agents are contraindicated [39]. It may be An open-label pilot study involving 20 patients considered a third-line systemic traditional syswith psoriasis (PASI >10) assessed the efficacy temic treatment [39]. of enteric-coated mycophenolate sodium 720 mg twice daily for 6 weeks followed by 360 mg twice daily for another 6 weeks. By week six, none of Dosage the patients achieved PASI 75. However, 8/20 (40%) of patients achieved PASI 50. By week 12, 6TG is given two to three times per week to 2/8 (25%) of patients achieved PASI 75. Some reduce the risk of myelosuppression [42, 43]. The patients in the cohort did relapse (4/13 by week starting dose is 80 mg twice per week; the dose 12). Twenty-five percent (2/8) achieved a PASI is increased by 20 mg every 2–4 weeks [15]. The 75% in week 24 [40]. maximum dose is 160 mg given up to three times Another study compared the efficacy of MMF per week. to cyclosporine in 80 patients with chronic plaque psoriasis via a prospective, sequential, cross-­ over, non-randomized, two-phase, open-label Adverse Events study. Both treatments were efficacious, resulting in decreased in PASI scores from baseline. According to various reports, 22–68% of psoriaHowever, cyclosporine was faster to produce sis patients given 6TG develop myelosuppression results and produced more significant decreases [43, 44]. Up to 12% of patients given the drug in PASI scores (45.7%, 60.2% and 60.5% at 3, reported gastrointestinal adverse events, includ8 and 16 weeks respectively for mycophenolate ing nausea, vomiting, aphthous ulcers, gastric mofetil compared to 89.7%, 95.3% and 95.3%, ulcers, gastroesophageal reflux, and dysgeusia respectively, for cyclosporin) [41]. [15, 43, 44]. One quarter of patients treated with 6TG for psoriasis experienced LFT elevations [15, 43]. Patients treated with this agent also 6-Thioguanine (6TG) developed headaches, fatigue, photodermatitis, herpes zoster, multiple warts, hyperuricemia, hepatic veno-occlusive disease, portal hypertenFDA-Approved Indication(S) sion, and non-melanoma skin cancer [15, 43, 44]. 6-thioguanine (6TG) is FDA-approved to induce Psoriasis patients treated with 6TG have rarely and sustain remission in patients with acute non-­ developed hepatotoxicity; in the event that this lymphocytic leukemia, most commonly in com- adverse event does occur, stopping treatment bination with other chemotherapeutic agents [6]. leads to resolution [6, 15, 42, 43]. It is not recommended for long-term treatment due to a significant risk of hepatotoxicity. Recommended Monitoring Mechanism of Action 6TG is a purine nucleotide analogue that interferes with nucleic acid synthesis, curtailing cellular proliferation [6]. The clinician should obtain a history and physical examination.Laboratory evaluation entails CBC with differential, serum chemistry panel, LFTs, hepatitis B and C tests, tuberculosis screening, and pregnancy test (if indicated) before initiating V. Ramachandran et al. 130 6TG therapy [6, 15]. After starting treatment, a CBC with differentialshould be checked every 2–4 weeks, serum chemistries every 3 months, and a physical examination focusing on lymph nodes and skin cancer biannually; periodic pregnancy tests should be performed, if indicated [15]. Perils and Pitfalls 6-thioguanine is contraindicated in patients with known hypersensitivity to the drug. It is also contraindicated in patients with liver disease, immunosuppression, anemia, leukopenia, and/ or thrombocytopenia. Pregnant patients and nursing mothers are also contraindicated from taking 6TG [6, 12]. The cytoplasmic enzyme thiopurine methyltransferase (TPMT) metabolizes 6TG and other thiopurines [6, 44]. TPMT activity in Caucasians varies based on genetic polymorphisms. Specifically, 10% of Caucasians have intermediate TPMT activity and one in approximately 300 Caucasian individuals has no TPMT activity [44]. Lower TPMT activity increases the risk of myelosuppression in patients taking 6TG. Aminosalicylate derivatives, such as olsalazine, mesalazine, and sulfasalazine, may inhibit TPMT [6, 15]. Life-threatening infections due to 6TG-induced granulocytopenia have been reported [3]. It has been recommended that physicians measure TPMT activity in all psoriasis patients before starting 6-thioguanine to determine initial dosage and assess the risk of myelosuppression [37]. Strength of Evidence A retrospective 4-year review of the treatment of 18 patients with moderate-to-severe plaque psoriasis treated with 6-thioguanine. Overall, 14/18 (78%) patients had more than 90% improvement; 3 patients (17%) had 50–90% improvement while only 1 patient had less than 50% ­improvement [44]. An open-label study of 14 patients with severe, recalcitrant plaque psoriasis investigated the efficacy of pulse-dosing 6TG. Two-to-threetimes per week (120 mg twice a week to 160 mg 3 times a week) resulted in marked improvement in 10 of 14 (71%) patients [43]. A retrospestive study of 81 patients (76 plaque psoriasis, 6 palmoplantar pustulosis) treated with 6TG was reported on to assess this medication’s efficacy [45]. Overall, 78% of patients had complete or almost complete clearing, 11% had some improvement, and 11% had little or no change in their psoriatic lesions. Four out of five patients with palmoplantar pustulosis had complete or almost complete clearing. Patients maintained their response for 3–145 months (mean = 33). The most commonside effect was myelosuppression [45]. Case reports corroborate these findings, albeit with limited strength of evidence [42, 46]. Systemic Tacrolimus FDA-Approved Indication(s) Oral tacrolimus is FDA-approved, in conjunction with corticosteroids, to prevent organ rejection in patients with kidney, liver, or heart transplants [7]. Mechanism of Action Tacrolimus inhibits the phosphatase activity of calcineurin and thereby suppresses T cell activation and proliferation via DNA replication [7]. When Best to Use Systemic tacrolimus is best used to treat severe, recalcitrant, chronic plaque psoriasis [47]. Evidence also exists for its use in lieu of cyclosporine in patients with increased cardiovascular risk [47, 48]. 10 Traditional Systemic Therapy II: Retinoids and Others Dosage Systemic tacrolimus is started at a dose of 0.05 mg/kg/day [7, 15, 47]. The dose can be increased to 0.10 mg/kg/day after 3 weeks and to 0.15 mg/kg/day after 6 weeks [47, 48]. Adverse Events The adverse effects associated with oral or intravenous (i.e. systemic) tacrolimus include: insomnia, tremors, headaches, altered sensory function, muscle pains, pruritus, malaise or fatigue, photophobia, nausea, and diarrhea [47]. Tremor, nausea, and abnormal renal function tests are commonly reported in transplant patients whereas hyperglycemia (and diabetes mellitus), hyperkalemia, hypertension, myocardial hypertrophy, elevated LFTs, leukocytosis, dyspnea, anemia (including red cell aplasia), edema, fever, nephrotoxicity, neurotoxicity and arthralgias were less commonly reported in these patients when given systemic tacrolimus [7, 15, 47]. Recommended Monitoring At baseline, the provide should obtain a detailed medical history and perform a physical examination. Baseline and repeat serum creatinine, potassium, and fasting glucose should be obtained and evaluated regularly. Monitoring of metabolic and blood components should be clinically guided [47]. Since tacrolimus can increase blood sugar levels when administered systemically, repeat blood sugars (serum concentration and hemoglobin A1c must be obtained periodically. A CBC count with differential, serum BUN and creatinine levels, LFTs, and pregnancy test (if indicated) should all be obtained before starting systemic tacrolimus [9]. After starting treatment, serial evaluations should be done of the patient’s blood pressure, serum chemistries, BUN and creatinine, LFTs, and pregnancy test (if indicated) [7, 15, 47]. A monitoring frequency has not been firmly established. Even if asymptomatic, patients should be monitored for signs and 131 s­ ymptoms of hypertension alongside repeat measurements [15]. Perils and Pitfalls Systemic tacrolimus is contraindicated in patients with known hypersensitivity to tacrolimus or to its metabolites and in nursing mothers [7, 15]. It causes fetal harm in pregnant animals [7, 15]. Transplant patients treated with tacrolimus have an increased risk of developing lymphoma and skin malignancies and an increased susceptibility to infection [7]. Since psoriasis patients already have an increased baseline risk of lymphoma, this particular potential problem needs to be followed closely. The clinician must advise patients to take oral tacrolimus consistently either with or without food, as the presence of food affects the drug’s bioavailability. The cytochrome P450 system metabolizes systemic tacrolimus, and it therefore interacts with many drugs [15]. One should not give systemic tacrolimus concurrently with cyclosporine; discontinue one drug at least 24 hours before starting the other. Strength of Evidence A randomized, controlled trial evaluated the safety and efficacy of systemic tacrolimus (0.1 mg/kg/d titrated to response and side effects) rcompared to placebo in 50 patients with severe recalcitrant plaque psoriasis over the course of nine weeks [48]. At the conclusion of the study period, PASI scores decreased by 83% on average in the tacrolimus group compared to 47% in the placebo group (P < 0.02) [48]. The most common side effects were diarrhea and paresthesias. An open-label pilot study of 26 patients with severe refractory plaque psoriasis investigated the efficacy of oral tacrolimus (0.1 mg/kg/day divided equally into two doses per day). Overall, 21/26 (80.37%) of participants hadimprovement in mean PASI scores at the end of the trial. Of these 21 patients with improvement, 19 achieved at least PASI 75 and, of those, 11 scored PASI 90. The most common adverse events experienced V. Ramachandran et al. 132 were: diarrhea, abdominal pain, acral paraesthesia, and myalgia [49]. The combination of systemic evirolimus and tacrolimus was shown to be effective in completely clearing refractory chronic plaque psoriasis in one post-transplant patient [50]. Leflunomide FDA-Approved Indication(s) Leflunomide is FDA-approved to treat active rheumatoid arthritis in adults [8]. Mechanism of Action Leflunomide inhibits de novo pyrimidine synthesis and thereby suppresses cell proliferation and inflammation [8]. When Best to Use Leflunomide can be considered for patients with treatment-resistant, widespread, chronic plaque psoriasis as a second-line systemic therapy or psoriatic arthritis as a disease modifying antirheumatic agent [15, 51]. Dosage Leflunomide should be started at 100 mg/day for 3 days, and then maintained at a dose of 20 mg/ day [8, 15, 51]. Adverse Events The most common adverse events in patients taking leflunomide are diarrhea, nausea, upper respiratory tract infection, headache, hypertension, and alopecia. Baseline LFTs arer recommended as some patients may uncommonly experience elevated liver enzymes [39]. Rarely, patients taking leflunomide have rarely developed pancytopenia, agranulocytosis, thrombocytopenia, Stevens-Johnson syndrome, and toxic epidermal necrolysis [8]. Recommended Monitoring The clinician should obtain history and perform a physical examination. Additionally, at baseline, providers should obtain CBC with differential and LFTs. Pregnancy test (if indicated) may also be obtained before starting leflunomide [39] After starting treatment, a CBC with differential and LFTs should be checked monthly for 6 months, then every 6–8 weeks [8]. If indicated, ongoing pregnancy tests should be checked [8]. Perils and Pitfalls Leflunomide is contraindicated in patients with hypersensitivity to leflunomide and in pregnant or nursing mothers [8]. Hepatotoxicity and fatal liver failure have been reported in patients treated with leflunomide, and it is well worth noting that concurrent methotrexate therapy increases the risk of hepatotoxicity. Concurrent rifampin use leads to increased and potentially toxic peak levels of leflunomide’s active metabolite [8]. Strength of Evidence A randomized, placebo-controlled trial evaluated the safety and efficacy of leflunomide compared to placebo in 109 patients with active psoriatic arthritis and psoriasis with ≥3% BSA involvement … Seventeen percent of the leflunomide-­ treated group achieved PASI 75 compared to 8% in the placebo group (P = 0.048). Additionally, 59% of leflunomidetreated patients achieved 20% in Psoriatic Arthritis Response Criteria compared to 30% in the placebo group (P < 0.0001) [51]. 10 Traditional Systemic Therapy II: Retinoids and Others 133 Penicillin V and Erythromycin Recommended Monitoring FDA-Approved Indication(S) No specific hematologic or biochemical monitoring is recommended for short-term therapy with penicillin V or erythromycin [9, 10]. These medications have good safety profiles. Among other indications, penicillin V and erythromycin are both approved to treat mild to moderate Streptococcus pyogenes-associated infections [9, 10]. Mechanism of Action Penicillin V inhibits biosynthesis of cell-wall mucopeptides during cellular multiplication in penicillin-sensitive microorganisms [10]. Erythromycin inhibits protein synthesis in susceptible microorganisms by binding to the 23S ribosomal RNA molecule within the 50S subunit, preventing elongation of peptide chains [9]. When Best to Use Penicillin V or erythromycin can be used to treat guttate psoriasis when the condition appears related to or associated with a bacterial infection, usually of an upper respiratory nature (classically Streptococcal pharyngitis) [52–54]. If patients present well after the episode of possible infection preceding guttate psoriasis antibiotic treatment may not be appropriate. Dosage Both antibiotics (penicillin V or erythromycin) are given orally in a dosage of 250 mg four times daily, for 14 days [54]. Adverse Events Patients given penicillin V sometimes report nausea, vomiting, abdominal pain, diarrhea, and black hairy tongue [10]. Patients taking erythromycin often develop nausea, vomiting, abdominal pain, diarrhea, and anorexia [9]. Perils and Pitfalls Penicillin is contraindicated in patients with hypersensitivity to the drug; reported hypersensitivity reactions have been fatal [10]. Patients receiving penicillin V have developed Clostridium difficile associated diarrhea. Highdose penicillin is rarely associated with leukopenia, anemia, thrombocytopenia, nephropathy, and neuropathy [10]. Erythromycin is contraindicated in patients taking terfenadine, astemizole, pimozide, or cisapride due to increased risk of fatal ventricular arrhythmia and also in patients with hypersensitivity to erythromycin [9]. Patients taking erythromycin have developed LFT abnormalities, hepatitis, pseudomembranous colitis, Q-T interval, ventricular arrhythmias, erythema multiforme, Stevens-Johnson syndrome, toxic epidermal necrolysis, pancreatitis, convulsions, and reversible hearing loss [9]. While the association of Streptococcal infection of the pharynx is a well-known, forgotten in examination may be perianal source of infection which may lead to withheld antibiotics [55]. Strength of Evidence A few antibiotic regimens have been studied in the contex of guttate psoriasis, including pencillin V, erythromycin, rifampin (proposed to act via anti-inflammatory effects), or combination of these treatments (erythromycin plus rifampin). These studies are largely limited to small and uncontrolled investigations [52–59]. In 2019, a Cochrane Database systematic review sought to assess the effects of antistreptococcal interventions for guttate and chronic V. Ramachandran et al. 134 plaque psoriasis. The conclusion of this study was that there was a major lack of well-designed studies, limiting evidence to low-quality studies with potential for various biases. Overall, the efficacy and safety of antistreptococcal therapy in the manangement of guttate and chronic plaque psoriasis are uncertain and cannot be strongly recommended at present [60]. Apremilast FDA-Approved Indication(s) Apremilast (OTEZLA®) is FDA-approved to treat adults with active psoriatic arthritis and patients with moderate-to-severe plaque psoriasis who are candidates for phototherapy or systemic therapy [61]. Mechanism of Action Apremilast is a selective inhibitor of phosphodiesterase (PDE) 4, the predominant PDE in inflammatory and immune cells, resulting in increased intracellular levels of cyclic adenosine monophosphate (cAMP) [62]. The increased levels of cAMP modulate the expression of inflammatory cytokines involved in the pathogenesis of psoriasis. Inducible nitric oxide synthase, tumor necrosis factor alpha (TNF-α), interferon gamma (IFN-γ), interleukin (IL)-17A/F, IL-22, and IL-23 are all decreased whereas IL-10, an anti-­ inflammatory cytokine, is increased [62, 63]. When Best to Use There are several benefits to apremilast for plaque psoriasis, manifesting in some general principles for its best use. Since apremilast is not immunosuppressive, it is useful in patients with underlying/active infectious diseases (e.g. viral hepatitis, tuberculosis) or those at increased risk for infection (e.g. liver cirrhosis, diabetes, intravenous drug users). In patients with pre-existing disease-­associated (e.g. human i­mmunodeficiency virus, acquired immune deficiency syndrome, malignancy) or iatrogenic immunocompromised states (e.g. chemotherapy), apremilast is a valuable treatment [12]. Furthermore, apremilast may be a consideration in patients for whom the side effects or contraindications of other biologic regimens (e.g. infection, heart failure, demyelinating disorders) exclude them from consideration [64]. Patients with psoriasis may have various comorbidities necessitating numerous daily medications [65]. In such patients, apremilast may be beneficial due to its minimal interactions with cytochrome P450. Serum levels are not increased inpatients taking medications that inhibit cytochrome P450 [66, 67]. Additionally, unlike other disease-modifying agents antirheumatic drugs, aprelimast does not exclude patients with hepatic impairment [68]. In obese patients, the side effect of weight loss may be beneficial in treatment [61]. Apremilast’s oral formulation may also improve adherence in patients with needle phobia (or “needle-fatigue”), elderly patients, or those facing impediments to social life or work activity secondary to injections [69]. Dosage Regardless of use, the FDA-approved daily dosage of apremilast is 30 mg twice daily [39]. This final recommended dosage is supported by clinical phase II trials, which assessed clinical response to differing doses and frequency of apremilast, showing that 30 mg twice daily was the optimal regimen [70–72]. To improve the tolerability, gradual uptitration is advised in achieving the final recommended dose. The following regimen is suggested [73]: • • • • • • Day 1: 10 mg in the morning Day 2: 10 mg in morning and 10 mg in evening Day 3: 10 mg in morning and 20 mg in evening Day 4: 20 mg in morning and 20 mg in evening Day 5: 20 mg in morning and 30 mg in evening Day 6 and thereafter: 30 mg in morning and 30 mg in evening 10 Traditional Systemic Therapy II: Retinoids and Others 135 Adults with severe renal impairment (end-­ not differ from the extremely low rates observed stage renal disease stage 3b and above) are rec- in the ESTEEM 1 and 2 studies [77]. These ommended to take 30 mg once daily. During the short- and long-term findings were corroborated titration stage at the start of therapy, only the and validated by the LIBERATE studies [78, 79]. morning dose should be given [74]. Real-world investigation outside of clinical trials also validate the strong safety profile found in phase 3 trial data [80] (Table 10.4). Adverse Events Overall, the safety data does not seem to show significant increase in serious infections, opporSince apremilast functions as an anti-­inflammatory tunistic infections, major cardiac events, organ rather than an immunosuppressant, no increased damage, or malignancies. Additionally, there was risk of opportunistic infections has been observed no change in laboratory abnormalities [75–79]. [68]. In the Efficacy and Safety Trial Evaluating the Effects of Apremilast in Psoriasis (ESTEEM) phase III trials (ESTEEM 1 and 2), adverse Recommended Monitoring effects were compared in the 0–16-week period (placebo-controlled) and assessed in the solely Apremilast has the added benefit of no required apremilast exposure period (0–52 weeks) laboratory monitoring, such as liver and kid(Table 10.3) [75, 76]. ney function test, due to lack of organ-specific The most common adverse events were diar- or cumulative toxicities with this medication rhea, nausea, upper respiratory tract infection, [47]. However, monitoring patients for adverse nasopharyngitis, and headaches (Table 10.2). sequelae of adverse effects is warranted. Of these, the adverse events most different from Although the risk is low, patients with a history the placebo group were: diarrhea and nausea. of depression, suicidal ideation, or history of suiLong-­term (0 to ≥156 weeks) investigation of cide should be closely monitoring for worsenpooled data from ESTEEM 1 and 2 showed no ing symptoms during therapy [69, 81]. Advising new adverse events affecting ≥5% of the cohort patients, family members, and caregivers of these were observed [77]. Overall, adverse events, seri- side effects is recommended. Additionally, the ous adverse events, and adverse events leading to risk of weight loss warrants periodic body weight drug discontinuation did not increase with long-­ checks, especially in underweight patients. term drug use. Furthermore, the rates of major Clinically significant weight loss should prompt cardiac events, depression, and malignancy did medication discontinuation [68, 81]. Table 10.3 Pooled adverse event data from the ESTEEM 1 and 2 phase III clinical trials for apremilast. AE = adverse events ≥1 AE ≥1 severe AE ≥1 serious AE ≥1 AE leading to drug withdrawal ≥1 AE leading to death Placebo-controlled period (0–16 week) Apremilast Placebo Placebo ESTEEM 1 ESTEEM 1 ESTEEM 2 (n = 560) (n = 136) (n = 282) 157 82 (60.3%) 388 (69.3%) (55.7%) 9 (3.2%) 6 (4.4%) 20 (3.6%) 8 (2.8%) 3 (2.2%) 12 (2.1%) 9 (3.2%) 7 (5.1%) 29 (5.2%) Apremilast ESTEEM 2 (n = 272) 185 (68.0%) Apremilast-exposure period (0–52 weeks) Apremilast Apremilast ESTEEM 2 ESTEEM 1 (n = 380) (n = 804) 633 (78.7%) 296 (77.9%) 12 (4.4%) 5 (1.8%) 15 (5.5%) 48 (6.0%) 34 (4.2%) 59 (7.3%) 33 (8.7%) 18 (4.7%) 27 (7.1%) 1 (0.4%) 0 (0.0%) 1 (0.1%) 0 (0.0%) 0 (0.0%) 1 (0.2%) V. Ramachandran et al. 136 Table 10.4 Pooled side effect data from the ESTEEM 1 and 2 phase III clinical trials for apremilast Diarrhea Upper respiratory tract infection Nausea Nasopharyngitis Headache Tension headaches Placebo-controlled period (0–16 week) Apremilast Placebo Placebo ESTEEM 1 ESTEEM 1 ESTEEM 2 (n = 560) (n = 136) (n = 282) 20 (7.1%) 8 (5.9%) 105 (18.8%) 21 (7.4%) 6 (4.4%) 57 (10.2%) Apremilast ESTEEM 2 (n = 272) 43 (15.8%) 13 (4.8%) Apremilast-exposure period (0–52 weeks) Apremilast Apremilast ESTEEM 2 ESTEEM 1 (n = 380) (n = 804) 150 (18.7%) 55 (14.5%) 143 (17.8%) 35 (9.2%) 19 (6.7%) 23 (8.2%) 13 (4.6%) 12 (4.3%) 50 (18.4%) 20 (7.4%) 17 (6.3%) 14 (5.1%) 123 (15.3%) 108 (13.4%) 52 (6.5%) 52 (6.5%) Perils and Pitfalls 9 (6.6%) 6 (4.4%) 1 (0.7%) 2 (1.5%) 88 (15.7%) 41 (7.3%) 31 (5.5%) 31 (5.5%) 63 (16.6%) 55 (14.5%) 22 (5.8%) 29 (7.6%) mean change (−6.6%), and pruritus visual analogue scale (VAS) score decrease (−31.5%) Patients with known hypersensitivity to apre- were significantly different in apremilast-treated milast or any excipients in its formulation are patients compared to placebo-treated patients contraindicated from this medication [82]. (5.3%, 17.0%, 3.9%, −2.1%, and − 7.3%, respec­ Concurrent use of cytochrome P450 activators tively) (all P < 0.0001) [75]. (e.g. rifampin) can lead to loss of efficacy [82]. Similarly, another phase III, multicenter, The effects of apremilast on pregnancy, labor, double-­blind, placebo-controlled study (ESTEEM delivery, and lactation in humans is unknown. 2) randomized 274 adults (2:1) to apremilast of Although cases reports exist regarding its use in placebo. By week 16, the percentage of patients pediatric patients, apremilast has not been stud- apremilast-treated patients achieving PASI-75 ied in this population and is off-label in this pop- (28.8%), PASI-50 (55.5%), sPGA score of 0 or ulation [82]. 1 (20.4%), DLQI mean change (−6.7%), and Dose titration is important as the gastroin- pruritus VAS score decrease (−33.5%) were sigtestinal side effects are much prevalent when nificantly different in apremilast-­treated patients not performed, leading to higher rates of drug compared to placebo-­ treated patients (5.8%, withdrawal [73]. Xanthines, which are found in 19.7%, 4.4%, −2.8%, and − 12.2%, respectively) caffeine, can increase cAMP through PDE inhi- (all P < 0.0001) [55]. The LIBERATE studies bition. This can lead to increased risk of diarrhea validate these prior two studies’ findings over the or other side effects [83]. long-term (104 weeks) [78, 79]. There is some evidence that apremilast is Apremilast does not appear to be as efficacious more effect in biologic-naïve patients reflected as other biologics as demonstrated by its lower by a higher efficacy in this group of patients in PASI-75 scores compared to other biologics the ESTEEM trials [75, 76]. [84, 85]. The German Society of Dermatology’s consensus psoriasis treatment guidelines noting the drug as the least efficacious biologic [86]. Strength of Evidence Nevertheless, real-world multi-center studies cite apremilast as a valuable part of the dermaA phase III, multicenter, double-blind, placebo-­ tologists’ armamentarium against psoriasis. One controlled study (ESTEEM 1) randomized 844 such retrospective study of 148 patients noted a adults (2:1) to apremilast or placebo. By week greater proportion of apremilast monotherapy 16, Psoriasis Area and Severity Index (PASI)- patients achieving response (PASI-75) (26/59, 75 (33.1%), PASI-50 (58.7%), static Physician’s 44.1%) compared to patients undergoing comGlobal Assessment (sPGA) score of 0 or 1 binational therapy (apremilast plus one of: pho(21.7%), dermatology life quality index (DLQI) totherapy, systemic therapy, biologic therapy) 10 Traditional Systemic Therapy II: Retinoids and Others 137 4. Soriatane [package insert]. Coral Gables: Stiefel (33/89, 37.1%) (P = 0.396). However, the authors Laboratories, Inc; 2011. note this is partially due to the patients on com5. Hydrea (hydroxyurea). Food and Drug Administration. binational therapy having more extensive/refracAccessed at https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/016295Orig1s047,s048Lbl. tory disease or psoriatic arthritis (P < 0.001) [80]. pdf. March 2016. Retrieved on June 28, 2019. The percentage of PASI-75 patients in this real-­ 6. TABLOID [package insert]. Research Triangle Park: world study is similar to the ESTEEM 1 and 2 GlaxoSmithKline; 2004. trial data [75, 76]. 7. Prograf [package insert]. Deerfield: Astellas Pharma US, Inc; 2011. Nevertheless, apremilast may be effectively be 8. Arava [package insert]. Bridgewater: Sanofi-Aventis used in the real world as monotherapy or comU.S. LLC; 2011. binational therapy, especially in biologic-naïve 9. Ery-Tab [package insert]. Thousand Oaks: Rebel patients. This notion is complemented by evidence Distributors Corp; 2010. for apremilast use resulting in lower per-­patient 10. Penicillin V Potassium [package insert]. Sellersville: Teva Pharmaceuticals USA; 2011. per-month costs than biologic treatment [87]. Conclusion Several factors should be considered when selecting a regiment for a patient with psoriasis including pre-existing comorbidities, special patient populations, resource-limited settings, and adherence. Many treatments, particularly, systemics have contraindications based on preexisting comorbidities or risks [12]. In this chapter, we provide a clinically-oriented summary of second-­line traditional systemic agents to navigate the myriad of treatment options. Nevertheless, an important general principle is that the best t­reatment is one the patient will be adherent to [88–91]. This concept may be considered in deciding treatments. Additionally, combinational treatments with other modalities (e.g. phototherapy) are often more efficacious. References 1. Dogra S, Mahajan R. Psoriasis: epidemiology, clinical features, co-morbidities, and clinical scoring. Indian Dermatol Online J. 2016;7(6):471–80. 2. Schlager JG, Rosumeck S, Werner RN, Jacobs A, Schmitt J, Schlager C, Nast A. Topical treatments for scalp psoriasis. Cochrane Database Syst Rev. 2016 Feb 26;2:CD009687. 3. Le Moigne M, Sommet A, Lapeyre-Mestre M, Bourrel R, Molinier L, Paul C, Montastruc JL. Healthcare cost impact of biological drugs compared with traditional systemic treatments in psoriasis: a cohort analysis in the French insurance database. J Eur Acad Dermatol Venereol. 2014 Sep;28(9):1235–44. 11. Warren RB, Griffiths CE. Systemic therapies for psoriasis: methotrexate, retinoids, and cyclosporine. Clin Dermatol. 2008 Sep-Oct;26(5):438–47. 12. Kaushik SB, Lebwohl MG. Psoriasis: which therapy for which patient: focus on special populations and chronic infections. J Am Acad Dermatol. 2019 Jan;80(1):43–53. 13. Cather JC, Crowley JJ. Use of biologic agents in combination with other therapies for the treatment of psoriasis. Am J Clin Dermatol. 2014;15(6):467–78. 14. Smith J, Cline A, Feldman SR. Advances in psoriasis. South Med J. 2017 Jan;110(1):65–75. 15. Menter A, Korman NJ, Elmets CA, Feldman SR, Gelfand JM, Gordon KB, Gottlieb AB, Koo JY, Lebwohl M, Lim HW, Van Voorhees AS, Beutner KR, Bhushan R. Guidelines of care for the management of psoriasis and psoriatic arthritis: section 4. Guidelines of care for the management and treatment of psoriasis with traditional systemic agents. J Am Acad Dermatol. 2009 Sep;61(3):451–85. 16. Ormerod AD, Campalani E, Goodfield MJD, et al. British Association of Dermatologists guidelines on the efficacy and use of acitretin in dermatology. Br J Dermatol. 2010;162:952–63. 17. Murray HE, Anhalt AW, Lessard R, Schacter RK, Ross JB, Stewart WD, Geiger JM. A 12-month treatment of severe psoriasis with acitretin: results of a Canadian open multicenter study. J Am Ac ad Dermatol. 1991 Apr;24(4):598–602. 18. Werner B, Bresch M, Brenner FM, Lima HC. Comparative study of histopathological and immunohistochemical findings in skin biopsies from patients with psoriasis before and after treatment with acitretin. J Cutan Pathol. 2008;35:302–10. 19. Italian Multicenter Study Group on Cyclosporin in Psoriasis. Cyclosporin versus etretinate: Italian multicenter comparative trial in severe plaque-form psoriasis. Dermatology. 1993;187(Suppl 1):8–18. 20. Lassus A, Geiger JM. Acitretin and etretinate in the treatment of palmoplantar pustulosis: a double-blind comparative trial. Br J Dermatol. 1988;119:755–9. 21. Lowe NJ, Prystowsky JH, Bourget T, Edelstein J, Nychay S, Armstrong R. Acitretin plus UVB therapy for psoriasis. Comparisons with placebo plus 11 Apremilast Jerry Bagel and Elise Nelson Abstract In 2014 apremilast was the first oral FDA approved therapy in almost 20 years. Although the efficacy of apremilast, PASI 75—33% @ week 16 was inferior to the biologic agents approved in its era, i.e. ustekinumab, Adalimumab, PASI 75—71%, the safety of apremilast resulted in many dermatologists prescribing apremilast to their patients with psoriasis. The reason for the expansive use of apremilast is at least partially due to the following factors, no risks of malignancy, serious infections, no opportunistic infections, no laboratory monitoring required. Many psoriatics, in part to direct to consumer marketing, have been fearful of the side effects of biologic agents, yet since topical therapy was not providing adequate efficacy they wanted a different option. In addition to showing efficacy for plaque psoriasis, apremilast has also been studied and revealed efficacy in psoriatic arthritis (FDA approved), psoriatic onychodystrophy, scalp psoriasis, and palmar plantar psoriasis. J. Bagel (*) · E. Nelson Psoriasis Treatment Center of Central New Jersey, East Windsor, NJ, USA The Mount Sinai School of Medicine, New York, NY, USA The University of Pisa, Pisa, Italy © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_11 Key Learning Objectives 1. To understand the mechanism of action of apremilast 2. To understand the appropriate use and efficacy of the apremilast 3. To understand the safety issues of apremilast In 2014 the FDA approved apremilast for the treatment of psoriasis and psoriatic arthritis. Acetretin and cyclosporine were FDA approved in 1996 and 1997 respectively. In this time frame there were six biologic therapies approved, however, apremilast was the first new oral therapy in almost 20 years. As opposed to biologic agents which bind extracellular targets i.e. TNF-alpha, IL-12, IL-23, IL-17, apremilast is a small molecule which acts intracellularly. The mechanism by which apremilast acts is by inhibiting the activity of phosphodiesterase 4 (PDE4). In inflammatory cells, PDE4 catalyzes the conversion of cAMP to AMP. The ratio of cAMP to AMP regulates the transcription of either anti-inflammatory mediators or pro-inflammatory mediators. Higher intracellular concentrations of AMP result in a greater transcription of pro-inflammatory mediators i.e. IL-17, IL-22, TNF alpha and accordingly, less transcription of anti-inflammatory mediators. More specifically, an increase in intracellular 141 142 cAMP activates Protein Kinase A (PKA). PKA activates certain transcription factors, but inhibits NF-KB which is thought to decrease the amount of pro-inflammatory mediators. Hence, by inhibiting PDE4, apremilast increases intracellular cAMP and promotes anti-inflammatory transcription via the activation of the cAMP-response element binding protein. The FDA approval of apremilast was based on two phase III clinical trials, ESTEEM 1 and 2. The ESTEEM 1 and 2 randomized, placebo-­ controlled studies evaluated apremilast 30 mg BID in adult patients with chronic moderate-to-­ severe plaque psoriasis, PASI ≥ 12, BSA ≥ 10, PGA ≥ 3. Patients with a history of prior phototherapy, systemic, and biologic therapy were eligible to participate in the trial. The primary endpoint was PASI-75 response at week 16. In ESTEEM 1, 844 patients participated (562 apremilast, 282 placebo), with a mean age of 46 who had psoriasis for approximately 20 years. The mean PASI = 19, BMI = 31, and over half had prior systemic or biologic therapy. At week 16, PASI-75 was achieved by 33.1% in the apremilast cohort vs placebo which was 5.3%. In addition, the PASI-50, PASI change from baseline, Scalp PGA = 0–1, NAPSI-50, and DLQI % response was 59, 52, 47, 33, 70 in the apremilast cohort vs 17, 17, 18, 15, 34 for placebo. Evaluating psoriatic onychodystrophy there was a 22% improvement in NAPSI scores within 16 weeks. PASI-75 response was maintained through week 32. In those patients who obtained a PASI-­ 75 at week 32, there was an 80% improvement in PASI through Week 52. The most common adverse events reported in >5% of patients in the apremilast cohorts were diarrhea, nausea, upper respiratory tract infections, and headaches. There were no serious opportunistic infections, the rates of malignancy and laboratory abnormalities were comparable between the apremilast cohort and placebo. The LIBERATE study compared apremilast to placebo and had an active control in etanercept. At week 16, PASI-75 for placebo, apremilast, and etanercept was 12%, 40%, and 48% respectively. In this study apremilast was con- J. Bagel and E. Nelson tinued through week 104 without any increase in adverse events noted. There have been reports of weight loss associated with apremilast. In view of the fact that over 35% of patients have a BMI > 30, weight loss is generally not a concern. However, for those thinner individuals and elderly, weight loss may be an issue. In addition diarrhea and/or vomiting may result in electrolyte imbalance especially in those taking diuretics. Diarrhea is listed as a warning, it tends to occur in the first two weeks and resolve over time with continued dosing. It can be severe, reduce dose/discontinue, if needed. Many of the nuisance side effects i.e. headaches, diarrhea, nausea, and vomiting may be ameliorated by prolonging the tapering period of apremilast at the initiation of therapy i.e. instead of a 5 day taper starting at 10 mg on day 1, advancing to 30 mg bid on day 6, a 10 or 15 day taper significantly decreases the incidence of these nuisance side effects. There is a warning related to depression, so it is important to exclude patients with a history of major depressive episodes. Based on the phase 3 Palace 1–3 clinical trials apremilast was FDA approved for psoriatic arthritis. At week 16 apremilast yielded ACR 20 = 32%–41%, placebo = 19%. At week 260, the ACR 20, 50, 70 for apremilast was 67%, 44%, and 27% respectively. A significant number of these subjects had been biologically experienced and biologic-failures. ACR 20 = 23% response rates in biologic failures was lower than biologically naive = 43% at week 16. Overall swollen joint counts and tender joint counts decreased by 87% and 80% respectively over 260 weeks. In addition 80% and 54% of subjects obtained dactylitis count = 0 and enthesitis score = 0 at 260 weeks of treatment. While apremilast is not the most efficacious treatment for psoriasis, it is popular due to its safety profile. Based on its mechanism of action, apremilast does not suppress any of the extracellular pro-inflammatory molecules as much as monoclonal antibodies. By not suppressing as much, there is no increase in serious infections, hence, apremilast acts as an immunomodulating agent not as an immunosupressive. There were 11 Apremilast no serious opportunistic infections, no increase in malignancy with cumulative experience, laboratory anomalies were infrequent and transient, and in fact no laboratory monitoring is required. In addition, a phase IV study evaluated apremilast 30 mg bid in adults with moderate-to-­ severe palmoplantar psoriasis. At week 32, 24% patients randomized to apremilast were clear or almost clear. In a post-hoc analysis of ESTEEM 1 and 2 and a phase II study approximately 48% (n = 92) were clear or almost clear at week 16, however the placebo rate was rather high with an average of 27% (n = 52). Baseline PPPGA ≥ 3. In a Phase III study in patients with moderate to severe scalp psoriasis (SSA ≥ 20%) 43% of patients treated with apremilast at week 16 were clear or almost clear compared to placebo = 14%. Scalp itch and whole body itch response was seen in 47% and 45% of patients respectively. In my experience, many patients who opt for apremilast tend to be hesitant of biologic risk or much less commonly needle phobic. However, based on the data, if patients do not capture a 25% improvement within 8 weeks there is not much likelihood of treatment success. Expectations with apremilast need to be defined letting patients know that there are other more efficacious options should apremilast not work for them. Apremilast has not been evaluated in combination with biologics. Ustekinumab may clear psoriasis, but not psoriatic arthritis as well. By adding apremilast instead of methotrexate better efficacy with less toxicity can be obtained. A similar scenario with adalimumab, where the joints may do well, but in patients who don’t achieve full skin clearance, adding apremilast instead of methotrexate may be helpful. In addition combining apremilast with ultraviolet B has shown synergistic efficacy. Bibliography 1. Bagel J, Nelson E, Keegan BR. Apremilast and narrowband ultraviolet-B combination therapy for treating moderate-to-severe plaque psoriasis. J Drugs Dermatol. 2017 Oct 1;16(10):957–62. 2. Crowley J, Thaçi D, Joly P, Peris K, Papp KA, Goncalves J, Day RM, Chen R, Shah K, Ferrándiz 143 C, Cather JC. Long-term safety and tolerability of apremilast in patients with psoriasis: pooled safety analysis for ≥156 weeks from 2 phase 3, randomized, controlled trials (ESTEEM 1 and 2). J Am Acad Dermatol. 2017 Aug;77(2):310-7.e1. https://doi. org/10.1016/j.jaad.2017.01.052. Epub 2017 Apr 14. 3. Danesh MJ, Beroukhim K, Nguyen C, Levin E, Koo J. Apremilast and adalimumab: a novel combination therapy for recalcitrant psoriasis. Dermatol Online J. 2015 Jun 16;21(6). pii: 13030/qt5gf406zs. 4. Duffin KC, Papp KA, Bagel J, Levi E, Chen R, Gottlieb AB. Evaluation of the physician global assessment and body surface area composite tool for assessing psoriasis response to apremilast therapy: results from ESTEEM 1 and ESTEEM 2. J Drugs Dermatol. 2017 Feb 1;16(2):147–53. 5. Kavanaugh A, Mease PJ, Gomez-Reino JJ, Adebajo AO, Wollenhaupt J, Gladman DD, Hochfeld M, Teng LL, Schett G, Lespessailles E, Hall S. Longterm (52-­ week) results of a phase III randomized, controlled trial of apremilast in patients with psoriatic arthritis. J Rheumatol. 2015 Mar;42(3):479–88. https://doi. org/10.3899/jrheum.140647. Epub 2015 Jan 15. 6. Kavanaugh A, Gladman DD, Edwards CJ, Schett G, Guerette B, Delev N, Teng L, Paris M, Mease PJ. Long-term experience with apremilast in patients with psoriatic arthritis: 5-year results from a PALACE 1-3 pooled analysis. Arthritis Res Ther. 2019 May 10;21(1):118. https://doi.org/10.1186/ s13075-019-1901-3. 7. Langley A, Beecker J. Management of common side effects of apremilast. J Cutan Med Surg. 2018 Jul/Aug;22(4):415–21. https://doi.org/ 10.1177/1203475417748886. Epub 2017 Dec 31. Review. 8. Nguyen CM, Leon A, Danesh M, Beroukhim K, Wu JJ, Koo J. Improvement of nail and scalp psoriasis using apremilast in patients with chronic psoriasis: Phase 2b and 3, 52-week randomized placebo-controlled trial results. J Drugs Dermatol. 2016 Mar;15(3): 272–6. 9. Papp K, Reich K, Leonardi CL, Kircik L, Chimenti S, Langley RG, Hu C, Stevens RM, Day RM, Gordon KB, Korman NJ, Griffiths CE. Apremilast, an oral phosphodiesterase 4 (PDE4) inhibitor, in patients with moderate to severe plaque psoriasis: results of a phase III, randomized, controlled trial (Efficacy and Safety Trial Evaluating the Effects of Apremilast in Psoriasis [ESTEEM] 1). J Am Acad Dermatol. 2015 Jul;73(1):37–49. https://doi.org/10.1016/j. jaad.2015.03.049. 10. Paul C, Cather J, Gooderham M, Poulin Y, Mrowietz U, Ferrandiz C, Crowley J, Hu C, Stevens RM, Shah K, Day RM, Girolomoni G, Gottlieb AB.Efficacy and safety of apremilast, an oral phosphodiesterase 4 inhibitor, in patients with moderate-to-severe plaque psoriasis over 52 weeks: a phase III, randomized controlled trial (ESTEEM 2). Br J Dermatol. 2015 Dec;173(6):1387–99. https://doi.org/10.1111/ bjd.14164. Epub 2015 Nov 7. 12 Etanercept Andrew F. Alexis and Charlotte M. Clark Abstract Key Learning Objectives Etanercept is a soluble dimeric fusion protein 1. To understand the mechanism of action that was the first anti-tumor necrosis factor of Etanercept (TNF-α(alpha)) drug to be approved for the 2. To understand the appropriate use and treatment of psoriasis and psoriatic arthritis. It efficacy of the Etanercept is administered subcutaneously by self-­ 3. To understand the safety issues of injection. As with other biologic drugs for Etanercept psoriasis, etanercept offers a targeted approach to treatment that lacks end organ side effects of traditional systemic therapies such as methotrexate, cyclosporine, or acitretin. With over Introduction 8 years of postmarketing experience in psoriasis (and over 14 years since approval for modEtanercept was the first anti-tumor necrosis facerate to severe RA in 1998), a large body of tor (TNF-α(alpha)) drug to be approved for the safety data exists for etanercept. Here, we treatment of psoriasis and psoriatic arthritis. review the safety and efficacy of etanercept in As a TNF-α(alpha) blocking agent, etanercept the treatment of plaque psoriasis and psoriatic modulates inflammatory processes of innate and arthritis, with emphasis on published Phase 3 extrinsic immune responses, cell trafficking, and and Phase 4 clinical trial data. Safety considacute and chronic inflammation that are abererations, recommended monitoring, and studrant in psoriasis. In this chapter, the safety and ies of combination therapy are also discussed. efficacy of etanercept in the treatment of plaque psoriasis and psoriatic arthritis will be reviewed, with emphasis on published Phase 3 and Phase 4 clinical trial data. A. F. Alexis (*) Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA e-mail: aalexis@chpnet.org C. M. Clark Department of Dermatology, Columbia University Medical Center, New York, NY, USA © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_12 Background Psoriasis is a chronic inflammatory disease, characterized by hyperkeratotic epidermal lesions formed in response to T cell activation and the 145 146 associated release of proinflammatory cytokines [1]. Although the exact pathogenesis of psoriasis remains to be fully elucidated, TNF appears to play a key role in the inflammatory cascade associated with psoriasis and psoriatic arthritis. When compared to uninvolved skin, greater concentrations of TNF-α are expressed in the stratum corneum of psoriatic lesions [2]. Etanercept is a TNF-α inhibitor and FDA approved drug for the treatment of adult patients with chronic moderate to severe plaque psoriasis who are candidates for systemic therapy or phototherapy. Etanercept is also approved for the treatment of other TNF mediated inflammatory diseases such as rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, and polyarticular juvenile idiopathic arthritis. Structure and Mechanism of Action Etanercept is a soluble dimeric fusion protein that includes two TNF-α receptors fused to the constant region (Fc) of human IgG1, allowing it to bind specifically with non-membranous bound TNF-α [1, 3, 4]. Etanercept binds to and inactivates TNF [3]. Furthermore, etanercept modifies responses induced by TNF activity, such as adhesion molecule expression (needed for leukocyte migration) and blood cytokine levels [5]. Therapy is administered as a subcutaneous injection by patients at home. The peak absorption of etanercept is at 51 h, with a mean half-life of 68 h [4]. Etanercept in the Treatment of Psoriatic Arthritis Psoriatic skin lesions typically precede the onset of joint symptoms and therefore, dermatologists can potentially diagnose psoriatic arthritis in early stages through careful history and examination. Elevated TNF-α levels have been found in psoriatic arthritis joint fluid in comparison A. F. Alexis and C. M. Clark to osteoarthritis controls [2]. Psoriatic arthritis patients treated with etanercept have shown to have significant reductions in cutaneous psoriasis lesions as well as improvement of their arthritis [1, 6]. Etanercept (25 mg SC twice-weekly) has been shown to be efficacious in the treatment of psoriatic arthritis as evidenced by significantly greater percentages of subjects achieving a 20% or greater reduction in American College of Rheumatology criteria (ACR20) scores [6]. Significant inhibition of radiographic disease progression (measured by mean change in modified total Sharp score) has also been shown in controlled clinical trials of etanercept in the treatment of psoriatic arthritis [6, 7]. Etanercept in the Treatment of Psoriasis The safety and efficacy of etanercept has been shown in large double blinded placebo-controlled trials [8–10]. Based on US and Global phase III trial data the dosage approved by the US FDA for the treatment of plaque psoriasis is 50 mg SC twice weekly for 12 weeks followed by 50 mg SC thereafter. Statistically significant proportions of etanercept treated subjects achieved a 75% or greater reduction in Psoriasis Area and Severity Index (PASI 75) scores compared to placebo (Table 12.1) [6, 7]. Clinically meaningful improvements in the quality of life have also been demonstrated in psoriasis patients treated with etanercept [6]. After discontinuation of therapy, a gradual onset of disease recurrence is observed. In one study, median time to disease relapse (measured as ≥50% loss of PASI improvement achieved from baseline after 24 weeks of therapy) was 3 and 2 months, in PASI 50 and PASI 75 responders, respectively [8]. Successful discontinuation and re-treatment with etanercept has also been shown [8, 9]. Therapeutic efficacy after dosage reduction at 12 weeks from 50 mg SC twice weekly to 50 mg once weekly is maintained by a majority of patients as demonstrated by the percentage of sub- 12 Etanercept 147 Table 12.1 The US and Global Phase III psoriasis clinical trials; study design, baseline characteristics, and clinical outcomes [6, 7] Study design Number of patients Duration Drug regimen US Phase III clinical trial [6] Multicenter (47 sites), randomized, double-blinded, placebo-controlled, parallel-group 672 patients were randomized, 652 participated in study treatment 24 weeks 1. Placebo (n = 166) for 12 weeks, then etanercept 25 mg BIW for 12 more weeks 2. Low dose (25 mg QIW) (n = 160) 3. Medium dose (25 mg BIW) (n = 162) 4. High dose (50 mg BIW) (n = 164) Efficacy endpoints Demographics and baseline clinical characteristics Duration of psoriasis (years) PASI score Affected body surface area (%) Efficacy Results PASI 50 PASI 75 PASI 90 Mean PASI improvement Results PASI 50 PASI 75 PASI 90 Mean PASI improvement a Placebo group (mean) Primary endpoint: PASI 75 at week 12 Secondary endpoints: 1. PASI 50 and 90 at week 12 2. Physician global assessment 3. Patient global assessment 4. Dermatology life quality index High dose Medium Low dose dose group group group (mean) (mean) (mean) 18.4 ± 0.9 19.3 ± 0.9 18.5 ± 0.9 18.6 ± 0.9 18.3 ± 0.6 18.2 ± 0.7 18.5 ± 0.7 18.4 ± 0.7 28.8 ± 1.4 27.7 ± 1.5 28.5 ± 1.6 29.9 ± 1.6 Placebo group Low dose group Medium dose group 12 weeks 14% 4% 1% 14.0 ± 2.6 41%a 14%a 3%a 40.9 ± 2.4a 24 weeks NA 33% NA NA 58% 25% 6% 50.3 ± 2.5 17.5 (range 1.4–51.2) Etanercept 25 mg BIW group (median) 21.5 (range 0.8–64.6) Etanercept 50 mg BIW group (median) 18.1 (range 0.8–60.5) 16.0 (range 7.0–62.4) 20.0 (range 10.0–95.0) 16.9 (range 4.0–51.2) 23.0 (range 7.8–95.0) 16.1 (range 7.0–57.3) 25.0 (range 10.0–80.0) High dose group Placebo group Etanercept 25 mg BIW group Etanercept 50 mg BIW group 58%a 34%a 12%a 52.6 ± 2.7a 74%a 49%a 22%a 64.2 ± 2.4a 12 weeks 9% 3% 1% NA 64%b 34%b 11%b NA 77%b 49%b 21%b NA 70% 44% 20% 62.1 ± 2.5 77% 59% 30% 71.1 ± 2.2 24 weeks NA 28% NA NA NA 45% NA NA NA 54% NA NA P < 0.001 for comparison with placebo at week 12 P < 0.0001 for comparison with placebo at week 12 b Global Phase III clinical trial [7] Multicenter (50 sites), randomized, double-blinded, placebo-controlled, parallel-group 611 patients were randomized, 583 participated in study treatment 24 weeks 1. Placebo (n = 193) for 12 weeks, then etanercept 25 mg BIW for 12 more weeks 2. Etanercept 25 mg BIW (n = 196) for 24 weeks 3. Etanercept 50 mg BIW (n = 194) for 12 weeks, then etanercept 25 mg BIW for 12 more weeks Primary endpoint: PASI 75 at 12 weeks Secondary endpoints: 1. PASI 50 and 90 at 12 weeks 2. Patient global assessment Placebo group (median) 148 jects maintaining a PASI 50 or greater after “stepdown” therapy [8]. In addition, approximately 30% of the non-responders (those not achieving a PASI 75) after 12 weeks of dose- reduction were shown to achieve a PASI 75 by week 24, despite continued reduced-dose therapy [8]. The long-term safety and efficacy of etanercept 50 mg twice weekly in patients with psoriasis has been investigated in a 96 week open-label extension trial [11]. Significant improvement in psoriasis associated depression in patients treated with etanercept (compared to placebo) was demonstrated in a study that included assessment of the Beck depression inventory (BDI) and Hamilton rating scale for depression (HAM-D) [12]. A recent retrospective study investigating potential racial and ethnic differences found no differences in safety and efficacy variables (including adverse event rates and improvements in body surface area of involvement) between Caucasians, African-Americans, and Hispanics [13]. Combination Therapy Etanercept has been used safely in combination with other agents to enhance or maintain efficacy. Adjunctive topical calcipotriene 0.005% and betamethasone dipropionate 0.064% ointment [14], narrow-band UVB phototherapy [15], or methotrexate [16, 17], has been reported in published trials. The above adjunctive therapies can be considered in cases of waning or insufficient efficacy with etancercept monotherapy. However, potential risks and benefits must be considered and appropriate monitoring is advised when combining therapies (especially those with potential immunosuppressive and/or malignancy risks). Safety A general outline recommending risk assessment prior to commencing etanercept therapy, as well as recommendations for specific monitoring of symptoms and routine laboratory tests when initiating and maintaining etanercept therapy, is provided (Table 12.2) [4, 5, 18, 19]. A. F. Alexis and C. M. Clark Table 12.2 Recommended risk assessment prior to introduction etanercept therapy (a) and recommended symptom and laboratory monitoring prior to initiating and while maintaining etanercept therapy (b) [4, 5, 18, 19] (a) Contraindications Concurrent live vaccination History of etanercept-induced hypersensitivity reactions Use in those with Wegener’s granulomatosis receiving immunotherapy (associated with higher incidence of solid malignancies and does not improve clinical outcome if compared to standard therapy alone) Septicemia, or active infection Relative contraindications/cautions Pregnancy (pregnancy category B), inadequate data for risk assessment for breast feeding, caution advised Caution if CHF, especially CHF grade III–IV new York heart association (NYHA) Caution in patients with or at risk for demyelinating disorders Caution if HBV carrier Personal history of frequent or recurrent infections, including history of chronic open wounds. Caution if TB risk, history of latent TB, or if patient travels or resides to regions with endemic TB or mycoses Caution if moderate to severe alcoholic hepatitis Personal history of uncontrolled diabetes mellitus History of malignancy within the past 5 years or in patients with increased malignancy risk History of blood disorders or myelosuppression Caution in patient >65 years of age Caution use in patients with concurrent immunosuppressants- anakinra or abatacept is not recommended with etanercept therapy In patients with a significant exposure to varicella virus, etanercept therapy should be temporarily discontinued and considered for prophylactic treatment with varicella zoster immune globulin Caution in patients with latex allergy- the needle cover of prefilled syringes and needle cover within needle cap contain latex-derived components (b) Baseline monitoring PPD (baseline and annually) or Quantiferon gold (for latent TB) is required, along with CXR for exclusion of active TB Liver function test (baseline and every 2–6 months), hepatitis B and C testing, complete blood count (baseline and every 2–6 months), basic chemistry (baseline and every 2–6 months), and optional baseline antinuclear antibodies (ANA) Monitoring of signs and symptoms Discontinue etanercept therapy during active infection. Consider empiric anti-fungal treatment for those at risk for invasive fungal infections or for patients residing or travelling to regions where mycoses are endemic 12 Etanercept Table 12.2 (continued) Discontinue etanercept 1–2 weeks prior to surgery, and reinitiation of etanercept therapy 2 weeks after uncomplicated surgical procedures Discontinue etanercept therapy 4 weeks prior to and reinitiate 4 weeks after vaccination (to prevent decreased vaccination efficacy) Discontinue etanercept therapy if malignancy detected, with the exception of cutaneous basal cell carcinoma Periodic evaluation of signs and symptoms of opportunistic infections Yearly examination for skin cancer detection If pregnancy occurs, discuss risks versus benefits (pregnancy category B drug) Consider risks versus benefits in patients with cardiovascular co-morbidities Provide annual inactivated influenza vaccination, preferably prior to biologic therapy initiation Monitor hepatitis B carriers for reactivation during and several months after initiating etanercept therapy. If reactivation occurs, consider discontinuing etanercept and beginning anti-viral therapy CHF congestive heart failure, PPD purified protein derivation, CXR chest X-ray As with other anti-TNF therapies, serious and opportunistic infections, (including bacterial, mycobacterial, fungal, viral, parasitic) have been observed in patients receiving etanercept. Reactivation of latent tuberculosis (TB) is a risk for this class of biologics and therefore screening for TB is required at baseline and annually [4, 5, 18, 19]. Fatal cases of reactivation of hepatitis B virus have also been reported and as such, baseline screening for viral hepatitis is required before initiation of treatment with etanercept [4, 5, 18, 19]. Other non-infectious considerations include congestive heart failure (CHF) and demyelinating disorders. Worsening of CHF and rare new onset cases of CHF have been reported in patients taking etanercept [3–5, 19, 20]. In addition, there are rare reports of exacerbation or new onset of demyelinating disorders among etanercept patients [4, 5, 19]. Therefore, TNF inhibitors, including etanercept should be considered with caution in patients with pre-existing CHF or demyelinating disorders [4, 5, 19]. 149 Malignancies Malignancies have been reported in patients using etanercept and other anti-TNF agents. Rates of malignancies among patients with anti-­TNF therapy have been analyzed using clinical trial data and large rheumatologic disease and biologic therapy databases [21–25]. Reported malignancies include lymphomas, non-­ melanoma skin cancers (NMSC), leukemia, and rare cases of Merkel Cell carcinoma [26]. Among adult psoriasis patients treated with etanercept in clinical trials (n = 4410) up to 36 months, the observed rate of lymphoma was comparable to that in the general population [5]. No cases were observed in the etanercept- or placebo-­ treated patients during the controlled portions of these trials. However, in the controlled portions of etanercept trials in adult rheumatology patients - with rheumatoid arthritis (RA), ankylosing spondylitis (AS), and psoriatic arthritis (PsA)—two lymphomas were observed among etanercept treated patients versus 0 among control patients [5]. In combined data of controlled and uncontrolled portions of clinical trials of etanercept for adult rheumatology patients (RA, AS, and PsA) representing 12,845 patient years of therapy, the rate of lymphoma observed (0.10 per 100 patient-years) was threefold higher than that expected in the general US population based on the Surveillance, Epidemiology, and End Results (SEER) Database [5]. However, higher rates of lymphoma (up to several-fold) have been reported in the RA patient population compared to the general population [5]. Higher rates of non-melanoma skin cancer have been observed among psoriasis patients treated with etanercept. In controlled clinical trials of adult psoriasis patients (n = 1245), the rate of NMSC observed was 3.54 cases per 100 patient-years among those treated with etanercept versus 1.28 cases per 100 patient-years among control patients [5]. Rare cases of Merkel cell carcinoma have been reported in the post-­ marketing period [5]. For all etanercept patients at risk for skin cancer, full body skin examinations are recommended [5]. 150 Excluding lymphoma and NMSC, no difference in exposure-adjusted rates of malignancies between etanercept and placebo-treated patients have been observed in the controlled portions of etanercept clinical trials (across all indications) [5, 27]. In the pediatric population, malignancies including lymphoma and leukemia have been reported, particularly among children or adolescents taking concomitant immunosuppressive agents [5]. References 1. Krueger JG. The immunologic basis for the treatment of psoriasis with new biologic agents. J Am Acad Dermatol. 2002;46(1):1–23. quiz 23–6 2. Gratch N, Alexis AF. Etanercept in dermatology and off-label use. In: Weinberg JM, Buchholz R, editors. TNF-alpha inhibitors. Basel: Birkhauser Verlag part of Springer Science and Business Media; 2006. p. 55–63. 3. Weinberg JM. An overview of infliximab, etanercept, efalizumab, and alefacept as biologic therapy for psoriasis. Clin Ther. 2003;25(10):2487–505. 4. Papp KA. The safety of etanercept for the treatment of plaque psoriasis. Ther Clin Risk Manag. 2007;3(2):245–58. 5. Enbrel: Enbrel® (etanercept) prescribing information. Manufactured by Immunex Corporation. Marketed by Amgen, Inc. and Pfizer, Inc. Thousand Oaks: Amgen; 2012. 6. Mease PJ, Kivitz AJ, Burch FX, et al. Etanercept treatment of psoriatic arthritis: safety, efficacy, and effect on disease progression. Arthritis Rheum. 2004;50(7):2264–72. 7. Mease PJ, Kivitz AJ, Burch FX, et al. Continued inhibition of radiographic progression in patients with psoriatic arthritis following 2 years of treatment with etanercept. J Rheumatol. 2006;33(4):712–21. 8. Papp KA, Tyring S, Lahfa M, et al. A global phase III randomized controlled trial of etanercept in psoriasis: safety, efficacy, and effect of dose reduction. Br J Dermatol. 2005;152(6):1304–12. 9. Leonardi CL, Powers JL, Matheson RT, et al. Etanercept as monotherapy in patients with psoriasis. N Engl J Med. 2003;349(21):2014–22. 10. Gordon KB, Gottlieb AB, Leonardi CL, et al. Clinical response in psoriasis patients discontinued from and then reinitiated on etanercept therapy. J Dermatolog Treat. 2006;17(1):9–17. 11. Tyring S, Gordon KB, Poulin Y, et al. Long-term safety and efficacy of 50 mg of etanercept twice weekly in patients with psoriasis. Arch Dermatol. 2007;143(6):719–26. A. F. Alexis and C. M. Clark 12. Tyring S, Gottlieb A, Papp K, et al. Etanercept and clinical outcomes, fatigue, and depression in psoriasis: double-blind placebo-controlled randomised phase III trial. Lancet. 2006;367(9504):29–35. 13. Shah SK, Arthur A, Yang YC, Stevens S, Alexis AF. A retrospective study to investigate racial and ethnic variations in the treatment of psoriasis with etanercept. J Drugs Dermatol. 2011;10(8): 866–72. 14. Kircik LH. Topical calcipotriene 0.005% and betamethasone dipropionate 0.064% maintains efficacy of etanercept after step-down dose in patients with moderate-­ to-severe plaque psoriasis: results of an open label trial. J Drugs Dermatol. 2011;10(8): 878–82. 15. Gambichler T, Tigges C, Scola N, et al. Etanercept plus narrowband ultraviolet B phototherapy of psoriasis is more effective than etanercept monotherapy at 6 weeks. Br J Dermatol. 2011;164(6):1383–6. 16. Strober BE. Successful treatment of psoriasis and psoriatic arthritis with etanercept and methotrexate in a patient newly unresponsive to infliximab. Arch Dermatol. 2004;140(3):366. 17. Strober BE, Clarke S. Etanercept for the treatment of psoriasis: combination therapy with other modalities. J Drugs Dermatol. 2004;3(3):270–2. 18. Lebwohl M, Bagel J, Gelfand JM, et al. From the medical Board of the National Psoriasis Foundation: monitoring and vaccinations in patients treated with biologics for psoriasis. J Am Acad Dermatol. 2008;58(1):94–105. 19. 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(5): 826–50. 20. van Lumig P, Driessen R, Berends M, Boezeman J, van de Kerkhof P, de Jong E. Safety of treatment with biologics for psoriasis in daily practice: 5-year data. J Eur Acad Dermatol Venereol. 2012;26(3): 283–91. 21. Strangfeld A, Hierse F, Rau R, et al. Risk of incident or recurrent malignancies among patients with rheumatoid arthritis exposed to biologic therapy in the German biologics register RABBIT. Arthritis Res Ther. 2010;12(1):R5. 22. Wolfe F, Michaud K. Biologic treatment of rheumatoid arthritis and the risk of malignancy: analyses from a large US observational study. Arthritis Rheum. 2007;56(9):2886–95. 23. Wolfe F, Michaud K. The effect of methotrexate and anti-tumor necrosis factor therapy on the risk of lymphoma in rheumatoid arthritis in 19,562 patients during 89,710 person-years of observation. Arthritis Rheum. 2007;56(5):1433–9. 24. Pallavicini FB, Caporali R, Sarzi-Puttini P, et al. Tumour necrosis factor antagonist therapy and cancer development: analysis of the LORHEN registry. Autoimmun Rev. 2009;9(3):175–80. Adalimumab for Psoriasis 13 Cooper B. Tye and Jennifer C. Cather Key Learning Objectives 1. To understand the mechanism of action of Adalimumab 2. To understand the appropriate use and efficacy of the Adalimumab 3. To understand the safety issues of Adalimumab Introduction Psoriasis is an immune-mediated inflammatory disease that affects 2–3% of the population worldwide [1]. Psoriasis patients experience episodic flares with few spontaneous remissions. Psoriasis is characterized by scaling, erythema, and thickened plaques on the skin. In addition to physical symptoms, psoriasis patients often experience psychosocial issues and have poor health related quality of life (HRQoL) [2]. Psoriasis is a complex disease, and onset results from a combination of genes and the environment [3]. Additionally, some patients with psoriasis develop psoriatic C. B. Tye UT Health San Antonio School of Medicine, San Antonio, TX, USA e-mail: tye@livemail.uthscsa.edu J. C. Cather (*) Mindful Dermatology, Modern Dermatology and Modern Research Associates, Dallas, TX, USA © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_13 arthritis, a painful and potentially debilitating inflammatory arthritis (6–42% depending on the population studied) [4]. Psoriasis is also associated with a number of comorbidities including obesity, cardiovascular disease, diabetes (Type II), psychiatric disorders, and metabolic syndrome [5–7]. Individuals with psoriasis may also be at risk for other inflammatory diseases, such as Crohn’s disease and Hidradenitis Suppurativa [8, 9]. A variety of treatment options are available and patients with moderate to severe psoriasis are candidates for systemic therapy [10, 11]. A number of biologic agents have been approved for psoriasis and additional molecules are currently being studied in clinical trials [12]. Here we review adalimumab (brand name Humira), a fully human monoclonal IgG1 antibody that blocks tumor necrosis factor alpha (TNF-a), a cytokine elevated in inflammation [13]. Adalimumab binds both soluble and membrane-bound TNF-a and blocks TNF-a interactions at the p55 and p75 TNF cell surface receptors with high affinity. Adalimumab Adalimumab was first approved in 2002 for rheumatoid arthritis. Today, it is approved for 10 indications which include ankylosing spondylitis, Crohn’s disease, fingernail psoriasis, hidradenitis suppurativa (HS), juvenile idiopathic arthritis, plaque psoriasis, psoriatic arthritis, rheumatoid 153 C. B. Tye and J. C. Cather 154 arthritis, ulcerative colitis, and uveitis [14, 15]. Adalimumab is given by subcutaneous injection, with dosing dependent on indication. Adalimumab is available as a single-use prefilled pen (40 mg/0.8 mL) or a single-use prefilled syringe (40 mg/0.8 mL or 20 mg/0.4 mL). Recently, a citrate free formulation was approved which provides for a better patient injection experience. Adalimumab was the third TNF-inhibitor approved for psoriatic arthritis and psoriasis and other approved TNF-inhibitors for these indications include etanercept (brand name Enbrel), infliximab (brand name Remicade), and certolizumab (brand name Cimzia). Golimumab (brand name Simponi), is a TNF-inhibitor only approved for psoriatic arthritis. A number of guidelines of care for psoriasis have been published, including American Academy of Dermatology guidelines, [16–18] German guidelines, [19, 20] Canadian guidelines, [21] European guidelines, [22, 23], Italian guidelines, [24] and British guidelines [25]. Consensus statements have also explored monitoring and vaccinations with biologics [26] and monitoring comorbidities, [27] while others have examined psoriasis treatment in a case-based manner [28, 29]. A chapter in this textbook is dedicated to exploring the guidelines of care. Here we provide an overview of adalimumab in psoriasis and psoriatic arthritis, as well as in comorbidities including fingernail psoriasis, hidradenitis suppurativa, and include a very brief mention of inflammatory bowel disease. We review the pivotal phase III clinical trials for psoriasis, psoriatic arthritis, fingernail psoriasis, and hidradenitis suppurativa, and ongoing safety and monitoring considerations. We also provide insight into treatment of special populations as well as biologic comparator studies. Biosimilar data is included elsewhere in this text. Adalimumab in Clinical Trials for Psoriasis and Psoriatic Arthritis A number of clinical trials have been conducted for psoriasis and psoriatic arthritis. REVEAL (Randomized controlled evaluation of adalim- umab every other week dosing in moderate to severe psoriasis trial), CHAMPION (Comparative study of adalimumab versus methotrexate versus placebo in psoriasis patients), and ADEPT (Adalimumab efficacy and safety in psoriatic arthritis). Additionally, a number of biologic comparator trials have been performed which we will briefly describe; however, full details are provided in separate chapters in this book. REVEAL The pivotal phase III psoriasis trial, REVEAL, was a 52-week, prospective multicenter, randomized, double-blind placebo-controlled trial of 1212 moderate to severe psoriasis patients, designed to examine efficacy, safety, and tolerability of adalimumab [13]. The criteria for enrollment was similar to many biologics trials, where individuals were at least 18 years old, had been diagnosed with moderate to severe psoriasis for at least 6 months, and had stable psoriasis for at least the past 2 months. Here, moderate to severe psoriasis was defined as 10% or more body surface area affected, a psoriasis area severity index (PASI) of 12 or greater, or a physician’s global assessment (PGA) of at least moderate severity at the baseline visit. Treatment washout periods were required and similar to other biologic trials: topical therapy (2 weeks), phototherapy (2 weeks for UVB phototherapy and 4 weeks for PUVA), systemic therapies (4 weeks), and biologic therapies (12 weeks, or 6 weeks for efalizumab). Low to mid-potency topical corticosteroids were permitted on the palms, soles, face, and intertriginous areas. All potential participants were screened for latent tuberculosis, and if latent disease was present, individuals could enroll as soon as appropriate chemoprophylaxis for tuberculosis was started. Patients with a history of chronic recurrent infections, demyelinating disease, cancer, or lymphoproliferative disease were excluded. The 52-week study trial design included 3 different treatment periods. During the first 16 weeks (Period A), patients were randomized 2:1, adalimumab to placebo. The adalimumab 13 Adalimumab for Psoriasis group received 80 mg adalimumab at week 0 followed by 40 mg adalimumab at week 1 and then 40 mg every other week thereafter for 15 weeks, while the other group received placebo. Period B was defined as weeks 16–32 and at week 16, patients in the placebo arm received 80 mg adalimumab, followed by 40 mg adalimumab every other week beginning in week 17. Also, at week 16, all patients achieving a 75% or greater response rate were entered into a 17-week open label extension, receiving 40-mg adalimumab every other week. Those who did not achieve PASI 75 were entered into a different open label extension, receiving 40 mg every other week. Period C, weeks 33–52, was FDA mandated to examine loss of adequate response in patients who had achieved PASI 75 at weeks 16 and 33. In Period C this set of PASI75 responders were re-­ randomized 1:1 to adalimumab or placebo. Time to loss of response and retreatment data were obtained with this patient population. At week 16, the primary efficacy endpoint of at least a 75% improvement in PASI score (PASI75) was reported as 71% of patients in the adalimumab group compared to 7% of placebo using intent to treat (ITT) analysis. In ITT, those lost to follow-up are considered non-responders in the analysis. PASI 90 and PASI 100 were achieved by 45% and 20%, respectively, in the adalimumab group at 16 weeks, compared to only 2% and 1% in the placebo group. At week 24, 70% of all patients had achieved PASI 75. At week 33, the loss of adequate response phase was initiated. Patients randomized to placebo lost response more frequently (28% compared to 5% in the adalimumab group), and the time to loss of adequate response was shorter in the placebo group. At 33 weeks in the REVEAL study, patients who maintained a PASI75 while randomized to adalimumab treatment (n = 290) at the primary endpoint were further randomized to treatment with adalimumab (n = 250) or placebo (n = 240). At the end of the secondary endpoint of 52 weeks, 68% of individuals randomized to the adalimumab treatment group maintained clear or minimal diseased based on PGA, as opposed to 28% of individuals in the placebo group [13]. 155 Additionally, when looking at PASI scores, 79% of adalimumab treated patients achieved PASI75 at 52 weeks, 54% achieved PASI90, and 32% achieved PASI100. In the control group, 43% of individuals achieved PASI75, 18% achieved PASI90, and 8% achieved PASI100. The REVEAL trial had an open label extension (OLE) period that enabled participants to receive adalimumab therapy for approximately 3 years [30]. For those who achieved PASI75, efficacy was well maintained over the 3-year period, using a last observation carried forward (LOCF) analysis. A subanalysis examined consequences of when therapy was stopped and restarted with adalimumab [30]. Efficacy was slightly higher for those who received continuous adalimumab therapy compared to the retreatment group (75% compared to 73% at week 108 using LOCF). Retreatment response was greatest in patients who had experienced PASI50 or greater at the time retreatment was started. Additionally, individuals who were part of the REVEAL clinical trial, along with 3 other clinical trials, who achieved PASI <75 by 16 weeks, achieved PASI50 to PASI <75 by 33 weeks, and lost response at the end of the trial were eligible to enroll in a long term OLE study. Patients who completed 52 weeks of REVEAL were also eligible for this study. During the REVEAL OLE study, patients received adalimumab 40 mg twice a month for at least 108 weeks, and their responses were analyzed relative to the REVEAL baseline. At the end of the REVEAL OLE study, 88% of treated patients achieved a PASI75, 57% achieved a PASI90, and 38% achieved PASI 100. Also, 71% of these treated patients achieved a PGA of clear or minimal. Adalimumab was well-tolerated in the REVEAL trail. The majority of adverse effects (AEs) were mild to moderate. Less than 2% of patients discontinued use due to adverse events and there were no deaths. Infections included upper respiratory tract infections, nasopharyngitis, and sinusitis. Injection site reactions were more common in the adalimumab group, occurring in 3.2% of the adalimumab group compared to 1.8% of the placebo group. Serious adverse C. B. Tye and J. C. Cather 156 events (SAEs) were comparable in both groups at 16 weeks. Because 16 weeks is not sufficient for identifying AEs, a 3-year extension provides more insight into potential safety concerns. During the 3-year open label extension, adalimumab was well tolerated [30]. There were 2 cases of tuberculosis, 5 candidiasis infections, and 2 deaths (coronary artery disease in a 75-year old man and unknown cause of death in a 47-year old man). There were no cases of lymphoma, lupus-like syndrome, or demyelinating disorders during this period. One of the limitations of the open label extension is that individuals whose dose escalated to 40- mg every week were considered off protocol, and LOCF prior to dose escalation was used in the analysis, although they were still receiving adalimumab. In addition to the physical symptoms of psoriasis, psoriasis can also impact an individual’s ability to work [31]. The effect of adalimumab on work productivity was examined using the Work Productivity and Activity Impairment (WPAI) questionnaire during the REVEAL trial [32]. At 16 weeks, adalimumab decreased psoriasis related work productivity and activity impairment, and improvements in WPAI were seen in the adalimumab treated group compared to the placebo group, 15.5% and 11.1%, respectively. Individuals who were unemployed and had high scores of total work productivity impairment and total activity impairment also had measurably more severe psoriasis. When psoriasis symptoms worsened, patients described increases in pain, increased WPAI scores, and greater impairment in mental and physical component summaries, respectively [33]. A number of subanalyses were also conducted from the REVEAL trial. One analysis examined if there were any differences in adalimumab efficacy or safety between different patient subgroups during the initial 16 weeks of the trial [34]. While improvement in psoriasis was seen consistently across most subgroups, decreased response to adalimumab occurred in patients with greater weight or body mass index. This was most apparent in obese individuals (BMI ≥ 30), where 65% achieved PASI75, compared to 75% of overweight individuals (BMI ≥25 but <30), and 79% of individuals with a normal BMI (BMI 18.5 < 25). There were no significant differences in SAEs between adalimumab and placebo across subgroups. Subgroups examined included age, sex, race, baseline weight intervals, baseline body mass index, disease duration, baseline severity, prior treatments and comorbidities. Another subanalysis explored the effects of comorbidities (e.g. hypertension, psoriatic arthritis, hyperlipidemia, obesity, depression, arthritis, diabetes mellitus, and cardiovascular disease) on patient reported outcomes [35]. Comorbidities were associated with greater impairment in HRQoL and work productivity, measured by the Dermatology Life Quality Index (DLQI), Short Form 36 (SF-36) health survey, and WPAI questionnaire. There were consistent improvements in DLQI, SF-36 physical component summary score, SF-36 mental components summary score, and WPAI in the group receiving adalimumab at 16 weeks. In an analysis of a phase II study, patients receiving adalimumab also experienced a reduction of symptoms of depression compared to the placebo group, measured by the Zung Self-­ rating Depression score [36]. Champion Another psoriasis trial of interest, CHAMPION, was conducted in Europe and Canada and included a comparator arm of methotrexate as well as a traditional placebo arm [37]. Patient inclusion criteria and washout periods were similar to the REVEAL trial (described in detail above) except patients were required to be methotrexate and TNF-inhibitor naïve. CHAMPION was a randomized, double blind, double dummy placebo-controlled trial, and 217 patients were randomized 2:2:1 to adalimumab (40 mg every other week after an 80 mg loading dose), methotrexate (7.5–25 mg weekly), or placebo. The primary endpoint was patients achieving a 75% improvement in PASI score at 16 weeks. At week 16, 79.6% of the adalimumab group achieved PASI75, compared to 35.5% of the methotrexate group and 18.9% of the placebo 13 Adalimumab for Psoriasis group. In addition, 16.7% of the adalimumab group, 7.3% of the methotrexate group, and 2% of the placebo group achieved PASI100. This study has been acknowledged for using the traditional systemic therapy methotrexate as an active comparator. It has also been criticized, because the methotrexate arm was conducted for such a short time period, and a successful methotrexate regimen typically takes a longer period of time and has additional dose modifications. Many patients reported adverse events, including 73.8% of the adalimumab group, 80.9% of the methotrexate group and 79.2% of the placebo group. Most adverse events were mild. Some (5% or 15 patients) withdrew from the study, and 8 of the withdrawals were for adverse events, including 1 from the adalimumab group, 6 from the methotrexate group, and one from the placebo group. Elevated liver enzymes were more common in the methotrexate group compared to the adalimumab or placebo groups, 9.1%, 1.9%, and 7.5% respectively. An additional analysis of the CHAMPION trial examined adverse event free days during the comparator arm of the trial [38]. Those in the adalimumab group experienced more adverseevent free days (36.9 days) compared to those in the methotrexate (8.3 days) or placebo (6.7) groups over the 16-week period. While these data are encouraging, a limitation is the short duration of the comparator arm, allowing only those adverse events occurring within the first 16 weeks to be captured. An analysis of the week 16 efficacy scores of adalimumab compared to methotrexate and placebo stratified by baseline body mass index has been published. At week 16, normal weight (<25 kg/m2), overweight (25 to <30 kg/m2), and obese (> = 30 kg/m2) patients on adalimumab had PASI75 responses of 85%, 85.7%, and 61.3% compared to 43.4%, 29.3%, and 26.1% with methotrexate and 28.6%, 16.7%, and 0% for placebo. Additionally, at week 16, the PASI90 results were 70%, 53.6%, and 35.5% with adalimumab; 26.7%, 7.3%, and 8.7% with methotrexate, and 9.5%, 16.7%, and 0% for placebo. The greatest DLQI improvements were seen in the adalimumab treatment arms [39]. 157 ADEPT Adalimumab has also been studied in psoriatic arthritis. ADEPT was a phase III randomized, double-blind, parallel-group, placebo controlled trial examining adalimumab efficacy and safety in psoriatic arthritis patients [40]. Like other psoriatic arthritis trials, patients who had moderately to severely active psoriatic arthritis and a history of inadequate response to non-steroidal anti-­ inflammatory drugs (NSAIDS) were eligible to participate. Patients were stratified by methotrexate use and psoriasis skin involvement (≥ 3% BSA or < 3%). Approximately half of patients were taking methotrexate at baseline. Patients in the adalimumab arm received 40 mg adalimumab subcutaneously every other week for 48 weeks. If improvement was not seen at week 12 (defined as at least a 20% decrease in both swollen and tender joint counts on 2 consecutive visits), participants could receive corticosteroids or disease modifying anti-rheumatic drugs (DMARDS). Primary endpoints were the American College of Rheumatology 20% improvement (ACR20) response at week 12 and changes in modified total Sharp score (mTSS) of structural damage at week 24. In addition, HRQoL was measured in all patients. For those patients who also had 3% or more body surface area of psoriasis, skin improvement was also measured. At 12 weeks, 58% of the adalimumab group achieved ACR20, compared to 14% of the placebo group. Patients in the adalimumab group had a modified Psoriatic Arthritis Response Criteria (PsARC) response rate of 62% at week 12, and 60% at week 24, compared to placebo rates of 26% at week 12 and 23% at week 24. Adalimumab also inhibited joint destruction, as is seen with the TNF-inhibitors etanercept and infliximab [41]. Radiographs were taken at baseline, 24, and 48 weeks. The mean change in mTSS at 24 weeks was −0.1 for patient receiving adalimumab compared to 0.9 for those receiving placebo. At 48 weeks, patients on adalimumab showed a mean change in mTSS of −0.2. Erosion scores were examined, and there was improvement in those receiving adalimumab (mean change 0) compared to placebo (mean change 158 0.6). When joint space narrowing scores were examined, there was a mean change of −2 in the adalimumab group compared to 0.4 in those receiving placebo. Skin symptoms also improved in those patients who also had psoriasis. Improvements in health-related quality of life, fatigue, and disability, measured using the DLQI, SF-36, Functional Assessment of Chronic Illness Therapy - Fatigue (FACIT-Fatigue) Scale and HAQ DI were also seen [42]. HAQ DI scores improved significantly in patients receiving adalimumab compared to the control group. At 24 weeks, patients with psoriatic arthritis treated with adalimumab saw improvement of their HAQ DI score to 0.4, as opposed to a score of 0.9 in the placebo group. In addition, patients treated with 40 mg of adalimumab every other week showed a reduction rate in their HAQ DI burden of 47% and 49% respectively at 12 and 24 weeks, while those in the placebo group had rates of 1% and 3% during the same time frame. Regarding safety data from the ADEPT trial, adverse events were similar between the adalimumab and placebo groups at 24 weeks [40]. There were 12 serious adverse events, 7 in the adalimumab group and 5 in the placebo group. Treatment was discontinued by 4 individuals because of adverse events, 3 in the adalimumab group and 1 in the placebo group. Alanine aminotransferase levels (ALT) were elevated more frequently in patients in receiving adalimumab, and in most cases ALT elevations were transient, resolving without discontinuing adalimumab. Elevated ALT levels were observed in individuals taking concomitant methotrexate, isoniazid, or alcohol. At the end of the 24-week trial, participants could continue into a 24-week open-label extension [43]. Adalimumab was given 40 mg subcutaneously every other week after week 24 for 24 weeks. Radiographs were taken at week 48, and safety data was collected. Many (n = 285) enrolled in the open-label extension, including 138 from the adalimumab arm and 147 from the placebo arm. ACR responses were maintained at 48 weeks. Those on continuous adalimumab therapy had ACR20, ACR50, and ACR 0 rates of 48%, 24%, and 20%, respectively at week 48. C. B. Tye and J. C. Cather ACR scores were obtained using an ITT analysis in the open label extension. Skin improvement was also maintained at 48 weeks in the adalimumab group, with response rates of 67%, 58%, 46%, and 33% for PASI50, PASI75, PASI90, and PASI100, respectively. In the group receiving placebo until week 24, improvement was seen at week 48, with response rates of 61%, 54%, 33%, and 31% achieving PASI50, PASI75, PASI90, and PASI100. Improvements in disability measurements were also maintained at 48 weeks. When considering joint progression, patients receiving adalimumab the entire time had a mean mTSS of −0.1 at week 24, and − 0.1 at week 48, indicating sustained control of progression. For those in the placebo group until week 24 that then received adalimumab until week 48, mTSS were 0.9 and 1.0, respectively, indicating potential halting of progression at week 48. Improvements were also seen in joint erosion scores and joint progression scores through 48 weeks. Adalimumab was well tolerated during weeks 24–48, with the most common adverse events being upper respiratory tract infections, nasopharyngitis, and injection site reactions. One serious infection was reported (gastroenteritis) and 10 other SAEs were reported. Elevated serum transaminase levels were also reported in 9 patients. During the first 48 weeks there were no deaths and no reports of tuberculosis or granulomatous infections, demyelination, lymphoma or carcinoma, new antinuclear antibody formation, drug-induced lupus, or congestive heart failure. 2-year data has also been published from the ADEPT trial, providing a better understanding of efficacy and safety in patients with psoriatic arthritis [42]. The open-label extension of the ADEPT trial was continued for 120 weeks, and data were available for 144 weeks after the beginning of the ADEPT trial. ACR20, ACR50, and ACR70 scores at week 104 were 57.3%, 27.8%, and 29.9% respectively. PsARC response rates were 65.9% at week 48 and 63.5% at week 104. All analyses used LOCF. Improvements in HRQoL and disability were also maintained. Inhibition of radiographic progression was also seen throughout the 144 weeks. 13 Adalimumab for Psoriasis From baseline to week 48, 87% of adalimumab treated patients had no progression, and 79% of patients had no radiographic progression at week 144, as evaluated with mTSS. At week 24, the mean change from baseline of mTSS was −0.1 for the adalimumab treated group compared to 0.8 of the placebo group. The adalimumab treated group also had mean changes of mTSS of 0.1 and 0.5 at 48 and 144 weeks respectively. A subanalysis of the ADEPT trial examined risk factors that predict radiographic progression [44]. Elevated baseline C-reactive protein (CRP) (> = 1.0 mg/dl: odds ratio = 3.28) was a strong independent risk factor for joint progression, and treatment with adalimumab reduced the risk of progression five-fold. Individuals taking adalimumab also experienced lowering of CRP levels compared to controls. Over the 2 years, adalimumab was well tolerated, and adverse events were similar to what is expected in rheumatoid arthritis patients. Throughout 2 years of exposure, there were 5 opportunistic infections, 1 patient had peritoneal tuberculosis and 4 patients experienced oral candidiasis. Additionally, there was one case of non-­ Hodgkin’s B-cell lymphoma, 2 basal cell carcinomas, and 1 neuroendocrine carcinoma of the skin. There were no reports of central nervous system demyelinating disease, lupus-like syndrome, congestive heart failure, or adalimumab-­ related allergic reactions. Adalimumab for Special Populations Most clinical trials are conducted in adults, and there is limited data about TNF-inhibitor use in women of childbearing potential during pregnancy, in children, in the elderly, and in rare situations. Use of adalimumab in these special populations is reviewed below. Pregnancy and Lactation Pregnancy can affect the severity of psoriasis, as one-third of women experience improvement, 159 one-third experience worsening of disease, and the remaining third see no change in their psoriasis [45]. Experts recommend using adalimumab with caution during pregnancy [46]. The benefit/risk ratio is acceptable in first and second trimester for moderate to severe plaque patients and must be re-evaluated in the third trimester. Infants exposed to adalimumab in utero should avoid live vaccines for 6 months. Women who become pregnant while on adalimumab, or any biologic, are encouraged to enroll in a pregnancy registry. Additionally, TNFinhibitors are not typically used during lactation [47]. Interestingly, adalimumab has been studied during in vitro fertilization (IVF). Women with lower levels of TNF-a had better success rates with IVF, and there were no increases in birth defects due to adalimumab treatment, although the study was small (100 pregnant women, 136 babies) [48, 49]. Pediatric Psoriasis in children can be challenging to treat, as few treatments have been studied in children. Etanercept has been studied in a pediatric psoriasis population and is reviewed in this textbook [50]. Adalimumab is approved in Europe for pediatric psoriasis in ages 4 and older; however, in the US, it is only approved for four pediatric indications (Crohn’s Disease, Hidradenitis Suppurativa, Uveitis and Juvenile Idiopathic Arthritis) [15, 51]. A phase 3 double-blind, multiperiod trial was performed to evaluate the safety and efficacy of adalimumab compared to weekly methotrexate in children aged 4 up to 18 years of age with severe psoriasis unresponsive to topical therapies. Children received adalimumab based on their weight with significant improvement in PASI75 [52]. A real-life retrospective multicenter analysis of children treated with adalimumab over a 52-week period found that at week 16, 29.6% of patients achieved a PASI90, 55.5% achieved a PASI75. Results were sustained over 24 weeks and did not differ between biologic naïve and biologic exposed patients. No serious infections were observed [53]. 160 Elderly C. B. Tye and J. C. Cather Geriatric psoriasis patients pose additional management challenges. In the elderly, adalimumab can be used as a first line therapy in patients with more extensive disease [54]. It is important that elderly patients are properly monitored, due to the higher general incidence of comorbidities which include cardiovascular disease, metabolic syndrome, nonalcoholic fatty liver disease, infections and malignancies. Lymphoma rates in patients with psoriasis 65 years of age or older appear to be 3x higher than in age matched controls without psoriasis [55]. Additionally, ­nonmelanoma skin cancers are more common in this demographic so skin cancer screening is important. The main endpoint for the study was the number of patients achieving a PGA-F score of ≤1 with a ≥ 2 grade improvement from their baseline [56]. Another secondary endpoint was the amount of patients achieving a mNAPSI 75 [56]. Also, beginning at week 16, patients whose baseline BSA increased ≥25 were taken out of the study to an escape period [56]. At 26 weeks in the trial, patients’ mNAPSI and PGA-F scores were evaluated. The study found that 49% of patients receiving adalimumab achieved a PGA-F score of clear or minimal, as compared to 7% of the placebo group. 47% of adalimumab patients also achieved a mNAPSI75 compared to only 3% of the placebo group [15, 57]. A decrease in nail pain in adalimumab treated patients was also demonstrated in this study [15]. Fingernail Psoriasis Hidradenitis Suppurativa Adalimumab was studied for its effectiveness in treating fingernail psoriasis using a phase 3, multicenter, double-blinded, randomized, placebo-­controlled trial with parallel-arm. To be eligible for the study, patients must have had moderate or worse severity plaque psoriasis, ranked on the PGA scale, as well as moderate or worse severity fingernail psoriasis, which was graded on a 5-point Physician’s Global Assessment of Fingernail Psoriasis (PGA-F) scale. Eligible patients also had to have a Modified Nail Psoriasis Severity Index (mNAPSI), a scale that judges patients’ fingernail psoriasis based on the presence and severity of pitting, onycholysis, oil-drop dyschromia, splinter hemorrhages, and hyperkeratosis, ≥8 with a BSA involvement of ≥10%, or else have a mNAPSI of ≥20 with a BSA involvement of ≥5% [56]. During the trial, 217 patients were randomized to receive either adalimumab (n = 109) or to a placebo group (n = 108). At week 0, patients in the adalimumab group were given a loading dose of 80 mg adalimumab, and then from week 1 on all patients in this group received 40 mg of adalimumab subcutaneously every other week [15, 56]. Hidradenitis Suppurativa is a chronic inflammatory disease characterized by recurrent, painful sinus tracts, abscesses, and nodules [58]. Similar to chronic psoriasis, HS is associated with increased levels of inflammatory cytokines, including TNF alpha [59], a delay in diagnosis, and significant psychological and physiological comorbidities [60]. Individuals with psoriasis have also been shown to be more likely to also be affected with HS [8]. Adalimumab has been studied for HS in the PIONEER 1 and 2 studies which were the first completed Phase 3 trials for HS [15, 61]. Briefly, the induction dosing for patients with HS is the same as inflammatory bowel disease patients and the maintenance dosing, which starts on day 29, is a weekly 40 mg subcutaneous injection. Adalimumab is the only FDA approved therapy for HS. The safety profile of adalimumab in the PIONEER trials were consistent with other trials [15, 61]. Generalized Pustular Psoriasis In Japan, an open-label 52-week study examined the safety and efficacy of adalimumab in 10 13 Adalimumab for Psoriasis patients with generalized pustular psoriasis. Induction was similar to current psoriasis dosing; however, if needed the patients could increase to 80 mg every other week. Seven of the patients (including 3 who dose escalated to 80 mg) achieved clinical response at week 16 and 5 achieved clinical response at week 52 [62]. Inflammatory Bowel Disease (IBD) Psoriasis and inflammatory bowel disease, especially Chron’s disease share several epidemiologic factors, genetic similarities, and inflammatory cytokine profiles [63–65]. A ­retrospective US claims based study involving 5492 pairs of patients with moderate to severe psoriasis and their age, gender, and geographic based matched controls found that psoriasis patients had approximately 3.5 fold increase prevalence of IBD [66]. Additionally, a prospective study of women in the Nurses’ Health Study 1 and 2 (n = 174,476) evaluated the incidence of psoriasis and IBD. There were 2752 women in these two studies and in a pooled analysis the women were found to have a four-fold increase in Crohn’s disease but no increase incidence of ulcerative colitis [67]. Patients with severe psoriasis and psoriatic arthritis have the highest risk for Crohn’s disease and ulcerative colitis [68]. There are no known trials dedicated to the treatment of psoriasis patients who have concomitant IBD. However, both adalimumab and infliximab have demonstrated efficacy in psoriasis, psoriatic arthritis, Crohn’s disease and ulcerative colitis [69]. Hemodialysis A case report involving a patient with psoriasis being treated with infliximab required transition to adalimumab due to pulmonary edema thought secondary to intravenous fluids at time of infliximab infusion [70]. 161 Biologic Comparator Trials Briefly, we will discuss some of the important plaque psoriasis trials in which a newer biologic has been compared to adalimumab. More complete details of these trials can be found in the chapters that focus on the comparator drug. 1. Guselkumab Voyage 1 and 2 Trials compared the safety and efficacy of guselkumab (an anti-IL23 monoclonal antibody) to adalimumab. Additionally, data on withdrawal, retreatment, adalimumab non-responders treated with guselkumab, and maintenance data out to 1 year was obtained. Guselkumab demonstrated superior efficacy compared to adalimumab [71, 72]. A secondary analysis of the trial results has been published for the difficult to treat body regions, such as scalp, hands, feet, and nails. Guselkumab was not superior to adalimumab when looking at efficacy in nails; however, it was superior for other difficult to treat areas examined [73]. 2. Ixekizumab Two trials evaluated the efficacy of ixekizumab versus adalimumab for the treatment of psoriatic arthritis: SPIRIT-P1 and SPIRIT-­ H2H. Full details are described in the Ixekizumab chapter in this text. In the SPIRIT-P1 trial, adult biologic naïve patients with psoriatic arthritis achieved comparable ACR results at week 24 with ixekizumab or adalimumab. There was no added value when methotrexate was used in combination with ixekizumab; however, adalimumab patients did better on combination therapy [74]. SPIRIT-H2H directly compared ixekizumab and adalimumab with endpoint measures of ACR50 and PASI100 at week 24. Again, ixekizumab scores for ACR50 and PASI100 were not improved with concurrent methotrexate use. However, adalimumab again demonstrated improved efficacy in C. B. Tye and J. C. Cather 162 these measures when combined with methotrexate [75]. 3. Risankizumab IMMvent, a phase 3, randomized, double-­ blind, active-comparator-controlled trial investigated adalimumab vs risankizumab for the treatement of moderate-severe plaque psoriasis [76]. Risankizumab showed significantly greater efficacy in providing skin clearance compared to adalimumab. Practical Considerations for TNF-Inhibitors There are a number of safety and practical considerations for all TNF-inhibitors, including adalimumab [10]. TNF-inhibitors are contraindicated in patients with active, serious infections. Testing for tuberculosis should be performed on all patients who will be treated with ­ TNF-­ inhibitors. The Centers for Disease Control (CDC) has released guidelines regarding testing for tuberculosis [77]. Additionally, live vaccines should not be used in patients receiving TNF-­inhibitors. Demyelinating events, new onset or exacerbation of symptoms, may also occur under TNF-blockade. Anti-TNF therapy should not be used in patients with a history of demyelinating disease (e.g. multiple sclerosis, optic neuritis, and peripheral demyelinating disease such as Guillain-Barre syndrome). Caution should be used in patients with congestive heart failure. Patients should also be screened for hepatitis B prior to receiving therapy, as TNF-blockers may reactivate hepatitis B in patients who are carriers of the disease. Many individuals experience injection site reactions, although most are minor and the citrate free formula has improved patients’ injection experience. Malignancies, hepatotoxicity, and new onset psoriasis with anti-TNF therapy are also considerations. Prior to initiating systemic therapy, including adalimumab, a physical examination should be performed and a full medical history taken. It is important to ask about age appropriate cancer screening (pap smears, mammograms and colonoscopies), cancer history, infection history, vac- cination history, as well as personal or family history of inflammatory bowel disease history and arthritis. A total body skin examination should also be performed. Clinicians should also inquire about social and lifestyle history, and family planning if relevant. A number of labs can also be requested, including a complete blood count (CBC) and comprehensive metabolic panel (CMP), Hepatitis screen (B and C), a human immunodeficiency virus (HIV) screen if warranted, and a tuberculosis test upon initiation and yearly. Once systemic therapy is initiated, monitoring patients on therapy is vital. Patients should have periodic total body skin examinations as well as a yearly evaluation for tuberculosis [10]. Safety Update Leonardi et al. analyzed the long-term safety of adalimumab from 18 clinical trials involving 3727 adults with plaque psoriasis. The cumulative exposure was 5429.7 patient years. The adverse events remained stable with no new safety signals [78]. In a recent review of safety registries involving adults with psoriasis, Strober et al. found that across multiple registries the adalimumab safety data was consistent with the long-term safety data reported from the clinical trial experience [79]. An analysis explored adalimumab safety in 23,457 participants across 71 global trials, rheumatoid arthritis (36 trials, n = 14,109), juvenile idiopathic arthritis (3 trials, n = 212) ankylosing spondylitis (4 trials, n = 1684), psoriatic arthritis (4 trials, n = 837), psoriasis (13 trials, n = 3010), and Crohn’s disease (11 trials, n = 3606) [80]. This analysis represented 12 years of adalimumab exposure, with 52.5% and 48.7% of patients receiving concomitant immunosuppressant agents or concomitant systemic steroids, respectively. The majority of patients (60%) were rheumatoid arthritis patients, and approximately two-thirds received concomitant therapy. The most frequently reported serious adverse events were infections, and these were most often seen 13 Adalimumab for Psoriasis in the rheumatoid arthritis and Crohn’s disease trials. Malignancy rates were as expected in adalimumab-treated patients compared with the general population. There was an increase in lymphoma incidence in patients with rheumatoid arthritis, but this increase was in the expected range of rheumatoid arthritis patients not on anti-­ TNF-­therapy. Non-melanoma skin cancer rates were elevated in rheumatoid arthritis patients, psoriasis patients, and Crohn’s disease patients. While there may be some distinct differences in patient populations, no new safety signals were revealed. lack Box Warnings: Infection B and Malignancy A series of warnings are detailed on the prescribing information of adalimumab, including black box warnings for serious infections and malignancies [81]. Patients taking adalimumab are at increased risk of serious infection, and many who developed serious infections while taking adalimumab were also taking concomitant immunosuppressants (e.g. methotrexate or corticosteroids). Tuberculosis is a concern, including active tuberculosis and reactivation of latent tuberculosis. All patients should be screened for latent tuberculosis prior to initiating therapy. If latent tuberculosis is present, anti-TB therapy should be initiated prior to beginning adalimumab or any TNF-inhibitor. Because anti­TB therapy can increase liver function tests, and in rare cases adalimumab can too, we wait a month after beginning anti-TB therapy before starting adalimumab. Compliance with tuberculosis prophylaxis is key. Consultation with an infectious disease clinician for managing patients with latent tuberculosis is recommended. Invasive fungal infections (e.g. histoplasmosis, coccidioidomycosis, candidiasis, aspergillosis, blastomycosis, and pneumocystosis) and opportunistic bacterial (e.g. legionella, listeria) or viral infections have been reported. Patients presenting with a serious infection should discontinue adalimumab. 163 In the controlled portions of adalimumab trials across all indications, adalimumab treated patients had a rate of serious infections of 4.3 per 100 patient years (n = 7973) compared to a rate of 2.9 per 100 patient years in placebo-treated patients (n = 4848) [15]. These same trials showed a reported active TB rate of 0.2 per 100 patient years. The rate of positive purified protein derivative conversion in this same study was 0.09 per 100 patient years [15]. Through 16 weeks of the REVEAL RCT trial, a rate of serious infections of 2.8% was observed in adalimumab treated patients (n = 814) vs. a rate of 3.3% in the placebo group (n = 398). From weeks 33–52, the rates of infection among adalimumab treated patients and the placebo group were 1.1% (n = 250) and 2.4% (n = 240) respectively [13]. Tuberculosis rates for this study were 0.0% for both groups at the 16-week mark, and 0.0% and 1.2% for the adalimumab and placebo treated groups, respectively, from 33–52 weeks [13]. In 30 psoriasis clinical trials, there was a rate of serious infection of 1.37% across 1403 adalimumab patients, as well as a rate of active tuberculosis of 0.18% [82]. The ESPIRIT rate of serious infection per 100 patient years was 1.0 (n = 6045) and the rate of active tuberculosis was <0.1 [83]. Malignancies have been reported in patients taking TNF-inhibitors, including hepatosplenic T-cell lymphoma (HSTCL), a rare and usually fatal T-cell lymphoma almost exclusively reported in adalimumab-treated males with Crohn’s disease taking concomitant azathioprine or 6-mercaptopurine. In the controlled phase of adalimumab trials across all indications, an equal number of malignancies, including lymphomas, were seen in the adalimumab group compared to the control group. These trials showed 0.7 malignancies per 100 patient years among 7973 adalimumab treated patients and 4848 placebo treated patients. Lymphoma rates in these trials were 0.11 per 100 patient years [15]. In the REVEAL RCT, both adalimumab and placebo treated patients showed similar incidences of malignancies, with an incidence of 0.8 through 16 weeks and 0.0 from weeks 33–52. No lymphoma was seen in this RCT [13]. The ESPIRIT 8-year post C. B. Tye and J. C. Cather 164 marketing malignancy incidence was 1.1 in adalimumab treated patients (n = 6405) and the lymphoma rate was <0.1 [83]. A publication evaluating 13 adalimumab clinical trials for treatment of moderate to severe plaque psoriasis with treatment for up to 5 years did not show cumulative toxicity and adverse event rates were stable or decreased over time [82]. There was an increased rate of non-melanoma skin cancer in adalimumab trials and it was more prevalent in those patients with previous immunosuppressant therapy and psoriasis patients previously treated with PUVA. When considering lymphoma, more cases of lymphoma were observed in the TNF-blocker patients compared to controls. In adalimumab trials, 3 lymphomas occurred in 6693 adalimumab treated patients compared to 1 in 3749 patients in the control group, across 32 global trials. In 45 trials, the rate of lymphoma was 0.11 per 100 patient years, approximately three-fold higher than that of the general population. Patients with chronic inflammatory disease states such as rheumatoid arthritis, psoriasis, psoriatic arthritis, hidradenitis and inflammatory bowel disease may be at a higher risk for lymphomas than the general population, even in the absence of therapies. In fact, several studies have suggested that there is no increased risk for lymphoma when using TNF inhibitors to treat these disease states [84–87]. More data is needed at this time as post-marketing cases of acute and chronic leukemia have also been observed. Most patients who developed malignancies were also receiving concomitant immunosuppressants. It has been challenging to determine the relationship between infection and malignancy and TNF-inhibitors. A meta-analysis examined 20 randomized, placebo-controlled psoriasis trials of TNF-inhibitors, etanercept, infliximab, adalimumab, golimumab and certolizumab, with 6810 patients total [88]. There was a small increased risk of infection (odds ratio 1.18) but no increased risk of serious infection (odds ratio 0.7). There was an increased risk of malignancy in the TNF-­ inhibitor group compared to the control group (odds ratio 1.48 for all malignancies and 1.26 when nonmelanoma skin cancer was excluded). Post-Marketing Safety Information A number of additional safety signals have been observed in post-marketing surveillance of adalimumab [81]. Hypersensitivity reactions may occur, and anaphylaxis or angioneurotic edema may be rare events. Allergic reactions were seen in ~1% of patients taking adalimumab. There have also been rare reports of hematologic reactions, including cytopenia, with TNF-therapy. Adalimumab treatment may result in autoantibody formation, and in rare cases, the development of a lupus-like syndrome [89]. Demyelinating disease [90], reactivation of hepatitis B infections, and worsening of congestive heart failure have been reported, underscoring the importance of appropriate screening. Elevations in liver enzymes were also reported. TNF-inhibitors are acceptable for patients with Hepatitis B and C as long as appropriate monitoring and/or antiviral therapy is in place with hepatology and/or infectious disease consultation [91, 92]. A number of adverse events have also been reported in post-marketing surveillance [81]. These include gastrointestinal disorders (diverticulitis, large bowel perforations including perforations associated with diverticulitis and appendiceal perforations associated with appendicitis, pancreatitis, ulcerative colitis [93], liver failure, sarcoidosis, nervous system disorders (demyelinating disorders and cerebrovascular accident), respiratory disorders (interstitial lung disease, including pulmonary fibrosis, pulmonary embolism), skin reactions (Stevens Johnson Syndrome, cutaneous vasculitis, erythema multiforme, new or worsening psoriasis (all sub-types including pustular and palmoplantar) [94, 95], alopecia [96], and vascular disorders (systemic vasculitis and deep vein thrombosis). Adalimumab Pearls 1. Dose adjustments may be required. There are 10 different indications for adalimumab and multiple different dosing regimens. Ryan et al. evaluated in patients with psoriasis or hidradenitis the safety of weekly 13 Adalimumab for Psoriasis versus every other week dosing and found similar adverse event rates [97]. Additionally, optimizing therapy with temporary dose escalation followed by de-escalation permitted the achievement of long-term disease control in patients treated out to 252 weeks [98]. Dose escalation was not associated with additional safety concerns. Individualization of therapy with adalimumab has been attempted by evaluating serum trough levels in 51 patients over time. Excellent responses were seen with serum trough levels at 6.46 micrograms per mL [99]. 2. Adalimumab is the market leader for the treatment of psoriasis with concomitant psoriatic arthritis but newer agents are also effective. Historically, TNF-inhibitors have been the treatment of choice for individuals with both psoriasis and psoriatic arthritis [4]. An indirect comparison using individual patient data from randomized clinical trials involving adalimumab (REVEAL and CHAMPION) as well as trials comparing etanercept to placebo found adalimumab significantly outperformed etanercept in PASI, DLQI, Patient global assessment, symptom and lesion resolution, and adverse event [100]. TNF-inhibitors show synergy with methotrexate, and patients with higher BMIs tend to do better on the monoclonal TNF inhibitors, including adalimumab [101, 102]. Rotation within the TNF-inhibitor class is possible, but there may be diminishing returns after trying two different TNF- inhibitors. A number of small studies have examined rotation within the TNF-inhibitor class. In one study, 30 patients who failed etanercept were transitioned to adalimumab [103]. Efficacy was examined at weeks 12, 24, and 48, and 27%, 36%, and 54% achieved PASI75. Adalimumab was well tolerated, and there was no increase in adverse events for patients receiving adalimumab who had previously received etanercept. Another study examined 14 patients with inadequate responses to etanercept who were transitioned to adalimumab [104]. At 16 weeks, 9/14 (64%) achieved 165 PASI 50, (of these 4 achieved PASI 75 and 1 achieved PASI 90). No serious adverse events were reported. A sub-analysis of BELIEVE was also conducted to examine efficacy of adalimumab in patients treated previously with anti-TNF agents [105]. BELIEVE was a double-blind, randomized, controlled trial where patients received 80 mg at week 0 and 40 mg every other week of adalimumab with topical vehicle or topical calcipotriol/betamethasone dipropionate once daily for 4 weeks and then as needed [106]. The trial enrolled 703 patients, with 38.6% having prior anti-TNF therapy compared to 61.4% who were naïve to anti-TNF therapy. Nearly two-thirds (61.7%) of patients with prior anti-TNF therapy achieved PASI 75 at week 16 compared to 71.17% of anti-TNF naïve patients. Adalimumab was well tolerated, and adverse events were similar between those with previous anti-TNF therapy and those without. Another open-label phase IIIb trial, PRIDE (an open-label access program to evaluate the safety and effectiveness of adalimumab when added to inadequate therapy for the treatment of psoriasis), was conducted in Canada, and examined adalimumab response in patients who had not responded previously to other psoriasis therapies [107]. The trial enrolled 203 patients in the 24-week trial. Patients received 80 mg loading dose at week 0 and 40 mg every other week beginning at week 1, for 23 weeks. At week 16, 70.9% achieved PASI 75. Adalimumab was generally well tolerated with 9 patients reporting serious adverse events. As more experience and data is accumulated with the IL-17 inhibitors, there will be erosion of the TNF-inhibitor market share since they also address both psoriasis and psoriatic arthritis. Biomarkers are being evaluated as predictors for response to both TNF-inhibitors and IL-17 antagonists [108, 109]. 3. Adalimumab is my treatment of choice for psoriasis patients or psoriatic arthritis patients with hidradenitis suppurativa 166 Case series report success using adalimumab for patients with psoriasis and concomitant HS [110]. Remember, HS requires a much higher dose of adalimumab so even if HS is mild and psoriasis is the major reason for therapy, consider the dosing regimen for HS to ensure adequate disease control for both diseases. 4. Adalimumab is my treatment of choice for psoriasis and or psoriatic arthritis patients with concomitant inflammatory bowel disease As described above, TNF-inhibitors are the treatment of choice for patients with psoriasis and concomitant psoriatic arthritis and inflammatory bowel disease. 5. Pediatric data is available for plaque psoriasis and adalimumab is approved in the EU; however, it is not approved for psoriasis in the US at this time. 6. Clinic experience with biologics are different than clinical trial experience. As with all psoriasis therapies, it is important to understand real-world patient experiences. A real-world drug survival meta-analysis has been published (cut off for study inclusion was Oct 2017). Thirty-seven studies with 32,631 subjects were included and drug survival for all biologics decreased over time dropping from 66% at year 1 to 41% at year 4 for etanercept, from 69% to 47% for adalimumab, from 61% to 42% for infliximab and from 82% to 56% for ustekinumab [111]. A recent study examined patient reported reasons for discontinuing treatment [112]. Patients (n = 1095) with moderate to severe psoriasis who received systemic treatment were interviewed. Of these, 200 patients received adalimumab in the past and were asked why they discontinued it. The top three reasons for discontinuing adalimumab were because it did not work well enough (34%), worked well at first but stopped working well (22%), or non-lifethreatening side effects (14.5%). C. B. Tye and J. C. Cather Complications of Not Treating Psoriasis The majority of moderate to severe psoriasis patients are not being treated as suggested by guidelines. A National Psoriasis Foundation survey showed that nearly 40% of patients surveyed were not in treatment for their psoriasis [113]. Of those who were in treatment, 57% of patients with severe psoriasis were on topical therapy alone, and only 3% of these patients had ever tried phototherapy, a systemic therapy, or a biologic. In a chart review of dermatologists treating 10 or more psoriasis patients per month, 40% of patients with severe disease received topical therapy alone [114]. Additionally, individuals with psoriatic arthritis are also undertreated, with 59% of patients diagnosed receiving no medication, biologic or topical, for their psoriatic arthritis [115]. There are complications of not treating psoriasis. Extensive psoriasis leaves an unresolved inflammatory burden in skin. In addition, there is an elevated systemic inflammatory burden that impacts comorbidities. New data suggests that treatment with TNF-inhibitors can reduce the risk of myocardial infarction [116]. Gkalpakiotis et al. have reported the results of their pilot study looking at the beneficial effects adalimumab has on biomarkers connected to cardiovascular disease [117]. A decrease in both E-Selectin and IL-22 were seen after 3 months on adalimumab— both are linked with cardiovascular disease and more studies are needed to elucidate the importance of this affect. In addition to physical complications, there are also psychosocial implications. Untreated or inadequately controlled psoriasis can impact quality of life and physical functioning. Finally, untreated psoriasis may have an economic impact through time lost from work and reduced productivity while at work. There are a number of appropriate treatments for individuals with moderate to severe psoriasis. Adalimumab is one of these treatments and is a viable option for some. 13 Adalimumab for Psoriasis Conclusion Adalimumab, a member of the anti-TNF class of biologics, is an appropriate treatment for individuals with psoriasis, with or without psoriatic arthritis. Patients with psoriasis who have hidradenitis suppurativa or inflammatory bowel disease can address all diseases with one medication. Numerous clinical trials (e.g. REVEAL, CHAMPION, ADEPT, SPIRIT-P1 and others) have shown adalimumab to be efficacious and well tolerated with a safety profile similar to others in the class. Adalimumab treatment also improved HRQoL. As with all immunosuppressive therapies, appropriate screening and monitoring of patients on adalimumab is required. Adalimumab is an important option within the psoriasis treatment armamentarium. References 1. Nestle FO, Kaplan DH, Barker J. Psoriasis. N Engl J Med. 2009;361(5):496–509. 2. Stern RS, Nijsten T, Feldman SR, Margolis DJ, Rolstad T. Psoriasis is common, carries a substantial burden even when not extensive, and is associated with widespread treatment dissatisfaction. J Investig Dermatol Symp Proc. 2004;9(2):136–9. 3. Capon F, Burden AD, Trembath RC, Barker JN. Psoriasis and other complex trait dermatoses: from loci to functional pathways. J Invest Dermatol. 2012;132(3 Pt 2):915–22. 4. Gottlieb A, Korman NJ, Gordon KB, Feldman SR, Lebwohl M, Koo JY, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis: section 2. Psoriatic arthritis: overview and guidelines of care for treatment with an emphasis on the biologics. J Am Acad Dermatol. 2008;58(5):851–64. 5. Davidovici BB, Sattar N, Prinz J, Puig L, Emery P, Barker JN, et al. Psoriasis and systemic inflammatory diseases: potential mechanistic links between skin disease and co-morbid conditions. J Invest Dermatol. 2010;130(7):1785–96. 6. Gelfand JM, Neimann AL, Shin DB, Wang X, Margolis DJ, Troxel AB. Risk of myocardial infarction in patients with psoriasis. JAMA. 2006;296(14):1735–41. 7. Neimann AL, Shin DB, Wang X, Margolis DJ, Troxel AB, Gelfand JM. Prevalence of cardiovascular risk factors in patients with psoriasis. J Am Acad Dermatol. 2006;55(5):829–35. 167 8. Kridin K, Shani M, Schonmann Y, Fisher S, Shalom G, Comaneshter D, et al. Psoriasis and Hidradenitis Suppurativa: A Large-scale Population-based Study. J Am Acad Dermatol. 2018. 9. Najarian DJ, Gottlieb AB. Connections between psoriasis and Crohn’s disease. J Am Acad Dermatol. 2003;48(6):805–21. quiz 22-4 10. Menter A, Gottlieb A, Feldman SR, Van Voorhees AS, Leonardi CL, Gordon KB, 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(5):826–50. 11. Pariser DM, Bagel J, Gelfand JM, Korman NJ, Ritchlin CT, Strober BE, et al. National Psoriasis Foundation clinical consensus on disease severity. Arch Dermatol. 2007;143(2):239–42. 12. Johnson-Huang LM, Lowes MA, Krueger JG. Putting together the psoriasis puzzle: an update on developing targeted therapies. Dis Model Mech. 2012;5(4):423–33. 13. Menter A, Tyring SK, Gordon K, Kimball AB, Leonardi CL, Langley RG, et al. Adalimumab therapy for moderate to severe psoriasis: a randomized, controlled phase III trial. J Am Acad Dermatol. 2008;58(1):106–15. 14. Miller I, Lynggaard CD, Lophaven S, Zachariae C, Dufour DN, Jemec GB. A double-blind placebo-­ controlled randomized trial of adalimumab in the treatment of hidradenitis suppurativa. Br J Dermatol. 2011;165(2):391–8. 15. AbbVie Inc. (North Chicago I. HUMIRA Injection Package Insert. 16. Coates LC, Kavanaugh A, Mease PJ, Soriano ER, Laura Acosta-Felquer M, Armstrong AW, et al. Group for Research and Assessment of psoriasis and psoriatic arthritis 2015 treatment recommendations for psoriatic arthritis. Arthritis Rheumatol. 2016;68(5):1060–71. 17. Elmets CA, Leonardi CL, Davis DMR, Gelfand JM, Lichten J, Mehta NN, et al. Joint AADNPF guidelines of care for the management and treatment of psoriasis with awareness and attention to comorbidities. J Am Acad Dermatol. 2019;80(4):1073–113. 18. Menter A, Strober BE, Kaplan DH, Kivelevitch D, Prater EF, Stoff B, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with biologics. J Am Acad Dermatol. 2019;80(4):1029–72. 19. Nast A, Kopp I, Augustin M, Banditt KB, Boehncke WH, Follmann M, et al. German evidence-­based guidelines for the treatment of psoriasis vulgaris (short version). Arch Dermatol Res. 2007;299(3):111–38. 20. Nast A, Kopp IB, Augustin M, Banditt KB, Boehncke WH, Follmann M, et al. Evidence-based (S3) guide- Infliximab, Golimumab, and Certolizumab Pegol 14 Jacob A. Mojeski and Robert E. Kalb Abstract Psoriasis and psoriatic arthritis are common chronic inflammatory disorders that cause substantial physical, financial, and psychosocial burdens for patients. Biologic therapy for these diseases has led to a major improvement in the therapeutic armamentarium, and subsequently, patient quality of life. The tumor necrosis factor alpha (TNFα) blocking class is one family of biologic therapy that has provided significant relief for patients. Infliximab was the first TNFα inhibitor approved for use in the United States in 1998 while golimumab and certolizumab pegol are more recent additions. With over 20 years of clinical experience using TNFα inhibitors, physicians may employ these agents in a manner that maximizes efficacy while decreasing risks of potential side effects. This chapter will provide a comprehensive review of the published data on the safety and efficacy of infliximab, golimumab, and certolizumab. J. A. Mojeski · R. E. Kalb (*) Department of Dermatology, School of Medicine and Biomedical Sciences, State University at of New York at Buffalo, Buffalo, NY, USA e-mail: jacobmoj@buffalo.edu; kalb@buffalo.edu © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_14 Key Learning Objectives 1. To understand the mechanism of action of Infliximab, Golimumab, and Certolizumab Pegol 2. To understand the appropriate use and efficacy of the Infliximab, Golimumab, and Certolizumab Pegol 3. To understand the safety issues of Infliximab, Golimumab, and Certolizumab Pegol Introduction The treatment of psoriasis and psoriatic arthritis has undergone a revolution with the advent of biologic therapy providing patients with more treatment choices and greater hope for sustained symptomatic relief [1, 2]. Infliximab, golimumab and certolizumab are all part of the Anti-TNF class of drugs. Although these drugs have similar profiles, each have specific therapeutic qualities that can be taken advantage of in the clinical setting. Infliximab and golimumab are known for their rapid-onset of clinical efficacy while certolizumab has the added benefit of use during pregnancy as minimal to no drug crosses the placenta or into breast milk. 173 174 J. A. Mojeski and R. E. Kalb Infliximab (Remicade) is one of the more nent of the typical IgG antibody structure, cerwidely used tumor necrosis factor (TNF)-alpha tolizumab contains only a monovalent Fab inhibitors, and has extensive clinical experience fragment of humanized anti-TNFa antibody [15]. in various immunologic conditions. It is a chime- Additionally, situated on the hinge region of the ric human-murine monoclonal antibody, consist- drug are two polyethylene glycol chains, which ing of human constant and murine variable confer its pegylated component (PEG) [16, 17]. regions and binds to both soluble and membra- Similar to other anti-TNF agents, Certolizumab nous forms of TNF-alpha, thereby neutralizing has been FDA approved for multiple indications the cytokine’s effect [3–6]. It was first approved in the treatment of various inflammatory disby the U.S. Food and Drug Administration (FDA) eases. The current list of approved conditions for the treatment of moderate to severe Crohn’s includes moderate-severe rheumatoid arthritis, disease in August of 1998. Its use was later axial spondyloarthritis, psoriatic arthritis, psoriaexpanded to include the following indications: sis, and Crohn’s disease [18]. Dosing is via sub-­ rheumatoid arthritis (11/1999), ankylosing spon- cutaneous injection and is typically standard dylitis (12/2004), psoriatic arthritis (05/2005), throughout its indications. For psoriasis, it is ulcerative colitis (09/2005), pediatric Crohn’s administered 400 mg Q2W and for psoriatic disease (05/2006), psoriasis (09/2006), and pedi- arthritis dosing varies between 200 mg Q2W or atric ulcerative colitis (09/2011) [7, 8]. For the 400 mg Q4W. treatment of psoriasis and psoriatic arthritis the drug is usually administered intravenously over 2 h, at a dosage of 5 mg/kg on weeks 0, 2, and 6, Mechanism of Action followed by maintenance infusions every 8 weeks thereafter [3, 9]. While the intravenous adminis- The specific mechanisms of immune-mediated tration can be inconvenient, it is administered inflammatory diseases have yet to be fully eluciinfrequently and has the benefit of rapidly achiev- dated, however, the over-expression of tumor ing high serum concentrations, therefore offering necrosis factor (TNF) is believed to play a pivotal the potential for rapid and sustained improve- role. This is documented by extensive investigament [4, 6, 10, 11]. tion and confirmed by the efficacy of anti-TNF Golimumab (Simponi, Simponi Aria) is biologics in treating these disorders [14, 19, 20]. another member of the TNF antagonist family Numerous in vitro and in vivo studies have conand available in subcutaneous injection (Simponi) tributed to the knowledge base behind TNF-alpha or IV formulation (Simponi Aria). It is a fully inhibitors, and it has been proposed that apoptohuman IgG1k monoclonal antibody that targets sis, reduction of pro-inflammatory cytokines, both transmembrane and soluble forms of TNF-­ down-regulation of adhesion molecules, and alpha with high affinity and specificity [12, 13]. increases in circulating T regulatory cells are the As of 2017, both formulations of golimumab main mechanisms of action behind the efficacy of have been FDA approved for use in numerous TNF-alpha inhibition [3, 13]. Infliximab, goliminflammatory diseases, including moderate-­ umab and certolizumab exert their anti-­ severe rheumatoid arthritis, ankylosing inflammatory effects by binding to the soluble ­spondylitis, and psoriatic arthritis either alone or and transmembrane forms of TNF-alpha. This in combination with conventional agents. leads to inhibition of the induction of the inflamSubcutaneous golimumab has an additional indi- matory cascade and ultimately results in the rapid cation for the treatment of ulcerative colitis [13, reduction in the number of cells at the site of 14]. Golimumab is self-administered as a 50 mg inflammation. Infliximab and golimumab have or 100 mg subcutaneous injection once each the additional function of being able to induce month or intravenously at 2 mg/kg for at weeks 0 complement-dependent cytotoxicity and and 4 then every 8 weeks thereafter. antibody-­ dependent cell-mediated cytotoxicity Certolizumab Pegol (Cimzia) is a biologically through their Fc portions [20, 21]. Current develunique anti-TNF agent. Lacking the Fc compo- opments in the biology of the TNF receptor path- 14 Infliximab, Golimumab, and Certolizumab Pegol way have suggested pleotropic effects. There are two antagonizing TNF receptor pathways including the TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2) pathways. The TNFR1 pathway signals most of TNF’s effects including inflammation and cell death and is expressed ubiquitously whereas the TNFR2 pathway is only expressed through immune cells, endothelial cells, and multiple CNS cells and provides homeostatic function [22, 23]. Recently, alternative strategies in blocking TNF receptors are being developed that target the TNF receptor-1 (TNFR1) pathway and augment the TNF receptor-­2 (TNFR2) pathway [22]. Importantly, the Fc region of an antibody, which infliximab and golimumab contain, prolongs half-life and prevents premature degradation of biologic drugs. Certolizumab does not contain this Fc portion. The covalent linking of its PEG moiety to the hinge region of the antibody increases the half-life of the drug and decreases protease sensitivity [24]. Certolizumab’s safety profile during pregnancy and breastfeeding is thought to be due to its distinctive lack of an Fc component. Because there is no Fc region, the drug is unable to bind the 175 neonatal FcRn-mediated transfer across the placenta. Subsequently, minimal to no drug is delivered to the fetus of mothers being treated with a certolizumab regimen [24–28]. This is a significant advantage if anti-TNF therapy is required in a woman of child bearing potential. fficacy for Psoriasis and Psoriatic E Arthritis Infliximab The first documented treatment response with infliximab was published in a case report of a patient with a long-standing history of Crohn’s disease with concomitant severe psoriasis [29]. The patient received a single infusion of infliximab (5 mg/kg) and 4 weeks later there was a dramatic improvement in the severity of the psoriasis. Subsequently, a double-blind, randomized trial was conducted in 2001 by Dr. Alice Gottlieb’s research group (Fig. 14.1) [30]. The trial enrolled 33 patients with clinically-defined moderate-severe plaque psoriasis who were ran- Infliximab Therapy Week 0 Week 10 PASI 42 PASI 1.8 Fig. 14.1 Before and after 10 weeks of intravenous infliximab therapy. A significant reduction in plaques is demonstrated providing promising data of the benefits of infliximab in decreasing disease burden J. A. Mojeski and R. E. Kalb 176 domized to receive either intravenous placebo or 5 mg/kg, or placebo administered at weeks 0, 2, intravenous infliximab in either 5 mg/kg or and 6 [31]. At week 10, 72% and 88% of the 10 mg/kg dosages at weeks 0, 2, and 6. Three patients treated with 3 mg/kg and 5 mg/kg of infpatients withdrew from the study, one from each liximab, respectively, achieved a 75% or greater treatment group. At week 10, 9 of the 11 (82%) improvement using the PASI evaluating system patients in the infliximab 5 mg/kg group and 10 in comparison to only 6% of patients in the plaof the 11 (91%) patients in the infliximab 10 mg/ cebo group (p < 0.001). Furthermore, 46% of kg group were considered responders (clear or patients treated with 3 mg/kg and 58% treated almost clear rating according to the PGA) com- with 5 mg/kg reached a 90% improvement in pared to only 2 of 11 (18%) receiving placebo PASI score compared to only 2% of the patients (p < 0.01). Additionally, nine of 11 (82%) in the in the placebo group (p < 0.001). Quality of life infliximab 5 mg/kg group and 8 of 11 (73%) in scores were also improved with the use of inflixthe infliximab 10 mg/kg group achieved at least a imab. This study was notable for rapid clinical PASI 75 compared with only 2 of 11 (18%) in the improvement after only 2 weeks in each treatplacebo group (p < 0.05). In both infliximab-­ ment group [31]. treated groups, the median time to response was In 2005 and 2007, data from two large phase only 4 weeks. This trial provided promising data III multicenter, randomized, double-blind for the high degree of clinical benefit and rapid placebo-­controlled studies emerged -- EXPRESS time to response in the treatment of moderate-­ I and EXPRESS II (Fig. 14.2) [32, 33]. These severe plaque psoriasis with infliximab. studies further demonstrated the dramatic effecNumerous randomized controlled trials followed tiveness of infliximab in patients suffering from to confirm this initial success. moderate-severe plaque psoriasis. In EXPRESS In the multicenter, double-blind phase II I, patients were allocated to receive infusions of SPIRIT trial by Gottlieb et al. in 2004, patients either 5 mg/kg of infliximab or placebo at weeks with severe plaque psoriasis were randomly 0, 2, and 6, and then every 8 weeks thereafter to assigned to receive infusions of either 3 mg/kg, week 46 [33]. Beginning at week 24, the patients Infliximab phase III study efficacy EXPRESS II EXPRESS 82 % of Patients 80 57 78 58 78 61 56 45 55 43 34 n=301 n=276 n=281 PASI 75 n=150 n=141 n=134 PASI 90 Fig. 14.2 PASI response to IV infliximab therapy at weeks 10, 24, and 50. At week 10, approximately 80% of patients in the EXPRESS and EXPRESS II trials achieved PASI 75. This response was largely maintained through week 24 with 82% of patients in the EXPRESS trial achieving PASI 75 and 78% achieving PASI 75 in the EXPRESS II trial 14 Infliximab, Golimumab, and Certolizumab Pegol in the placebo group crossed over to receive infliximab treatments. Although remarkable clinical improvements were noted at week 6 (after only two infusions), by the end of the induction period (week 10) 80% of patients treated with infliximab achieved PASI 75, with 57% reaching at least a 90% improvement (PASI 90), and 26% of patients achieving complete clearing of the skin (a score of 0 in PASI). This is in comparison to 3%, 1%, and 0% in the placebo group, respectively (p < 0.0001). These responses were maintained through week 50, with 82% and 61% of patients attaining a PASI 75 at weeks 24 and 50 (Figs. 14.3 and 14.4) [32]. Additionally, infliximab therapy was noted to have a significant effect on nail manifestations of psoriasis, a traditionally treatment-resistant disease. Nail ­ Psoriasis Severity Index (NAPSI) improvements were noted as early as week 10 in infliximabtreated patients. At week 24, there was a 56% mean decrease of the NAPSI in the infliximab 177 group, and this response was also maintained through week 50 (p < 0.0001 for week 24) (Fig. 14.5) [32]. The EXPRESS II trial also documented the efficacy of infliximab in a placebo-controlled protocol (Fig. 14.2) [33]. This trial compared the efficacy of continuous therapy (every 8 weeks) versus intermittent (as needed) maintenance regimens. Patients were randomized to induction therapy at weeks 0, 2, and 6 with infliximab 3 mg/kg, 5 mg/kg, or placebo. At week 14, the infliximab group was then further randomized to continuous or maintenance regimens at their originally designated dose. Through week 50, this study demonstrated that the clinical response to treatment was best maintained using continuous infliximab therapy compared to an as-needed basis in both dosage groups. The median of the average percent improvement in PASI from week 16 through week 50 was 89.6% in the 5 mg/kg every 8-week group compared to 76.4% in the Median Serum concentration of infliximab through week 50 in week 10 responders Infliximab 5 mg/kg Median Serum Infliximab Concentration (µg/mL) 100 10 1 < 0.1 0 2 Week Infusion 6 10 14 22 30 38 46 50 Responders at Week 10 and Maintain Response at Week 50 (n = 56) Responders at Week 10 and Do Not Maintain Response at Week 50 (n = 19) Fig. 14.3 Difference in median serum infliximab concentration between responders and non-responders in EXPRESS and EXPRESS II trials. As an independent predictor of treatment response, patients that responded to infliximab therapy had a significantly higher median serum concentration of infliximab than nonresponders J. A. Mojeski and R. E. Kalb 178 Infliximab therapy Baseline Week 24 Fig. 14.4 Clinical response to Infliximab. Another example of disease improvement on IV infliximab therapy during the EXPRESS trials. A patient with significant baseline disease burden achieves almost a complete response after 24 weeks of infusions as-needed group (p < 0.001). Similar results were obtained for the 3 mg/kg group as well, with 80.6% improvement in the continuous group versus 72.4% in the as-needed group (p < 0.001). Furthermore, the authors showed that a 5 mg/kg dosage regimen achieved superior induction and maintenance scores compared to the lower dosage. Patient assessments across treatment groups were consistent with the PASI scores, as the continuous infusion of 5 mg/kg group reported the greatest improvements in their DLQI quality of life scores. Also in 2005 and 2007, the IMPACT I and II trials were conducted as randomized, ­double-­blind placebo-controlled studies enrolling patients with psoriatic arthritis in whom prior therapy with at least one disease-modifying anti-­rheumatic drug (DMARD) had previously failed [34, 35]. In IMPACT I, patients were assigned to receive infusions of either 5 mg/kg of infliximab or placebo at weeks 0, 2, 6, and 14. After week 16, patients initially assigned to receive placebo were crossed over to receive infliximab, and all groups received 5 mg/kg of infliximab every 8 weeks through week 50. At week 16, 65% of patients treated with infliximab attained an ACR20 response, compared to only 10% of placebo-­treated patients (p < 0.001). In addition, 46% of the infliximab group produced an ACR 50 response and 29% an ACR 70 response, where no placebo-treated patients achieved either of these end points (p < 0.001). Furthermore, at the 16-week evaluation 75% of infliximab-treated patients were improved according to the PsARC, compared to only 21% of placebo-controlled patients (p < 0.001). Additionally, although secondary endpoints the infliximab group showed substantial improve- 14 Infliximab, Golimumab, and Certolizumab Pegol 179 Infliximab therapy Baseline Week 24 Fig. 14.5 Infliximab in patients with nail disease. Nail Psoriasis Severity Index (NAPSI) improvements were noted during the EXPRESS and EXPRESS II trials. By week 24, a 56% mean decrease in NAPSI was achieved in the infliximab treatment group ments in in psoriatic skin disease as well as the percentages of patients suffering from the common complications of psoriatic arthritis, namely dactylitis and enthesitis [34, 35]. The IMPACT II trial was a 54-week multicenter study designed to expand the published clinical response data from the initial, 24-week, double blind placebo-controlled period [36] with an expansion of the number of enrolled patients [35]. As previously reported, these results demonstrated significant improvements in the signs and symptoms of psoriasis and psoriatic arthritis, as well as quality of life and physical functioning in patients through 1 year of treatment. Another outcome assessed was attainment of a “major clinical response,” which is defined as ACR 70 improvement for 24 consecutive weeks. At week 54, major clinical response was achieved by 12.1% of the infliximab-treated group. The authors also demonstrated that 5 mg/kg is a suf- ficient dose for a majority of patients. Dose escalation from 5 mg/kg to 10 mg/kg was assessed in 15 non-responders (patients who did not achieve ACR 20). Interestingly, in the patients requiring dose escalation, patients who had not achieved an ACR 20 score before escalation did not achieve ACR 20 despite doubling of the drug dosage. Furthermore, dose escalation did not appear to significantly improve PASI responses. Out of the 15 patients who receive dose escalations, only 12 had a baseline BSA of 3% or greater and were therefore included in the PASI analysis. Five of these patients achieved a PASI 75 at week 38 and maintained that response through week 54. Conversely, the seven patients who did not achieve a PASI 75 response at week 38 were unable to achieve the response after dose escalation. Overall, these studies show that infliximab therapy significantly reduces the signs and symptoms of psoriatic arthritis in patients that were 180 resistant to other treatment modalities and these benefits were sustained through 1 year of therapy. In August 2011, the first documented head-to-­ head, randomized trial (RESTORE 1) comparing the efficacy and safety of infliximab versus methotrexate was published [37]. 868 methotrexate-­ naïve patients were randomized to receive 5 mg/ kg of infliximab at weeks 0, 2, 6, 14, and 22 or 15 mg weekly methotrexate with a dose increase to 20 mg weekly at week 6 if the PASI response was <25%. Patients with less than a PASI 50 response were allowed to switch treatment groups at week 16. At week 16, a PASI 75 response was achieved by 508/653 (78%) of infliximab-treated patients compared to 90/215 (42%) of patients receiving methotrexate, and this response was maintained throughout the 26-week study (p < 0.001). Key secondary endpoints, including PGA, DLQI, and PASI 90 were likewise achieved by a greater proportion of infliximab-treated patients. More impressively, 46/63 (73%) patients who switched from methotrexate to infliximab at week 16 demonstrated a PASI 75 at week 26. Although the incidence of severe adverse events was slightly higher in the infliximab group (7% vs. 3%), a majority of the serious adverse events were infusion-related reactions, and the overall adverse event incidence was comparable between groups. The study demonstrated that infliximab was both well tolerated and more efficacious than methotrexate in patients with moderate-severe plaque psoriasis. Moreover, infliximab was proven to be a successful alternative agent in patients who had previously failed methotrexate therapy. While all biologics have performed well in short-term clinical trials, very limited direct comparisons between agents are available, particularly for psoriasis. However, since most studies used similar designs and end points, limited comparisons from randomized controlled trials are possible. Adalimumab and infliximab appear to have similar efficacy, with 70–80% of patients achieving a PASI 75 score at 12 weeks [9]. According to a systematic review by Rodgers et al., infliximab is associated with the highest probability of response on joint (ACR and J. A. Mojeski and R. E. Kalb PsARC) and skin (PASI) outcomes from 12–14 weeks [38]. Etanercept appears to be slightly less efficacious than its counterparts, demonstrating 50% of patients attaining a PASI of 75 at 12 weeks. Authors of a recent network meta-analysis concluded that, anti-TNF agents may be slightly less efficacious compared to anti­IL17 and anti-IL12/23 inhibitors, but are more effective than anti-metabolites such as methotrexate [39]. Anti-TNF agents have the advantage in terms of a well-known long term side effect based on over 20 years of experience. Promising data was recently published in The Lancet pertaining to the efficacy of a biosimilar to infliximab known as CT-P13 which was approved by the FDA in 2016 for all indications of infliximab after its patent expired in 2015. A treatment switch was performed in a Norwegian 52-week randomized, double-blind study known as NOR-SWITCH. Patients were enrolled from each inflammatory condition that infliximab is currently indicated for including plaque psoriasis, psoriatic arthritis, Crohn’s disease, ulcerative colitis, rheumatoid arthritis, and ankylosing spondylitis. In a 1:1 ratio, patients were either continued on infliximab or switched to CT-P13 treatment with unchanged dosing regimen. The study showed that switching therapy to this biosimilar was not inferior to continued treatment with infliximab. However, the study was not powered sufficiently to suggest non-inferiority in individual disease processes. With a cost-saving of up to 69% when compared to infliximab, the use of this drug and introduction of other biosimilars can significantly improve access to biologic therapies and lessen the financial burden of these medications [40]. One of the major advantages of infliximab is that it has a weight-based dosing on a milligram per kilogram basis unlike a majority of the TNF antagonists used to treat psoriasis [41, 42]. This allows for a more individualized patient-tailored therapy, which is particularly important as patients with psoriasis are typically above average weight [41, 42]. Additionally, a few studies have demonstrated the continued efficacy of infliximab regardless of body mass index, whereas the efficacy of fixed-dose TNF antagonists may 14 Infliximab, Golimumab, and Certolizumab Pegol 181 Infliximab: BMI and efficacy 100 Percent of Patients 78.3 77.5 80 74.4 60 40 20 5.0 0 1.7 n=231 n=60 Normal (<25) n=121 n=373 Overweight (25-30) 2.6 n=155 n=519 Obese (≥30) Body Mass Index Placebo Combined Infliximab Groups Fig. 14.6 Infliximab and BMI. There is no significant difference between normal weight patients and obese patients in achieving an efficacious response with fixeddose infliximab suggesting that there may be no need for weight-based dosing be compromised [41–43]. In one study in JAAD (Fig. 14.6), there was no significant difference between the number of patients who achieved an efficacious response with a BMI greater than or equal to 30 (74.4%) compared to those of normal body weight (77.5%) [43]. Combination Therapy Currently, there is some evidence to suggest that patients with psoriasis may obtain an improved response from combination therapy with infliximab and another therapeutic agent such as methotrexate, cyclosporine, acitretin, narrow-band UVB (nb-UVB) phototherapy, azathioprine, and apremilast [44–49]. A retrospective analysis of 23 patients receiving infliximab in combination with methotrexate or azathioprine showed PASI 50 improvements by 91.3% of patients. A prospective study performed by Goedkoop et al. showed that infliximab plus methotrexate therapy decreased PASI score and biopsy findings from the patients showed decreased angiogenesis and cellular infiltration at 4 weeks of therapy [49]. Additionally, an open-label study (RESPOND study) was conducted comparing the efficacy and safety of methotrexate alone versus methotrexate in combination with infliximab in patients suffering from psoriatic arthritis [50]. 115 methotrexate-­ naïve patients were randomly assigned to receive either 15 mg/week of methotrexate plus infliximab 5 mg/kg at weeks 0, 2, 6, and 14, or methotrexate therapy alone (15 mg/week). At week 16, 86.3%, 72.5%, and 49.0% of patients receiving combination therapy compared to 66.7%, 39.6%, and 18.8% of patients receiving methotrexate alone achieved ACR 20 (p = 0.021), 50 (p = 0.0009), and 70 (p = 0.0015) responses, respectively. With regards to skin disease, patients whose baseline PASI was 2.5 or greater, 97.1% of those on combination therapy and 54.3% of those on methotrexate alone experienced a 75% or greater improvement in PASI scores (p < 0.0001). Importantly, combination therapy was determined to be generally well-tolerated by the patient population. In the infliximab plus methotrexate group, 46% had treatment-related adverse 182 events (2 serious AEs) while the methotrexate alone group experienced 24% AEs (no serious AEs). While the overall incidence of adverse events was higher in the combination therapy group, a majority of the events were mild to moderate. Overall, this study suggested potential benefit of combination therapy for patients with psoriatic arthritis. In 2010, the New England Journal of Medicine published a randomized, double-blind trial comparing the efficacy of infliximab monotherapy, azathioprine monotherapy, and the combination of the two drugs in patients with Crohn’s disease [51]. 508 patients with moderate to severe Crohn’s disease were randomized to receive either 5 mg/kg infliximab at weeks 0, 2, 6, and then every 8 weeks thereafter, 2.5 mg/kg oral azathioprine daily, or combination therapy with the two medications (with the monotherapy groups also receiving either oral or infusion placebos). At week 26, 56.8% of the patients receiving combination therapy were in steroid-free clinical remission compared to 44.4% of those receiving infliximab alone (p = 0.02) and 30.0% receiving azathioprine alone (p = 0.006 for comparison with infliximab and p < 0.001 for comparison with combination therapy). At week 50, a similar trend was obtained with 46.2% of patients on combination therapy, 34.9% on infliximab alone (p = 0.04), and 24.1% on azathioprine alone (p = 0.03 for comparison with infliximab and p < 0.001 for comparison with combination therapy) maintaining clinical remission status. Safety data was generally similar among groups with serious infections occurring in 3.9% of the combination group compared to 4.9% in the infliximab group and 5.6% in the azathioprine group. While this study documented the incremental benefit of combination therapy for Crohn’s disease, controlled data are lacking in psoriasis. Only two randomized control trials have been performed to date assessing efficacy of combination therapy in plaque psoriasis with both involving etanercept with methotrexate. Each showed superior efficacy compared to etanercept monotherapy. Another trial known as OPTIMAP is underway currently that will assess the performance of adalimumab and methotrex- J. A. Mojeski and R. E. Kalb ate combination therapy [52]. Whether combination therapy with all TNF antagonists, including infliximab, will be more effective in improving or maintaining response are important questions to be answered for patients with psoriasis. As previously mentioned, maintaining the high initial response with infliximab therapy for a chronic disease such as psoriasis remains a challenge. The mechanisms for loss of response include antibodies to infliximab, tolerance to the drug, and drug metabolism for individual patients [7, 53–58]. Predictors of continued response include serum levels of infliximab at the time of infusion (Fig. 14.3) [32], as well as effective trough plasma concentrations [57]. Approximately 10% of patients with Crohn’s disease and rheumatoid disease in clinical trials have developed antibodies [59], and reports of up to one-third of patients with psoriatic disease [33, 60]. Studies have established relationships among antibody formation and low serum drug levels or failure and loss of response in patients with rheumatic and inflammatory bowel disease [61–65]. In psoriasis, elevated levels of antibodies were discovered in non-responders, and authors concluded that they appeared to play a role in the lack or regression of response [33, 66, 67]. In one trial in particular, patients with an initial response to infliximab positive for antibodies to the drug at week 10 were less likely to maintain a good response at 1 year than their antibody-negative counterparts [33]. Recently, a retrospective cohort study of 93 patients receiving infliximab therapy for psoriasis was performed to assess dosing techniques for prolonged infliximab response. The authors confirmed that concomitant methotrexate use was associated with increased time to discontinuation or dose escalation, likely due to its ability to decrease the formation of anti-infliximab antibodies. Additionally, increased dosing frequency, as compared to increasing the size of the dose, lead to statistically significant increase in time to discontinuation or dose escalation [58]. Thus, in order to maintain response, physicians may either start with methotrexate in combination with infliximab or add methotrexate during therapy. 14 Infliximab, Golimumab, and Certolizumab Pegol Golimumab Subcutaneous and intravenous golimumab have shown impressive efficacy results in patients with psoriatic arthritis. Although one may expect that the effects of golimumab in the treatment of plaque psoriasis are comparable to other TNF antagonists with a similar benefit in psoriatic arthritis, there are no published trials directly assessing the effects of this medication in plaque psoriasis to date. Therefore, it is not known how golimumab compares to other TNF inhibitors in the treatment of moderate to severe plaque psoriasis. With regards to psoriatic arthritis, In a randomized, placebo-controlled, multicenter clinical study (GO-REVEAL), 405 patients with active psoriatic arthritis were enrolled and randomly assigned to receive subcutaneous injections of placebo, golimumab 50 mg, or golimumab 100 mg every 4 weeks [68]. At week 14, 51% and 45% of patients receiving golimumab 50 mg and 100 mg respectively, achieved an ACR 20 response, compared with only 9% of patients receiving placebo therapy (p < 0.001). At week 24, an ACR 20 response was observed in 52% of patients in the 50 mg group and 61% in the 100 mg group, versus 12% of patients receiving placebo (p < 0.001). The trial continued and an impressive 279 patients completed five total years of therapy. At the five-year conclusion of the study, ACR20, 50, and 70 scores were 62.8–69.9%, 43.4–50.7%, and 30.8– 35.6%, respectively. This showed that golimumab not only made significant clinical improvements for patients, but the therapy also had significant longevity. Improvement in dactylitis and enthesitis scores, as well as nail psoriasis severity was evident. Patient’s PASI scores were determined as a secondary endpoint and showed improvement against placebo. In patients who were suffering from at least 3% BSA involvement of skin psoriatic disease, 40% in the 50 mg golimumab group and 58% in the 100 mg golimumab group had at least 75% improvement in their skin disease (PASI 75) by 183 week 14 [68]. This was compared to only a 3% PASI 75 response in placebo-treated patients (p < 0.001). This beneficial response was maintained through week 24 in both golimumab groups (56% and 66% for 50 mg and 100 mg) whereas only 1% of patients in the placebo group reached a PASI 75 at this time marker (p < 0.001). At week 104, data revealed a PASI 75 of 68.8% of patients in the 50 mg group and 76% in the 100 mg golimumab group [12, 68, 69]. However, the trial failed to show consistent difference in PASI score in patients treated with a golimumab regimen compared to patients with baseline methotrexate use [69]. This study gave significant evidence that subcutaneous golimumab improved the clinical signs and symptoms of psoriatic arthritis, along with associated skin and nail disease, as well as physical functioning and quality of life [12, 68, 69]. Intravenous golimumab has had similar efficacy results as evidenced by the GO-VIBRANT trial. In the phase III, randomized, double-blind, placebo-controlled trial, patients with psoriatic arthritis were assigned to IV placebo or golimumab at 2 mg/kg at weeks 0, 4, 12, and 20. A primary endpoint of ACR20 response was assessed and was achieved by 75.1% of patients in the golimumab group compared to 21.8% of patients in the placebo group (p < 0.001). Additionally, greater mean changes were observed in each of the secondary endpoints of the study. ACR50 and ACR70 responses were found in 43.6% and 24.5% of golimumabtreated patients compared to only 6.3% and 2.1% in ­ placebo group, respectively. Additionally, 59.2% of golimumab-treated patients enrolled in the study achieved PASI75 response compared to only 13.6% of placebo patients. This study suggests comparable efficacy of both formulations of golimumab in the treatment of psoriatic arthritis. Again, although this gives some evidence toward the use of golimumab for the treatment of plaque psoriasis, a randomized control study would be more effective to determine golimumab’s place in the management of the condition [70]. J. A. Mojeski and R. E. Kalb 184 Certolizumab tions every 2 weeks and patients were assessed at 16 weeks and 48 weeks. At week 16, patients Certolizumab has significant promise in the man- enrolled in CIMPASI I and II achieved PASI 75 agement of both moderate-severe plaque psoria- responder rates of 75.8% and 82.6%, respectively sis and psoriatic arthritis. With regards to plaque in the certolizumab 400 mg group and 66.5% and psoriasis, there have been three phase 3 trials 81.4%, respectively in the certolizumab 200 mg (CIMPACT I, CIMPASI I, and CIMPASI II). The group. When compared to patients receiving only CIMPACT trial compared certolizumab against placebo therapy, 6.5% of patients in CIMPASI-I etanercept and placebo while CIMPASI I and II and 11.6% of patients in CIMPASI-II receiving were solely placebo-controlled studies. The placebo achieved PASI 75 (P < 0.001). These CIMPACT trial results showed that the use of results were largely maintained throughout the certolizumab for patients with chronic plaque 48-week trial (Fig. 14.8). Additionally, pooled psoriasis was associated with a significant differ- analysis of both trials showed impressive PASI ence in PASI 75 scores compared to placebo. 100 response rates of 34.5% for certolizumab Additionally, when directly compared to its anti- 400 mg and 28.5% for certolizumab 200 mg. TNF counterpart etanercept, certolizumab End-points of Dermatology Life Quality Index 400 mg was superior to and 200 mg was non-­ and Physician Global Assessment also showed significant improvement [71]. inferior to etanercept (Fig. 14.7) [28]. Certolizumab’s use in the management of psoIn the randomized-control CIMPASI I and II trials patients were enrolled and assigned to 2:2:1 riatic arthritis was studied in a 4-year trial treatment groups consisting of certolizumab (RAPID-PsA) performed in patients treated with 400 mg, certolizumab 200 mg (with loading and without concomitant DMARD therapy. The doses of 400 mg at 0, 2, and 4 weeks) and pla- study was a double-blind and placebo-controlled cebo. Each group received subcutaneous injec- study through 24 weeks, dose-blind to 48 weeks 100 Placebo (N = 57) CZP 200 mg Q2W (N = 165) CZP 400 mg Q2W (N = 167) 80 ETN (N = 170) 74.7% Responder rate (%) 66.7% †‡ 68.2% † † 61.3% †§ 60 † 40 53.3% † 20 * 5.0% * 3.8% 0 0 2 4 8 12 16 Week Fig. 14.7 Percentage of responders (PASI 75) in placebo, certolizumab and etanercept groups within the CIMPACT trial. Certolizumab displays superiority to etanercept at 400mg Q2W dosing and non-inferiority at 200mg Q2W dosing 14 Infliximab, Golimumab, and Certolizumab Pegol 185 CIMPASI-1 Placebo (N=51) CZP 200 mg Q2W (N=95) CZP 400 mg Q2W (N=88) Responder rate (%) 100 87.1% 75.8%† 80 † 60 * 40 * † * 20 0 * 0 2 4 6.5% 8 12 Placebo (N=49) CZP 200 mg Q2W (N=91) CZP 400 mg Q2W (N=87) Responder rate (%) 100 CIMPASI-2 67.2% 66.5%† † 80 † 60 0 20 24 28 32 40 82.6%† 81.3% † 81.4% 78.7% * * * 0 2 4 11.6% 8 12 16 20 100 24 28 Week 32 40 CZP 200 mg Q2W (N=186) CZP 400 mg Q2W (N=175) Responder rate (%) Placebo (N=100) 80 48 83.6% 82.0% † Pooled 48 † 40 20 † 16 † † 60 † 76.7%† 70.7% † 40 † 20 0 9.9% * 0 2 4 8 12 16 20 24 28 Week 32 40 48 Fig. 14.8 CIMPASI-I and CIMPASI-II randomized control trail results. Patients randomized to both the certolizumab 200mg Q2W and certolizumab 400mg Q2W achieve significant benefit compared to patients in placebo group. Results were maintained throughout 48-week trial period and then open label until 216 weeks [72–76] In the trial, 409 patients were randomized to complete 24 weeks of treatment with either certolizumab 200 mg every 2 weeks, 400 mg every 4 weeks, or placebo. ACR 20 response was significantly greater at 12 weeks (58.0% and 51.9% vs. 24.3%, respectively (p < 0.001)) and PsARC achievement was also significant against placebo group at 24 weeks (78.3% and 77.0% vs. 33.1% (p < 0.001) [72]. The study was continued over 4 years with exciting results. Of the 67% of patients who completed 216 weeks of therapy, 60.4% achieved a Disease Activity Index for Psoriatic Arthritis low disease activity or remission with an increase to 66.3% at 216 weeks. Additionally, at four-years, resolution rates for enthesitis, dactylitis, and nail psoriasis were 71%, 81%, and 65% respectively [73, 74, 76]. Safety Considerations Throughout their nearly two decades of clinical use, the safety profile of TNF-inhibitors is well characterized [2]. In clinical trials, infliximab, golimumab, and certolizumab have been proven to be generally well tolerated, however, due to their down-regulatory effects on the immune sys- 186 J. A. Mojeski and R. E. Kalb tem, all TNF antagonists have labeled warnings among patients involved in psoriasis trials (393 about the potential development of bacterial, patients with psoriatic arthritis and 1112 patients viral, and fungal infections during treatment with psoriasis) were the lowest of all inflamma(9,77–79). While the most common adverse tory conditions examined. When examining these events are mild, consisting of nausea, headache, data it is also important to note that inflammatory upper respiratory tract infections, abdominal conditions have been associated with adverse pain, fatigue, and fever, serious and sometimes reactions regardless of the effect of anti-TNF fatal events have been reported [7, 77–79]. agents such as psoriasis and cardiovascular risk Kavanaugh et al. reported that only 8.6% of [75]. patients treated with golimumab experienced a serious adverse event up to 104 weeks [12]. Infusion-related reactions are also possible and Infusion-Reactions were reported in about 20% of patients receiving infliximab compared to only 10% in placebo Infusion-related reactions are unique to inflixgroups [31, 59]. These reactions included hyper- imab and golimumab as these are currently the tension, hypotension, bronchospasm, chest pain, only TNF antagonists that are administered intradyspnea, pruritis, and fever [80, 81]. Infusion-­ venously. Infliximab infusions can be adminisrelated reactions may occur with all intravenously-­ tered within a hospital or a community setting administered biologic agents, but because [82]. Typically, infliximab is administered over a infliximab is a human-mouse antibody, anaphy- 2 hour time span, however, a prospective cohort laxis is possible, although uncommon [2]. For study demonstrated that infliximab infusion can golimumab, injection site reactions occurred in be safely administered over 1 hour in patients 8.9% of golimumab treated patients, but only with no past history of significant infusion reacwith 0.7% of all golimumab injections over tion [84]. IV Golimumab is administered over a 104 weeks of treatment [12]. Malignancy, auto- 30-minute period [70, 85]. A majority of the immune disease, demyelinating disease, and con- infusion-­reactions are mild, consisting of flushgestive heart failure [13] serve as additional ing, dizziness, nausea, sweating, and increase in concerns with the use of these agents, however temperature, typically occurring within the first with appropriate screening and selection of 2 hours after treatment [31, 86–88]. However, patients, the potential for development of these patients may develop antibodies to golimumab more serious conditions declines. Also of impor- and infliximab which may lead to more serious tant note, the adverse event data of many of the reactions, although rare, including shortness of biologics are derived primarily from patients breath, hypo/hypertension, chest tightness, sympwith rheumatoid arthritis (RA) or inflammatory toms of anaphylaxis such as urticaria, and bronbowel disease (IBD) as they have the most chospasms. Additional reactions of arthralgias, ­long-­term and extensive data available [1, 38]. myalgias, headache, fatigue, and influenza-like The generalizability of these findings in patients symptoms may occur as well ([32, 85, 86, 89, suffering from psoriasis and psoriatic arthritis 90]). It has been estimated that infusion reactions remains unclear. More recent data suggests the occur in 3–22% of patients receiving treatment side effect profile may be more favorable in with infliximab for psoriasis [87]. Golimumab patients with psoriatic diseases [1, 77, 82, 83]. appears to have a lower incidence of infusion-­ For certolizumab specifically, a study performed related reactions with 0.8% of patients experiencby Curtis et al. analyzed the side-­effect profile of ing a reaction through 24 weeks of the the drug used on 11,317 patients across a multi- GO-VIBRANT study and 1.1% of patients tude of inflammatory conditions including psori- treated with a combination of methotrexate and asis, psoriatic arthritis, axial spondyloarthritis, golimumab in the GO-FURTHER study [85, 90] rheumatoid arthritis, and Crohn’s disease [77]. Typically, symptoms can be monitored and will These data show that the rates of adverse events resolve with minor analgesics or antihistamines. 14 Infliximab, Golimumab, and Certolizumab Pegol If a severe reaction develops, infliximab should be discontinued immediately with commencement of appropriate treatment [88]. Generally, infliximab and golimumab treatment can be continued after a mild or moderate reaction. Attempts have been made to reduce the probability of infusion-­related reactions. Various agents have been administered as pre-medications, including intravenous steroids, acetaminophen, and antihistamines, however, most trials have failed to demonstrate a protective effect of premedications and lead to doubt on whether their risk of potential side effects is justifiable [84, 90–93]. Co-medication with disease-modifying therapies [84, 94–96] and ensuring a reliable maintenance schedule (opposed to an as-needed schedule) [88] have been shown to reduce the risk. Infection Infections are serious complications that can result from the use of TNF antagonists. While the most common types reported are upper respiratory tract infections and urinary tract infections, more serious infections such as cellulitis, pneumonia, abscesses, skin ulceration, pyelonephritis, cholecystitis, and sepsis have occurred [12, 59, 68, 79]. Although observational studies and meta-analyses of rheumatoid arthritis and inflammatory bowel disease randomized controlled trials have indicated an increased risk of serious and non-serious infections [97–103], clinical safety data specific for psoriasis and psoriatic arthritis demonstrated only a very small increased risk of overall infection with the short-term use of biologics, without an increased risk in the development of more serious infections [1]. Furthermore, the authors even suggested that the increased risk of overall infection may be attributable to variations in follow-up time between the treatment and placebo groups. Rheumatoid arthritis and IBD patient populations are typically treated with the concomitant use of additional immunosuppressants and have a higher background incidence of infection. These factors are the likely explanations for an increased infection rate in these groups [97, 98]. By design, 187 patients with psoriasis receive monotherapy in clinical trials [104–108]. Although the overall risk for infection reached statistical significance, it may have limited clinical implications as 97.6% of the reported infections were non-serious, with a large majority represented by upper respiratory infections [1]. More surprisingly, Dommasch et al. found a marginally statistically significant decreased risk of serious infection [1]. There were three reported cases of cellulitis in the placebo group, versus only one in the treatment group. It is postulated that an improvement in skin disease and a decreased amount of excoriations and breaks in the skin barrier are possible explanations for this unexpected finding. A significant concern of health practitioners is the potential for the reactivation of tuberculosis [7, 59, 60]. The risk of reactivation of latent TB is, of course, dependent on the incidence of latent infection [4]. From January 1998 to September 2002, the reported cumulative incidence for patients in the USA was estimated at 54/100,000 for infliximab [109]. However, the reality of this serious risk has lead to the introduction of pre-­ treatment screening procedures, which have successfully reduced the number of cases [99, 110]. It has been reported that reactivation occurs at the greatest frequency within the first 12 weeks of treatment [111, 112]. The mechanism by which TB reactivation occurs is not fully understood. Anti-TNF agents likely lead to apoptosis of active CD8+ T cells. These cells, along with the cytokine TNF-alpha, are crucial for the maintenance of granulomas and the lysis of host cells harboring intracellular invaders, such as Mycobacterium tuberculosis. The anti-TNF agent etanercept is associated with a lower risk of TB reactivation in comparison to other anti-TNF agents. This may be due to less inhibition of TNF-a as etanercept binds solely soluble TNF-a whereas other anti-­ TNF agents bind tmTNF [3, 19]. There is also some data to suggest that psoriasis itself is a risk factor for the risk of TB infection [113]. Screening for TB infection should include a full history and physical exam. Additionally, patients require a tuberculin skin test (TST), and/or interferon-­ gamma release assay (IGRA) with chest radiography occurring after positive screen [13, 188 113–115]. The CDC guidelines suggest that 3 months of isoniazid/rifapentine is the treatment of choice in countries such as the USA with low to moderate levels of latent infection. As of 2018, this recommendation has been expanded from only adults to patients aged 2–17 years and patients with HIV infection and taking antiretroviral medications [116]. In the setting of active TB, anti-TNF agents should be discontinued immediately. There is still no conclusive evidence on when to re-start or initiate anti-TNF therapy after treatment of active or latent TB infection. A multi-disciplinary panel, the Italian multidisciplinary task force for screening of tuberculosis before and during biologic therapy (SAFEBIO) reviewed the available evidence from phase III randomized control trials in which anti-TNF agents were studied. The panel evaluated latent TB infection, identification of individualized level of risk of TB reactivation, and management of patients when TB reactivation occurred. The panel recommended that biologic agents could be initiated or resumed 1 month after the treatment of latent or active TB. Since the TST and IGRA will not be helpful for the diagnosis of TB reactivation in these patients, they need consistent clinical monitoring for signs and symptoms of active TB [113]. Additional rare severe, opportunistic infections have also been reported, such as listeriosis, coccidioidomycosis, and histoplasmosis along with infections caused by Cryptococcus, Aspergillus, and Pneumocystis [2, 79]. Although the incidence of fungal infections were not significantly increased in clinical trials, based on case reports and post-marketing surveillance data, a fungal infection should be suspected if a patient on biologic therapy develops a fever [13]. Viral infections are an additional concern, as there have been reports of reactivated hepatitis B, and worsening of hepatitis C [2]. All anti-TNF agents carry a boxed warning regarding the reactivation of hepatitis B. Although these complications exist, a recent recommendation from a JAAD literature review concluded grade C evidence for the use of hepatitis B and C screening. Therefore, this can be left to clinical judgement J. A. Mojeski and R. E. Kalb and assessment of risk factors [117]. In the case of a positive result, anti-TNF therapy should be initiated only in combination with hepatitis treatment and close monitoring of liver enzymes and viral DNA levels under the supervision of a specialist [118–121]. There is currently limited evidence on the safety of infliximab treatment in HIV positive individuals as most of these patients are excluded from trials with biologic agents [13, 117]. Caution is therefore advised when considering anti-TNF therapy in these high-risk individuals [122]. In addition to the aforementioned screening measures, no live vaccinations should be administered to a patient undergoing TNF antagonist therapy, and these agents should be withheld if patients are given antibiotics and completely discontinued in the presence of severe infections, as these patients have a higher propensity to develop serious bacterial complications [123–125]. Despite the risk of infectious complications, the overwhelming majority of these infections are minor, consisting mainly of upper respiratory infections [1]. JAMA published a multi-center, retrospective study examining whether the use of biologic agents was associated with an increased risk of serious infections requiring hospitalization in comparison to non-biologic agents [126]. They determined that TNF-antagonists were not associated with an increased risk of hospitalization for serious infections across multiple autoimmune diseases, including psoriasis and psoriatic arthritis. However, it was noted that for rheumatoid arthritis patients, infliximab was associated with a higher rate of serious infection when compared alone to non-biologics (adjusted hazard ratio: 1.25, 95% confidence interval: 1.07–1.48) and when compared to the other TNF agents, etanercept (aHR: 1.26; 95% CI: 1.07– 1.47) and adalimumab (aHR: 1.23; 95% CI: 1.02–1.48). Subgrouping of the TNF antagonists was not performed for psoriasis specifically, however the rate of serious infections for the biologics as a group compared to the non-biologics was not significantly different. Baseline use of glucocorticoids, however, was associated with a significantly increased risk of serious infection 14 Infliximab, Golimumab, and Certolizumab Pegol and hospitalization compared to no baseline use of steroids. An additional concern surrounding the increased risk of infections is whether or not this risk will further increase perioperatively, and whether or not infliximab infusions should be held for a certain period of time before a major or minor surgery. Current recommendations from many professional societies recommend withholding therapy prior to surgery. The therapy may be withheld for 3–5 half-lives of the drug or individualized to patient characteristics and surgery type. However, this is not without risk and disease flares can lead to adverse functional outcomes and impair rehabilitation [127]. A retrospective study was conducted to assess the safety of preoperative infliximab use before restorative proctocolectomy and ileal pouch-anal anastomosis (IPAA) in patients suffering from ulcerative colitis (UC) [128]. Although controversy exists surrounding the risks of preoperative infliximab [129–132], it was determined that short-term postoperative and infectious complications were similar between the group that had received infliximab within 12 weeks of the surgery (44.8%), and those who had not (44.2%) [133]. In fact, a trend toward lower rates of wound infection was observed for the infliximab group (3.5%) compared to controls (19.2%). Another recent retrospective cohort performed on patients receiving infliximab within 4 weeks of elective hip or knee arthroplasty were not at an increased risk within 1 year compared to patients withholding therapy [134]. While these results are promising, there is still a need for prospective studies. Malignancy Similar to infections, meta-analyses and observational studies with TNF antagonists in the RA population demonstrated an increased risk of malignancy [97, 98, 135, 136], although there is conflicting evidence [137–144]. Again, the same argument holds true as above where the different disease states and the combination of immunosuppressants in this population may have a syner- 189 gistic effect resulting in an increased risk of infection and malignancy that may not be applicable to the psoriatic patient population [104– 108]. In fact, in a meta-analysis the authors concluded that there was no statistically significant increased risk of malignancy in patients with psoriatic disease on short-term biologic therapy [1]. Overall, in the malignancies observed, 70.6% were non-melanoma skin cancers [1]. Whether this was an artifact of increased recognition as the psoriatic lesions healed is uncertain. Additionally, it has been proposed that a patient’s psoriasis may inherently increase their risk for developing lymphoma, further complicating the analysis of biological agents [144, 145]. In the golimumab trials, non-melanoma skin cancers were also the most common, however colon cancer, prostate cancer, and small cell lung carcinoma were also observed, although infrequently [12]. Furthermore, the results of randomized controlled trials show that 26% of malignancies occur within 12 weeks from enrollment in patients receiving TNF inhibitors, suggesting pre-existence of the cancers before initiation of biologic therapy [146]. Therefore, it is prudent that patients undergo age and risk adequate screening before initiation of treatment. Another meta-analysis was performed on patients enrolled on certolizumab therapy regimens for all approved indications. This study determined a standardized incidence ratio (SIR) of malignancy to be 1.03 (95% CI 0.87 to 1.20) using the SEER database and an SIR of 1.45 (95% CI 1.23 to 1.69) when using the worldwide GLOBOCAN healthy population data set for patients taking certolizumab. The authors suggest that, although the SEER data base is typically used in biologic safety studies, GLOBOCAN data may be more representative of the population and caution should be exercised when dealing with biologics and malignancy risk [18]. Overall, the short-term risk-benefit profile of biologics in patients with psoriatic disease is favorable. However, continued long-term studies with larger patient populations are necessary in order to adequately assess the risk of cancer and J. A. Mojeski and R. E. Kalb 190 serious infection with chronic use of infliximab, golimumab, and certolizumab. Laboratory Data/Autoimmune Disease use of TNF antagonists [9, 13]. Because of this increased risk, patients with MS along with their first-degree relatives have been cautioned against the use of the drugs [123, 124]. Dermatologic Some studies have reported abnormalities in laboratory data associated with the use of infliximab. Various skin complications have been reported In a randomized study by Reich et al., a signifi- with the use of anti-TNF agents, some of which cant increase in liver enzymes, aspartate and ala- include, delayed hypersensitivity type reactions, nine aminotransferases was observed [32]. lupus-like syndrome, bullous skin lesions, Anti-TNF agents have been associated with a eczematous-­like purpura, annular lichenoid erupnumber of cases of liver injury, such as choles- tion, and leucocytoclastic vasculitis [79]. In additatic disease and hepatitis, with some life-­ tion, several eczematous eruptions have occurred threatening cases reported [147–149]. with the use of infliximab, however most reacAdditionally, an association has been made tions resolve with discontinuation of the offendbetween anti-TNF agents and the development of ing agent [155]. autoimmune hepatitis type 1 reactions. After disInterestingly, there are an increasing number continuation of the anti-TNF agent, liver enzymes of cases documenting the paradoxical formation return to normal values and harmful reactions of psoriasis during treatment with TNF inhibitors such as hepatitis typically regress. Interestingly, [3, 134, 156–160], primarily in patients treated anti-TNF agents have also been anecdotally used for other autoimmune diseases, such as Crohn’s as rescue therapy in treatment-resistant primary disease and rheumatoid arthritis [133, 161]. It is biliary cholangitis and autoimmune hepatitis likely that the pathophysiology involves the dis[147]. Other laboratory abnormalities including equilibrium of cytokines caused by the inhibition hematological disturbances have also been noted, of TNF-alpha. It has been suggested that TNF-a such as rare cases of aplastic anemia and pancy- normally inhibits plasmacytoid dendritic cell topenia, which can further predispose the patient secretion of interferon-alpha (IFN-a), a known to serious infections [2]. inducer of psoriasiform lesions. The upregulation Additionally, due to its chimeric nature, inflix- of IFN-a that occurs in patients on anti-TNF-a imab has been shown to result in the formation of therapy may result in the formation of skin disantibodies [79]. Most commonly, antinuclear ease in predisposed individuals [160, 162]. antibodies (ANA), antibodies to double-stranded Various treatments have been reported to be useDNA, and anti-cardiolipin antibodies have been ful in this situation, such as aggressive topical reported [150–152]. Studies of Crohn’s disease therapy, the addition of methotrexate, acitretin, show that 44% develop ANAs at some point dur- cyclosporine, phototherapy, or switching the ing the course of their treatment with infliximab TNF agent [160, 163]. Most frequently used [7, 59]. While the development of systemic lupus agents have been glucocorticoids and vitamin D erythematosus (SLE) in patients undergoing bio- analogs for topical therapy and methotrexate, logic treatment is rare, it is postulated that the glucocorticoids, and phototherapy for systemic important role TNF plays in autoregulation may treatment. Complete discontinuation of all biobe linked with new signs of autoimmune disease logics is often unnecessary, and a change in the in patients receiving anti-TNF agents [13, 153, TNF antagonist used should be considered if con154]. It is also noted that withdrawal of therapy ventional treatment fails. Ultimately, 30.7–100% typically results in resolution of symptoms. of patients were able to continue on anti-TNF-a Several demyelinating and neurologic events, therapy with adjuvant topical or systemic treatincluding exacerbations of pre-existing multiple ment [160, 162]. sclerosis (MS), have also been reported with the 14 Infliximab, Golimumab, and Certolizumab Pegol Treatment Switches There is evidence that a gradual decline in therapeutic efficacy occurs in some patients with TNF antagonists, whether through decreased bioavailability of the drug or as a biological adaptation to chronic blockade, such as the development of anti-drug antibodies [7, 56, 164]. However, switching to a different biologic agent has proven effective in treatment-refractory patients. In 2007, JAAD published a retrospective study involving the efficacy of infliximab in patients who previously failed treatment with etanercept [164]. Infliximab was initiated at 5 mg/kg and administered on weeks 0, 2, 6, 14, and every 8 weeks thereafter. After only 12 to 14 weeks of infliximab therapy, 17 of 19 (89%) patients showed improvements in their PGA and BSA. Fifteen (79%) still maintained adequate control on infliximab at the time of study publication, although 10 patients required infliximab dose escalation, with a majority of patients requiring infusions every 6 weeks to maintain continued response. In addition, safety data was obtained and compared between the two TNF-­ antagonists. The use of infliximab was associated with a possible increased incidence in adverse events compared to etanercept (16 versus 5 events, respectively). However, a majority of these events were considered minor. A multicenter, open-label prospective study (PSUNRISE) was conducted to evaluate the clinical response of an etanercept-to-infliximab switch in patients with psoriasis unresponsive to, or with a loss of response to, etanercept therapy [55]. Patients were included who had a PGA score of at least 2 despite 4 or more months of treatment with etanercept. Patients received intravenous infusions of infliximab 5 mg/kg at weeks 0, 2, 6, 14, and 22. At week 10, 65.4% of patients achieved a PGA score of 0 (clear) or 1 (minimal) and 61.3% of patients maintained this response throughout the 26 weeks. Moreover, there were no unexpected side effects or safety concerns experienced during this study. This trial demonstrates that patients with an inadequate response to etanercept may achieve substantial benefit after switching to infliximab. 191 As an important aside, treatment switches from anti-TNF therapy for non-medical reasons, such as job loss or change of insurance, has been associated with deteriorating clinical outcomes. In a 2017 study, disease flares, disease control, and health care utilization was compared between patients who were switched/discontinued and those that continued their anti-TNF agent. Switchers were further classified by those that switched to another biologic therapy and those that switched to conventional therapy. The data included patients with inflammatory conditions including plaque psoriasis, psoriatic arthritis, spondylarthritis, IBD, and rheumatoid arthritis. Importantly, the authors found that switchers/discontinuers had worse clinical outcomes, even if the agent that was switched to was another biologic agent (165). This study suggests that physicians need to be careful in the management of their patients with inflammatory conditions and have considerable knowledge about the insurance formularies and compliance of their respective patients. Conclusion Infliximab has been clinically available for over two decades and a large body of data exists proving its generally increased efficacy and fewer adverse events over traditional agents in numerous inflammatory disorders, including psoriasis and psoriatic arthritis. Although newer anti-IL17 and anti-IL12/IL23 agents may have similar or slightly superior efficacy in the treatment of psoriasis, research has proven the effectiveness of infliximab, golimumab, and certolizumab in treating signs and symptoms of psoriasis, and demonstrated their rapid and prolonged suppression of inflammation preventing long-term disease progression, especially in debilitating disease such as psoriatic arthritis. Infliximab and golimumab are unique in the IV formulations providing a rapid onset of action. Certolizumab has a unique treatment niche in women of child bearing potential. No new or unexpected safety issues have emerged over the years, and infliximab, golimumab, and certolizumab have been shown to be well-tolerated when clinicians 192 appropriately select patients and adhere to adequate screening and monitoring guidelines. As for long-term efficacy, there is a great need for the development of predictive biomarkers to predict response to biologic therapy, and more data is needed in order to fully characterize the role of neutralizing anti-drug antibodies. Overall, the risk-benefit profile of these medications is favorable, and at this time, cost appears to be the main barrier to the more widespread and extensive use of these biological agents. The development and use of biosimilars may have the potential to reduce these costs for patients. References 1. Dommasch ED, Abuabara K, Shin DB, Nguyen J, Troxel AB, Gelfand JM. The risk of infection and malignancy with tumor necrosis factor antagonists in adults with psoriatic disease: a systematic review and meta-analysis of randomized controlled trials. J Am Acad Dermatol. 2011;64:1035–50. https://doi. org/10.1016/j.jaad.2010.09.734. 2. Smolen JS, Emery P. Infliximab: 12 years of experience. Arthritis Res Ther. 2011;13(Suppl 1):S2. https://doi.org/10.1186/1478-6354-13-S1-S2. 3. Silva LC, Ortigosa LC, Benard G. Anti-TNF-alpha agents in the treatment of immune-mediated inflammatory diseases: mechanisms of action and pitfalls. Immunotherapy. 2010;2:817–33. https://doi. org/10.2217/imt.10.67. 4. Tak PP, Kalden JR. Advances in rheumatology: new targeted therapeutics. Arthritis Res Ther. 2011;13(Suppl 1):S5. https://doi. org/10.1186/1478-6354-13-S1-S5. 5. Gupta AK, Skinner AR. A review of the use of infliximab to manage cutaneous dermatoses. J Cutan Med Surg. 2004;8:77–89. https://doi. org/10.1177/120347540400800202. 6. Gall JS, Kalb RE. Infliximab for the treatment of plaque psoriasis. Biologics: Targets & Therapy. 2008;2:115–24. 7. Drosou A, Kirsner RS, Welsh E, Sullivan TP, Kerdel FA. Use of infliximab, an anti-tumor necrosis alpha antibody, for inflammatory dermatoses. J Cutan Med Surg. 200(7):382–6. https://doi. org/10.1177/120347540300700503. 8. Services U S D o H a H. FDA Approved Drug Products, Remicade. In, 2011. 9. Herrier RN. Advances in the treatment of moderate-­ to-­ severe plaque psoriasis. Am J Health Syst Pharm. 2011;68:795–806. https://doi.org/10.2146/ ajhp100227. J. A. Mojeski and R. E. Kalb 10. Scallon B, Cai A, Solowski N, et al. Binding and functional comparisons of two types of tumor necrosis factor antagonists. J Pharmacol Exp Ther. 2002;301:418–26. https://doi.org/10.1124/ jpet.301.2.418. 11. Smeets TJ, Kraan MC, van Loon ME, Tak PP. Tumor necrosis factor alpha blockade reduces the synovial cell infiltrate early after initiation of treatment, but apparently not by induction of apoptosis in synovial tissue. Arthritis Rheum. 2003;48:2155–62. https:// doi.org/10.1002/art.11098. 12. Weger W. Current status and new developments in the treatment of psoriasis and psoriatic arthritis with biological agents. Br J Pharmacol. 2010;160:810–20. https://doi.org/10.1111/j.1476-5381.2010.00702.x. 13. Sfikakis PP. The first decade of biologic TNF antagonists in clinical practice: lessons learned, unresolved issues and future directions. Curr Dir Autoimmun. 2010;11:180–210. https://doi. org/10.1159/000289205. 14. Services U S D o H a H. FDA Approved Drug Products, Simponi and Simponi Aria. In, 2013. 15. Lee JU, Shin W, Son JY, Yoo KY, Heo Y. Molecular basis for the neutralization of tumor necrosis factor a by Certolizumab Pegol in the treatment of inflammatory autoimmune diseases. Int J Mol Sci. 2017;18 https://doi.org/10.3390/ijms18010228. 16. Rivkin A. Certolizumab pegol for the management of Crohn’s disease in adults. Clin Ther. 2009;31:1158– 76. https://doi.org/10.1016/j.clinthera.2009.06.015. 17. Bourne T, Fossati G, Nesbitt A. A PEGylated fab’ fragment against tumor necrosis factor for the treatment of Crohn disease: exploring a new mechanism of action. Bio Drugs. 2008;22:331–7. https://doi. org/10.2165/00063030-200822050-00005. 18. Services USD. O H a H. FDA approved drug products. Cimzia. 19. Bianchi L, Costanza G, Campione E, Ruzzetti M, et al. Biomolecular index of therapeutic efficacy in psoriasis treated with anti-TNF-a agents. G Ital Dermatol Venereol. 2018;153:316–25. https://doi. org/10.23736/S0392-0488.16.05427-4. 20. Mitoma H, Horiuchi T, Tsukamoto H, Ueda N. Molecular mechanisms of action of anti-TNF­a agents -- comparison among therapeutic TNF-a antagonists. Cytokine. 2018;101:56–63. https://doi. org/10.1016/j.cyto.2016.08.014. 21. Blandizzi C, Gionchetti P, Armuzzi A. The role of tumour necrosis factor in the pathogenesis of immune-mediated disease. Int J Immunopathology and Pharmacology. 2014;27:1–10. https://doi.org/10 .1177/03946320140270S101. 22. Kalliolias GD, Ivashkiv LB. TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat Rev Rheumatology. 2016;12:49–62. https://doi.org/10.1038/nrrheum.2015.169. 23. Tracey D, Klareskog L, Sasso EH, Salfeld JG, Tak PP. Tumor necrosis factor antagonist mechanisms of action: a comprehensive review. Pharmacol 15 Ustekinumab George Han, Caitriona Ryan, and Craig L. Leonardi Abstract Ustekinumab is a fully human IgG antibody to the common p40 subunit of interleukin-12 (IL12) and IL-23, which has shown considerable efficacy in the treatment of psoriasis and psoriatic arthritis. This review examines the efficacy and safety of ustekinumab for the treatment of psoriasis in clinical studies to date. Ustekinumab was shown to be highly effective in the treatment of moderate-to-­severe psoriasis with sustained response for up to 5 years in the majority of patients. Adverse events in clinical studies to date have been for the most part, mild and similar to that in placebo-treated patients. Similarly, ustekinumab has shown efficacy in psoriatic arthritis and in adolescent psoriasis in patients ages 12 and up. While early questions about major adverse cardiovascular events existed in relation to this class of medication, the burden of evidence over the clinical trial program and over a decade in clin- G. Han (*) Department of Derrmatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA e-mail: george.han@mountsinai.org C. Ryan Department of Dermatology, Baylor University Medical Center, Dallas, TX, USA C. L. Leonardi Department of Dermatology, Saint Louis University School of Medicine, St. Louis, MO, USA © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_15 ical practice has borne out that ustekinumab remains a safe and effective treatment for psoriasis and psoriatic arthritis. Key Learning Objectives 1. To understand the mechanism of action of ustekinumab. 2. To understand the appropriate use and efficacy of the ustekinumab. 3. To understand the safety issues of ustekinumab. Introduction Ustekinumab (Janssen Biotech, Philadelphia, PA) is a fully human immunoglobulin G1/kappa (IgG1/κ) monoclonal antibody to the p40 subunit common to interleukin-12 (IL-12) and IL-23, which has shown efficacy in the treatment of moderate-to-severe psoriasis and psoriatic arthiritis [1]. It was first approved for treatment of plaque psoriasis in adults in the United States in 2009 and subsequently received approval to treat psoriatic arthritis in 2013 and psoriasis in adolescents in 2017. It is also approved in Europe and multiple other countries for psoriasis and psoriatic arthritis; and for use in psoriasis for ages 6 and up by the European Medicines Agency [2]. IL-12 and IL-23 are heterodimeric cytokines 201 202 which possess a common p40 subunit linked by a disulfide bond to a unique chain, IL-12p35 and IL-23p19, respectively [3, 4]. The p40 subunit binds to the IL-12 receptor-beta1 (IL-12Rb1) on the surface of T lymphocytes and natural killer cells. Interleukin-12 is a potent inducer of interferon-­ gamma (IFN-g), which promotes T-cell differentiation toward a Th1 lineage, while interleukin-23 is the major regulator of Th17 CD4 cells, a subset of T helper cells distinct from Th1 and Th2 cells, defined by their ability to produce IL-17 [5]. Interleukin-23 stimulates IL-17A, IL-17F and IL-22 production from Th17 cells and mediates its effects through Janus kinase-2 (JAK2) and STAT3, leading to hyperproliferation of keratinocytes and production of chemokines, angiogenic factors (VEGF) and pro-­inflammatory mediators such as tumor necrosis factor-alpha (TNF-a), nitric oxide and IL-1b [6, 7]. As psoriasis was originally thought to predominately be a Th1-cell mediated disease, ustekinumab was specifically developed to target IL-12; indeed, IL-23 was not even identified until after ustekinumab was submitted to the FDA for approval. The dichotomy of these two cytokines lies in their upregulation of Th1 cells (for IL-12) and Th17 cells (for IL-23) [6, 8]. Interestingly, it has since been shown that IL-12 may have little to do with the pathogenesis of psoriasis [9, 10] and actually inhibits IL-23 itself [11]. The concomitant inhibition of IL-23 was thus, a fortuitous result of targeting the p40 subunit [12, 13]. It is worthwhile to note that a similar monoclonal antibody targeting the p40 subunit, briakinumab, was under development but was withdrawn from clinical trials due to safety concerns relating primarily to adverse cardiovascular risk [14]. Pharmacokinetics In phase I studies in psoriasis, ustekinumab showed linear pharmacokinetics with both ascending single intravenous doses and ascending single subcutaneous doses of the drug [15]. After a single subcutaneous dose, ustekinumab was slowly absorbed, reaching maximum concentration (tmax) between 7 and 14 days [16]. The G. Han et al. absolute bioavailability (F) of ustekinumab was estimated to be 57.2% after a single subcutaneous dose, with an apparent volume of distribution of 79-161 ml/kg at the terminal phase [17]. Steady-state concentrations were achieved by week 28 in phase III studies in psoriasis, with trough steady-state serum concentrations (ctrough) showing dose proportionality. The median ctrough in those taking 90 mg every 12 weeks was twice that of those taking 45 mg in two phase III psoriasis studies (0.47 vs 0.21ug/ml in PHOENIX 1 and 0.49 vs 0.26ug/ml in PHOENIX 2), with no evidence of accumulation with either dosage regimen. The metabolic pathway of ustekinumab has not yet been fully elucidated. As a human IgG monoclonal antibody, ustekinumab is most likely degraded into small peptides and amino acids by the reticuloendothelial system in the same manner as endogenous IgG. A combined analysis of phase III studies in psoriasis calculated the mean half-life to be 21.6 days [18]. A population based approach was used to further characterize the pharmacokinetic profile of ustekinumab based on two phase III studies [1, 18, 19]. Mean values for apparent clearance, apparent volume of distribution and absorption rate constant were 0.465 L/ day, 15.7 L and 0.354/day, respectively. Based on the known bioavailability of ustekinumab, the volume of distribution of ustekinumab was calculated to be approximately 8.9 L in a 90 kg psoriasis patient, suggesting that ustekinumab is confined to the intravascular system, with limited tissue distribution. Factors influencing variation in apparent clearance and apparent volume of distribution included body weight, diabetes mellitus (independent of weight) and anti-drug antibodies to ustekinumab [18]. The most significant effect was caused by body weight, with an apparent clearance and apparent volume of distribution approximately 55% and 37% higher, respectively, in those with a bodyweight of greater than 100 kg compared with those of 100 kg or less. This emphasizes the importance of dose adjustment in those who have a higher bodyweight to achieve a similar efficacy. None of the 28 concomitant medications analyzed had a significant effect on the pharmacokinetic profile. 3.2% of patients developed antibodies to ustekinumab, 15 Ustekinumab which was associated with a mean increase in apparent clearance of 35.5% in these patients. In the US, the current licensed dosage regimen is 45 mg of ustekinumab at baseline, 4 weeks and every 12 weeks in those less than 100 kg in weight, and 90 mg of ustekinumab at the same intervals for those heavier than 100 kg. These dosage recommendations were made based on analyses of 10-Kg increments with a presumed ideal cutoff point to separate response between the lower and higher dose regimens [20]. 203 of these cytokines at week 1 compared to baseline, the mRNA expression of IL-8, IL-18 and IFN-γ was significantly decreased in the lesional skin of those who had a sustained response of at least 70% improvement in PASI at 3 separate time-points (week 8, 12 and 16), compared with those without sustained PASI improvement. The effects of ustekinumab on lesional skin and peripheral blood in a subset of psoriasis patients were examined in a phase II psoriasis study [22]. At week 12, a significant decrease in median epidermal thickness correlated with a reduction in cellular proliferation (Ki67) and Pharmacodynamics T-cell infiltration (CD3) by 84.3% and 70.7%, respectively, in the combined ustekinumab group. Two phase I studies have examined the pharma- Surprisingly, the level of IL-12p40, the target of codynamics of ustekinumab in lesional psoriatic ustekinumab, increased 13-fold from baseline to skin. The first examined the effect of intravenous week 12, before slowly decreasing to near baseustekinumab in 18 patients. There was a signifi- line levels by week 32. This was postulated to be cant reduction in the expression of IFN-g, TNF-­ due to decreased clearance of non-functional cirα, IL-8, IL-10, IFN-γ-inducible protein-10 and culating complexes of ustekinumab-bound monocyte chemotactic protein-1 (MCP-1) within IL-12p40. The expression of cutaneous lympho2 weeks of administration of the drug before clin- cyte antigen (CLA), which facilitates homing of ical response and histological changes were evi- activated T cells to the skin, significantly dent [21]. A significant reduction in total CD3+ T decreased from baseline in ustekinumab-treated cells was observed in responders (those achiev- patients compared with placebo, while there was ing a PASI-75 response) but not in non-­responders no change in expression of CD45RA, CD45RO, by week 2. Levels of TNF-α were significantly CXCR3, CD25 or HLA-DR on T cells. The sysreduced in responders but not in poor responders, temic effects of ustekinumab were also investiwhile levels of IL-12p40 and IL-23p19 were gated in vitro in healthy donors using isolated reduced in both populations, particularly in peripheral blood mononuclear cells in the absence responders. When baseline genetic expression of or presence of recombinant IL-12 or IL-23 [22]. responders was compared to that of non-­ Ustekinumab inhibited up-regulation of IL-12R, responders, mRNA expression of TNF-α was sig- IL-2Rα (CD25) and the co-stimulatory receptor nificantly higher in responders and correlated CD40L, while reducing IL-12 and IL-23-induced with the percentage improvement in PASI, sug- secretion of the pro-inflammatory cytokines IFN-­ gesting that this may be a predictor of treatment γ, TNF-α, IL-2 and IL-17A. response. The second phase I study examined the effect of subcutaneous ustekinumab in 21 patients. Punch biopsies were obtained from Clinical Efficacy lesional skin 24 hours before, and 1 week after IL-12/23 antibody administration, and the mRNA Two large scale phase 3, multicenter, randomexpression of various cytokines, including TNF-­ ized, double-blind, placebo-controlled, parallel α, IFN-γ, IL-8, IL-18, IL-12/23p40 subunit, studies (PHOENIX 1 and PHOENIX 2) were IL-23p19 subunit, IL-12p35 subunit, IL-10, performed to evaluate the subcutaneous adminisIP-10, RANTES and CCL-2 was measured using tration of ustekinumab in patients with moderate-­ real-time polymerase chain reaction. Although to-­severe psoriasis [1, 19]. In PHOENIX 1, 766 there was no significant change in the expression patients were randomized to receive 45 mg or 204 90 mg of ustekinumab at baseline, week 4, followed by every 12 weeks, or to receive placebo at week 0 and 4, with crossover to receive either 45 mg of ustekinumab or 90 mg of ustekinumab at week 12 [1]. Those in the initial ustekinumab group who achieved a sustained PASI-75 at both week 28 and 40 were re-randomized at week 40 to ongoing treatment or withdrawal from treatment for another 36 weeks until loss of response (weeks 40–76). These patients were retreated when they lost at least 50% of PASI improvement. At week 12, PASI-75 was achieved in 67.1% of those receiving 45 mg, and 66.4% of those receiving 90 mg of ustekinumab compared with 3.1% of those taking placebo, while PASI-­ 90 was achieved in 41.6%, 36.7% and 2% of these groups, respectively. Those receiving placebo, achieved similar response rates after crossover to active treatment. Maximum efficacy was achieved at week 24, with PASI-75 responses of 76.1% and 85% for the 45 mg and 90 mg ustekinumab groups, respectively. A significantly higher proportion of those receiving maintenance treatment maintained a PASI-75 response compared to those from whom treatment was withdrawn (p < 0.0001). Response rates stayed stable up to 76 weeks in the maintenance group, with a median time to loss of PASI-75 of 15 weeks. No rebound flare was observed on discontinuation of ustekinumab (defined as a PASI greater than 125% of baseline). Of the 195 patients who recommenced ustekinumab after losing response, 85.6% reestablished a PASI-75 response within 12 weeks. Patient-determined quality of life, as determined by DLQI, showed a similar improvement to objective assessments of disease severity, with median changes in ustekinumab-treated patients significantly greater than placebo-treated patients at week 12. A subsequent analysis showed that at 3 years, 79.8% of patients (601 of 753 patients) who received one or more dose of ustekinumab remained in the study and that 80.9% (45 mg) and 82.7% (90 mg) of week 40 responders continuing treatment every 12 weeks achieved a PASI-75 response [23]. In PHOENIX 2, a similar schedule was followed over 52 weeks, with a total of 1230 G. Han et al. patients randomized to receive 45 mg or 90 mg of ustekinumab or placebo at baseline, week 4, and then every 12 weeks or placebo at baseline and week 4, followed by a placebo crossover to receive 45 mg or 90 mg of ustekinumab (week 12–28) [19]. At week 28, partial responders (those who had achieved greater than PASI50 but less than PASI-75), were randomized to escalate dosing to every 8 weeks at their originally assigned dose, or to continue receiving ustekinumab every 12 weeks (week 28–52). At week 12, 66.7% of patients receiving 45 mg ustekinumab, and 75.7% receiving 90 mg ustekinumab achieved PASI-75, compared with 3.7% receiving placebo, while PASI-90 was achieved in 42.3%, 50.9% and 0.7% of these groups, respectively. There was also a highly significant improvement in DLQI in the ustekinumab treated patients compared with the placebo group at week 12 (p < 0.0001). Maximum PASI-75 response rates were attained at week 20 (74.9% of the 45 mg group and 83.5% of the 90 mg group). At week 28, 22.7% of patients in the 45 mg ustekinumab group were partial responders compared with 15.8% in the 90 mg group. Independent predictors of partial response included increased bodyweight, a history of non-­ response to biologic agents, longer duration of psoriasis and the presence of psoriatic arthritis. Partial responders had a significantly higher incidence of antibodies to ustekinumab (12.7%) compared to responders (2%), and serum trough drug levels were two to three times lower than those of responders. Escalation from a 12 weekly to an 8 weekly dosing regime resulted in a four to five times increase in the mean trough serum concentrations in partial responders between week 28 and week 52. In partial responders who received dosing intensification of ustekinumab 90 mg every 12 weeks to ustekinumab 90 mg every 8 weeks, there was a significant increase in those achieving PASI75, compared to those who remained at a dose of ustekinumab 90 mg every 12 weeks, with response rates of 68.8% and 33.3%, respectively. There was no significant difference in 15 Ustekinumab response, however, in those randomized to receive 45 mg of ustekinumab every 8 weeks, compared to those who continued to receive 45 mg every 12 weeks. Further data were gathered out to 5 years of follow-up, with patients randomized to either 45 mg or 90 mg groups where dosage and interval adjustments (from q12 weeks to q8 weeks; from 45 mg to 90 mg) were allowed at the discretion of the investigator. At week 244, the proportions of patients achieving PASI 75 were 76.5% and 78.6% for those who started in the 45 mg and 90 mg groups, respectively; PASI 90 was achieved in 50.0% and 55.5% of patients in the 45 mg and 90 mg groups, respectively [24]. The first study to directly compare the efficacy of two biologic agents in psoriasis was the ACCEPT study, a single-blind randomized, parallel phase III study of 903 psoriasis patients comparing the safety and efficacy of ustekinumab with etanercept [25]. Patients were randomized to receive ustekinumab 45 mg or 90 mg at baseline and at week 4 or etanercept 50 g twice weekly for 12 weeks. There was no placebo arm in this study. At week 12 PASI-75 was achieved in 67.5% of those receiving 45 mg ustekinumab, 73.8% of those receiving 90 mg of ustekinumab and 56.8% of those receiving etanercept, while PASI-90 was achieved in 36%, 45% and 23% of these groups, respectively. A PGA of cleared or minimal was achieved in 65.1%, 70.6% and 49% of the groups. Psoriatic Arthritis A randomized, double-blind, placebo-controlled, Phase III study of ustekinumab was performed in 615 patients with active psoriatic arthritis (PSUMMIT-1) [26]. Patients who previously used methotrexate and those who were using disease-­ modifying antirheumatic drugs (DMARDs) or NSAIDs were allowed in the study but patients with a history of TNF-α inhibitor use were not. The primary endpoint was ACR20 at week 24. At the end of 24 weeks, 22.8% patients on placebo achieved ACR20 compared to 42.4% and 49.5% 205 of patients on ustekinumab 45 mg and 90 mg, respectively. About half of the patients took methotrexate during the study, which did not influence the likelihood of attainment of ACR20 for ustekinumab vs. placebo, though the benefit was likely to be slightly greater in patients who were not using concomitant methotrexate. A later trial was conducted (PSUMMIT-2) which was a randomized, double-blind, placebo-­ controlled, Phase III study of ustekinumab in psoriatic arthritis, with the notable difference from PSUMMIT-1 that patients who had used TNF-α inhibitors previously were now allowed into the study [27]. A total of 312 patients were studied, including 180 with prior exposure to TNF-α inhibitors. For TNF-α-naïve patients, ACR20 rates were similar to PSUMMIT-1—with 43.7% and 43.8% of patients achieving this threshold for ustekinumab 45 mg and 90 mg, respectively. However, the response rates for patients who had previously been on TNF-α inhibitors was somewhat lower—36.7% and 34.5% for ustekinumab 45 mg and 90 mg, respectively. Notably, the study was not powered to assess for a statistically significant difference in response rates between the two groups (TNF-α experienced or naïve). Also interestingly, the patients’ skin disease seemed to track along with the psoriatic arthritis response, where patients previously exposed to TNF-α inhibitors experienced a little over 10% lower achievement of PASI-75 than patients who had never been on that class of biologic. Adolescent Patients A randomized, double-blind, placebo-controlled Phase III study was completed in adolescent patients from 12–17 years of age [28]. The study was conducted with a primary endpoint of PGA 0/1 and PASI 75 at week 12, with a long-term extension to 1 year. A total of 110 patients were studied and assigned to different dosing regimens, including a half-dose cohort. Patients under 60Kg received 0.75 mg/Kg in the standard dose (SD) group or 0.375 mg/Kg in the half-­ G. Han et al. 206 standard dose (HSD) group. Patients from greater than 60 Kg to 100 Kg received 45 mg (SD) or 22.5 mg (HSD) and patients greater than 100 Kg received 90 mg (SD) or 45 mg (HSD). Overall, 78.4% of patients in the HSD and 80.6% in the SD group achieved PASI 75 compared to 10.8% for placebo. 54.1% of patients in the HSD group achieved PASI 90 compared to 61.1% of patients in the SD group and 5.4% of patients in the placebo group. In the placebo-controlled period (through 12 weeks), 56.8% of placebo, 51.4% of HSD, and 44.4% of SD patients reported adverse events. No major safety issues were observed different from the side effects seen from ustekinumab in adults. Based on this data, the FDA approved ustekinumab for patients above 12 years of age at the standard dosing regimen. Adverse Effects There is a good deal of safety data for ustekinumab available from a pooled longitudinal analysis of phase II and phase III clinical trials in moderate-­to-­severe psoriasis [29–31] as well as longer term registry databases, including the Psoriasis Longitudinal Assessment Registry (PSOLAR). Initial safety data was based on all safety data available from the Phase 2 study, PHOENIX 1 and 2 and the ACCEPT study, with a total of 5 years’ of safety data. A total of 3117 patients had been followed for up to 4 years (6791 patient-­ years). During the combined 12 week placebo-­controlled period of these studies, 50.4%, 57.6% and 51.6% of patients treated with placebo, 45 mg of ustekinumab and 90 mg of ustekinumab, respectively, experienced at least one adverse event, while 1.4%, 1.6% and 1.4% of patients in these groups experienced a serious side effect and 1.9%, 1.1% and 1.4% were withdrawn from the study due to adverse effects. The most common side effects were nasopharyngitis, upper respiratory tract infections, headache and arthralgias. Rates of adverse events, serious adverse events, infections or adverse events requiring discontinuation of treatment did not increase with increased duration of treatment and were comparable between ustekinumab doses. Serious Infections Based on animal models of cytokine deficiency and patients with genetic mutations encoding IL-12 and IL-23 or their receptors, there was particular concern about a theoretical increase in the risk of serious infections or malignancy with the advent of these agents. In animal models, IL-12 has been shown to be important in prevention against mycobacteria, salmonellosis and toxoplasmosis and patients with a genetic deficiency of the IL-12 receptor or the IL-12p40 subunit have an increased risk of severe infections with intracellular pathogens such as mycobacteria and salmonella [32–36]. Experimental animal models of herpes viral infection, viral encephalitis, viral hepatitis and aquired immunodeficiency syndrome (AIDS), have also demonstrated a central role for IL-12 in protecting against viral infection [37–40]. Interleukin-12 also appears to be important in defense against opportunistic fungal infections, with increased susceptibility to Cryptococcus neoformans infection observed in p35−/− or p40−/−knockout mice and prevention of infection following the administration of IL-12 [41, 42]. Similarly, IL-12p40−/− knockout mice did not control fungal proliferation and dissemination and succumbed to infection following intravenous inoculation of yeast cells of paracoccidioidomycosis. Interleukin-23 is also believed to contribute to immune protection in the skin, lung and gut. Interleukin-23 and IL-17, however, have been shown to be only marginally involved in primary infections with pathogens that require TH1 immunity [43–47]. One study showed that blocking IL-23 alone does not increase bacterial burden in immunocompetent mice after BCG infection but that blocking TNF-α or the p40 subunit results in increased infectious burden [48, 49]. Interleukin-­ 23p19 deficient mice showed significant mortality following a sub-lethal dose of intrapulmonary Klebsiella pneumoniae, however, while IL-12p40-deficient, IL-12p35- deficient and IL-17R-deficient mice also show increased susceptibility to infection following inoculation [50]. Administration of IL-17 restored the normal infectious response in IL-23p19-deficient mice, 15 Ustekinumab 207 but not completely in p40-deficient mice, sug- 55]. Patients with congenital deficiencies of gesting the additional role of IL-12-induced IL-12p40 or IL-12R, however, do not appear to IFN-g production in the immune defense to have an increased incidence of malignancy [35]. Klebsiella infection. Another experimental study Interleukin-23, in contrast, appears to promote showed that IL-23 plays a critical role in the host tumor growth, suggesting that inhibition of this defense to Pneumocystis Carinii [51]. Individuals cytokine may inhibit carcinogenesis [56]. with abnormalities of Th17 cell function, such as There were no differences in the incidence patients with hyper-IgE (Job’s) syndrome and of non-melanoma skin cancer (NMSC) or other chronic mucocutaneous candidiasis, are more malignancies during the placebo-controlled prone to infection with Staphylococcus aureus phases of usekinumab studies [31]. The rate of and candida albicans. NMSC was 0.7/100PY in the 45 mg group and However, clinical studies to date have not 0.53/100PY in the 90 mg group (34 BCC and shown an increase in serious infections with the 10 SCC) and higher among patients previously use of ustekinumab [31]. In the pooled analysis, treated with psoralen-UVA [31, 57]. Long-term the frequency of infection during the placebo-­ follow-up data showed rates of malignancies controlled period of the studies in patients receiv- other than NMSC of 0.63/100PY in the 45 mg ing placebo, 45 mg of ustekinumab or 90 mg group and 0.61/100PY in the 90 mg group, with of ustekinumab, was 23.2%, 27% and 24.1%, a total of 42 malignancies over the course of the respectively, while the rates of serious infection studies [31]. These rates were consistent with were similar between the placebo (1.70/100PY) age-, race- and gender-matched rates expected in and 90 mg (1.97/100PY) groups but lower in the the normal United States population according to in the 45 mg group (0.49/100PY) [31]. The rates the Surveillance, Epidemiology, and End Results of overall infection, serious infection and infec- (SEER) database. Furthermore, analysis of the tions requiring antibiotic treatment remained PSOLAR database showed no increased maligstable or decreased over the 4 years, with rates nancy risk for ustekinumab in matched-control serious infection of 0.8/100 patient years (PY) cohort. and 1.32/100PY for patients treated with 45 mg and 90 mg of ustekinumab respectively and were consistent with expected rates in psoriasis patients Major Adverse Cardiovascular Events using the Marketscan Database. No cases of tuberculosis were reported in the pooled analysis, The withdrawal of a similar biologic treatment but one man who had a previously negative puri- mechanistically, briakinumab, from clinical studfied protein derivative and QuantiFERON-TB ies generated some interest and concern regardGold screening tests, had a tuberculosis reactiva- ing the potential adverse cardiovascular effects of tion after two doses of ustekinumab in a RCT of anti-IL-12p40 agents. A total of five MACE Asian psoriasis patients [52]. (0.3%), including one cardiovascular death, were reported in 1582 ustekinumab-treated patients compared with no events in 732 placebo recipients (0%) during the placebo-controlled phases Malignancy of the pooled analysis [31]. All of these events Animal studies have also suggested increased occurred in patients with three or more cardiotumorigenicity following inhibition of IL-12 vascular risk factors. All cardiovascular events [53–55]. Interleukin-12 has shown anti-tumor were re-adjudicated externally at the four-year and anti-metastatic activity in murine tumor follow-up safety analysis and a total of 34 MACE models of melanoma, renal cell carcinoma and were identified, with four cardiovascular deaths breast cancer, while IL-12 deficient mice have an [31]. The rate of MACE was 0.56/100PY and increased incidence of UVB-induced skin tumors 0.46/100PY in the 45 mg and 90 mg groups, and malignant transformation of papillomas [53– respectively. This was reported to be within the 208 G. Han et al. expected range of such events for both the gen- showed a significantly higher risk of MACE in eral US population and the psoriatic population, patients treated with anti-IL-12/23 agents comleading the sponsoring company to conclude that pared with placebo (odds ratio = 4.23, p = 0.04). there was no apparent change in cardiac risk with The discrepancy between these meta-analyses this agent. However, it is clear that numerically, a results from the use of different statistical methhigher number of MACE were observed in the ods to compare MACE rates. Risk difference is placebo-controlled period in the ustekinumab-­ considered to be a more conservative measure treated cohort compared to control. in comparing rare events but has the potential to Two subsequent meta-analyses have been introduce false negative results. This was chosen performed to evaluate the effect of anti-IL-12/23 over the Peto odds ratio method by the authors agents on cardiovascular risk [14, 58]. The first of the first study because 16 of the 22 studies was an independent meta-analysis of 22 random- had zero events and would have been excluded ized controlled trials (RCTs) comprising 10,183 from the analysis if the odds ratio was used, patients to evaluate the effect of biologic agents possibly leading to selection bias [59]. A later on cardiovascular risk in psoriasis patients [14]. meta-­analysis of a number of biologics including Double-blind, placebo-controlled RCTs of anti-­ ustekinumab also used the Peto odds ratios, and IL12p40 agents (ustekinumab and briakinumab) concluded that there was not a significant risk of and TNF-a inhibitors (infliximab, etanercept and MACE with ustekinumab [60]. Somewhat reasadalimumab) were compared and absolute risk suringly, further studies of ustekinumab use in the differences were used as an effect measure, mea- real world have not shown a significant increase suring the excess probability of major adverse in MACE; a cohort study of over 60,000 patients cardiovascular events (MACE, a composite end- from multiple databases was studied and no sigpoint of myocardial infarction, cerebrovascular nificant difference was found in the risk of develaccident, or cardiovascular death) in those receiv- oping atrial fibrillation or MACE after initiation ing active treatment compared to those receiving of therapy with ustekinumab vs. a TNF-α inhibiplacebo. During the placebo-controlled phases tor [61]. While there is some disagreement in the of the anti-IL-12p40 studies, 10 of the 3179 literature, the majority of currently-­available evipatients treated with anti-IL-12p40 therapies dence—after over a decade on the market—suphad a MACE compared with zero events in 1474 ports that there is no significant safety signal with patients treated with placebo (risk difference 1.2 MACE for ustekinumab. events/100PY, p = 0·12). In anti-TNF-a trials, only one of 3858 patients treated with anti-TNF­a treatments had a MACE compared with one of Other Adverse Events 1812 treated with placebo (risk difference 0.05 events/100PY, p = 0.94). Although the apparent Although there was initial concern that IL-12 increase in MACE observed with patients receiv- blockade could result in skewing towards a Th2 ing anti-IL-12p40 antibodies was not statistically type cytokine profile, there was no evidence of significant, the findings did raise questions about exacerbation of any atopic disease in these studthe cardiovascular safety of the anti-IL-12p40 ies. No cases of demyelination were reported agents and the authors recommended heightened apart from a possible case in a patient who was vigilance when commencing patients with car- retrospectively diagnosed with human immunodiovascular risk factors on ustekinumab. The sec- deficiency virus and profoundly lymphopenic at ond meta-analysis examined the rate of MACE the time. Two cases of reversible posterior leukein patients in randomized, placebo-controlled, ncephalopathy syndrome were reported as well; monotherapy studies of IL-12/23 antibodies one in a 65 year old with a history of alcohol using the Peto one-step odds-ratio, an alterna- abuse who recovered fully from the episode and a tive statistical method, as an effect measure. This 58 year old with a positive urine drug screen for 15 Ustekinumab MDMA, benzodiazepenes, barbiturates, and marijuana who also recovered fully [62, 63]. Laboratory Abnormalities In phase I studies of ustekinumab in psoriasis, transient asymptomatic decreases in CD4+ T cells and CD16+/CD56+ natural killer cells were observed in some patients but this was not seen in phase II or III studies [15, 16]. In phase II and III studies in psoriasis and in psoriatic arthritis, the incidence of haematological and biochemical abnormalities was low and similar between ustekinumab-treated and placebotreated patients. In the phase II study of psoriasis, however, there was a non-significant trend of elevated non-fasting glucose levels in ustekinumabtreated patients (10%) compared with controls (4%) (p = 0.23) [64]. This was not observed in subsequent phase III studies in psoriasis and ustekinumab was shown to have no effect on haemoglobin A1c levels. Pregnancy and Lactation Ustekinumab is pregnancy category B. The product prescribing information states that ustekinumab should be used during pregnancy only if the potential benefit justifies the potential risk to the fetus [65]. Toxicology studies on cynomolgus monkeys revealed no maternal or fetal abnormalities following administration of intravenous and subcutaneous doses of up to 50 mg/kg during the period of organogenesis [65]. Post-­marketing data is an important source of information for pregnancy risks and outcomes. A publication from 2019 reported the outcomes of 478 maternal pregnancies across all indications of ustekinumab (including Crohn’s disease and ulcerative colitis)—the majority resulted in live births and the rates of live births, spontaneous abortion, and congenital anomalies were consistent with the general population [66]. The product prescribing information for ustekinumab advises caution in administration 209 of ustekinumab to nursing mothers, where the unknown risks to the infant from gastrointestinal exposure to ustekinumab should be weighed against the known benefits of breast-feeding [65]. Ustekinumab is excreted in the milk of lactating monkeys, and as endogenous IgG is excreted in human milk, it is expected that ustekinumab will be also be present. It is not known if ustekinumab would be absorbed systemically through the immature neonatal gastrointestinal tract after ingestion. Cautions for Patients Treated with Anti-IL-12p40 Agents There are no absolute contraindications to treatment with ustekinumab [65]. Treatment should be deferred in patients with clinically important active infections until the infection resolves or is appropriately treated [65]. No increase in serious infections has been observed with the use of ustekinumab, however, despite initial concerns regarding longer half-life of the drug [31]. Patients with evidence of active or latent tuberculosis should be treated with anti-tuberculosis treatment prior to initiating treatment [65]. Current evidence does not support use or avoidance of ustekiniumab within populations at risk for major adverse cardiovascular events. Ustekinumab has no currently known drug interactions. Patients should receive all age-­ appropriate immunizations recommended by current guidelines prior to commencing ustekinumab and live vaccines should not be administered during treatment. The Bacillus Calmette-Guérin (BCG) vaccine should not be administered for 1 year prior to or after discontinuation of treatment [65]. Conclusions The field of psoriasis research has been revolutionized by the increased understanding of the pivotal role of the Th-17/IL-23 axis in disease pathogenesis. As the first non-TNF-α inhibiting biologic for the treatment of psoriasis, ustekinumab signaled 210 G. Han et al. the arrival of new therapeutic options for psoria- References sis patients, offering more targeted treatment with 1. Leonardi CL, Kimball AB, Papp KA, et al. Efficacy better efficacy and favorable side effect profiles. and safety of ustekinumab, a human interleukin-12/23 Ustekinumab, the only licensed anti-IL-12p40 monoclonal antibody, in patients with psoriasis: agent, still plays an important role in the current 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1). Lancet. therapeutic armamentarium for psoriasis although 2008;371:1665–74. newer agents focused on inhibiting only IL-23 are 2. See URL: http://www.clinicaltrials.gov/ct2/results? available. The infrequency of dosing afforded by term=psoriasis; last accessed 21 June 2012. the long half-life of the drug is particularly appeal3. Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev ing to patients. Adverse events in clinical studies to Immunol. 2003;3:133–46. date have been for the most part, mild and similar 4. Oppmann B, Lesley R, Blom B, et al. Novel p19 proto that in placebo-treated patients. Long-term post-­ tein engages IL-12p40 to form a cytokine, IL-23, with marketing data has not revealed any significant biological activities similar as well as distinct from IL-12. Immunity. 2000;13:715–25. concerns for the use of ustekinumab, including in 5. Yao Z, Painter SL, Fanslow WC, et al. Human IL-17: pregnant patients. The approval of ustekinumab in a novel cytokine derived from T cells. J Immunol. adolescents is another strong selling point of the 1995;155:5483–6. medication, speaking to its efficacy, but also bring6. Nestle FO, Kaplan DH, Barker J. Psoriasis. N Engl J Med. 2009;361:496–509. ing a better dosing regimen to this population of 7. Lowes MA, Kikuchi T, Fuentes-Duculan J, et al. young adults who certainly appreciate that aspect Psoriasis vulgaris lesions contain discrete populaof treatment. Long-term follow-up safety data and tions of Th1 and Th17 T cells. J Invest Dermatol. the use of registries are important in examining the 2008;128:1207–11. 8. Nestle FO, Turka LA, Nickoloff BJ. Characterization incidence of cutaneous and non-­cutaneous maligof dermal dendritic cells in psoriasis. Autostimulation nancy in ustekinumab-treated patients, and curof T lymphocytes and induction of Th1 type cytorent evidence points to no risk with ustekinumab. kines. J Clin Invest. 1994;94:202–9. Taken together, there continues to be a role for 9. Lee E, Trepicchio WL, Oestreicher JL, et al. Increased expression of interleukin 23 p19 and p40 in lesional ustekinumab in our armamentarium for treating skin of patients with psoriasis vulgaris. J Exp Med. psoriasis and psoriatic arthritis given its favour2004;199:125–30. able dosing regimen, proven long-term safety track 10. Chan JR, Blumenschein W, Murphy E, et al. record, and reliable efficacy. IL-23 stimulates epidermal hyperplasia via TNF Conflicts of Interest G Han has served as an consultant, advisor, speaker, or investigator for Abbvie, Athenex, Boehringer Ingelheim, Bond Avillion, Bristol-­Myers Squibb, Celgene, Eli Lilly, Novartis, Janssen, LEO Pharma, MC2, Ortho Dermatologics, PellePharm, Pfizer, Regeneron, Sanofi Genzyme, SUN Pharmaceuticals, and UCB. C Ryan has served as a speaker, advisory board member, or investigator for Jansen-Cilag, Pfizer, Galderma, and Abbott. C Leonardi has served as a consultant, investigator, or speaker for: Abbott, Amgen, Anacor, Celgene, Janssen, Eli Lilly, Galderma, Glaxo Smith Kline, Incyte, Maruho, Merck, Pfizer, Sandoz, Schering-Plough, Sirtris, Stiefel, Leo, Novartis, Tolmar, Novo Nordisk, Vascular Biogenics, Warner Chilcott and Wyeth. and IL-20R2-dependent mechanisms with implications for psoriasis pathogenesis. J Exp Med. 2006;203:2577–87. 11. Langrish CL, Chen Y, Blumenschein WM, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med. 2005;201:233–40. 12. Murphy CA, Langrish CL, Chen Y, et al. Divergent pro- and antiinflammatory roles for IL-23 and IL-12 in joint autoimmune inflammation. J Exp Med. 2003;198:1951–7. 13. Cua DJ, Sherlock J, Chen Y, et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature. 2003;421:744–8. 14. Ryan C, Leonardi CL, Krueger JG, et al. Association between biologic therapies for chronic plaque psoriasis and cardiovascular events: a meta-analysis of randomized controlled trials. JAMA. 2011;306: 864–71. 15. Kauffman CL, Aria N, Toichi E, et al. A phase I study evaluating the safety, pharmacokinetics, and clinical response of a human IL-12 p40 antibody in 16 Guselkumab Deep Joshipura, Brooke Rothstein, and David Rosmarin Abstract Introduction Guselkumab is a fully human IgG antibody to the p19 subunit of interleukin-23 (IL-23). Guselkumab has potent efficacy in the treatment of plaque psoriasis with a favorable safety profile, providing patients with a sustained response for 3 years in long term studies. In addition, there is growing evidence that guselkumab is effective at treating psoriatic arthritis. This review discusses the efficacy and safety of guselkumab in the treatment of plaque psoriasis and provides an update on the published data available from clinical trials. Guselkumab (Janssen Pharmaceuticals, Inc., Philadelphia, PA) is a fully human monoclonal IgG1λ antibody that selectively binds to the p19 subunit of interleukin 23 (IL-23) [1] (Fig. 16.1). It is currently FDA approved for the treatment of moderate-to-severe plaque psoriasis. IL-23 is a heterodimeric cytokine composed of subunits p40 and p19 and found at high levels in the serum and plaques of patients with psoriasis. Ustekinumab (Janssen Pharmaceuticals, Inc., Philadelphia, PA) binds to the p40 subunit which is common to both IL-12 and IL-23. In contrast, guselkumab binds to the p19 subunit only, and therefore IL-12 is not suppressed. IL-12 is a promoter of T-cell differentiation to Th1 cells whereas IL-23 promotes the development of Th17 cells which produce IL-17A, IL-17F, and IL-22 [1]. These cytokines are crucial in the pathogenesis of psoriasis and signal through the Janus kinase (JAK) and signal transducers and activators of transcription (STAT) pathway. Key Learning Objectives 1. To understand the mechanism of action of Guselkumab 2. To understand the appropriate use and efficacy of the Guselkumab 3. To understand the safety issues of Guselkumab Structure and Mechanism D. Joshipura · B. Rothstein · D. Rosmarin (*) Tufts Medical Center, Boston, USA e-mail: djoshipura@tuftsmedicalcenter.org; BRothstein@tuftsmedicalcenter.org; drosmarin@ tuftsmedicalcenter.org © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_16 Pharmacodynamics and Pharmacokinetics Structurally guselkumab is a human monoclonal IgG1λ antibody that selectively binds to the p19 213 D. Joshipura et al. 214 IL-23 Guselkumab p19 Dendritic cells T17 cell p40 a IL-17A IL-17F Keratinocyte proliferation b T22 cell IL-22 IL-23 Receptor Macrophages Naive T cell membrane When guselkumab binds to the p19 subunit of IL-23, and blocks its interaction with its receptor, the IL-23- mediated signaling pathway and release of pro-inflammatory cytokines are consequently blocked. Fig. 16.1 The mechanism of action of guselkumab in plaque psoriasis. Release of IL-23 by activated dendritic cells and macrophages. Once IL-23 binds to its cell surface receptor, it triggers differentiation, proliferation and survival of T cells subsets. T17 cells include Th17, Tc17, ILC3 and γδ T cells (reprinted with permission from Springer nature) subunit which is shared by the cytokines interleukin 23 (IL-23), and IL-39 [2]. IL-23 is a heterodimeric cytokine composed of p19 and p40 secreted by activated dendritic cells and macrophages. Once activated through interaction with its IL-23 receptor on T-lymphocytes, it induces differentiation of Th-17 and Th-22 cells (Fig. 16.1). Activation of Th-17 cells leads to secretion of IL-17 and IL-22 causing epidermal hyperplasia and inflammation in psoriasis [3]. In patients with psoriasis, administration of guselkumab reduced serum levels of IL-17A, IL-17F and IL-22 relative to pre-treatment levels on exploratory analysis of the pharmacodynamic markers [2]. The role of IL-39 in psoriasis, if any, is unclear. The pharmacokinetics, pharmacodynamics, safety, and tolerability of guselkumab were analyzed in a phase I clinical trial of 24 patients with moderate-to-severe plaque psoriasis and 47 healthy controls [4]. The healthy participants were administered a single intravenous (0.03– 10 mg/kg) or subcutaneous (10–300 mg) dose of guselkumab, and the subjects with psoriasis were administered a single subcutaneous dose of placebo or guselkumab (10, 30, 100, 300 mg). Guselkumab exhibited linear pharmacokinetics for both intravenous and subcutaneous doses tested in healthy participants and subjects with psoriasis. Mean terminal half-life (t1/2) was 15–17 days in patients with psoriasis and 12–19 days in healthy controls. Both the intravenous and subcutaneous forms of administration were well tolerated in this study. Following subcutaneous administration of guselkumab 100 mg at 0, 4 and every 8 weeks, the mean guselkumab steady-state trough serum concentration was approximately 1.2 mcg/mL [2]. After a single 100 mg subcutaneous injection, the absolute bioavailability was estimated to be approximately 49% in healthy subjects. The exact pathway through which guselkumab is metabolized is not well characterized. As a human IgG monoclonal antibody, guselkumab is expected to be degraded similar to endogenous IgG into small peptides and amino acids [2]. Development of anti-drug antibodies occur due to normal immune response to a foreign protein. Formation of anti-drug antibodies can affect half-life, increase elimination of the drug, and decrease efficacy. A study done by Zhu et al., evaluated anti-drug antibodies (ADA) through 100 weeks of drug exposure from the pivotal VOYAGE 1 and VOYAGE 2 trials. It was determined that 8.5% of subjects in these trials had anti-drug antibodies [5]. Most of these patients had low titers and the anti-drug antibodies were often transient, with serum guselkumab concen- 16 Guselkumab trations being comparable between ADA+ and ADA- patients. Anti-drug antibodies did not affect the efficacy of guselkumab. The incidence of injection site reactions was low in patients with and without anti-drug antibodies. Clinical Efficacy Guselkumab has been FDA approved since 2017 for the treatment of moderate-to-severe plaque psoriasis in patients who are candidates for systemic therapy or phototherapy. This approval was based on efficacy demonstrated in three large pivotal phase III clinical trials VOYAGE 1 [6], VOYAGE 2 [7], and NAVIGATE [8]. The findings above were also supported by other trials such as a phase I pilot study of guselkumab by Sofen H et al. [9], a phase II (X-PLORE) [10] trial, and a phase III trial conducted in Japan [11]. In a phase I trial of 24 plaque psoriasis patients who were randomized to placebo or guselkumab 10, 30, 100 or 300 mg, PASI75 was achieved at week 12 in 50% of patients on guselkumab 10 mg, 60% on 30 mg, 60% on 100 mg, and 100% on 300 mg. On skin immunohistochemistry, there was a reduction in epidermal thickness as well as T-cell and dendritic cell infiltration. There were also serum reductions in IL-17A levels in responsive patients [12]. In the phase II trial [10] of 293 moderate-to-­ severe psoriasis patients randomized to receive guselkumab (5 mg at weeks 0 and 4 and every 12 weeks thereafter, 15 mg every 8 weeks, 50 mg at weeks 0 and 4 and every 12 weeks thereafter, 100 mg every 8 weeks, or 200 mg at weeks 0, 4 and every 12 weeks thereafter), placebo, or adalimumab, significant improvement was observed with guselkumab. At week 16, the proportion of patients with a PGA score of 0 or 1 was 34%, 61%, 79%, 86%, and 83% in the guselkumab groups, respectively, 7% in the placebo group, and 58% in the adalimumab group. The proportion of patients achieving a PASI75 at week 16 was 44%, 76%, 81%, 79%, and 81% in the guselkumab groups, respectively, 5% in the placebo group, and 70% in the adalimumab group. The proportion of patients achieving a PASI90 at 215 week 16 was 34%, 34%, 45%, 62%, and 57% in the guselkumab groups, respectively, 2% in the placebo group, and 44% in the adalimumab group. Guselkumab was effective through week 40; however, there was some loss of efficacy right before the scheduled dose of guselkumab when the injection was dosed every 12 weeks. Guselkumab was well-tolerated and adverse events were similar among the treatment groups. This successful phase II study showed promising evidence of the clinical efficacy of IL-23 inhibitors for the treatment of plaque psoriasis leading to further studies in a phase 3 program. VOYAGE 1 VOYAGE 1 was a phase III, randomized, double-­ blind, placebo- and active comparator- controlled multinational trial (n = 837) conducted to confirm the efficacy and safety of guselkumab compared to adalimumab and placebo for the treatment of plaque psoriasis [6]. Patients ≥18 years old who were candidates for systemic therapy or phototherapy with moderate-to-severe plaque psoriasis (characterized by Investigator Global Assessment (IGA) score ≥ 3, Psoriasis Area and Severity Index (PASI) score ≥ 12, and body surface area (BSA) ≥ 10) for at least 6 months were included. Excluded patients were those with severe progressive medical conditions or cancers (except nonmelanoma skin cancer) diagnosed within 5 years, history of active tuberculosis, and those who had received guselkumab or adalimumab at any time. Patients were randomized to receive guselkumab, adalimumab, or placebo for 16 weeks; at week 16 the patients on placebo were crossed-­ over to receive guselkumab through week 48. Co-primary endpoints were the proportions of patients achieving an IGA score of clear/minimal disease (IGA 0/1) and 90% or greater improvement in PASI score from baseline (PASI90) at week 16 [6]. Clinical response of guselkumab was demonstrated against placebo for all the co-primary and major secondary endpoints (all p < 0.001). At week 16, 85.1% of patients in the gusel- 216 kumab group achieved an IGA score of 0/1 compared to 6.9% for placebo. Additionally, at this same timepoint, 73.3% of patients in the guselkumab group achieved PASI90 compared to 2.9% in the placebo group. At week 16, guselkumab was superior to adalimumab in IGA 0/1 scoring (85.1% versus 65.9%), PASI 90 scoring (73.3% versus 49.7%), and achievement of PASI 75 (91.2% versus 73.1%) [6]. Guselkumab was also superior to adalimumab at week 24 in IGA 0/1 (84.2% versus 61.7%) and PASI90 (80.2% versus 53.0%) scoring. The superior efficacy of guselkumab was also observed at week 48 with a significantly higher proportion of guselkumab patients achieving scores of IGA 0/1 (80.5% versus 55.4%) and PASI90 (76.3% versus 47.9%). Patients randomized to placebo achieved similar results to the guselkumab group once they were crossed over [6]. D. Joshipura et al. and then guselkumab was re-initiated in the placebo group once 50% clinical response was lost. Adalimumab responders were switched to placebo at week 28 and guselkumab was initiated once 50% clinical response was lost, whereas adalimumab non-­responders stopped treatment at the end of week 23 and guselkumab was initiated at week 28 [7] (Table 16.1). The co-primary end points were the proportions of patients on guselkumab achieving IGA 0/1 and PASI90 at week 16 as compared to ­placebo. At week 16, guselkumab was superior to placebo with higher IGA 0/1 scoring (84.1% versus 8.5%) and PASI90 scoring (70.0% versus 2.4%) (Table 16.2). Similar to VOYAGE I, guselkumab was superior to adalimumab at week 16 both in IGA 0/1 (84% versus 68%) and PASI90 scoring (70.0% versus 46.8%). From weeks 28 to 48, better persistence of response was observed in the guselkumab maintenance versus withdrawal group, with 88.6% of patients in the mainVOYAGE 2 tenance group sustaining a PASI90 response compared to 36.8% of those in the withdrawal VOYAGE 2, a phase III, multicentered, random- group (p < 0.001). The median time to loss of ized, double-blind, placebo- and adalimumab PASI90 response was 23 weeks after the last comparator-controlled study (n = 992), evaluated guselkumab dose. Of adalimumab non-­ the efficacy and safety of interrupted guselkumab responders who switched to guselkumab, 66.1% treatment, as treatment gaps frequently occur in achieved PASI90 by the end of the study [7]. clinical practice [7]. In addition, VOYAGE 2 In a pooled analysis of VOYAGE 1 and assessed the transition from adalimumab to VOYAGE 2, guselkumab was effective at treating guselkumab, providing clinically relevant infor- disease-specific areas such as the scalp, hands, mation about patients who switch biologics. The and fingernails [15]. inclusion/exclusion criteria were similar to At week 16, significantly more patients on VOYAGE 1 as stated above. The study consisted guselkumab compared to placebo achieved a of a placebo-controlled period (weeks 0–16), an scalp-specific Investigator Global Assessment active comparator-controlled period (weeks (ss-IGA) score of 0 or 1 (81.8% versus 12.4%). 0–28), and a randomized withdrawal and re-­ At week 16, 75.5% of patients on guselkumab treatment period (weeks 28–72). Patients were achieved a Physician Global Assessment of hands initially randomized to guselkumab 100 mg at and/or feet (hf-PGA) score of 0 or 1 compared to weeks 0, 4, 12, and 20; placebo at weeks 0, 4, and 14.2% of placebo patients. Guselkumab was 12, then guselkumab at weeks 16 and 20; or superior to adalimumab at week 24 in achieving adalimumab 80 mg at week 0, 40 mg at week 1, a score of 0 or 1 in both ss-IGA (85% versus and every 2 weeks thereafter through week 23 68.5%) and hf-PGA (80.4% versus 60.3%). At (Fig. 16.2 and Table 16.1). At week 28, patients week 16, more patients on guselkumab than plawho achieved PASI90 on guselkumab were re- cebo achieved a fingernail Physician Global randomized to continue guselkumab or start pla- Assessment (f-PGA) score of 0 or 1 (46.7% vercebo (to simulate an interruption in treatment) sus 15.2%). Similar proportions of patients 16 Guselkumab 217 Active-comparator period Placebo-controlled period Randomized withdrawal and retreatment period Placebo-crossover period Placebo → retreatment R Continue guselkumab 100 mg q8w Guselkumab 100 mg weeks 0, 4. then q8w NR Continue guselkumab 100 mg q8w R R Placebo Guselkumab 100 mg Placebo → retreatment NR Continue guselkumab 100 mg q8w R Adalimumab 80 mg at week 0 → 40 mg at week 1, then q2w Week 0 R 16 PE SE = Randomization q2w = every 2 weeks Responders PE = Primary endpoint Placebo → guselkumab 100 mg 0, 4, then q8w* NR Initiate guselkumab 100 mg 0, 4, then q8w* 24 SE 28 48 SE SE = Secondary endpoint q8w = every 8 weeks R = 90% or greater improvement in Psoriasis Area and Severity Index (PASI 90) Nonresponders NR = < PASI 90 response Fig. 16.2 Schema showing randomization of patients at baseline in VOYAGE II (reprinted with permission from K. Reich et al) achieved an f-PGA of 0 or 1 on guselkumab and adalimumab (60.0% and 64.3% p = 0.11). Improvement in difficult-to-treat areas such as the scalp, palms, and soles, with about 70% achieving complete clearance, led to significant improvement in health-related quality of life. In a pooled analysis of the 1829 patients from VOYAGE 1 and VOYAGE 2, guselkumab was superior to placebo and adalimumab in almost all subpopulations of patients who were stratified based on age, gender, weight, body mass index, ethnicity, baseline disease features, and previous treatments [16]. Guselkumab showed a more consistent clinical response across all weight quartiles compared to adalimumab. In patients in the highest weight quartile of >100 kg, 78.3% of guselkumab patients achieved an IGA of 0 or 1 compared to only 45.2% of adalimumab patients. Improvements in the Dermatology Life Quality Index (DLQI) were greater for guselkumab versus placebo at week 16 and for guselkumab versus adalimumab at week 24 [13]. The proportion of patients who achieved a DLQI of 0 or 1, indicating no impact on quality of life, at week 24 was higher with guselkumab compared to adalimumab (58.9% versus 40.2%). D. Joshipura et al. 218 Table 16.1 Dosing regimens from phase 3 VOYAGE 1 and VOYAGE 2 trials permission (reprinted with permission from Springer nature) Trial VOYAGE 1 [13] Active comparator (ADA) period PL-controlled period (wks 0–16) GUS wks 0 and 4 → q8w PL wks 0, 4 and 12 ADA 80 mg wk 0, 40mg wk 1 → 40 mg q2w VOYAGE 2 [14] GUS wks 0 and 4 → q8w PL wks 0, 4 and 12 ADA 80 mg wk 0, 40mg wk 1 → 40 mg q2w Randomized WD and re-TX perioda (wks 28–72) PL crossover periodb GUS q8w continued to wk 44 GUS wks 16 and 20 → q8w to wk 44 ADA 40 mg q2w continued to wk 47 GUS at wk 20 GUS at wks 16 and 20 ADA 40 mg q2w to wk 23; no TX wks 24–28 NA NA NA R: PLc or GUS q8w NR: GUS q8w R: PLc NR: GUS q8w R: PLc Maintenance and Recapture Based on VOYAGE 1, clinical responses were maintained through 100 and 156 weeks for patients on guselkumab, with 82.1% and 82.8% of patients achieving a PASI90 at these time points, respectively. Similarly, IGA 0/1 was maintained by 83.3% and 82.1% of guselkumab patients at week 100 and 156, respectively. Patients did have improvement in DLQI and Psoriasis Symptoms and Signs diary scores at the same time points [14]. In VOYAGE 2, patients with ≥90% PASI scores were re-randomized to continue guselkumab versus withdrawal and placebo. The treatment with guselkumab was restarted upon losing ≥50% of PASI at week 28 or at week 72. Loss of PASI75 response was associated with increased serum levels of IL-17A from week 40, IL-17F from week 36, and IL-22 from week 44 onwards. Maintenance of response was associated with suppression of serum levels of IL-17A, IL-17F, and IL-22. If there is an interruption in treatment with guselkumab, there is a slow relapse of psoriasis and restarting guselkumab can recapture the initial response in most patients [17]. NAVIGATE NR: GUS study-wks 28 and 32 → q8w ADA adalimumab, GUS guselkumah 100 mg, NA not applicable, NR PASI 90 non-responder, PASI 90 ≥ 90% improvement in Psoriasis Area and Severity Index, PL placebo, pts patients, qxw every x weeks, R PASI 90 responder, re(TX) re(treatment), WD withdrawal, wk/s week/s, → indicates thereafter a VOYAGE 1 wks 16–48; VOYAGE 2 wks 16–28, prior to randomized withdrawal and re-Tx through to wk 72 b GUS-treated pts were re-randomized based on PASI response at wk 28; NR = PASI < 90 and R ≥ PASI 90 c Upon loss of ≥ 50% of the 28-wk PASI response, pts were switched to GUS (100 mg at wk 0 and 4 → GUS 100 mg q8w) In the NAVIGATE phase III trial, the efficacy of guselkumab was analyzed in patients who did not adequately respond to ustekinumab [8]. Inclusion/ exclusion criteria were similar to VOYAGE 1 except patients who had received ustekinumab (as opposed to adalimumab) within 3 months of study onset were excluded. The study started with a 16-week open-label period where all the randomized patients received on-label dosing of ustekinumab (weight based dosing of either 45 mg or 90 mg) at week 0 and 4. At week 16, inadequate responders (IGA ≥ 2) were randomized to guselkumab 100 mg at weeks 16, 20 and every 8 weeks thereafter, or to continue 16 Guselkumab 219 Table 16.2 Efficacy of guselkumab along with HR-QOL outcomes at week-16 in pivotal VOYAGE 1 and VOYAGE 2 trials (reprinted with permission from Springer nature) Trial VOYAGE 1 [5, 13] VOYAGE 2 [5, 14] Regimen (no. of PASI (% pts) pts) 75b 90c GUS (329) 91*† 73*† IGA 0/1 (% pts)b ssIGA 0/1 (% pts)a 85*† 83* ADA (334) PL (174) 73 6 50 3 66 7 70 15 GUS (496) 86*† 70*† 84*† 81* ADA (248) 69 47 68 67 PL (248) 8 2 9 11 HR-QOL outcomes mean change from BL [BL] PSSD symptomd DLQId * –42 [54] –11* [14] –35 [54] –9 [14] –3 [48] <–1 [13] –40* [54] –11* [15] –33 [54] –10 [15] –8 [59] –3 [15] ADA adalimumab, BL baseline value, DLQI Dermatology Life Quality Index, GUS guselkumab, HR-QOL health-­ related quality of life, IGA Investigator Global Assessment (0 = cleared; 1 = minimal), PASI Psoriasis Area and Severity Index, PASI x improvement of ≥ x% from BL in PASI score, PL placebo, PSSD Psoriasis Symptoms and Signs Diary, pts patients, qxw every x weeks, ssIGA scalp-specific IGA (0 = absence; 1 = very mild), UST ustekinumab * p ≤ 0.001 vs. PL; †p < 0.001 vs. ADA a Major secondary outcome GUS vs. PL. Assessed in pts with BL ss-IGA score of ≥2 who had a ≥ 2-grade improvement; included 277, 286 and 145 pts in the GUS, ADA and PL groups, respectively, in VOYAGE 1 and 408, 194 and 202 pts in VOYAGE 2. No statistical comparisons performed for GUS vs. ADA b Major secondary outcome for GUS vs. ADA c Co-primary endpoint for GUS vs. PL at wk 16 in the VOYAGE trials; major secondary endpoint for GUS vs. ADA d Major secondary outcome for GUS vs. PL: mean change from BL in PSSD symptom score assessed in 249, 274 and 129 pts in the GUS, ADA and PL groups, respectively, in VOYAGE 1 and 411, 201 and 198 pts in VOYAGE 2; mean change from BL in DLQI score assessed in 322, 328 and 170 pts in the GUS, ADA and PL groups, respectively, in VOYAGE 1 and 496, 248 and pts in VOYAGE 2 ustekinumab at week 16 and every 12 weeks thereafter, while responders with IGA 0/1 continued to receive ustekinumab. The primary end point of the trial was the number of visits at which patients achieved an IGA score of 0 or 1 and at least a two-grade improvement relative to week 16 from week 28 through week 40 [8]. Among randomized patients, the guselkumab group had a significantly higher mean number of visits at which patients had an IGA score of 0 or 1 and at least a two-grade improvement relative to week 16 from week 28 through week 40 (primary end point) compared with the ustekinumab group (1.5 versus. 0.7; p < 0.001). Furthermore, at week 52, 51.1% of guselkumab patients compared to 24.1% of ustekinumab patients reached a PASI90 [8]. ECLIPSE A phase III, multicenter, randomized, double-­ blind, active comparator controlled study named ECLIPSE compared the efficacy and safety of guselkumab and the anti-IL17a inhibitor, secukinumab, in adult patients with moderate-to-­ severe plaque psoriasis [18]. 1048 adult patients with moderate-to-severe plaque psoriasis were randomized to receive guselkumab or secukinumab in a 1:1 ratio for 44 weeks. The primary endpoint was the proportion of patients who achieved a ≥ 90% improvement in Psoriasis Area Severity Index score (PASI90) at week 48. At week 48, guselkumab was superior to secukinumab in achieving PASI90 (84.5% versus 70.0%; p < 0.001). Major secondary end- D. Joshipura et al. 220 points were not met such as the superiority in proportion of patients achieving a PASI75 in the guselkumab group compared to the secukinumab group at both week 12 and 48 (84.6% versus 80.2%; p = 0.062) [18]. Other Studies Guselkumab was evaluated in a phase III randomized, double-blind, placebo-controlled study in Japanese patients with moderate-to-severe plaque psoriasis. Results were similar to VOYAGE 1 and VOYAGE 2 in which approximately 70% of patients on guselkumab achieved a PASI90 at week 16 [11]. Guselkumab was also evaluated in 159 Japanese patients with palmoplantar pustulosis and showed superiority over placebo [19]. Safety/Adverse Events Placebo 422 Patients treated, n 15.9 Average weeks of follow-up 128 Total patient-years of follow-up 198 (46.9) At least on adverse events Common advers eventsa Nasopharyngitis 33 (7.8) 19 (4.5) Upper respiratory tract infection Headache 14 (3.3) Arthralgia 9 (2.1) Hypertension 8 (1.9) Injecton site 3 (0.7) erythema Diarrhoea 4 (0.9) Pruritus 17 (4.0) Injection site bruising 4 (0.9) Back pain 8 (1.9) Cough 5 (1.2) Fatigue 6 (1.4) Gastroehteritis 3 (0.7) Adverse events leading 5 (1.2) to discontinuation Infections 90 (21.3) Infections requiring 30 (7.1) treatment Serious adverse events 6 (1.4) Guselkumab Adalimumab 823 16.2 581 16.1 255 179 407 (49.5) 291 (50.1) 65 (7.9) 41 (5.0) 54 (9.3) 20 (3.4) 38 (4.6) 22 (2.7) 18 (3.1) 10 (1.7) 20 (2.4) 16 (1.9) 12 (2.1) 26(4.5) 13 (1.4) 13 (1.6) 13 (1.4) 13 (1.6) 11 (1.3) 10 (1.2) 10 (1.2) 9 (1.1) 11 (1.3) 10 (1.2) 10 (1.2) 9 (1.1) 9 (1.1) 10 (1.2) 9 (1.1) 10 (1.2) 193 (23.5) 56 (6.8) 145 (25.0) 43 (7.4) 16 (1.9) 12 (2.1) Overall guselkumab is well tolerated, and side Data are presented as n (%). aCommon adverse event are defined effects are usually mild. In a pooled analysis of as those occurring in at least 1% of patients in the guselkumab VOYAGE 1 and VOYAGE 2 through 100 weeks, group. safety of guselkumab was assessed [20]. The Fig. 16.3 Common adverse events noted in the pooled most common adverse events (AE) reported were analysis of VOYAGE 1 and VOYAGE 2 phase 3 clinical nasopharyngitis, upper respiratory tract infec- trials on guselkumab (Reprinted with permission from tion, and headache. Arthralgias were reported in Reich et al.) 2.7% of patients on guselkumab compared with 2.0% and 2.1% of patients on adalimumab and tively. The proportion of patients with injection placebo, respectively [20]. There were no differ- site reactions was higher in the adalimumab ences in serious AEs between the groups group before the crossover to guselkumab (8.8% (Fig. 16.3). versus 0.8%). No events of Crohn’s disease were Through week 52, guselkumab and adalim- reported and the rates of vulvovaginal (0.4%) and umab treated patients had few adverse events and skin (0.2%) candidiasis and neutropenia (0.1%) serious adverse events (262.45/100 PYs versus were low [20]. 328.28/100 PYs and 6.2/100 PYs versus 7.77/100 PYs) (Fig. 16.4) Serious infections, non-­ melanoma skin cancers (NMSC), malignancy Pediatrics and Breastfeeding excluding NMSC, and major adverse cardiovascular events (MACEs) were low for patients There have been reports of pediatric patients treated with guselkumab and no increased signal treated with guselkumab [21, 22], though there was seen with treatment from weeks 0 to 100 are no large studies evaluating safety in this compared to just weeks 0 to 52 [20]. patient population. There is a lack of safety data Injection site reactions were noted in 2.8% for patients who are pregnant receiving guseland 2.8% of patients at week 52 and 100, respec- kumab. Guselkumab was not detected in the milk 16 Guselkumab 221 Guselkumaba Treated patients. n Average weeks of follow-up Total PYs of follow-up Adverse events Observed number of events Incidence per 100 PYs 95% confidence interval Adverse events leading to discontinuation Observed number of patients Incidence per 100 PYs 95% confiedence interval Infections Observed number of events Incidence per 100 PYs 95% confiedence interval Infections requiring treatment Observed number of events Incidence per 100 PYs 95% confiedence interval Serious adverse events Observed number of events Incidence per 100 PYs 95% confiedence interval Adalimumab Through Week 52 Through Week 100 Before crossover b After crossover c 1221 45.4 1221 89.0 581 45.1 500 51.7 1065 2084 502 496 2795 4384 1647 794 262.45 252.81–272.37 210.41 204.23–216.73 328.28 312.62–344.23 160.15 149.20–171.69 26 2.46 38 1.83 23 4.63 8 1.62 1.60–3.60 1.30–2.51 2.93–6.94 0.70–3.19 1070 100.47 1703 81.74 77.90–85.71 540 107.63 98.75–117.11 353 71.20 495 23.76 145 28.90 95 19.16 21.71–25.95 24.39–34.01 15.50–23.42 66 6.20 131 6.29 22 4.44 4.79–7.88 5.26–7.46 39 7.77 5.53–10.63 94.54–106.68 300 28.17 25.07–31.55 63.96–79.03 2.78–6.72 aIncludes patients randomized to guselkumab at baseline (n = 825) and those initially randomized to placebo who crossed over to receive guselkumab at week 16 (n = 398) in VOYAGE 1 and VOYAGE 2. bInclude patents randomized at baseline to adalimumab before crossover to guselkumab at the end of the active-comparator perod (week 52) in VOYAGE 1 and before crossover to guselkumab during the randomized withdrawal and retreatmet period (starting at week 28) (28-08.20) in VOYAGE 2. cIncludes patients randomized at baseline to adalimumabafter crossover to guselkumab at the end of the active-comparator perod (week 52) in VOYAGE 1 and after crossover to guselkumab during the random- ized withdrawal and retreatmet period (starting at week 28) in VOYAGE 2. Fig. 16.4 Summary of adverse events per 100 patient years through 100 weeks of follow up. Data from the pooled analysis from VOYAGE 1 and 2 (reprinted with permission from Reich et al.) of lactating monkeys, though IgG is known to be present in human milk [2]. It is not known if guselkumab would be absorbed systemically through an immature neonatal gastrointestinal tract after breastfeeding. Tuberculosis, HIV, and Live Vaccines Patients should be screened for tuberculosis (TB) prior to starting guselkumab. For patients being treated for latent TB, they can be safely started on guselkumab at the same time as the patient receives the first dose of antitubercular treatment. There is one case report of a patient who is HIV positive who received guselkumab with stable CD4 counts at 12 months [23]. As for all psoriasis biologics, live vaccines are contraindicated since there is an absence of safety data. Malignancy Guselkumab does not target the activity of IL-12/ Th-1 helper cells. Furthermore, in certain situations IL-23 may promote tumor growth [24], so in theory, use of guselkumab may be safe in patients with cancer as guselkumab inhibits the activity of IL-23; however there is an absence of evidence in the literature on the use of guselkumab in patients with malignancy. Therefore, guselkumab should be used with caution in this patient population pending more data. Inflammatory Bowel Disease Guselkumab treatment is not associated with new-onset or exacerbations of inflammatory bowel disease (IBD) and may serve as a future treatment modality [25]. The anti-IL12/IL23 D. Joshipura et al. 222 inhibitor ustekinumab [26] and anti-IL23 blocker risankizumab have both been shown to improve inflammatory bowel disease [27]. Psoriatic Arthritis Hidradenitis Suppurativa Hidradenitis suppurativa (HS) is a chronic inflammatory debilitating skin condition characterized by development of painful abscess, cysts followed by development of sinus tracts and scarring. An overactivity of IL-23/Th-17 pathway has been hypothesized to play a role in the pathogenesis of HS [29]. Two case series evaluating the use of guselkumab in patients with HS reported benefit in some of the patients [22, 30]. Guselkumab and other IL-23 inhibitors are currently under further investigation as a treatment option for patients with HS. The efficacy of Guselkumab was studied in a phase 2 clinical trial done in patients with active psoriatic arthritis [28]. 149 patients were included with ≥3 tender and swollen joints, with C-reactive protein levels ≥3 mg/L and ≥ 3% BSA of plaque psoriasis. Patients were randomized to 2:1 to receive either guselkumab 100 mg at week 0, 4 and every 8 weeks through week 44 or placebo. Patients receiving placebo were subsequently crossed over to Patient Controlled Injector receive guselkumab at week 24, 28 then every 8 weeks through week 44. An early escape to In the placebo-controlled double-blind phase III open label ustekinumab was enabled in patients ORION study which evaluated guselkumab with <5% improvement from baseline in both administered through a patient-controlled injecswollen and tender joints. Primary endpoint was tor (One-Press), the primary endpoints were ACR20 response at week 24 which was achieved PASI90 and IGA of 0 or 1 at week 16 [31]. in 58% of patients on guselkumab versus 18.4% Patients also filled out the self-injection assessof placebo. 34% of patients on guselkumab ment questionnaire (SIAQ) and patient-­controlled achieved ACR50 versus placebo at week 24 injection device questionnaire. Guselkumab was compared to 10.2% of placebo patients, and superior to placebo in terms of PASI90 (75.8% 14% of patients had ACR70 response at week 24 versus 0.0%) and IGA of 0 or 1 (80.6% versus compared to 2.0% of placebo patients. 60% of 0.0%). Injection site reactions were similar for patients with prior anti-TNF-alpha exposure both placebo and guselkumab treated subjects. achieved ACR20 versus 57.8% of anti-TNF-­ SIAQ post-dose scores were also favorable with alpha naive patients at week 24.56.6% of patients rating the one-press device as easy/very patients on guselkumab had complete resolution easy to use from weeks 0 to 28 (87%–100%) and of enthesitis at week 24 versus 29% in the pla- were satisfied/very satisfied with the device cebo group. Similarly, 55.2% of patients in the (81%–100%). guselkumab group had complete resolution of dactylitis at week 24 versus 17.4% in the placebo group. 23% of patients had achieved mini- Conclusion mal disease activity at week 24 compared to 2.0% of patients on placebo [28]. While there is The IL-23 inhibitor guselkumab offers a targeted some promising early data on the use of gusel- therapy for patients with moderate-to-severe kumab for patients with psoriatic arthritis, there plaque psoriasis. The ability to achieve high effiis no evidence that guselkumab reduces radio- cacy with minimal safety risk and infrequent graphic progression of disease. Furthermore, injections are major advantages of guselkumab. more data is needed to determine the relative Therefore, the development of anti-p19 agents is efficacy of guselkumab against first line FDA a significant advancement in the treatment of approved agents for psoriatic arthritis which tar- psoriasis. The ability of guselkumab to treat psoget TNF-alpha and IL-17. riatic arthritis and other comorbidities of psoria- 16 Guselkumab 223 placebo- and active comparator. J Am Acad Dermatol [Internet]. 2017;76(3):405–17. Available from: doi:https://doi.org/10.1016/j.jaad.2016.11.041. 7. Reich K, Armstrong AW, Foley P, Song M, Wasfi Y, Randazzo B, et al. Efficacy and safety of guselkumab, an anti-interleukin-23 monoclonal antibody, compared with adalimumab for the treatment of patients with moderate to severe psoriasis with randomized withdrawal and retreatment: Results from the phase III, double-blind, p. J Am Acad Dermatol [Internet]. Conflict of Interest 2017;76(3):418–31. Available from: doi:https://doi. David Rosmarin has received honoraria as a conorg/10.1016/j.jaad.2016.11.042. sultant for AbbVie, Celgene, Dermavant, 8. Langley RG, Tsai TF, Flavin S, Song M, Randazzo Dermira, Janssen, Lilly, Novartis, Pfizer, B, Wasfi Y, et al. Efficacy and safety of guselkumab in patients with psoriasis who have an inadequate Regeneron, and Sanofi; has received research response to ustekinumab: results of the randomsupport from AbbVie, Bristol Meyers Squibb, ized, double-blind, phase III NAVIGATE trial. Br J Celgene, Dermira, Incyte, Janssen, Lilly, Merck, Dermatol. 2018;178(1):114–23. Novartis, Pfizer, and Regeneron Pharmaceuticals 9. Sofen H, Smith S, Matheson RT, Leonardi CL, Calderon C, Brodmerkel C, et al. Guselkumab (an Inc.; and has served as a paid speaker for AbbVie, IL-23-specific mAb) demonstrates clinical and Celgene, Janssen, Lilly, Novartis, Pfizer, molecular response in patients with moderate-to-­ Regeneron Pharmaceuticals Inc., and Sanofi. severe psoriasis. J Allergy Clin Immunol [Internet]. 2014;133(4):1032–40. Available from: doi:https://doi. org/10.1016/j.jaci.2014.01.025. No Conflict of Interest 10. Gordon KB, Duffin KC, Bissonnette R, Prinz JC, Deep Joshipura and Brooke Rothstein. Wasfi Y, Li S, et al. A Phase 2 Trial of Guselkumab versus Adalimumab for Plaque Psoriasis. N Engl J Med. 2015;373(2):136–44. 11. Ohtsuki M, Kubo H, Morishima H, Goto R, Zheng References R, Nakagawa H. Guselkumab, an anti-interleukin-23 monoclonal antibody, for the treatment of moderate to 1. Al-Salama ZT, Scott LJ. Guselkumab: A Review severe plaque-type psoriasis in Japanese patients: effiin Moderate to Severe Plaque Psoriasis. Am J Clin cacy and safety results from a phase 3, randomized, Dermatol [Internet]. 2018 Dec 22 [cited 2019 Aug double-blind, placebo-controlled study. J Dermatol. 4];19(6):907–18. Available from: http://www.ncbi. 2018; nlm.nih.gov/pubmed/30467781. 2. Janssen Biotech. Tremfya (Guselkumab)[package 12. Sofen H, Smith S, Matheson RT, Leonardi CL, Calderon C, Brodmerkel C, et al. Guselkumab (an insert]. US Food and Drug Administration website. IL-23-specific mAb) demonstrates clinical and https://www.accessdata.fda.gov/drugsatfda_docs/ molecular response in patients with moderate-to-­ label/2017/761061s000lbl.pdf. Revised 7/2017. severe psoriasis. J Allergy Clin Immunol. 2014; Accessed 06/2019. 3. Puig L. The role of IL 23 in the treatment of psoriasis. 13. Armstrong AW, Reich K, Foley P, Han C, Song M, Shen YK, et al. Improvement in Patient-Reported Expert Rev Clin Immunol. 2017; Outcomes (Dermatology Life Quality Index 4. Zhuang Y, Calderon C, Marciniak SJ, Bouman-Thio and the Psoriasis Symptoms and Signs Diary) E, Szapary P, Yang TY, et al. First-in-human study with Guselkumab in Moderate-to-Severe Plaque to assess guselkumab (anti-IL-23 mAb) pharmacoPsoriasis: Results from the Phase III VOYAGE 1 and kinetics/safety in healthy subjects and patients with VOYAGE 2 Studies. Am J Clin Dermatol [Internet]. moderate-­to-severe psoriasis. Eur J Clin Pharmacol. 2019;20(1):155–64. Available from: doi:https://doi. 2016; org/10.1007/s40257-018-0396-z. 5. Zhu Y, Marini JC, Song M, Randazzo B, Shen YK, Li S, et al. Immunogenicity of Guselkumab Is Not 14. Griffiths C, Papp K, Song M, Randazzo B, Li S, Shen Y, et al. Maintenance of response with guselkumab Clinically Relevant in Patients with Moderate-to-­ for up to 3 years’ treatment in the phase 3 VOYAGE Severe Plaque Psoriasis. J Invest Dermatol. 2019; 1 trial of patients with plaque psoriasis. In: Journal 6. Blauvelt A, Papp KA, Griffiths CEM, Randazzo of the European Academy of Dermatology and B, Wasfi Y, Shen YK, et al. Efficacy and safety of Venereology [Internet]. 2019. p. 49–50. Available guselkumab, an anti-interleukin-23 monoclonal antifrom: http://www.embase.com/search/results?subacti body, compared with adalimumab for the continuous on=viewrecord&from=export&id=L627805046%0A treatment of patients with moderate to severe psohttp://dx.doi.org/10.1111/jdv.15514 riasis: Results from the phase III, double-blinded, sis such as inflammatory bowel disease still needs to be better elucidated. Additional long-term safety data is needed to gather further information on the maintenance of therapeutic response and rare adverse events that may be associated with guselkumab use. 224 15. Foley P, Gordon K, Griffiths CEM, Wasfi Y, Randazzo B, Song M, et al. Efficacy of guselkumab compared with adalimumab and placebo for psoriasis in specific body regions a secondary analysis of 2 randomized clinical trials. JAMA Dermatol. 2018;154(6): 676–83. 16. Gordon KB, Blauvelt A, Foley P, Song M, Wasfi Y, Randazzo B, et al. Efficacy of guselkumab in subpopulations of patients with moderate-to-severe plaque psoriasis: a pooled analysis of the phase III VOYAGE 1 and VOYAGE 2 studies. Br J Dermatol. 2018; 17. Gordon KB, Armstrong AW, Foley P, Song M, Shen Y-K, Li S, et al. Guselkumab efficacy after withdrawal is associated with suppression of serum IL-23-­ regulated IL-17 and IL-22 in psoriasis: VOYAGE 2 study. J Invest Dermatol. 2019; 18. Johnson J and. No Title [Internet]. 2018. Available from: https://www.jnj.com/new-phase-3-data-demonstrate-superiority-of-tremfya-guselkumab-vs-cosentyx-secukinumab-in-delivering-pasi-90-responses-inthe-treatment-of-moderate-to-severe-plaque-psoriasis-at-week-48 19. Terui T, Kobayashi S, Okubo Y, Murakami M, Zheng R, Morishima H, et al. Efficacy and Safety of Guselkumab in Japanese Patients With Palmoplantar Pustulosis. JAMA Dermatology [Internet]. 2019 Jul 3 [cited 2019 Aug 1]; Available from: http://www.ncbi. nlm.nih.gov/pubmed/31268476 20. Reich K, Papp KA, Armstrong AW, Wasfi Y, Li S, Shen YK, et al. Safety of guselkumab in patients with moderate-to-severe psoriasis treated through 100 weeks: a pooled analysis from the randomized VOYAGE 1 and VOYAGE 2 studies. Br J Dermatol. 2019;180(5):1039–49. 21. Kim SR, Kibbi N, Craiglow BG. Guselkumab for the treatment of severe refractory psoriasis in a pediatric patient. JAAD Case Reports [Internet]. 2019 Jun [cited 2019 Aug 4];5(6):552–4. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/31245518 22. Casseres RG, Kahn JS, Her MJ, Rosmarin D. Guselkumab in the treatment of hidradenitis suppurativa: a retrospective chart review. J American Academy Dermatology. 2019; 23. Bartos G, Cline A, Beroukhim K, Burrall BA, Feldman SR. Current biological therapies for use in HIV-positive patients with psoriasis: case report D. Joshipura et al. of gesulkumab used and review. Dermatol Online J. 2018; 24. Langowski JL, Zhang X, Wu L, Mattson JD, Chen T, Smith K, et al. IL-23 promotes tumour incidence and growth. Nature. 2006; 25. Grossberg LB. A Case Report of Successful Treatment of Crohn’s Disease and Psoriasis With Guselkumab. Inflamm Bowel Dis [Internet]. 2019 Jun 18 [cited 2019 Aug 2];25(7):e84–e84. Available from: http:// www.ncbi.nlm.nih.gov/pubmed/30863855 26. Sandborn WJ, Gasink C, Gao L-L, Blank MA, Johanns J, Guzzo C, et al. Ustekinumab Induction and Maintenance Therapy in Refractory Crohn’s Disease. N Engl J Med [Internet]. 2012 Oct 18 [cited 2019 Aug 1];367(16):1519–28. Available from: http://www. ncbi.nlm.nih.gov/pubmed/23075178 27. Feagan BG, Sandborn WJ, D’Haens G, Panés J, Kaser A, Ferrante M, et al. Induction therapy with the selective interleukin-23 inhibitor risankizumab in patients with moderate-to-severe Crohn’s disease: a randomised, double-blind, placebo-controlled phase 2 study. Lancet (London, England) [Internet]. 2017 Apr 29 [cited 2019 Aug 1];389(10080):1699–709. Available from: https://linkinghub.elsevier.com/ retrieve/pii/S0140673617305706 28. Deodhar A, Gottlieb AB, Boehncke W-H, Dong B, Wang Y, Zhuang Y, et al. Efficacy and safety of guselkumab in patients with active psoriatic arthritis: a randomised, double-blind, placebo-controlled, phase 2 study. Lancet [Internet]. 2018 Jun 2 [cited 2019 Aug 2];391(10136):2213–24. Available from: http://www. ncbi.nlm.nih.gov/pubmed/29893222 29. Schlapbach C, Hänni T, Yawalkar N, Hunger RE. Expression of the IL-23/Th17 pathway in lesions of hidradenitis suppurativa. J Am Acad Dermatol. 2011; 30. Kovacs M, Podda M. Guselkumab in the treatment of severe hidradenitis suppurativa. J Eur Acad Dermatology Venereol. 2019;33(3):e140–1. Available from: http://www.ncbi.nlm.nih.gov/ pubmed/30480844 31. Ferris LK, Ott E, Jiang J, Hong HCH, Li S, Han C, et al. Efficacy and safety of guselkumab, administered with a novel patient-controlled injector (one-press), for moderate-to-severe psoriasis: results from the phase 3 ORION study. J Dermatolog Treat. 2019; 17 Tidrakizumab George Han Abstract Tildrakizumab is a monoclonal antibody targeting the p19 subunit unique to the cytokine IL-23 that is FDA-approved in the treatment of plaque psoriasis. IL-23 is a central regulatory cytokine in the pathogenesis of psoriasis. Tildrakizumab was shown to have superior efficacy in treating psoriasis as compared to both placebo and an active comparator (etanercept) in two parallel pivotal phase III trials. This biologic medication has demonstrated efficacy, combined with strong evidence of durability and an excellent safety profile. Additionally, the fact that it is dosed every 12 weeks is an attractive feature to many patients. Early results for treatment of psoriatic arthritis are promising, and treatment seems to be consistent across patient weight categories and previous treatment characteristics. Numerous nuances exist in the approach to systemic therapy for psoriasis patients, and tildrakizumab is able to navigate many comorbidities to deliver a reasonable first-line biologic medication for patients with psoriasis. G. Han (*) Icahn School of Medicine at Mount Sinai, New York, NY, USA e-mail: george.han@mountsinai.org © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_17 Key Learning Objectives 1. To understand the mechanism of action of Tidrakizumab 2. To understand the appropriate use and efficacy of the Tidrakizumab 3. To understand the safety issues of Tidrakizumab Introduction Tildrakizumab (Tildrakizumab-asmn, SUN Pharmaceuticals US, Cranbury, NJ) is a high-­ affinity IgG1/κ monoclonal antibody targeting the p19 subunit of the cytokine IL-23. It represents the first biologic that was studied targeting IL-23 specifically, with phase II trials starting in 2010 and phase III trials starting in 2012 [1]. Initially developed by Schering-Plough (Kenilworth, NJ) and then Merck (Kenilworth, NJ), the rights to market this medication were bought by SUN Pharmaceuticals in 2014. Subsequently, a licensing agreement was made with Almirall (Barcelona, Spain) for marketing of the medication in Europe. Results from phase III trials were reported in 2017, with active comparator (Etanercept) and placebo controls. It is FDA approved in the US for the treatment of plaque psoriasis with studies currently under way for treatment of psoriatic arthritis. The dos225 226 ing of this medication is an injection of 100 mg at 0 weeks and 4 weeks, followed by injections every 12 weeks. It is indicated in the US for administration by a health care provider. G. Han agents (Mendelian susceptibility to mycobacterial disease) often caused by an autosomal recessive deficiency in IL-12. Furthermore, these patients are susceptible to Candida and Klebsiella infections [5]. With regards to tumor immunity, it is becoming clear that there are numerous roles Rationale of Targeting IL-23 of IL-12 in tumor immunity, with a role of IL-12 in both promoting cytotoxic Natural Killer IL-23 is a proinflammatory cytokine that func- (NK) cell function and in activating Interferon-­ tions via upregulation of the Th17 axis in inflam- gamma (IFN-γ) producing Th1 cells via the mation, with downstream effects carried out STAT4 pathway [6–8]. Furthermore, IL-12 can primarily via IL-17. Specifically, IL-23 induces effectively reprogram immune cells to act against differentiation of naïve CD4+ T cells into patho- tumor cells in certain environments [9]. Thus, genic Th17 cells, which then produces IL-17, there may be a role of IL-12 in protecting the host IL-6, and TNF-α [2]. Prior to the study of til- against the emergence of spontaneous tumors drakizumab, numerous medications had been [10] and it would be preferable to not block this developed targeting the IL-12/IL-23 pathway in cytokine in this context. On the contrary, IL-23 psoriasis. Ustekinumab and briakinumab are has been proposed as a disease marker in cancer, both monoclonal antibodies targeting the shared with numerous pro-tumor and metastasis funcp40 subunit of IL-12 and IL-23 [3]. While early tions that are appearing in varied malignancies studies focusing on examining the role of the p40 such as breast cancer, hepatocellular carcinoma, subunit seemed to show a role of this pathway in colorectal cancer, oral squamous cell carcinoma, psoriasis, subsequently, it was determined that and non-small cell lung cancer [11–16], among these medications are able to treat psoriasis pri- others [10]. Thus, it seems rather consistent that marily by blocking IL-23. A study with human blocking this pro-tumor molecule (IL-23) should skin biopsy samples showed that while the p40 not lead to any concerns about malignancy; subunit (shared by IL-12 and IL-23) and the p19 indeed, utilization of anti-IL-23 treatment in subunit (unique to IL-23) are elevated in lesional oncology has been proposed as a potential treatpsoriasis skin, the p35 subunit (unique to IL-12) ment in combination with other immunotherapies is not. On the contrary, levels of all three subunits [17]. It should be noted that in long-term registry are the same in non-lesional skin from psoriasis data, no evidence was found that blocking IL-12 patients. In normal skin, expression of all three and IL-23 with ustekinumab increased the risk subunits are the same or lower than in non-­ for cancer [18]. lesional skin from psoriasis patients [4]. Taken Previously, there was some concern that tartogether, these data were important in ­determining geting p40 (IL-12 and IL-23) was associated with that the inflammation underlying psoriasis pri- the development of major adverse cardiovascular marily hinges upon increased levels of IL-23 and events (MACE). One drug, briakinumab, was has little reliance on IL-12. withdrawn from clinical development due to the Aside from the theoretical rationale that sup- emergence of multiple MACE during Phase II ports targeting IL-23 rather than IL-12 in treating and III trials. While a statistically significant psoriasis, there are specific considerations that increase in risk of MACE is under debate and make targeting IL-23 much more attractive than multiple analyses have shown both risk and no IL-12. In terms of immunosuppression, it appears risk in this regard [19–22], the data does seem to that there are numerous functions of IL-12 in host support at least a trend towards MACE among defense. It has been shown that IL-12 appears to these studies. On the contrary, there seems to be be important in the immune response to no signal or trend towards MACE in any of the Mycobacterium and Salmonella infections, with medications targeting IL-23 [22]. The mechaa condition that causes susceptibility to these nisms whereby blockade of these interleukins 17 Tidrakizumab 227 can result in a cardiac event are unclear, but what- III parallel trials across sites in North America, ever minimal trend there seems to be with medi- Europe, Asia, and Australia (reSURFACE 1 and cations targeting both IL-12 and IL-23 together reSURFACE 2) [29]. The study was conducted seems to disappear with IL-23 blockade alone. with two doses of tildrakizumab (100 mg q12wk As compared to IL-17 blocking agents, one and 200 mg q12wk), which was likely a result notable difference among these agents is with of the dose-ranging phase IIb studies showing a regard to their roles in intestinal immunoregula- difference of about 10% in achievement of the tion. IL-17 inhibition exacerbates colitis, likely primary endpoint (PASI75 achievement, both at due to weakening intestinal epithelial barrier week 12 and week 16) [1]. While many medicafunction (subsequently allowing for microbial tions proceed with one tested dose in Phase III penetration and increased inflammation) while trials, the sponsor likely thought that there was IL-23 inhibition seems to promote an anti-­ enough of a benefit in the higher dose group to inflammatory milieu in the gut [23]. While IL-17 proceed with testing it in larger phase III trials. has been shown to exacerbate inflammatory bowel For one of the the Phase III trials (reSURFACE disease (IBD) [24], IL-23 blockade appears to be 2), an active comparator group treated with etana promising candidate in treatment of Crohn’s dis- ercept in addition to the control group was also ease [25]. Notably, the actual incidence of IBD in included. large analyses of IL-17 inhibitors is low [26, 27], Baseline characteristics of this trial were simibut it is a true, predictable result of the mechanism lar to most psoriasis trials [29]. The vast majority of action of this class of medication. of patients were white (65–92%, percentages given as proportion of patients in each arm) and male (65–72%). Average weight was close to 90 Pharmacokinetics/ Kg and average body surface area was slightly over 30%. The mean PASI at baseline was Pharmacodynamics roughly 20. In reSURFACE 1, percent treated Subcutaneous dosing of tildrakizumab results in previously with biologics was 23% across all slow systemic clearance, limited volume of dis- groups whereas in reSURFACE 2, that number tribution, and a long half-life in pharmacokinetic was between 12–13%. studies [28]. After a single subcutaneous injecThe co-primary endpoints of reSURFACE 1 tion of tildrakizumab, a dose-related increase in and 2 were achievement of PASI 75 and PGA or 0 serum concentration was observed, peaking at or 1 with at least a 2-point reduction in PGA from roughly 1 week and exhibiting a slow, linear baseline. Importantly, the primary endpoint outdecrease over time. The volume of distribution come measures were taken at week 12. At the was fairly low and the half-life was roughly time of these trials, there were only three approved 23–25 days for the doses studied. Bioavailability injectable biologics for psoriasis: etanercept, of tildrakizumab was estimated to approach 80% adalimumab, and ustekinumab. Likely due to the [28]. There is no weight-based dosing for til- rate of efficacy for TNF-α inhibitors, the paradrakizumab; although multiple doses were stud- digm at the time was to measure primary endied in Phase III trials, there was not enough of a points at week 12, even for ustekinumab [30]. difference in efficacy between doses to allow for Additionally, in the Phase II trials of tildrakiapproval of the higher dose. zumab, the rise in achievement of PASI 75 between weeks 12 and 16 was rather modest [1]. However, it has since become more clear that tarClinical Efficacy geting IL-12/23 and specifically IL-23 as well, leads to a somewhat slower rise towards peak effiThe efficacy of tildrakizumab in treating psoriasis cacy [31]. While tildrakizumab treatment was sigwas demonstrated in two large, multicenter, ran- nificantly better than placebo at week 12 in both domized, double-blind placebo-controlled phase co-primary endpoints, the numbers were perhaps G. Han 228 somewhat disappointing as they were found to be only comparable to previously reported numbers with ustekinumab. The percent achieving PASI 75 for tildrakizumab 200 mg was 62% and 66% for reSURFACE 1 and 2, respectively; for tildrakizumab 100 mg, 64% and 61%; for etanercept 48% (reSURFACE 2 only); and for placebo, 6% and 6%. The achievement of PGA 0 or 1, a somewhat more stringent criteria, for tildrakizumab 200 mg was 59% and 59% for reSURFACE 1 and 2, respectively; for tildrakizumab 100 mg, 58% and 55%; for etanercept 48%; and for placebo, 7% and 4% [29]. Thus, it appeared from the primary endpoint outcomes that there was not much of a difference between the two doses of tildrakizumab. Notably, the week 28 data was much better for tildrakizumab, with PASI 75 achieved by 79% of patients on tildrakizumab 200 mg and 77% of patients on tildrakizumab 100 mg. Clear or minimal PGA was attained by 67% of tildrakizumab 200 mg treated patients and 63% of tildrakizumab 100 mg treated patients at week 28. PASI 100 was achieved by 31% of patients on tildrakizumab 200 mg and 22% of patients on tildrakizumab 100 mg. As IL-23 serves as more of a regulatory cytokine in the pathogenesis of psoriasis than an effector molecule (such as IL-17), it is perhaps not surprising that it takes longer for these medications to reach peak efficacy, but it should be noted that durability does seem to be a strength of this drug (see below). At this point, 3-year data has been presented for tildrakizumab showing strong durability of effect [32]. At week 148, 91.2% of the observed cohort of responders (i.e. those who achieved PASI75 and entered long-term extension) maintained PASI75. It should be noted that while NRI data were presented (72.6% of responders maintained PASI75 response), the reporting of NRI data at 3 years or longer is abnormal owing to the likelihood that life events may necessitate someone’s withdrawal from a clinical trial. Other reports have generally used as observed data or other imputation methods for data points significantly longer than 100 weeks [33, 34]. Interestingly, over half of the NRI cohort (53.8%) and over 2/3 of the observed population (67.6%) achieved PASI90, implying that patients who achieve PASI75 early on in treatment are more likely than not to progress to a PASI90 response. Psoriatic Arthritis While psoriatic arthritis trials are ongoing for tildrakizumab and phase III trials have not been completed at the time of writing, the Phase IIb study of tildrakizumab for treating psoriatic arthritis has been conducted and the results have been reported. The results of this smaller study are quite interesting and represent a departure from the perceived weakness of the IL-23 inhibitor class, namely treating psoriatic arthritis. From a conceptual standpoint, it has been shown that there seems to be a different gene signature between psoriatic skin and synovium—the former is IL-17 dominant, while the latter tends to have a stronger TNF signature [35]. Furthermore, a report of a different IL-23 inhibitor, risankizumab, revealed that not only were primary endpoints for treating ankylosing spondylitis (AS) not met, there was no evidence of clinically meaningful improvements compared to placebo [36]. But the same does not seem to hold true for psoriatic arthritis in general, where it is expected that targeting IL-23 should offer efficacy in treating psoriatic arthritis. This was supported by a Phase II trial of another IL-23 inhibitor, guselkumab, where 58% of patients achieved ACR20 at week 24 [37]. This result was similar to the ACR20 response to adalimumab in the corresponding Phase III trial (ADEPT) [38]. However, when the psoriatic arthritis treatment data for tildrakizumab were released, it was an unexpected surprise. The ACR20 for tildrakizumab 100 mg (at the same dosing as for psoriasis, every 12 weeks) was reported to be 71.4%. In fact, ACR50 for this dose was 45.5% and ACR70 was achieved in 22.1% of patients [39]. Of course, these numbers have to be taken with a grain of salt as they are Phase II trials with only about 80 patients in each group, but the numbers are strikingly higher than reported Phase II trial data of a medication targeting the same pathway. It remains to be seen how these different medications in the same class end up distinguishing themselves, but 17 Tidrakizumab efficacy in psoriatic arthritis could potentially be a significant differentiator for tildrakizumab if the results hold up in Phase III trials. Safety/Adverse Events From a safety standpoint, the effects of tildrakizumab do not seem to be associated with any major safety concerns that stand out; in fact, the specific safety data profile appears to be one of the cleanest among all biologics. In the parallel trials, the rates of adverse events (one or more) were 42% and 49% for tildrakizumab 200 mg, 47% and 44% for tildrakizumab 100 mg, 48% and 55% for placebo, and 54% for etanercept. The rates of serious adverse reactions were low across all groups; ranging from 1–3% for all groups in the placebo-controlled period (with no consistent trend observed). The most common adverse events were nasopharyngitis and upper respiratory infection. The percentages of patients experiencing nasopharyngitis were 6% and 11% for tildrakizumab 200 mg, 8% and 13% for tildrakizumab 100 mg, 5% and 8% for placebo, and 12% for etanercept. Respiratory tract infection was only reported in reSURFACE 1 (likely an issue with coding the adverse events) and this was seen in 5% of patients on tildrakizumab 200 mg, 3% of patients on tildrakizumab 100 mg, and 6% of patients on placebo. Importantly, in part 2 of both trials, the rates of these infections remained stably low. There were very low rates and no signal for serious adverse events of interest, including severe infections, malignancies, non-melanoma skin cancer, major adverse cardiovascular events, and drug-related hypersensitivity reactions. Overall, the rates of adverse events with tildrakizumab are among the lowest reported with any biologic and must be considered as a significant strength of the medication. Immunogenicity Immunogenicity is a possibility as with any biologic. The specific rates of anti-drug antibody formation for tildrakizumab was about 6.5% 229 through 52–64 weeks, with about 2.5% of patients developing treatment-emergent, neutralizing antibodies, which are generally associated with reduced serum concentrations of the active drug and thus modestly reduced efficacy [40]. This was found to be true in a sub-analysis of the tildrakizumab trials, where treatment-emergent, neutralizing antibodies were associated with a modest decrease in PK of tildrakizumab and a mean PASI reduction of about 10–15%. It should be noted that comparatively, these are still low rates of immunogenicity and neutralizing antibodies were not found to be associated with any drug-related or serious adverse events [41]. Considerations in Treatment Approach When considering treatment options for psoriasis, there are numerous factors to consider. First, there is of course efficacy. However, it should be noted that the actual improvements in gross efficacy have started to cluster within a relatively narrow range for the majority of patients. Thus, other considerations may take a more prominent role in the selection of an appropriate medication for patients with psoriasis. Speed of Onset It should be noted that in general, the function of IL-12/23 and IL-23 inhibitors is on the slower side, especially as compared to IL-17 inhibitors. Patients may drift upwards slowly with any class of medication, but the rapid onset of action (i.e. time to achieve 50% reduction in baseline PASI) is the fastest for brodalumab, followed by ixekizumab and then secukinumab [42]. IL-23 inhibitors lag behind by a few weeks but the difference is not on the order of months. Similarly, all biologics take a little more time to cross successive PASI thresholds with plateauing of PASI 75 coming before that of PASI 90 and PASI 100 across the board, even with IL-17 inhibitors. When considering an important life event or other extenuating circumstances, if one wishes to optimize for G. Han 230 rapidity of onset of action, an IL-23 inhibitor would likely yield to an IL-17 inhibitor. Prior Treatment with Biologics One useful consideration is to evaluate whether patients who have previously been on other biologics will respond to a given treatment. For tildrakizumab, the percent achieving PASI 75 was very similar whether or not a patient was on a biologic previously. The number achieving PGA 0/1 was slightly lower in biologic-experienced individuals (46.2% vs. placebo for tildrakizumab 100 mg) than for biologic-naïve patients (52.5% vs. placebo for tildrakizumab 100 mg) [43]. Inflammatory Bowel Disease For patients with concerns about inflammatory bowel disease (either a personal history or a strong family history), a clear choice in this realm would be either a TNF-α inhibitor, an IL-12/23, or an IL-23 inhibitor. The former two classes of medications are approved for treatment of Crohn’s disease/IBD while IL-23 inhibitors have already reported promising Phase II data in the treatment of IBD. Conversely, IL-17 inhibitors have been found to exacerbate IBD, as previously outlined [24]. Metabolic Syndrome Given that metabolic syndrome is a common comorbidity afflicting patients with psoriasis, it should be noted that some reports dealing with tildrakizumab treatment have specifically looked at this subject. One report showed that patients with metabolic syndrome in the reSURFACE trials had a higher incidence of cardiovascular disorders (50.6% for patients with metabolic syndrome vs. 16.9% without) and psoriatic arthritis (21.5% for patients with metabolic syndrome vs. 14.8% without). Patients with metabolic syndrome were also heavier on average, with a mean weight at baseline of 106.9 Kg (vs. 82.9 Kg without metabolic syndrome). The responses of both groups of patients (with metabolic syndrome and without), notwithstanding the stark difference in body weight among other factors, was very similar in all outcome measures studied—PASI75, PASI90, PASI100, and mean PASI improvement [44]. Further data are needed about disease-specific modifying factors, as there is a logical argument that reducing levels of pro-­ inflammatory cytokines in the bloodstream by treating psoriasis with a biologic could reduce atherosclerosis and cardiovascular risk, which has been shown to hinge on similar cytokines and specifically IL-23 [44]. Withdrawal and Retreatment Tildrakizumab has been shown to have a relatively long duration of effect even after medication withdrawal, possibly due to the upstream/ regulatory function of IL-23 in psoriasis. In part 3 of the phase II trial, some patients were followed for 20 additional weeks off treatment after the 52-week treatment period—less than 10% relapsed in the tildrakizumab 100 mg group (maximum PASI improvement reduced by 50% or more) [1]. In a separate Phase III trial, 49% of patients on tildrakizumab 100 mg treatment maintained their PASI75 response 36 weeks after cessation of treatment [45]—this is a different measure and a higher bar than the previously defined relapse metric. Average time to relapse for tildrakizumab 100 mg treated patients was 226 days. In the same study, it was shown that retreatment with tildrakizumab 100 mg for at least 12 weeks was able to achieve PASI75 in 85.7% of patients. Pregnancy With regards to pregnancy, animal studies showed no evidence of embryotoxicity or teratogenicity, per the FDA label for tildrakizumab. The only data available for human pregnancy thus far comes from a total of 13 patients who became pregnant in the clinical trials who were then 17 Tidrakizumab removed from the study drug. Out of these patients, all pregnancies carried to full term resulted in normal live births and the rates of spontaneous abortion were similar to the general population (2 out of 13) [46]. Interestingly, given the above duration of effect for tildrakizumab where about half of patients maintain PASI75 response 36 weeks after discontinuation of treatment, it is likely that patients will continue receiving efficacy throughout most of their pregnancy. Given that IgG is transferred across the placenta only after week 13 of pregnancy, and increases to peak levels in the third trimester [47], there should be little concern in giving tildrakizumab to a woman of child-bearing age. If a patient finds out she is pregnant and wishes to stop the medication, it will be at least two half-­ lives of the medicine later that IgG can even be transferred across the placenta given the half-life of tildrakizumab. Conclusions Tildrakizumab is an effective treatment for plaque psoriasis that appears to have an extremely positive safety profile and good durability of treatment response. It is unfortunate that while this medication entered Phase III trials 2 years before any other IL-23 inhibiting medication, it was not the first to market and commercialization of the drug was delayed. In the context of other IL-23 targeted therapeutics, the efficacy is somewhat less when looking at PASI threshold attainment and clinical results may take somewhat longer to peak, but it should be noted that on average, improvements on this drug are still quite remarkable given what our expectations from biologics were even just a few years ago. The safety profile of tildrakizumab is certainly a strength of the medication, and allows for its confident usage in a wide array of patients. Further positive attributes of tildrakizumab center around its convenient dosing schedule and potential efficacy in psoriatic arthritis. It remains to be seen whether this drug will find a notable position in our treatment arsenal for psoriasis given the crowded marketplace, but there is no reason that 231 one should not think of tildrakizumab as a reasonable first-line option in treating psoriasis given its efficacy, safety, durability, and potential to treat psoriatic arthritis. References 1. Papp K, Thaci D, Reich K, Riedl E, Langley RG, Krueger JG, Gottlieb AB, Nakagawa H, Bowman EP, Mehta A, Li Q, Zhou Y, Shames R. Tildrakizumab (MK-3222), an anti-interleukin-23p19 monoclonal antibody, improves psoriasis in a phase IIb randomized placebo-controlled trial. Br J Dermatol. 2015;173(4):930–9. 2. Iwakura Y, Ishigame H. The IL-23/IL-17 axis in inflammation. J Clin Invest. 2006;116(5):1218–22. 3. Gandhi M, Alwawi E, Gordon KB. Anti-p40 antibodies ustekinumab and briakinumab: blockade of interleukin-12 and interleukin-23 in the treatment of psoriasis. Semin Cutan Med Surg. 2010;29(1): 48–52. 4. Chen X, Tan Z, Yue Q, Liu H, Liu Z, Li J. The expression of interleukin-23 (p19/p40) and interleukin-12 (p35/p40) in psoriasis skin. J Huazhong Univ Sci Technolog Med Sci. 2006;26(6):750–2. 5. de Beaucoudrey L, Samarina A, Bustamante J, Cobat A, Boisson-Dupuis S, Feinberg J, Al-Muhsen S, Janniere L, Rose Y, de Suremain M, Kong XF, Filipe-­ Santos O, Chapgier A, Picard C, Fischer A, Dogu F, Ikinciogullari A, Tanir G, Al-Hajjar S, Al-Jumaah S, Frayha HH, AlSum Z, Al-Ajaji S, Alangari A, Al-Ghonaium A, Adimi P, Mansouri D, Ben-­ Mustapha I, Yancoski J, Garty BZ, Rodriguez-Gallego C, Caragol I, Kutukculer N, Kumararatne DS, Patel S, Doffinger R, Exley A, Jeppsson O, Reichenbach J, Nadal D, Boyko Y, Pietrucha B, Anderson S, Levin M, Schandene L, Schepers K, Efira A, Mascart F, Matsuoka M, Sakai T, Siegrist CA, Frecerova K, Bluetters-Sawatzki R, Bernhoft J, Freihorst J, Baumann U, Richter D, Haerynck F, De Baets F, Novelli V, Lammas D, Vermylen C, Tuerlinckx D, Nieuwhof C, Pac M, Haas WH, Muller-Fleckenstein I, Fleckenstein B, Levy J, Raj R, Cohen AC, Lewis DB, Holland SM, Yang KD, Wang X, Wang X, Jiang L, Yang X, Zhu C, Xie Y, Lee PP, Chan KW, Chen TX, Castro G, Natera I, Codoceo A, King A, Bezrodnik L, Di Giovani D, Gaillard MI, de Moraes-Vasconcelos D, Grumach AS, da Silva Duarte AJ, Aldana R, Espinosa-­ Rosales FJ, Bejaoui M, Bousfiha AA, Baghdadi JE, Ozbek N, Aksu G, Keser M, Somer A, Hatipoglu N, Aydogmus C, Asilsoy S, Camcioglu Y, Gulle S, Ozgur TT, Ozen M, Oleastro M, Bernasconi A, Mamishi S, Parvaneh N, Rosenzweig S, Barbouche R, Pedraza S, Lau YL, Ehlayel MS, Fieschi C, Abel L, Sanal O, Casanova JL. Revisiting human IL-12Rbeta1 deficiency: a survey of 141 patients from 30 countries. Medicine (Baltimore). 2010;89(6):381–402. 18 Risankizumab Erica B. Lee, Deeti J. Pithadia, Kelly A. Reynolds, and Jashin J. Wu Abstract Risankizumab is a humanized IgG1 monoclonal antibody that targets the p19 subunit of interleukin-23 (IL-23). It has demonstrated substantial clinical efficacy in the treatment of psoriasis. This chapter reviews the efficacy and safety of risankizumab for the treatment of psoriasis as evidenced by the clinical trials to date. Four phase III trials have shown risankizumab to be more effective than ustekinumab and adalimumab in terms of PASI and sPGA scores with similar safety profiles to placebo. Thus far, no significant safety concerns have emerged for this drug, though further long-term studies will be critical in confirming this. E. B. Lee Department of Dermatology, University of Nebraska Medical Center, Omaha, NE, USA D. J. Pithadia Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA K. A. Reynolds Department of Internal Medicine, Kettering Hospital Network, Kettering, OH, USA e-mail: kpassabe@alumni.nd.edu J. J. Wu (*) Dermatology Research and Education Foundation, Irvine, CA, USA © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_18 Key Learning Objectives 1. To understand the mechanism of action of Risankizumab 2. To understand the appropriate use and efficacy of the Risankizumab 3. To understand the safety issues of Risankizumab Introduction Interleukin-23 (IL-23) is a heterodimer of a p40 subunit shared by IL-12 and IL-23 and a p19 subunit that is exclusive to IL-23 [1, 2]. Since its discovery in 2000, it has been identified as a key inflammatory cytokine involved the pathogenesis of psoriasis [2, 3]. Ustekinumab, an IL-12/23 inhibitor, has demonstrated effectiveness in treating psoriasis and other autoimmune diseases [4–7]. However, more recent evidence has shown that psoriatic skin lesions have elevated levels of the IL-23 p40 and p19 subunits but normal levels of the IL-12 specific subunit, p35 [8]. In a phase I study, Kopp et al. later confirmed that specific inhibition of IL-23 by tildrakizumab improves psoriatic symptoms [9]. Moreover, molecular and histopathologic profiles of skin from patients treated with an IL-23 inhibitor were shown to have “excellent improvement” in global histopathologic scores at significantly higher rates than patients treated with ustekinumab [10]. 235 E. B. Lee et al. 236 Risankizumab (AbbVie Pharmaceuticals, Inc., Chicago, IL) is a humanized IgG1 monoclonal antibody that specifically targets and inhibits the p19 subunit of interleukin (IL)-23 [11]. It is the third IL-23 inhibitor to be approved for the treatment of moderate-to-severe plaque psoriasis (following guselkumab and tildrakizumab) [12]. It is also being investigated for use in Crohn’s disease, ankylosing spondylitis, atopic dermatitis, ulcerative colitis, asthma, and hidradenitis suppurativa [13–20]. Pharmacology A phase I, single-rising-dose, placebo-controlled trial of patients with moderate-to-severe plaque psoriasis investigated the safety, efficacy, pharmacokinetics, and biomarker results of risankizumab [21]. Eighteen patients received a single intravenous (IV) dose of 0.01, 0.05, 0.25, 1, 3, or 5 mg/kg and 13 patients received a single dose of 0.24 or 1 mg/kg subcutaneously. In this study, risankizumab showed linear pharmacokinetics with mean clearance of 0.33 L/day and a mean terminal phase volume of distribution of 10.8 L. Risankizumab’s half-life ranged between 20 and 28 days. Maximal exposure ranged from 4 to 10 days in patients receiving risankizumab subcutaneously. Bioavailability was 59% for both subcutaneous and IV administration. These findings were confirmed by phase I and II trials in subjects with psoriasis and Crohn’s disease [22]. Based on the results of these analyses, clearance was estimated to be 0.35 L/day. Steadystate volume of distribution was estimated to be 11.7 L, and terminal-phase elimination half-life was calculated as 27 days. In patients receiving risankizumab subcutaneously, bioavailability was 72%. It was also found that higher body weight may have modest effect on risankizumab levels, particularly in patients greater than 100 kg. Upon incorporation of data from phase III clinical trials, clearance was found to be relatively similar to previous estimates at 0.31 L/ day [23]. Calculated steady-state volume of distribution and terminal-phase elimination halflife were also consistent at 11.2 L and 28 days, respectively. Bioavailability when given subcutaneously was markedly higher than previous findings at 89%. Steady-state plasma exposure was attained by week 16 of dosing. Dosing and Monitoring The standard dosing of risankizumab is 150 mg (two 75 mg injections) administered subcutaneously at week 0, week 4, then every 12 weeks. Prescribing information recommends that all patients undergo evaluation for tuberculosis infection prior to beginning risankizumab [24]. The guidelines for treatment with biologics published by the American Academy of Dermatology (AAD) and the National Psoriasis Foundation (NPF) also suggest that patients be screened with a complete blood count, complete metabolic profile, serologic tests for tuberculosis, hepatitis B and C, and possibly HIV testing at the discretion of the prescriber [25]. With regard to ongoing monitoring, the AAD/NPF guidelines suggest yearly tuberculosis screening in high-risk patients (patients in contact with individuals with active TB and in patients with an underlying medical condition). All other patients may be screened annually at the discretion of the provider. Patients should also have regular follow-up visits with their dermatologists, which should include screening for nonmelanoma skin cancer, hepatitis B and C, adverse effects, and infections [25]. Clinical Efficacy Phase I and II trials have demonstrated the efficacy of risankizumab in treating patients with moderate-to-severe psoriasis. In the previously mentioned phase I trial, 87% of patients treated with a single dose of risankizumab achieved at least a 75% decrease in the Psoriasis Area and Severity Index (PASI), with 58 and 16% achieving PASI 90 and PASI 100, respectively, by week 12 [21]. The phase II trial directly compared risankizumab to ustekinumab in 166 patients randomized to receive either risankizumab 18, 90, 18 Risankizumab or 180 mg (weeks 0, 4, and 16) or ustekinumab 45 or 90 mg (based on body weight, weeks 0, 4, and 16) [11]. At week 12, the proportion of patients achieving PASI 90 was 77% in those treated with risankizumab, compared to 40% of ustekinumab patients (P < 0.001). Moreover, 98% of the patients in the 90-mg risankizumab group achieved PASI 75, with 80% of patients reaching this endpoint as early as week 8. Four phase III trials have formally evaluated the safety and efficacy of risankizumab: UltIMMa-1, UltIMMa-2, IMMvent, and IMMhance. UltIMMa-1 and UltIMMa-2 were replicate, randomized, double-blind, placebo- and active-comparator-controlled trials [26]. Patients were randomized to receive either 150 mg risankizumab (week 0, 4, then every 12 weeks), 45 or 90 mg ustekinumab (based on weight per label), or placebo for the first 16 weeks of the study. From weeks 16 to 52, patients initially treated with placebo received risankizumab, while all others maintained their original treatment. Co-primary endpoints included the percentage of patients achieving PASI 90 and a static Physician’s Global Assessment (sPGA) of 0 or 1 at week 16. In UltiMMa-1, risankizumab outperformed both placebo and ustekinumab in terms of the primary endpoints. PASI 90 was achieved by 229 of 304 patients receiving risankizumab (75.3%) compared to 5 of 102 (4.9%) receiving placebo and 42 of 100 (42.0%) treated with ustekinumab at week 16 (P < 0.0001 vs. placebo and ustekinumab). By the first measured time point (week 4), the proportion of patients on risankizumab attaining PASI 90 was significantly higher than those treated with placebo, and by week 8, significantly higher than ustekinumab (P = 0.0001). Findings were similar in terms of sPGA scores, with 87.8% of patients reaching sPGA 0 or 1 versus 7.8 and 79.9% of patients receiving placebo and ustekinumab, respectively (P < 0.0001). Moreover, 35.9% of patients receiving risankizumab achieved PASI 100, or complete clearance of psoriatic lesions, by week 16, compared to 12% of patients receiving ustekinumab and 0% receiving placebo. At the end of the study period, 81.9% of patients on continuous risankizumab and 78.4% of patients who were switched from placebo at week 16 achieved PASI 90. 237 The results of UltiMMa-2 also demonstrated superior efficacy of risankizumab compared to placebo and ustekinumab. At week 16, 74.8% (220 of 294) of patients in the risankizumab group achieved PASI 90, versus 2.0% (2 of 98) and 47.5% (47 of 99) in the placebo and ustekinumab groups, respectively (P < 0.0001 vs. both placebo and ustekinumab). In addition, an sPGA 0/1 score was attained by 83.7% of risankizumab patients, 5.1% of placebo patients, and 61.6% of ustekinumab patients (P < 0.0001 vs. placebo and ustekinumab). In this trial, 50.7% of patients treated with risankizumab experienced complete clearance of psoriatic lesions at week 16; this increased to 59.5% in patients on continuous risankizumab by week 52. The IMMvent trial was a double-blind, active-­ comparator trial that assessed the safety and efficacy of risankizumab compared to the TNF inhibitor adalimumab [27]. In the first phase of the trial, patients were randomly assigned to therapy with either risankizumab 150 mg (subcutaneous injection at weeks 0, 4, then every 12 weeks) or adalimumab (80 mg at week 0, 40 mg at week 1, then 40 mg every other week thereafter). During weeks 16–52, those who were originally assigned to adalimumab were regrouped based on their response at 16 weeks: those with less than PASI 50 response switched to risankizumab, those with PASI 90 remained on adalimumab, and those with response between PASI 50 and PASI 90 were re-randomized to either initiate risankizumab or continue adalimumab. Patients originally assigned to risankizumab continued their treatment with dosing every 12 weeks. Although the data has not yet been formally published, it has been reported that risankizumab performs significantly better than adalimumab both in terms of the primary endpoints at week 16 (PASI 90 and sPGA 0/1), and the secondary endpoint, PASI 100 at week 44 [27]. Of 301 patients in the risankizumab group, 72% achieved PASI 90, compared to 47% of 304 patients in the adalimumab group at week 16. Eighty-four percent of risankizumab patients attained sPGA 0/1 at this time point, versus 60% of adalimumab patients. In addition, of those who switched to risankizumab at week 16, 40% achieved PASI 100 at 238 week 44. Preliminary results were not reported for patients who had been on risankizumab for duration of the entire trial. The fourth phase III clinical trial, IMMhance, was a placebo-controlled study comparing the safety and efficacy of continuous treatment and randomized withdrawal of risankizumab over the course of 104 weeks [28, 29]. In the initial phase, subjects were randomized to receive either risankizumab 150 mg or placebo for the first 28 weeks. The primary endpoints were PASI 90 and sPGA 0/1. In the second phase, risankizumab patients who achieved sPGA 0/1 were re-­randomized to continue risankizumab (maintenance) or placebo (withdrawal). Starting at week 32, any patient who experienced a relapse, defined as sPGA score of 3 or greater, was reinitiated on risankizumab (injection given immediately, 4 weeks later, then every 12 weeks). The primary endpoint for the second phase of the study was sPGA 0/1 at week 52. At week 16, 73% of patients assigned to risankizumab achieved PASI 90, and 84% attained sPGA 0/1, compared 2 and 7%, respectively, in the placebo group. At week 52, the proportion of patients in the continuous risankizumab group maintaining sPGA 0/1 was 87%, versus 71% in the withdrawal group (P < 0.001). These findings suggest that like other biologics, the efficacy of risankizumab is maximized with continuous treatment rather than periods of discontinuation and re-initiation. Safety As risankizumab is one of the newest biologics to be approved, safety data is limited to what has been reported in clinical trials. Adverse events (AEs) were reported in 65% (20 of 31) of patients followed for 24 weeks after a single dose of risankizumab in the phase I trial, though severity of adverse events did not appear to correlate with the dose received [21]. The most common adverse events reported were mild-to-moderate upper respiratory tract infections (10%), mild nasopharyngitis (13%), and mild-to-moderate headache (10%). Four serious AEs (SAEs) were reported concurrently with risankizumab ther- E. B. Lee et al. apy. These included alcoholic pancreatitis, ischemic thalamic stroke, transient ischemic attack, and polymyositis; investigators considered each of these independent of the study medication. Injection-site reactions were reported in 2 of 24 patients receiving risankizumab intravenously, while no reactions were reported in the subcutaneous administration group. The UltIMMa phase III trials demonstrated similar frequency of AEs among individuals treated with risankizumab and those treated with ustekinumab [26]. At the end of week 16 in UltIMMa-1, 49.7% of patients receiving risankizumab and 50.0% of patients receiving ustekinumab reported adverse events. Seven risankizumab patients reported SAEs in this time period, including one serious infection and one squamous cell carcinoma. Findings were similar in UltIMMa-2; 45.6 and 53.5% of patients reported any AE in the risankizumab and ustekinumab groups, respectively, by the end of week 16. Six SAEs were reported among risankizumab-­ treated patients, including three serious infections and one basal cell carcinoma. Across both trials, the most commonly reported adverse events were upper respiratory infection, psoriasis, and diarrhea. No tuberculosis, opportunistic infections, major adverse cardiovascular events (MACEs) or serious hypersensitivity reactions were reported. Overall rates of AEs per patient-years (PY) across the entire 52-week durations of UltIMMa-1 and UltIMMa-2 were 245.6 and 281.0 for risankizumab and ustekinumab, respectively [27]. Serious AEs occurred at a rate of 9.8 per 100 PY on risankizumab, compared to 1.9 per 100 PY on ustekinumab. Of these, serious infections occurred slightly less frequently in the risankizumab group (1.6 per 100 PY vs. 2.5 per 100 PY with ustekinumab), though MACE (0.5 per 100 PY vs. 0.0 per 100 PY), malignancy of any kind (1.5 per 100 PY vs. 0.5 per 100 PY), and death (0.3 per 100 PY vs. 0.0 per 100 PY) occurred with higher frequency in patients on risankizumab. One risankizumab patient died from sudden cardiac arrest 101 days after the last dose of risankizumab, and a second died of an unknown cause 161 days after the last 18 Risankizumab dose. Moreover, an additional risankizumabtreated patient in the IMMvent trial died of an acute myocardial infarction on study day 73. The IMMhance trial also reported death of one patient on risankizumab; this occurred on study day 263 and was ruled to be due to a MACE [29]. All of these patients had cardiovascular risk factors prior to initiation of the drug. It is important to consider that the sample sizes of the trials conducted so far are still relatively small; further research and larger population sizes are needed to determine whether risankizumab may have any serious long-term adverse effects. Pooled data from all phase III clinical trials found that 24% (263 of 1079) of patients treated with risankizumab were positive for anti-­risankizumab antibodies by week 52 [24]. Of these, about 57% (14% of all risankizumab-­ treated patients) were deemed to have neutralizing antibodies. However, higher antibody levels were associated with lower risankizumab concentrations and reduced clinical efficacy in only 1% of patients. Pregnancy and Lactation Available data regarding the safety of use in pregnancy and lactation is limited to studies in cynomolgus monkeys [24]. It is known that risankizumab may be transmitted from mother to fetus or infant, as human IgG is able to cross the placenta and is present in breastmilk. At doses that are 20 times the maximum recommended human dose (2.5 mg/kg based on administration of a 150 mg dose to a 60 kg individual), increased fetal or infant loss was found in pregnant monkeys. However, no effects on functional or immunological development related to risankizumab were observed in infant monkeys from birth through 6 months. Conclusion The elucidation of IL-23 as an important cytokine in the pathogenesis of psoriasis has led to great advances in the development of effective 239 biologic therapies. The advent of IL-23 inhibitors has provided psoriasis patients with novel options with exceptional clinical efficacy seen in the phase III clinical trials. However, it is important to consider the paucity of long-term efficacy and safety data. Further studies are undoubtedly necessary to specify the role of IL-23 inhibitors in the treatment of psoriasis.Conflicts of InterestDr. Wu is an investigator for AbbVie, Amgen, Eli Lilly, Janssen, Novartis; a consultant for AbbVie, Almirall, Amgen, Bristol-Myers Squibb, Celgene, Dermira, Dr. Reddy’s Laboratories, Eli Lilly, Janssen, LEO Pharma, Novartis, Promius Pharma, Regeneron, Sun Pharmaceutical, and UCB, Valeant Pharmaceuticals North America LLC; and a speaker for AbbVie, Celgene, Novartis, Regeneron, Sanofi Genzyme, Sun Pharmaceutical, UCB, Valeant Pharmaceuticals North America LLC. The rest of the authors have no conflicts to disclose. References 1. Oppmann B, Lesley R, Blom B, et al. Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12. Immunity. 2000;13:715–25. https://doi. org/10.1016/S1074-7613(00)00070-4. 2. Girolomoni G, Strohal R, Puig L, et al. The role of IL-23 and the IL-23/TH 17 immune axis in the pathogenesis and treatment of psoriasis. J Eur Acad Dermatol Venereol. 2017;31(10):1616–26. https://doi. org/10.1111/jdv.14433. 3. Chan JR, Blumenschein W, Murphy E, et al. IL-23 stimulates epidermal hyperplasia via TNF and IL-20R2-dependent mechanisms with implications for psoriasis pathogenesis. J Exp Med. 2006;203(12):2577–87. https://doi.org/10.1084/jem. 20060244. 4. Leonardi CL, Kimball AB, Papp KA, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 76-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 1). Lancet. 2008;371(9625):1665–74. https://doi.org/10.1016/ S0140-6736(08)60725-4. 5. Papp KA, Langley RG, Lebwohl M, et al. Efficacy and safety of ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with psoriasis: 52-week results from a randomised, double-blind, placebo-controlled trial (PHOENIX 2). Lancet. 2008;371(9625):1675–84. https://doi.org/10.1016/ S0140-6736(08)60726-6. Secukinumab for the Treatment of Inflammatory Skin and Joint Disease 19 Jason E. Hawkes and Avishan Vishi Hawkes Abstract and rheumatology. While the exact roles of interleukin-17 in the development of inflammatory diseases is not entirely clear, ongoing research and clinical studies are expanding our knowledge base and leading to the development of new treatments that can improve clinical outcomes for patients affected by these conditions. Psoriasis is a chronic inflammatory skin condition characterized by thick, erythematous, scaly plaques commonly found on the extensor surfaces, scalp, and trunk. While the etiology of this skin condition has not been fully elucidated, research has unequivocally shown that psoriasis represents a bona fide T cell-­ mediated disease that involves the skin, joints, and a myriad of other organs/tissues. The Abbreviations recent discovery of a set of pathogenic T cells that produce high levels of interleukin-17 in ACR 20 American College of Rheumatolresponse to interleukin-23 led to a major paraogy 20 score digm shift in the pathogenic model for this AS Ankylosing spondylitis condition resulting in numerous novel targeted ASAS 20 Assessment of Spondyloarthritis immune therapies. The robust phase 3 clinical International Society 20 (ASAS trial program for anti-interleukin-17A via 20) response secukinumab (sold under the brand axSpA Axial spondyloarthritis Cosentyx®) for the treatment of moderate-to-­ DLQI Dermatology Life Quality Index severe plaque psoriasis, psoriatic arthritis, and FDA US Food and Drug Administration ankylosing spondylitis provides a solid founIBD Inflammatory bowel disease dation supporting the high clinical efficacy Ig Immunoglobulin and safety of this medication in dermatology IL-17 Interleukin-17 IL-23 Interleukin-23 NAPSI NAil Psoriasis Severity Index J. E. Hawkes (*) nr-axSpA Nonradiographic axSpA Department of Dermatology, University of California PASI Psoriasis Area and Severity Index Davis and PCN-Rocklin, Sacramento, CA, USA e-mail: jehawkes@ucdavis.edu PK Pharmacokinetic PPP Palmoplantar psoriasis A. V. Hawkes Regeneron Pharmaceuticals, Inc., ppPASI Palmoplantar Psoriasis Area and Tarrytown, NY, USA Severity Index e-mail: Vishi.Hawkes@regeneron.com © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_19 243 J. E. Hawkes and A. V. Hawkes 244 PsA PSSI TNF Psoriatic arthritis Psoriasis Scalp Severity Index Tumor necrosis factor selective blockade of IL-17A and IL-23p19. This book chapter will focus on secukinumab, the first FDA-approved monoclonal antibody to selectively target IL-17A for the treatment of psoriatic disease. Key Learning Objectives 1. To understand the mechanism of action of Secukinumab 2. To understand the appropriate use and efficacy of the Secukinumab 3. To understand the safety issues of Secukinumab Secukinumab: Background and Pivotal Clinical Trial Data The rapid onset, clinical efficacy, and safety profile of secukinumab underscores the importance of the IL-23/IL-17 signaling pathway in the pathogenesis of psoriasis and inflammatory joint diseases. Originally developed in 1996 Introduction from HuMab® mice (Medarex, Inc.) using a portion of the human immunoglobulin repertoire, Psoriasis is a chronic, inflammatory skin dis- secukinumab (previously known as AIN457) is ease that is estimated to affect approximately a fully human immunoglobulin (Ig)G1/κ mono3% of the US population [1]. Plaque psoriasis, clonal antibody that was subsequently tested the most common disease subtype, is typified in clinical trials by Novartis Pharmaceuticals by well-­ demarcated, scaly, thickened plaques Corporation for the treatment of psoriasis, on the scalp, trunk, and extensor surfaces of the rheumatoid arthritis, and uveitis [4]. The FDA extremities. Inflammatory involvement of the approved secukinumab for the treatment of joints, known as psoriatic arthritis (PsA), occurs moderate-­ to-severe plaque psoriasis in adults in approximately one-third of patients with pre-­ on January 21, 2015. Secukinumab is currently existing skin disease or can be the presenting sold under the trade name Cosentyx® and selecsymptoms of psoriasis. Other less common pso- tively targets the IL-17A isoform. It was the firstriasis variants include erythrodermic, pustular, in-­ class FDA-approved monoclonal antibody palmoplantar, guttate, and inverse subtypes. that directly inhibits the IL-17 immune axis [5]. The pathogenesis of psoriasis is a complex, Secukinumab was subsequently approved for the T-cell mediated skin and joint disease primar- treatment of ankylosing spondylitis (AS) and PsA ily driven by upregulation of the interleukin-23 in adults on January 15, 2016. Below is a sum(IL-­23)/IL-17 signaling pathway [2, 3]. While mary of pivotal clinical trials for secukinumab the exact cause of psoriasis is still unknown, for each approved clinical indication. the onset of disease is clearly modified by several factors including psoriasis-associated genes or susceptibility loci, environmental exposures, Plaque Psoriasis diet, medications, and infections. In the last two decades, science and clinical trials have rapidly For plaque psoriasis, secukinumab is adminexpanded our knowledge of psoriatic disease, istered subcutaneously with a loading dose of leading to the development and US Food and 300 mg (two 150 mg injections) at weeks 0, 1, Drug Administration (FDA) approval of a broad 2, 3, and 4, then monthly thereafter. The 150 mg array of topicals, oral immunosuppressants, and dose at the same dosing interval is available and injectable immunotherapies. This therapeutic may be sufficient for some patients. Key phase armamentarium includes 11 monoclonal anti- 3 clinical trials provide evidence demonstrating bodies (or biologics) whose targets range from the efficacy of secukinumab in the treatment of inhibition of tumor necrosis factor (TNF) to moderate-­to-severe plaque psoriasis: ERASURE, 19 Secukinumab for the Treatment of Inflammatory Skin and Joint Disease FIXTURE, and CLEAR trials. Additional, subsequent phase 3 trials built upon and bolster the clinical data obtained from these three major trials. ERASURE was a double-blind, 52-week, placebo-­ controlled trial with 738 patients enrolled into secukinumab (300 or 150 mg weekly for 5 weeks, then monthly) versus placebo [6]. 75% improvement in the Psoriasis Area and Severity Index (PASI 75) for patients in the secukinumab treatment groups (300 or 150 mg) were 82 and 72%, respectively. Less than 5% of patients achieved a PASI 75 in the placebo arm. The FIXTURE trial was also a double-blind, 52-week, placebo-controlled with 1306 patients randomized to treatment with secukinumab (300 and 150 mg as above) or etanercept (50 mg twice weekly for 12 weeks, then weekly) versus placebo [6]. In this trial, a PASI 75 was achieved in 77 and 67% of patients by week 12 in the 300 and 150 mg treatment arms, respectively. In comparison, only 44% of patients achieved a PASI 75 with etanercept versus 5% in the placebo treatment arm. Importantly, a pooled analysis of the data from ERASURE and FIXTURE also showed ~95% of patients who had achieved a PASI 75 in the core clinical trials were able to achieve this same response by 12 weeks following secukinumab treatment interruption (data presented at the 2017 American Academy of Dermatology Annual Meeting in Orlando, Florida). The CLEAR study was a subsequent 52-week, double-blind, randomized controlled trial that enrolled 676 patients to treatment with secukinumab (300 mg weekly for 5 weeks, then monthly) versus ustekinumab (45 or 90 mg at weeks 0, 4, then every 12 weeks based on body weight) [7]. Due to the high efficacy shown for both treatments from previous studies, a more stringent primary study endpoint of PASI 90 instead of PASI 75 was used in the ERASURE and FIXTURE studies. The benchmark of PASI 90 as a primary study endpoint would continue to be used in subsequent studies for newer, highly effective biologic agents tested for the treatment of psoriasis. At week 16, 79% of psoriasis patients achieved a PASI 90 in the secukinumab group 245 compared to only 58% from the ustekinumab group. The distinct superiority of secukinumab compared to ustekinumab was still evident following 1 year of treatment [8], and secukinumab was strongly associated with a significantly higher Dermatology Life Quality Index. The SCULPTURE trial reported a similar, sustained clinical efficacy and safety for secukinumab through 5 years [9]. Finally, the JUNCTURE [10] and FEATURE [11] trials also demonstrated the efficacy and safety of secukinumab administered for auto-injector/pen and pre-filled syringes, and clinical results were comparable with previous studies. ifficult to Treat Psoriasis Subtypes: D Palmoplantar, Nail, and Scalp Psoriasis Most psoriasis clinical trials to date have been designed for plaque psoriasis or PsA, excluding less common disease subtypes and patients with disease that predominantly involves the scalp, palmoplantar, intertriginous, or genital skin. Scalp psoriasis, palmoplantar psoriasis (PPP), and nail abnormalities have historically been classified as more difficult to treat psoriasis areas. The exact reasons for the patterned expression or recalcitrant nature of disease in these specific skin areas is not currently known. However, disproportionate environmental triggers (e.g. increased trauma) and/or differences in resident immune cells in these areas of skin represent a possible explanation for these observations. Secukinumab was one of the first psoriasis biologics evaluated in trials specifically designed for patients with these difficult to treat subtypes in the GESTURE, TRANSFIGURE, and scalp psoriasis trials. GESTURE was a phase 3, randomized, double-­ blind, placebo-controlled, multicenter, trial conducted across 15 countries and consisted of 132 weeks of treatment for PPP patients [12]. 205 subjects were randomized to secukinumab 300 or 150 mg versus placebo. The primary endpoint was Palmoplantar Investigator’s Global Assessment (ppIGA) 0 (clear) or 1 (almost clear/ minimal) after 16 weeks of treatment. At week J. E. Hawkes and A. V. Hawkes 246 16, 33 and 22% of patients achieved clear or almost clear in the 300 and 150 mg treatments groups, respectively, compared to only 1.5% in the placebo treatment arm. This clinical response was accompanied by significant reductions in Palmoplantar Psoriasis Area and Severity Index (ppPASI) and higher Dermatology Life Quality Index (DLQI) 0/1 responses. Continued clinical improvement and a sustained efficacy was observed in moderate-to-severe PPP patients on continuous secukinumab treatment for up to 2.5 years [13]. In a very similar design as the GESTURE trial, TRANSFIGURE was conducted across 39 study locations and consisted of 132 weeks of secukinumab treatment for psoriatic nail involvement [14]. The primary study endpoint was improvement in the NAil Psoriasis Severity Index (NAPSI) after 16 weeks of secukinumab treatment. The primary endpoint was met demonstrating mean nail improvement in 73 and 64% of patients treated with secukinumab 300 and 150 mg, respectively. Improved NAPSI scores were sustained with continuous treatment over 2.5 years and was associated with multiple improved patient reported outcomes and Quality of Life indices. Finally, secukinumab was studied for the treatment of scalp psoriasis in a phase 3, double-­ blind, randomized trial of 102 psoriasis patients across 17 study locations [15]. In this 24-week study, patients were randomized to secukinumab 300 mg at weeks 0, 1, 2, 3, and 4 followed by monthly injections for 20 weeks versus placebo. The primary study measure was 90% improvement of Psoriasis Scalp Severity Index (PSSI 90) score from baseline to week 12. The proportion of patients achieving a PSSI 90 response at week 12 was 53% for the secukinumab 300 mg treatment group compared to only 2% of patients receiving placebo. More than one-third of patients receiving secukinumab achieved complete clearance of the scalp at week 12, ­ whereas 0% of patients receiving placebo had complete scalp clearance. Psoriatic Arthritis For PsA patients with coexistent skin disease, secukinumab is dosed and administered as above for moderate-to-severe plaque psoriasis. For PsA with no skin disease, 150 mg of secukinumab can be administered with a loading dose (150 mg at weeks 0, 1, 2, 3, and 4, then monthly) or without a loading dosage (150 mg every 4 weeks). 300 mg dosing should be considered for PsA patients with persistent or recalcitrant clinical disease and symptoms despite receiving the 150 mg dosing of secukinumab for an adequate period of time to allow symptoms to resolve (usually 3–4 months). Two initial phase 3 clinical trials (FUTURE 1 and 2) evaluated the efficacy of secukinumab for the treatment of PsA. FUTURE 1 was a 52-week, double-blind, placebo-controlled trial that randomized 606 patients with PsA to treatment with secukinumab (10 mg/kg dose at weeks 0, 2, and 4, then 150 or 75 mg subcutaneously every 4 weeks) versus placebo [16]. Approximately 50% of patients achieved at least a 20% improvement in the American College of Rheumatology 20 score (ACR 20) for both treatment arms compared to only 17% in the placebo arm. This response was observed and sustained through 1 year after initiation of secukinumab treatment. FUTURE 2 was a 4-year study that enrolled and randomized 397 PsA to secukinumab (300, 150, or 75 mg monthly after weekly injections at weeks 0, 1, 2, 3, and 4) versus placebo [17]. After 24 weeks of secukinumab treatment, 54% (300 mg), 51% (150 mg), and 29% (75 mg) of patients treated with secukinumab achieved an ACR 20 versus 15% of patients in the placebo arm. At week 128, patients with inadequate response to 150 or 75 mg dosing were escalated to 300 mg based on the judgment of the trial investigators. The ACR 20 and ACR 50 response rates were maintained around 70–75% and 40–50%, respectively, between weeks 26 through 208. PASI 75/90 scores were slightly lower for patients with PsA compared to plaque psoriasis, but similar to those observed in the pivotal phase 3 trials for plaque psoriasis. 19 Secukinumab for the Treatment of Inflammatory Skin and Joint Disease FUTURE 5 was another randomized, double-­ blind, placebo-controlled clinical trial and the largest ever conducted for PsA patients receiving treatment with any biologic [18]. In this study, 996 patients were randomized to 52 weeks of treatment in four different groups: secukinumab 300 or 150 mg monthly after loading doses at weeks 0, 1, 2, 3, and 4; secukinumab 150 mg monthly with no loading dose; or placebo. The primary endpoint was ACR 20 response at week 16 and was achieved in 63, 56, and 60% of patients in the secukinumab 300 mg + loading, 150 mg + loading, and 150 mg with no loading treatments groups, respectively. 27% of patients receiving placebo achieved an ACR 20 response at week 16. The ACR 20 response was sustained through week 52 in all treatments arms with superiority observed in the secukinumab 300 mg + loading dose group. Importantly, the study also evaluated multiple secondary endpoints including radiographic progression of joint disease at week 52, complete response of enthesitis, and complete response of dactylitis, which were achieved in roughly half of patients for all treatment groups. To date, most clinical trials have not discriminated between subtypes of PsA (e.g. axial PsA). This may be due in part to our evolving understanding and terminology around inflammatory arthropathies. Regardless, knowing whether these subtypes have differential responses to specific treatments is an important question to address. Novartis will attempt to address this via an ongoing phase 3b clinical trial (ClinicalTrials.gov Identifier: NCT02721966) evaluating the safety and efficacy of secukinumab 150 or 300 mg in the treatment of axial PsA patients who have failed at least 2 non-steroidal anti-­inflammatory drugs for at least 1 month. ASAS 20 response with secukinumab 300 mg versus placebo at Week 12 is the listed primary study end point of this trial. This and other studies designed to tease out PsA subtypes prior to the trial design will be of interest to clinicians treating this group of patients with complex disease. 247 Ankylosing Spondylitis Axial spondyloarthritis (axSpA) is an inflammatory, spinal arthritis which can be severely disabling for patients due to chronic back pain that often manifests before the age of 45. Patients with axSpA are characterized as having either AS (also known as radiographic axSpA) or nonradiographic axSpA (nr-axSpA). This condition is also associated with psoriasis and shares overlapping clinical features with PsA, such as sacroiliitis, synovitis, enthesitis, dactylitis, and the HLAB27 gene [19, 20]. It is not surprising, therefore, that a significant proportion of patients with AS and PsA respond to treatment with secukinumab. Secukinumab for the treatment of AS is administered as 150 mg with or without a loading dose: 150 mg every 4 weeks or 150 mg at weeks 0, 1, 2, 3, and 4 followed by monthly injections. The efficacy of secukinumab for the treatment of active AS was studied in four pivotal phase 3, double-blind, randomized, placebo-controlled trials: MEASURE 1-4. MEASURE 1 enrolled and randomized 371 AS patients for treatment with intravenous secukinumab (10 mg/kg dose) at weeks 0, 2, and 4 followed by 150 or 75 mg every 4 weeks versus placebo [21]. The primary endpoint for MEASURE 1 and 2 was the proportion of patients with at least 20% improvement in the Assessment of Spondyloarthritis International Society (ASAS 20) response criteria at week 16. At week 16, ASAS 20 response rates were 61 and 60% for the 150 and 75 mg doses, respectively, compared to only 29% in the placebo group. In MEASURE 2, a total of 219 AS patients were enrolled and randomized to treatment for 3 years with subcutaneous secukinumab (150 or 75 mg) at weeks 0, 1, 2, 3, and 4 followed by monthly injections versus placebo [21]. Patients in the placebo group were then randomly switched to subcutaneous secukinumab (150 or 75 mg) at week 16. ASAS 20 response rates at week 16 were 61 and 41% for subcutaneous secukinumab (150 and 75 mg), respectively, compared to only 248 28% of patients in the placebo group. ASAS 40 response rates at week 16 for the 150 mg treatment group were 36% and gradually improved to 49% through week 52. It is important to note that significant improvement in AS symptoms were observed for patients treated with 150 mg of secukinumab in all dosing groups, whereas 75 mg of secukinumab was only significant with a higher loading dose. MEASURE 3 was a 52-week trial that evaluated the safety and efficacy of subcutaneous secukinumab 300 and 150 mg monthly maintenance dosing for AS following intravenous loading (10 mg/kg at weeks 0, 2, and 4) versus placebo [22]. 61 and 58% of patients achieved the ASAS 20 response rate target at week 16 for the 300 and 150 mg treatment arms, respectively, compared to 36.8% in the placebo group. As with prior studies, improvements in clinical disease were maintained from week 16 through week 52. The clinical improvements and ASAS20 response rates observed in MEASURE 3 were essentially reproduced in MEASURE 4 [23], which evaluated the efficacy and safety of secukinumab 150 mg via a self-administered prefilled syringe (with or without a loading regimen) in AS patients for 104 weeks. However, due to a high placebo response rate, the results in the study were not statistically significant. Pharmacologic Properties and Safety J. E. Hawkes and A. V. Hawkes erties for this medication have been established across three major multiple clinical indications and in a highly diverse demographic of patients. Secukinumab has also been used safely in plaque psoriasis, PsA, and AS patients for more than 5 years. Therefore, the above referenced phase 3 clinical trials have provided a robust data set that establishes the high efficacy and excellent safety of secukinumab across multiple indications. The adverse events reported in clinical trials for these three indications have been remarkably similar and are summarized in Table 19.1. The most commonly reported adverse events in approximately 10% of patients was headache and nasopharyngitis or upper respiratory infections. Less common adverse events included non-serious infections or infestations, diarrhea, arthralgias, mucocutaneous candidiasis, hypertension, and hypercholesterolemia. Rare adverse events (occurring in less than 1% of patients) included transient neutropenia, major adverse cardiac event, inflammatory bowel disease (IBD), serious infections, uveitis, and malignancy or unspecific tumor formation. Mucocutaneous candidiasis and IBD are two particular adverse events of interest for this particular class of medication. The development of candida and other viral infections as a result of IL-17 inhibition reflects the importance Table 19.1 Summary of adverse events for secukinumab observed in phase 3 clinical trials for the treatment of plaque psoriasis, psoriatic arthritis (PsA), and ankylosing spondylitis (AS) Common Adverse Events: >10% of patients • Nasopharyngitis or upper respiratory infections The pharmacologic and biochemical properties • Headache of secukinumab and its mode of administration Less Common Adverse Events: 9–1% of patients determine its observed clinical efficacy and safety • Diarrhea in treated patients. As a human IgG1-based anti- • Mucocutaneous candidiasis body targeting IL-17A, secukinumab shares the • Non-serious infection/infestation • Hypertension stability and long half-life of other IgG1 antibod- • Hypercholesterolemia ies in humans [24]. These properties, as well as • Arthralgias its abundance in the human body, were factors Rare Adverse Events: <1% of patients that certainly influenced the decision underlying • Inflammatory bowel disease the design of secukinumab. The pharmacokinetic • Serious infections • Injection site reactions (PK) properties of secukinumab have also been • Major adverse cardiac event established with a known half-life of ~25 days, • Uveitis high absolute bioavailability, and no dose-­ • Neutropenia dependent clearance [25]. The favorable PK prop- • Malignancy or unspecified tumors 19 Secukinumab for the Treatment of Inflammatory Skin and Joint Disease of this specific cytokine in providing humans and the skin with protection against this infection. Humans with inborn errors of IL-17RA or IL-17F are also associated with recurrent candida infections, upper respiratory infections, and mild skin infections that provides a natural experiment in human nature that reflects complete blockade of IL-17 signaling in the skin [26]. The potential association between IL-17 blockade with secukinumab and the subsequent development of IBD is complicated for several reasons. First, the families and individuals with inborn errors in IL-17 signaling (i.e. no production of IL-17 in the body) do not go on to develop any major sequela, such as IBD or malignancy. This observation argues against the suggested link between suppression of IL-17 by secukinumab and the development of IBD. Second, secukinumab and brodalumab (an IL-17 receptor inhibitor) were tested for clinical efficacy in patients with IBD and no prior history of psoriatic disease. Some of the patients in these trials reported worsening of their gastrointestinal symptoms [27, 28]. This has led some clinicians to exercise caution with IL-17 inhibitors when treating patients with IBD and a concomitant inflammatory condition, such as psoriasis or PsA. However, patients with psoriatic disease already have an increased risk for developing IBD compared to the general population, which suggests a potential genetic rather than treatment-­associated link [29]. Attempts to prove a causative association between IL-17 inhibition and the development of new-onset IBD in humans or mice are ongoing, but have thus far been inconclusive. Understanding the role of IL-17 in the mucosa and gastrointestinal system are critically important as we are just beginning to understand the role of the IL-23/IL-17 signaling in tissues beyond the skin. uture Direction of IL-17 Blockade F for Chronic Inflammatory Conditions The development of secukinumab and other IL-17 inhibitors (e.g. ixekizumab, brodalumab, and bimekizumab) have transformed the way 249 psoriasis is treated. Nevertheless, patients with psoriatic disease who are unresponsive to secukinumab or other agents in this drug class underscore the complexity of this disease, especially since many of these patients are responsive to broader-acting agents such as TNF inhibitors, methotrexate, or cyclosporine. The rapid progress in the treatment of skin manifestations of psoriasis with IL-17 antagonists compared to first- and second-generation biologic treatments (TNF inhibitors or ustekinumab) is astonishing in comparison to the minimal progress we have made in the treatment of inflammatory arthropathies with these same agents. A side-by-side comparison of the PASI and ACR scores from phase 3 clinical trials for biologics used for psoriasis accentuates the treatment gap for the treatment of skin versus joint disease [2]. This has raised some experts in the field to suggest a future treatment strategy that includes the concomitant use of IL-17 inhibitors with a TNF inhibitor or the use or novel molecules that simultaneously target two cytokines (e.g. TNF and IL-17A) via bispecific antibodies [30]. Recent phase 2 clinical data in plaque psoriasis for bimekizumab, which targets both IL-17A and IL-17F, is very encouraging and provides an excellent proof of concept for this novel therapeutic strategy [31]. Additional data about the testing of bispecific antibodies is also available for review [32, 33]. The anticipated high cost and/or increased adverse events associated with two simultaneously administered biologics are the most likely deterrents for this treatment approach. Preclinical or early phase 1 clinical trial data for bispecific antibodies in the treatment of inflammatory conditions are just starting to emerge. As technology continues to advance, the promise of dual cytokine inhibition for the treatment of chronic inflammatory diseases is strong and worth ongoing investigative efforts. The continued development of novel bispecific molecules in conjunction with translational research projects that investigate the role of IL-17 on the immune cells unique to specific tissues will help advance our ability to better manage these complex inflammatory conditions. The role of secukinumab and other IL-17 antagonists for 250 the treatment of neutrophilic dermatosis, such as pyoderma gangrenosum or Sweet syndrome, will also be of interest given the role of this cytokine on the upregulation of molecules that enhance the recruitment of neutrophils into the skin. Conclusion The central role for the IL-23/IL-17 signaling pathway in psoriasis and associated inflammatory joint diseases has broadened our understanding of the dysregulated immune events driving chronic inflammatory conditions and has led to the development of highly effective, targeted biologic therapies. As a first-in-class inhibitor of IL-17A, secukinumab has proven to be a rapid acting, robust, sustainable, and safe treatment for psoriasis, PsA, and AS. For this reason, it has become a first-line therapy for many patients, especially those with comorbidities or reservations about initiating treatment with traditional systemic immunosuppressants. Clinical trials specifically designed to investigate the efficacy and safety of secukinumab for the treatment of difficult to treat disease, such as PPP, nail disease, and scalp psoriasis, is unprecedented in the field and gives clinicians additional empiric evidence to make more informed clinical decisions for the treatment of psoriatic disease and AS. Acknowledgements None. Conflict of Interest JEH currently serves on the medical board of the National Psoriasis Foundation and has been a consultant for AbbVie, Janssen, LearnSkin, Novartis, Pfizer, Regeneron-Sanofi, VisualDx, and UpToDate. AVH is a current employee of Regeneron Pharmaceuticals, Inc. and receives stock in the company as part of her employment. References 1. Rachakonda TD, Schupp CW, Armstrong AW. Psoriasis prevalence among adults in the United States. J Am Acad Dermatol. 2014;70(3):512–6. https://doi.org/10.1016/j.jaad.2013.11.013. 2. Hawkes JE, Yan BY, Chan TC, Krueger JG. Discovery of the IL-23/IL-17 signaling pathway and the treat- J. E. Hawkes and A. V. Hawkes ment of psoriasis. J Immunol. 2018;201(6):1605–13. https://doi.org/10.4049/jimmunol.1800013. 3. Martin DA, Towne JE, Kricorian G, Klekotka P, Gudjonsson JE, Krueger JG, et al. The emerging role of IL-17 in the pathogenesis of psoriasis: preclinical and clinical findings. J Invest Dermatol. 2013;133(1):17– 26. https://doi.org/10.1038/jid.2012.194. 4. Fishwild DM, O’Donnell SL, Bengoechea T, Hudson DV, Harding F, Bernhard SL, et al. High-­ avidity human IgG kappa monoclonal antibodies from a novel strain of minilocus transgenic mice. Nat Biotechnol. 1996;14(7):845–51. https://doi. org/10.1038/nbt0796-845. 5. Hueber W, Patel DD, Dryja T, Wright AM, Koroleva I, Bruin G, et al. Effects of AIN457, a fully human antibody to interleukin-17A, on psoriasis, rheumatoid arthritis, and uveitis. Sci Transl Med. 2010;2(52):52ra72. https:// doi.org/10.1126/scitranslmed.3001107. 6. Langley RG, Elewski BE, Lebwohl M, Reich K, Griffiths CE, Papp K, et al. Secukinumab in plaque psoriasis—results of two phase 3 trials. N Engl J Med. 2014;371(4):326–38. https://doi.org/10.1056/ NEJMoa1314258. 7. Thaci D, Blauvelt A, Reich K, Tsai TF, Vanaclocha F, Kingo K, et al. Secukinumab is superior to ustekinumab in clearing skin of subjects with moderate to severe plaque psoriasis: CLEAR, a randomized controlled trial. J Am Acad Dermatol. 2015;73(3):400–9. https:// doi.org/10.1016/j.jaad.2015.05.013. 8. Blauvelt A, Reich K, Tsai TF, Tyring S, Vanaclocha F, Kingo K, et al. Secukinumab is superior to ustekinumab in clearing skin of subjects with moderate-­to-severe plaque psoriasis up to 1 year: results from the CLEAR study. J Am Acad Dermatol. 2017;76(1):60–9. e9. https://doi.org/10.1016/j.jaad.2016.08.008. 9. Bissonnette R, Luger T, Thaci D, Toth D, Lacombe A, Xia S, et al. Secukinumab demonstrates high sustained efficacy and a favourable safety profile in patients with moderate-to-severe psoriasis through 5 years of treatment (SCULPTURE Extension Study). J Eur Acad Dermatol Venereol. 2018;32(9):1507–14. https://doi.org/10.1111/jdv.14878. 10. Paul C, Lacour JP, Tedremets L, Kreutzer K, Jazayeri S, Adams S, et al. Efficacy, safety and usability of secukinumab administration by autoinjector/pen in psoriasis: a randomized, controlled trial (JUNCTURE). J Eur Acad Dermatol Venereol. 2015;29(6):1082–90. https://doi.org/10.1111/jdv.12751. 11. Blauvelt A, Prinz JC, Gottlieb AB, Kingo K, Sofen H, Ruer-Mulard M, et al. Secukinumab administration by pre-filled syringe: efficacy, safety and usability results from a randomized controlled trial in psoriasis (FEATURE). Br J Dermatol. 2015;172(2):484–93. https://doi.org/10.1111/bjd.13348. 12. Gottlieb A, Sullivan J, van Doorn M, Kubanov A, You R, Parneix A, et al. Secukinumab shows significant efficacy in palmoplantar psoriasis: results from GESTURE, a randomized controlled trial. J Am Acad Dermatol. 2017;76(1):70–80. https://doi. org/10.1016/j.jaad.2016.07.058. 20 Ixekizumab Caitriona Ryan and Roisin O’Connor Abstract Ixekizumab is a fully human monoclonal antibody that selectively targets IL-17A which has shown to be highly effective in the treatment of psoriasis and psoriatic arthritis. This chapter reviews the efficacy and safety profile of ixekizumab for the treatment of psoriasis based on clinical studies to date. Studies demonstrate that ixekizumab is highly effective in the treatment of moderate-to-severe psoriasis and the majority of patients achieve high clearance rates. Uncommon adverse events reported during ixekizumab therapy, included neutropenia, candida infections, and inflammatory bowel disease, however long-term safety data beyond 5 years is not yet available. C. Ryan (*) Institute of Dermatologists Ireland, Dublin, Ireland Charles Institute of Dermatology, University College Dublin, Dublin, Ireland e-mail: cryanoffice@instituteofdermatologists.ie R. O’Connor Department of Dermatology, Children’s Health Ireland (CHI) at Crumlin, Dublin, Ireland © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_20 Key Learning Objectives 1. To understand the mechanism of action of Ixekizumab 2. To understand the appropriate use and efficacy of the Ixekizumab 3. To understand the safety issues of Ixekizumab Introduction Ixekizumab (Taltz, Eli. Lilly) is a humanised IgG4 monoclonal antibody that selectively binds to IL-17A and inhibits its interaction with the IL-17 receptor [1, 2]. It has been consistently shown to be effective in the treatment of moderate-­to-severe psoriasis [3]. Ixekizumab has been approved in adults for the treatment of moderate-­ to-severe plaque psoriasis and also psoriatic arthritis [4]. Ixekizumab is composed of two light chain polypeptides of 219 amino acids each, and two heavy chain polypeptides of 445 amino acids each [5]. Ixekizumab selectively blocks IL-17A which is a member of the IL-17 family of cytokines along with IL-17B, IL-17C, IL-17D, IL-17E and IL-17F [1]. Il-17A is produced by CD4+ T-cells called Th17 cells which represent a subset of CD4+ helper lymphocytes [6]. The family of IL-17 homodimeric proteins differ in 253 C. Ryan and R. O’Connor 254 amino acid sequence and consequently in spatial structure [1]. Ixekizumab binds specifically to IL-17A [1]. The active form of IL-17A occurs as a homodimer (A/A) or a heterodimer (A/F) with IL-17F [1]. The physiological response to IL-17A involves the release of cytokines and chemokines which are responsible for recruiting and activating neutrophils and memory T-cells to the site of inflammation, and maintaining a pro-­inflammatory state [1]. Il-17A has a key role in chronic inflammation and autoimmunity [1]. Current data suggests that IL-17A has three main functions including host defence, neutrophil homeostasis, and chronic pathogenic inflammation [1, 7, 8]. The discovery of IL-17, which plays a critical role in the development of psoriasis, has opened many new treatment options. The class of IL-17 inhibitors includes secukinumab, ixekizumab, brodalumab, bimekizumab, netakimab and M1095 [9]. Pharmacodynamics Ixekizumab targets the IL-17 cytokine pathway which plays a key role in the pathogenesis of psoriasis. Phase 1 data based on psoriatic skin biopsies demonstrate a dose-dependent reduction of epidermal thickness, a reduction in proliferating keratinocytes, T cells, dendritic cells and reductions in inflammatory markers including C-reactive protein from baseline to day 43 [9, 10]. Pharmacokinetics Mean peak concentrations occur between 4 and 7 days following the administration of one subcutaneous dose of ixekizumab in patients with psoriasis, with doses ranging from 5 to 160 mg [10]. Analyses suggest the average bioavailability of ixekizumab following subcutaneous injection is 54–0%. After the 160 mg starting dose, the mean (SD) maximum plasma concentration (Cmax) of ixekizumab is 19.9 (8.15) μg/ml. Steady state is achieved by week 8 following the initial starting dose of 160 mg and the 80 mg Q2W dosing sched- ule. Estimated mean (SD) of Cmax,ss is 21.5 (9.16) μg/ml, and Ctrough,ss is 5.23 (3.19) μg/ml. After swapping from the 80 mg Q2W dosing schedule to the 80 mg Q4W dosing regimen at Week 12, steady state is reached after approximately 10 weeks. Estimated mean (SD) of Cmax,ss, is 14.6 (6.04) μg/ml and Ctrough,ss is 1.87 (1.30) μg/ml [10]. Clinical Efficacy Three pivotal phase III trials, UNCOVER-1, UNCOVER-2 and UNCOVER-3 have evaluated the efficacy and safety of various dosing regimens of ixekizumab compared to placebo in patients with moderate to severe psoriasis [3]. UNCOVER-2 and UNCOVER-3 trials also compared ixekizumab to etanercept [11]. In UNCOVER-1 1296 patients were randomly assigned in a 1:1:1 ratio to receive 80 mg ixekizumab every 2 weeks (Q2W), 80 mg ixekizumab every 4 weeks (Q4W), or placebo. Patients assigned to the ixekizumab cohort received 160 mg starting dose followed by 80 mg Q2W or Q4W. The co-primary endpoints were Psoriasis Area and Severity Index (PASI) 75 and static Physician Global Assessment (sPGA) 0 or 1 at week 12. Patients who responded to ixekizumab treatment at 12 weeks (sPGA 0-1) were re-­ randomised to receive placebo, ixekizumab 80 mg Q4W, or ixekizumab 80 mg every 12 weeks with 48 weeks of follow up [3, 12]. By week 12, a significant superior response was reported to both ixekizumab regimens compared to placebo. The percentage of patients achieving PASI 75 was 89.1 and 82.6% for ixekizumab Q2W and Q4W compared to 3.9% in the placebo group (p < 0.001 compared to placebo) [12]. 81.8% achieved an sPGA of 0 or 1 with ixekizumab Q2W and 76.4% with ixekizumab Q4W compared to 3.2% in the placebo group (P < 0.001) [12]. Of the patients in the 2-week dosing cohort, 70.9% had a PASI 90 response, and 35.3% had a PASI 100 response at week 12 [12]. UNCOVER-1 demonstrated that this response was sustained through 60 weeks. 72.9% of ixekizumab-treated responders maintained sPGA of 0 or 1, 77.7% maintained or 20 Ixekizumab achieved PASI 75, and 52% maintained or achieved PASI 100 [11, 12]. In UNCOVER-2, 1224 patients were randomly assigned in a 2:2:2:1 ratio to receive 80 mg ixekizumab Q2W, 80 mg ixekizumab Q4W, etanercept 50 mg twice weekly or placebo [12]. The UNCOVER-1 and UNCOVER-2 trials shared similar induction designs. Patients in the ixekizumab groups received 160 mg starting dose followed by 80 mg Q2W or Q4W. At 12 weeks, the study showed ixekizumab was statistically superior compared to placebo [12]. The percentage of patients achieving PASI 75 was 89.7 and 77.5% for ixekizumab Q2W and Q4W respectively, compared to 2.4% in the placebo group (P < 0.0001) [12]. 83.2% of the Q2W group and 72.9% of the Q4W group achieved sPGA of 0 or 1, compared to 2.4% in the placebo group (P < 0.0001 compared to placebo) [12]. Both dosing schedules of ixekizumab were superior to placebo in achieving PASI 90, PASI 100, and improvement in DLQI score (P < 0.0001 compared to placebo) [12]. Ixekizumab 80 mg Q2W and Q4W regimens were also more efficacious in terms of number of patients reaching PASI 75 and sPGA 0 or 1 at week 12 when compared to etanercept (P < 0.001) [11, 12]. In UNCOVER-3 1346 patients were randomly assigned in a 2:2:2:1 ratio to receive 80 mg ixekizumab Q2W, 80 mg ixekizumab Q4W, etanercept 50 mg twice weekly, or placebo [12]. Patients in the ixekizumab groups received 160 mg starting dose followed by 80 mg dosing Q2W or Q4W [12]. UNCOVER-3 showed statistically better results for ixekizumab 80 mg Q2W and ixekizumab 80 mg Q4W than placebo [12]. 87.3% of the Q2W group and 84.2% of the Q4W group achieved PASI 75, compared to 7.3% in the placebo group (P < 0.0001) [12]. 80.5% of the Q2W and 75.4% of the Q4W group achieved an sPGA of 0 or 1, compared to 6.7% in the placebo group (P < 0.0001) [12]. Both ixekizumab dosing regimens were superior to placebo in achieving PASI 90, PASI 100 and improvements in DLQI (p < 0.0001) [11, 12]. Significantly more patients who had an itch numeric rating scale (NRS) score of 4 or more at baseline experienced an improvement of 255 4 or more points in both ixekizumab-treated groups compared to placebo and etanercept. Improvements were reported as early as week 1 for each ixekizumab dose versus placebo or etanercept (p < 0.0001) [11]. Pooled data from 2570 patients in UNCOVER-2 and UNCOVER-3 trials showed that ixekizumab treated patients achieved more rapid improvements both in health-related quality of life (HRQol) and itch compared to those receiving etanercept or placebo [2]. Ixekizumab-treated patients (both dosing groups) achieved the minimally clinical important difference (MCID) for the dermatology life quality index (DLQI) and itch numeric rating scale (NRS) scores at 2.1 weeks, while the median times for MCID in etanercept-treated patients were 4 weeks for DLQI and 8.1 weeks for NRS. The majority of patients in the placebo group did not achieve the MCID during the 12-week induction period [2]. Bodyweight Pooled data from three Phase 3 studies (UNCOVER-1, UNOCVER-2, UNCOVER-3) were used to evaluate the effect of bodyweight on the efficacy of ixekizumab treatment in patients with moderate-to-severe psoriasis [13]. 3855 patients were included in the analysis and patients were stratified into three bodyweight groups (<80 kg, 80 to <100 kg, ≥100 kg) [13]. A higher proportion of patients across all body weight groups treated with ixekizumab achieved PASI 75, PASI 90, or PASI 100 at week 12 compared to placebo or etanercept [13]. There was no significant difference in PASI 75 response rates across the different bodyweight groups [13]. Psoriatic Arthritis Two phase III clinical trials, SPIRIT-P1 and SPIRIT-P2, demonstrated the superiority of ixekizumab (80 mg Q2W and Q4W) over placebo in moderate-to-severe psoriatic arthritis (PsA) in patients who failed treatment with either 256 C. Ryan and R. O’Connor NSAIDs, conventional disease-modifying anti-­ twice weekly), or 80 mg ixekizumab Q2W or rheumatic drugs (csDMARDs), or tumour necro- Q4W after a starting dose of 160 mg [17]. At sis factor-α inhibitors (TNFi) for multiple joint week 12, all ixekizumab-treated patients were and cutaneous indices [14]. assigned to open-label to receive ixekizumab In SPIRIT-P1, 417 patients naive to biologic Q4W through to week 60 [17]. At week 12, the therapy with active psoriatic arthritis PsA were placebo group received a 160 mg starting dose randomised to receive subcutaneous injections of of ixekizumab, followed by IXE Q4W thereafplacebo, adalimumab 40 mg once every 2 weeks, ter [17]. A week 12, the etanercept group ixekizumab 80 mg Q2W, or ixekizumab 80 mg received placebo washout, then IXE Q4W at Q4W [15]. Both ixekizumab dosing regimens week 16 and thereafter [17]. included a 160-mg starting dose. The primary Of 1346 patients in this study, 796 had fingerendpoint was an American College of nail psoriasis at baseline and were included in Rheumatology (ACR) 20 response at week 24 this subgroup analysis [17]. The Nail Psoriasis compared to placebo [15]. More patients achieved Severity Index (NAPSI) was the assessment tool an ACR20 response with ixekizumab Q2W used to evaluate the severity of fingernail psoria(62.1%) and ixekizumab Q4W (57.9%) than pla- sis. A modified version of NAPSI was used and cebo (30.2%) (p ≤ 0.001) [15]. Significantly less toenail involvement was not assessed [17]. Each disease activity and functional disability was fingernail was divided into four quadrants and reported with both ixekizumab Q2W and Q4W NAPSI was used to assign a score to each nail for when compared to placebo at week 12 and 24 nail bed and nail matrix psoriasis [17]. The total [15]. Significantly less progression of structural NAPSI fingernail score ranged from 0 (no nail damage was reported at week 24 (p ≤ 0.01) with psoriasis) to 80 (severe nail psoriasis) [17]. ixekizumab treatment [15]. Ixekizumab was also Ixekizumab showed significant improvement superior in achieving PASI 75, 90 and 100 com- in fingernail psoriasis with promising results evipared to placebo in this study (p ≤ 0.001) [15]. dent as early as week 2, with greater NAPSI 363 patients with psoriatic arthritis for a min- improvement in the ixekizumab Q4W (5.1%) imum of 6 months who had an incomplete arm of the study compared to etanercept (−7.9%, response to, or were intolerant of, tumour necro- p = 0.024) [18]. By week 12, both ixekizumab sis factor inhibitors were recruited to SPIRIT-P2 groups exhibited greater NAPSI improvements [16]. 363 patients were randomly assigned from baseline in contrast to placebo and etaner(1:1:1) to receive 80 mg of ixekizumab Q2W or cept [17].This effect was maintained through Q4W after 160 mg starting dose, or placebo [16]. 60 weeks of treatment [17]. The primary endpoint was ACR-20 response at Patients that were switched to ixekizumab week 24 [16]. The percentage of patients who from placebo or etanercept at week 12 or 16 achieved ACR-20 response by week 24 was 53 respectively, demonstrated comparable improveand 48% for ixekizumab Q4W and Q2W, respec- ments to patients who received continuous ixekitively, compared to 20% of placebo-treated zumab treatment [17]. A higher proportion of patients [16]. patients in the ixekizumab Q4W (19.7%) and Q2W (17.5%) group achieved complete resolution of fingernail psoriasis compared to placebo Nail Psoriasis (4.3%) or etanercept (10.2%) at week 12 [17]. Irrespective of the initial stratification, more than Subgroup analysis in UNCOVER-3 evaluated 50% of patients achieved complete resolution by the efficacy of ixekizumab compared to placebo week 60 IXE Q4W treatment [17]. Complete or etanercept in patients with baseline finger- resolution rates in this study are significantly nail psoriasis [17]. Patients were randomized higher when compared to reports of clearance 1:2:2:2 to receive placebo, etanercept (50 mg rates with other biologics [17, 19–21]. 20 Ixekizumab Scalp The scalp is very often affected in psoriasis and is a particularly challenging area to treat [22]. There is limited information available on the efficacy of biologic therapy for scalp psoriasis [22]. A subgroup analysis was performed in a subset of patients with scalp psoriasis from UNCOVER-1, UNCOVER-2 and UNCOVER-3 trials to evaluate the efficacy of ixekizumab over 60 weeks [22]. The Psoriasis Scalp Severity Index (PSSI) was the assessment tool used to evaluate the severity of scalp psoriasis. PSSI is calculated as the sum of results for erythema, induration and desquamation multiplied by a score for the degree of scalp involvement [22]. Among patients with scalp psoriasis at baseline, (PSSI) 75, 90 and 100 were achieved at week 12 in a higher proportion of patients treated with ixekizumab Q2W (89.9, 81.7 and 74.6%) or Q4W (83.6, 75.6 and 68.9%) compared to placebo (12.7, 7.6 and 6.7%) or etanercept (67.6, 55.5 and 48.1%) [22]. These responses were sustained through week 60 of the maintenance (UNCOVER-1 and UNCOVER-2) and LTE arm of the study in patients who remained on ixekizumab Q4W [22]. Improvements occurred in patients with baseline scalp psoriasis in the UNCOVER-3 trial. The median time to PSSI 75, 90 and 100 were 2.1, 4.1 and 4.3 weeks, respectively in the ixekizumab Q2W group compared to 8.1, 8.3 and 12 weeks, respectively in etanercept group [22]. Palmoplantar Psoriasis Palmoplantar psoriasis is not only associated with considerable discomfort, and pain it also affects dexterity and mobility with significant quality of life implications [23–26]. Using pooled data from UNCOVER-1, UNCOVER-2 and UNCOVER-3 trials, the efficacy of ixekizumab was evaluated in a subpopulation of patients with moderate-tosevere plaque psoriasis and moderate-­ to-severe non pustular palmoplantar involvement [26]. The Palmoplantar Psoriasis Area and Severity Index (PPASI) was used to assess the severity of 257 psoriasis on the palms and soles. PPASI is calculated as the sum of results for erythema, induration and desquamation multiplied by a score for the degree of palm and sole involvement (range 0–72) [26]. 28.3% of patients from UNCOVER-1, UNCOVER-2 and UNCOVER-3 had evidence of palmoplantar psoriasis at baseline (PPASI ≥ 0). Of these, 9.1% of patients had moderate-to-­ severe palmoplantar psoriasis at baseline (PPASI ≥ 8) [26]. Among patients with palmoplantar psoriasis, 85.9, 73.9 and 48.9% treated with ixekizumab Q4W and 79.8, 69.3 and 51.8% treated with ixekizumab Q2W achieved PPASI 50, 75 and 100 respectively, at week 12, which is significantly more than placebo (32.9, 18.8 and 8.2%; p < 0.001) and etanercept (67.8, 44.1, and 32.2%; p < 0.05) [26].This response was maintained or improved in patients continuing on ixekizumab Q4W through week 60 [26]. Genital Psoriasis Genital psoriasis is a common, distressing and difficult-to-treat form of plaque psoriasis with a detrimental impact on quality of life and psychosexual functioning [27]. Ixora-Q was a phase IIIb, randomised, double-­ blind, placebo-controlled trial which appraised the efficacy of ixekizumab in patients with moderate-­to-severe genital psoriasis with ≥1% body surface area involved [27]. The static Physician’s Global Assessment of Genitalia (sPGA-G) was developed to assess the severity of genital psoriasis [27]. 149 patients with moderate-­ to-­severe genital psoriasis (sPGA-G) score ≥ 3), with BSA ≥ 1% were randomised 1:1 to receive placebo or ixekizumab (160 mg at week 0, then 80 mg every 2 weeks) [27]. The primary endpoint was to establish if ixekizumab was superior to placebo at week 12. This was measured by the percentage of patients achieving 0 or 1 (clear or minimal) on the sPGA-G [27]. At week 12, 73% of patients in the ixekizumab group achieved clear or almost clear genital skin (sPGA-G 0 or 1) compared to 8% in the placebo group. Fifty-six percent of patients achieved com- C. Ryan and R. O’Connor 258 plete clearance of genital psoriasis compared to placebo 5% (p < 0.001) [27]. In keeping with previous clinical studies of ixekizumab, 73% of patients achieved an overall sPGA 0 or 1 compared to placebo (3%, p < 0.001) [27]. Significant clinical improvement was reported as early as week 1 for sPGA-G 0 or 1, sPGA-G 0 and overall sPGA 0 or 1, and as early as week 4 for overall sPGA 0 [27]. Ixekizumab resulted in significant improvements in genital itch and HRQoL. 60% of patients treated with ixekizumab achieved a clinically meaningful (≥3-point) improvement from baseline in the genital itch numeric rating scale (NRS) compared to placebo (8%) at week 12 [27]. Almost 80% of patients treated with ixekizumab reported that the frequency of sexual activity was rarely affected by genital psoriasis by week 12 of treatment and significant improvement was already observed within the first week of treatment. Additionally, 45% of patients receiving ixekizumab reported no clinically significant impact on HRQoL at week 12 [27]. There are limited number of published open-label studies that have investigated the efficacy of therapeutic interventions for genital psoriasis [28–30]. Body Regions Pooled data from 3 trials (UNCOVER-1, UNCOVER-2, UNCOVER-3) through week 12 was used to assess the efficacy of ixekizumab on moderate-to-severe psoriasis affecting various body sites (head/neck, arms, trunk, legs) [18]. Ixekizumab Q2W was associated with significantly greater mean percent improvements in PASI from baseline compared to placebo for each body region. Improvements increased through week 12 with the highest on the trunk (92.8%), followed by the head/neck (91.4%), arms (89.9%) and legs (88.7%) (p < 0.001 vs. placebo) [18]. Erythrodermic and Pustular Psoriasis A phase 3, multicentre, single-arm, open-label, long-term study was carried out to evaluate the long term efficacy and safety of ixekizumab in Japanese patients with plaque psoriasis (n = 78), erythrodermic psoriasis (n = 8) and generalized pustular psoriasis (n = 5) [31]. 160 mg of ixekizumab was administered at week 0, followed by 80 mg every 2 weeks from weeks 2 to 12, and every 4 weeks from weeks 16 to 52 [31]. At week 52, PASI 75, PASI 90 and PASI 100 was achieved in 92.3, 80.8 and 48.7% of patients [31]. Patients with erythrodermic psoriasis or generalised pustular psoriasis responded favourably to ixekizumab therapy and 75 and 60% of patients achieved a sPGA 0 or 1 at week 52 [31]. Improvements in PASI, PSSI, DLQI and itch NRS were reported at week 12 and maintained through week 52 [31]. Adverse Effects Comprehensive ixekizumab safety data comes from the cumulative safety analysis covering up to 319 weeks of ixekizumab exposure combined from 11 controlled and uncontrolled ixekizumab studies on psoriasis, including three Phase 3 studies (UNCOVER-1, -2, and -3) [32]. Ixekizumab exhibited an acceptable safety profile and no novel safety findings compared to previous reports [3, 11, 32, 33]. 5689 patients treated with ixekizumab from 11 studies were included in the analysis, which accounted for 12 061.5 PY of exposure [32]. Treatment-emergent adverse events (TEAE) were reported by 83.9% of patients. 71% of patients reported mild-to-moderate TEAEs and 12.9% reported severe TEAEs [32]. A TEAE was an event that first occurred or increased in severity after baseline, on or prior to the final day, within the treatment time [32]. The most commonly (>6%) reported TEAE after 12 061.5 PY of ixekizumab therapy was: nasopharyngitis (22.9%), upper respiratory tract infection (13.5%), injection-site reaction (9.5%), headaches (7.8%) and arthralgia (7.1%) [32]. These results are similar to those previously reported with 6480 PY exposure [33]. Serious adverse events AEs were reported in 11.8% of patients and death occurred in 0.4% of patients (cardiac disorder (n = 11), neoplasm (n = 2), unknown cause (n = 5), respiratory failure (n = 2), severe 20 Ixekizumab cerebrovascular event (n = 1), severe dementia (n = 1) and injury (n = 1) [32]. 6.7% of patients reported TEAEs resulting in discontinuation from the study. There was no cases of confirmed tuberculosis (TB) or reactivation. However, 0.5 and 0.2% of patients reported a positive Mycobacterium tuberculosis complex test and a positive tuberculin test. These were the most commonly reported AE that lead to discontinuation from the study [32]. Research suggests a high rate of false-positive TB test results are commonly found in populations with a low risk of Mycobacterium tuberculosis infection which reflects the cohorts in these studies [34–36]. While there were no serious injection-site reactions reported, 0.2% of patients discontinued the study due to injection site reaction [36]. Adverse Events of Special Interest Ixekizumab exposure over 156 weeks showed no increase in incidence rates (IR) of adverse events of special interest (AESI) [32]. Infections, injection site reactions and allergic reactions/ hypersensitivities were the most commonly reported AESI [32]. Contact dermatitis, eczema, urticaria, dermatitis, rash and allergic rhinitis were the most common allergic events. Most of the injection-­site reactions were mild or moderate [32]. Infections In keeping with published literature to date infections are the most frequently AEs associated ixekizumab therapy [11, 33]. 60.8% of patients reported an infection while on ixekizumab therapy, however 25.4% were mild and 32.4% were moderate [32]. Only 3% were reported as severe infections [32]. Nasopharyngitis (22.9%), upper respiratory tract infection (13.5%), bronchitis (5.2%) and sinusitis (5.4%) are among the most commonly reported infections [32]. Serious infections were reported in 2.6% of patients, however none of these resulted in death [32]. Cellulitis was the most frequently reported seri- 259 ous infection affecting 0.5% of ixekizumab treated patients [32]. In keeping with IL-17A blockade and its role in mucosal immunity, mucocutaneous candidiasis was the most commonly observed opportunistic infection. This integrated analysis of 11 clinical studies showed no reports of endemic mycoses, systemic candidiasis, invasive aspergillus or other invasive fungal infections [32]. IBD Research has shown that the prevalence of Crohn’s disease and ulcerative colitis is increased in patients with psoriasis [37–40]. Pooled data from seven ixekizumab psoriasis trials reported that the incidence of Crohn’s disease and ulcerative colitis cases were uncommon (<1%) in ixekizumab treated patients [41]. The results are similar with those observed in the analysis of combined data of 11 ixekizumab psoriasis trials. Following 3 years of ixekizumab treatment the IRs of Crohn’s disease and ulcerative colitis were 0.0 and 0.1 per 100 PYs, respectively [32]. Malignancy Based on the combined data of 11 ixekizumab psoriasis trials, ixekizumab exposure up to 156 weeks was not associated with in increased risk malignancy [32]. Depression Depression is common in patients with psoriasis. The efficacy of ixekizumab was evaluated in patients with moderate-to-severe plaque psoriasis and moderately severe depressive symptoms [42]. Data were pooled from 3 randomised, double-­ blind, controlled phase 3 trials. Depressive symptoms and inflammation were evaluated at baseline and week 12 using a self-report questionnaire to quantify depressive symptoms (QIDS-SR16) and by measuring serum C-reactive protein (hs CRP) [42]. A sub- 260 group analysis was performed on patients with at least moderately severe depressive symptoms at baseline (QIDS-SR16 total score ≥ 11) [42]. The comorbid depressive symptom group was defined as patients who had a (QID-SR16) total score ≥ 11 at baseline [42]. Comorbid patients treated with ixekizumab Q2W and Q4W reported better responses in their QIDS-SR16 total score (−7.1 and −6.1) compared to placebo (−3.4; p < 0.001) [42]. Ixekizumab Q2W and Q4W was associated with higher rates of remission of depressive symptoms (45.2%, 33.6%) compared with placebo (17.8%; p ≤ 0.01) [42]. Ixekizumab treated patients was also associated with notable reductions in hs CRP and PASI compared to placebo [42]. Summary Ixekizumab selectively blocks IL-17A which is a member of the IL-17 family of cytokines. Numerous clinical trials (UNCOVER-1, UNCOVER-2, UNCOVER-3, and others) have shown Ixekizumab to be efficacious in treating psoriasis and psoriatic arthritis. Research has also shown ixekizumab to be well tolerated with a safety profile similar to others in the class. Recently published 5-year safety data, shows no new or unexpected safety concerns have emerged. However concern remains regarding the long-­term side effects of anti-IL17 agents until more long term data is available. The ongoing collection of data in registries will be important to allow for comprehensive safety data. References 1. Liu L, Lu J, Allan BW, Tang Y, Tetreault J, Chow CK, et al. Generation and characterization of ixekizumab, a humanized monoclonal antibody that neutralizes interleukin-17A. J Inflamm Res. 2016;9:39–50. 2. Leonardi CL, Blauvelt A, Sofen HL, Gooderham M, Augustin M, Burge R, et al. Rapid improvements in health-related quality of life and itch with ixekizumab treatment in randomized phase 3 trials: results from UNCOVER-2 and UNCOVER-3. J Eur Acad Dermatol Venereol. 2017;31(9):1483–90. C. Ryan and R. O’Connor 3. Gordon KB, Blauvelt A, Papp KA, Langley RG, Luger T, Ohtsuki M, et al. Phase 3 trials of ixekizumab in moderate-to-severe plaque psoriasis. N Engl J Med. 2016;375(4):345–56. 4. Blegvad C, Skov L, Zachariae C. Ixekizumab for the treatment of psoriasis: an update on new data since first approval. Expert Rev Clin Immunol. 2019;15(2):111–21. 5. https://www.accessdata.fda.gov/drugsatfda_docs/ label/2018/125521s009lbl.pdf 6. Miossec P, Korn T, Kuchroo VK. Interleukin-17 and type 17 helper T cells. N Engl J Med. 2009;361(9):888–98. 7. Stark MA, Huo Y, Burcin TL, Morris MA, Olson TS, Ley K. Phagocytosis of apoptotic neutrophils regulates granulopoiesis via IL-23 and IL-17. Immunity. 2005;22(3):285–94. 8. Cua DJ, Tato CM. Innate IL-17-producing cells: the sentinels of the immune system. Nat Rev Immunol. 2010;10(7):479–89. 9. Gisondi P, Geat D, Pizzolato M, Girolomoni G. State of the art and pharmacological pipeline of biologics for chronic plaque psoriasis. Curr Opin Pharmacol. 2019;46:90–9. 10. http://www.medicines.org.uk/emc/product/7233/ smpc 11. Griffiths CE, Reich K, Lebwohl M, van de Kerkhof P, Paul C, Menter A, et al. Comparison of ixekizumab with etanercept or placebo in moderate-to-­ severe psoriasis (UNCOVER-2 and UNCOVER-3): results from two phase 3 randomised trials. Lancet. 2015;386(9993):541–51. 12. Farahnik B, Beroukhim K, Zhu TH, Abrouk M, Nakamura M, Singh R, et al. Ixekizumab for the treatment of psoriasis: a review of phase III trials. Dermatol Ther. 2016;6(1):25–37. 13. Reich K, Puig L, Mallbris L, Zhang L, Osuntokun O, Leonardi C. The effect of bodyweight on the efficacy and safety of ixekizumab: results from an integrated database of three randomised, controlled phase 3 studies of patients with moderate-to-severe plaque psoriasis. J Eur Acad Dermatol Venereol. 2017;31(7):1196–207. 14. O’Rielly DD, Rahman P. A review of ixekizumab in the treatment of psoriatic arthritis. Expert Rev Clin Immunol. 2018;14(12):993–1002. 15. Mease PJ, van der Heijde D, Ritchlin CT, Okada M, Cuchacovich RS, Shuler CL, et al. Ixekizumab, an interleukin-17A specific monoclonal antibody, for the treatment of biologic-naive patients with active psoriatic arthritis: results from the 24-week randomised, double-blind, placebo-controlled and active (adalimumab)-controlled period of the phase III trial SPIRIT-P1. Ann Rheum Dis. 2017;76(1):79–87. 16. Nash P, Kirkham B, Okada M, Rahman P, Combe B, Burmester GR, et al. Ixekizumab for the treatment of patients with active psoriatic arthritis and an inadequate response to tumour necrosis factor inhibitors: results from the 24-week randomised, double-blind, 21 Brodalumab Annika S. Silfvast-Kaiser, Dario Kivelevitch, So Yeon Paek, and Alan Menter Abstract Key Learning Objectives Brodalumab is a recombinant, fully human 1. To understand the mechanism of action monoclonal receptor IgG2 antibody approved of brodalumab for the treatment of moderate-to-severe plaque 2. To understand the appropriate use and psoriasis. It inhibits the IL-17 receptor A efficacy of brodalumab (IL-17RA), differing from other biologics in 3. To understand the safety issues of the IL-17 class which target the IL-17A cytobrodalumab kine itself. It is a highly effective medication, with a rapid onset of action. Its superiority to placebo and to ustekinumab has been shown in clinical trials, reducing both the severity Introduction and the extent of psoriasis, with efficacy ­sustained up to 52 weeks. It has also shown Psoriasis affects approximately 2–3% of the superiority to placebo in patients with diffiworldwide population [1]. It is characterized by cult-to-treat nail and scalp psoriasis and early an extensive inflammatory infiltrate in the derevidence of efficacy in patients with psoriatic mis with excessive growth of the epidermal skin arthritis. Brodalumab is generally well-­ layer, presenting as well-demarcated, erythematolerated with a safety profile similar to that of tous plaques with overlying silvery scale [2]. In other biologics, with the addition of a label addition to skin manifestations, 30–35% of psowarning relating to suicidal ideation. Herein, riasis patients are affected by psoriatic arthritis we review the data behind the use of broda(PsA), usually presenting 10–15 years after the lumab for the treatment of psoriasis and psorionset of skin lesions [3]. Psoriasis is now largely atic arthritis. considered a systemic disease which can affect a multitude of organ systems including the cardiovascular system in addition to psychosocial health issues. Psoriasis is thought to result from initial antigenic or environmental stimuli in combination with predisposing genetic factors A. S. Silfvast-Kaiser · D. Kivelevitch · S. Y. Paek and dysregulation of the innate and adaptive A. Menter (*) Division of Dermatology, Baylor Scott & White, immune systems [4]. These antigenic stimuli Dallas, TX, USA activate native immune cells including T-helper e-mail: annikask@utexas.edu © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_21 263 A. S. Silfvast-Kaiser et al. 264 cells (Th1 and Th17), dendritic cells, and keratinocytes which then produce multiple pro-inflammatory cytokines including tumor necrosis factor (TNF)-alpha, interferon-alpha, IL-12, IL-17, and IL-23 [5, 6]. A complex interplay between immune cells and proinflammatory cytokines exists in this T-cell mediated chronic inflammatory skin disease. Ultimately, the cytokines released by Th1 and Th17 cells result in the characteristic inflammatory environment seen in psoriasis. Among others, Th17 cells release the full spectrum of cytokines IL-17A through IL-17F. IL-17A is the most influential, having been established as a factor in the development of psoriasis, multiple sclerosis, rheumatoid arthritis, and inflammatory bowel disease [7]. IL-17 is involved in neutrophil chemotaxis and stimulates fibroblast production of vascular endothelial growth factor, leading to endothelial cell proliferation and angiogenesis [8]. IL-17A also plays a role in the innate immune defense against fungal and bacterial pathogens [9]. Role of IL-17 in Psoriasis IL-17 and its related cytokines play an important role in the immunopathogenesis of psoriasis [10– 12]. This family of cytokines (IL-17A, IL-17B, IL-17C, IL-17D, IL-17E [IL-25], and IL-17F) has been shown to induce expression of psoriatic pro-inflammatory molecules in keratinocytes [9, 13]. IL-17 receptors are found on the surface of keratinocyte cells and promote signaling within the cell by receiving IL-17 cytokines. Elevated IL-17A, 17C, and 17F messenger RNA (mRNA) have been shown in the serum and plaques of psoriasis patients [14–16]. The success of IL-17 and IL-23 biologic targeting agents in the treatment of psoriasis has established the IL-17 cytokines as key players in psoriasis disease development [16, 17]. As our understanding of psoriasis continues to develop, an increasing number of systemic biologic agents that target these cytokines have been developed and are available to patients. Brodalumab is one such agent, targeting the IL-17 cytokines via blockage of the IL-17 receptor A. Brodalumab Brodalumab (Siliq™, Ortho-Dermatologics, United States; Kyntheum®, Leo Pharma, Europe) is a fully human IgG2 monoclonal antibody that differs from other anti-IL-17 agents (secukinumab and ixekizumab) by binding the subunit A of the IL-17 receptor rather than the IL-17 cytokine itself [18] (Fig. 21.1). By blocking IL-17RA, brodalumab manages to inhibit the downstream inflammatory activity of several interleukins (IL-17A, IL-17C, IL-17F, IL-17E [IL-25], as well as the IL17A/F heterodimer, preventing psoriatic inflammation [19, 20]. Brodalumab is the only biologic currently approved for the treatment of psoriasis with activity against IL-17C, a cytokine critical in the development of psoriasis, and the most abundant IL-17 cytokine found in psoriatic lesional skin [21–28]. Brodalumab was approved in February 2017 for the treatment of moderate-to-severe plaque psoriasis. Its superiority to placebo as well as to ustekinumab has been shown at 12 weeks for both PASI 75 and PASI 90 [29, 30]. It is not currently approved for the treatment of psoriatic arthritis, but is under investigation in clinical trials for this indication. osing, Storage, Administration, D and Pharmacokinetics Brodalumab is a subcutaneous, self-injectable medication dosed initially as one 210 mg/1.5 mL prefilled syringe at weeks 0, 1, and 2, and subsequently every 2 weeks [28, 31]. Storage of the syringe should occur at 2–8 °C or can be stored at room temperature (for up to 2 weeks, then it should be discarded). No re-refrigeration should occur. The medication should be allowed to reach room temperature for approximately 30 minutes, if the syringe has been refrigerated before injection [32]. If no or inadequate improvement is seen at 16 weeks, discontinuation of brodalumab should be considered as the likelihood of reaching greater disease improvement after this time point is unlikely [32]. Brodalumab reached maximum plasma concentration 2–4 days after one brodalumab 210 mg 21 Brodalumab Fig. 21.1 Mechanism of action of brodalumab. The heterodimeric IL-17RA is blocked by brodalumab, decreasing the downstream release of psoriasis-related inflammatory cytokines 265 Th17 cell IL-17A - IL-17F IL-17F IL-17A Brodalumab IL-17RA Brodalumab subcutaneous dose, with steady-state achieved after 10–12 weeks. After 20 weeks of administration the estimated accumulation ratio was 2.5-­ fold [33]. After subcutaneous injection, the estimated bioavailability was 57.6% [34]. The estimated half-life of brodalumab is 10.9 days [33]. Age, sex and race had no effect on the pharmacokinetics of brodalumab. The effect of renal or hepatic impairment on its pharmacokinetics has not been studied. Hepatic impairment and renal elimination are not expected to be clinically relevant as brodalumab is mainly eliminated via catabolism [33]. Although the area under the serum concentration-time curve at steady state was predicted to be two-fold higher in patients weighing less than 75 kg, no dosage adjustments for body weight are recommended [33]. Clinical Efficacy in Plaque Psoriasis Overall, brodalumab has shown similar efficacy to the other two IL-17 inhibitors. It exhibits an Target cell Brodalumab early response (seen as early as 4 weeks), maintenance of response (up to 52 weeks), as well as no loss of efficacy in patients with prior biologic exposure. The most common methods for clinical assessment in psoriasis clinical trials are the Psoriasis Assessment and Severity Index (PASI) and the Physicians Global Assessment (PGA). The PASI score is based on the quality of psoriatic lesions (i.e. erythema, induration, and scale) weighted by body surface area, on a scale of 0–72, with higher scores indicating more severe disease. Trial endpoints commonly assess the percentage of patients who achieve a certain reduction in PASI score compared to baseline (i.e. a 75% reduction in PASI score is termed PASI 75 with a 90% reduction termed as PASI 90). The PGA score is graded on a scale of 0–5 with 0 representing “clear” and 5 representing “severe” disease involvement. Brodalumab is indicated for the treatment of moderate-to-severe plaque psoriasis in adult patients who are candidates for systemic therapy 266 or phototherapy, having either had inadequate response to, or lost response to, other systemic therapies [28]. Below, we discuss the results of the phase I, II, and III clinical trials that led to brodalumab’s eventual approval. A. S. Silfvast-Kaiser et al. safety profiles of both the brodalumab and placebo cohorts were similar. Two patients (one from the 350 mg cohort and another from the 700 mg cohort) receiving brodalumab exhibited formation of non-neutralizing antibodies. Another phase I clinical trial in Japan evaluPhase I Clinical Trials ated the safety, tolerability, pharmacokinetics, Brodalumab’s safety and efficacy was initially and pharmacodynamics of brodalumab in 40 investigated in a double-blind, randomized, healthy volunteers and in 8 subjects with placebo-­controlled, ascending single-dose trial of moderate-­to-severe psoriasis [37]. Subjects with 25 healthy controls and patients with moderate-­ psoriasis had to meet inclusion criteria of to-­ severe psoriasis with a Psoriasis Area and PASI ≥ 10, BSA ≥ 10%, and have undergone at Severity Index (PASI) score ≥ 10 and Body least one previous phototherapy or systemic therSurface Area (BSA) ≥ 10%. Patients were fol- apy. In the healthy group, 8 subjects were ranlowed for 85 days with 20 of 25 patients receiv- domized in a 2:6 ratio to receive either placebo or ing a single dose of brodalumab (4 patients a single dose of brodalumab at 70 mg, 140 mg, received 140 mg subcutaneously (SC), 8 received 210 mg, or 420 mg SC or 210 mg IV. Psoriasis 350 mg SC, and 8 received 700 mg intravenously patients either received one dose of 140 mg SC (IV)). The 5 other patients received placebo. The brodalumab or 350 mg SC. Rapid, dose-­ majority of patients were men (76%) with a dependent improvement was seen in all psoriasis median age of 43 years and mean baseline PASI patients receiving brodalumab treatment. All 7 score of 14.1 [35, 36]. Rapid, dose-dependent patients in the 350 mg cohort reached PASI 50; 6 improvement in both PASI and static Physician of these patients achieved PASI ≥ 75; 3 of 6 Global Assessment (sPGA) scores within patients in the 140 mg cohort also achieved 2 weeks of dosing with either 350 mg SC or PASI ≥ 75. Safety profiles were similar between 700 mg IV brodalumab was seen. Two of four both the healthy patients and those with psoriasis. patients who received 140 mg SC achieved PASI Injection site erythema was the most commonly 50. All patients on 700 mg IV achieved PASI 50 reported AE for both placebo and brodalumab. by 4 weeks, with all but one achieving PASI 75 or Brodalumab antibodies were not detected in any greater by week 6. Three patients achieved PASI group up to 9 weeks. 90 by week 6. Six of eight patients who received 350 mg SC achieved PASI 50 and 3 of 8 patients Phase II Clinical Trials receiving the same dose reached PASI 75. None A total of 188 patients with moderate-to-severe of the 5 patients receiving placebo achieved PASI psoriasis with BSA ≥ 10% and PASI ≥ 12 were 50 [35]. enrolled and completed a phase II, randomized, Lesional and non-lesional skin biopsies were double-blind, placebo-controlled, dose-ranging performed on all subjects prior to treatment and study to evaluate the short term efficacy and then twice after dosing. The patients in the two safety of brodalumab. Sixty-six percent of the highest dose cohorts (300 and 700 mg) showed patients were male with a mean age of 43, and a significant reductions in epidermal thickness, mean PASI and BSA of 19 and 24%, respectively keratin 16, and Ki67-expresssing cells (as early [18]. Patients were randomly assigned to one of as week 1 after dosing of the two highest doses), five treatment groups (70 mg, 140 mg, or 210 mg along with lesional IL-17A, F and C mRNA lev- SC on day 1, week 1, 2, 4, 6, 8, and 10; or 280 mg els which returned to non-lesional levels over SC at day 1, week 4, and week 8; or placebo). 6 weeks [20, 35]. The most common adverse The percentage of improvement in PASI score at events (AEs) experienced by those receiving bro- week 12 compared to baseline was the primary dalumab were headache, gastroenteritis, and the endpoint. Secondary endpoints were attaining Koebner phenomenon at the biopsy site. The PASI 50, 75, 90, 100, as well as BSA and sPGA 21 Brodalumab scores at week 12. Compared to placebo, all brodalumab groups achieved significantly greater mean PASI improvement at week 12 (p < 0.001), with the earliest clinical improvement seen at week 2. In the various brodalumab dosing arms receiving 70, 140, 210, or 280 mg the mean PASI improvement was 45, 85.9, 86.3, and 76% respectively. The placebo arm had 16% mean PASI improvement. At week 12, PASI 75 or greater was achieved by 77% in the 140 mg brodalumab group and 82% in the 210 mg brodalumab group. In these two groups, PASI 90 or greater was seen in 72 and 75%, respectively. 38.3% of patients in the 140 mg arm and 80% of patients in the 210 mg arm reached PASI 100. A sub-analysis of this phase II data also showed that patients achieved clinical improvement and similar rates of AEs irrespective of whether self-reported psoriatic arthritis was present or not [38]. Compared to placebo, the brodalumab groups also showed significantly greater improvements in BSA, sPGA, Dermatology Quality of Life Index (DLQI) and SF-36 at week 12 [18]. Inflammatory markers and epidermal thickness also significantly improved in the brodalumab groups compared to placebo. The most commonly reported AEs in the brodalumab group were nasopharyngitis, upper respiratory i­ nfection, arthralgia, and injection site erythema. Three serious adverse events occurred with brodalumab: renal colic and two cases of grade 3 asymptomatic neutropenia (resolving after brodalumab withdrawal). Secondary analyses have also shown that at week 12, the brodalumab treatment groups demonstrated significant improvement in patient reported outcome measures including DLQI and Psoriasis Symptom Inventory (PSI) compared to placebo. Patients with prior biologic exposure and those naïve to prior biologic therapies showed no difference in the mean improvements in PASI scores [18, 38]. A 5-year open-label extension of this phase II trial enrolled 181 of the original 198 patients to evaluate the long-term safety and efficacy of brodalumab 210 mg every 2 weeks [39]. At 52 weeks, a dose reduction to 140 mg for patients ≤100 kg (n = 119) was implemented. Patients with inadequate response to this lower dose were then allowed 267 to increase their dose back to 210 mg (n = 19). Ninety percent of patients achieved sPGA 0/1 (clear to almost clear) at 12 weeks, 85% at 48 weeks, 76% at 96 weeks, and 72% at 144 weeks. At weeks 12 and 48, the mean BSA improvement from the baseline BSA of the parent study was 95 and 96%, respectively. The most frequently reported AEs occurring during open-­label extension were mild to moderate in severity and included nasopharyngitis, upper respiratory tract infection, arthralgia, back pain, and influenza. 14 patients reported AEs of grade 3 (severe) or greater and 9% of patients reported SAEs including cholecystitis, constipation (likely related), pyelonephritis, and benign parathyroid tumor. One death occurred due to rupture of an aortic aneurysm [38, 39]. In Japan, a phase II study randomized 151 moderate-to-severe plaque psoriasis patients to doses of 70, 140, 210 mg, or placebo at day 1, weeks 1, 2, 4, 6, 8 and 10 [40]. Efficacy and safety outcomes were similar to the phase II trial discussed above with the most common AEs in brodalumab patients being nasopharyngitis, diarrhea, upper respiratory tract inflammation, and folliculitis. At week 12, the mean improvement in PASI scores for the 70 mg, 140 mg, 210 mg, and placebo arms were 37.7, 82.2, 96.8, and 9.4%, respectively. PASI 75 was achieved at week 12 by 26, 78, and 95% of patients in the 70, 140, and 210 mg groups, respectively. PASI 90 or greater and PASI 100 was achieved by 15, 65, 92% and 2.6, 35, and 59% of these patients, respectively. DLQI scores of 0 or 1 were also significantly more common in patients in the 140 mg and 210 mg groups at week 12 (54.1 and 56.8%) compared to those on placebo (8.8%). Long-term brodalumab treatment also showed sustained clinical response and favorable safety profile in an open-label extension (OLE) of this study [41]. 133 of 145 patients (92%) completed the study with 94.4, 87.5, and 55.6% of patients in the 210 mg cohort achieving PASI 75, 90, and 100 by week 52 and 78.1, 71.2, and 43.8% of patients in the 140 mg group achieving PASI 75, 90, and 100, respectively. Nasopharyngitis (35.2%), upper respiratory tract inflammation (10.3%), and contact dermatitis (9.7%) were the most commonly reported AEs. A. S. Silfvast-Kaiser et al. 268 hase III Clinical Trials P or almost clear. Between 37 and 44% of patients Three phase III clinical trials (AMAGINE-1, in all AMAGINE trials achieved a PASI 100 AMAGINE-2, and AMAGINE-3) have been response by week 12 and approximately 70% of completed to assess the efficacy, safety, and patients achieved PASI 90 by week 12.81% of withdrawal-­ retreatment effect of brodalumab patients who had failed prior biologic therapy versus placebo in moderate-to-severe plaque pso- were able to attain PASI 75 response with 32% of riasis. AMAGINE-1 compared brodalumab only them achieving PASI 100. Of the PASI 100 to placebo. AMAGINE-2 and AMAGINE-3 com- responders from week 12 in the AMAGINE 2 and pared brodalumab’s efficacy and safety to that of 3 trials, 72% maintained PASI 100 response at ustekinumab in addition to placebo. Both of these week 52. studies were multicenter, randomized, double-­ blind, placebo-controlled, active comparator-­ AMAGINE-1 controlled, parallel group trials with a 12-week In AMAGINE-1, 661 patients with moderate-to-­ induction period, followed by maintenance with severe psoriasis were followed through a 12-week brodalumab up to week 52 (Table 21.1) [29, 30]. induction phase and then a withdrawal and treatAll phase III studies required a diagnosis of ment phase through 52 weeks [29]. Ninety six plaque psoriasis for ≥6 months, BSA ≥ 10%, percent of patients completed the induction phase PASI ≥ 12 and sPGA ≥ 3. The primary endpoint and 84.4% of patients continued through treatfor all three AMAGINE trials was PASI 75 and ment withdrawal and re-treatment through week sPGA 0/1 at week 12 as compared to placebo. 52. During the induction phase, patients received For AMAGINE-2 and AMAGINE-3, PASI 100 either brodalumab 140 mg, 210 mg, or placebo, for brodalumab versus ustekinumab was also a every 2 weeks for 12 weeks. After 12 weeks, primary endpoint. Between 76 and 80% of patients with sPGA score of 0/1 were re-­ patients in all three trials achieved sPGA of clear randomized to either continue with their current Table 21.1 Brodalumab phase III clinical trial outcomes [29, 30] Phase III trial AMAGINE-1 (n = 661) AMAGINE-2 (n = 1831) AMAGINE-3 (n = 1881) Brodalumab Study arms Placebo Brodalumab 140 mg Brodalumab 210 mg Placebo Ustekinumab Brodalumab 140 mg Brodalumab 210 mg Placebo Ustekinumab Brodalumab 140 mg Brodalumab 210 mg Phase III trial results Week 12 Primary & Secondary Outcomes (PASI 75/90/100) 2.7/0.9/0.5% 60.3a/42.5 a/23.3a% 83.3a/70.3a/41.9a% Week 12 sPGA 0/1 & sPGA 0 Week 52 Outcomes (PASI 75/90/100) 1.4 & 0.5% 53.9 & 23.3% 75.7 & 41.9% – –/66.7/43.9% –/78.3/67.5% 8.1/1.9/0.6% 70/47/21.7% 66.6a,b/49a,b/25.7a,b% 86.3a,b/69.9a,c/44.4a,c% 3.9 & 0.6% 61 & 21.7% 58 & 25.7% 78.6 & 44.8% – – – 80/75/56% 6/2.9/0.3% 69.3/47.9/18.5% 69.2a,b/52a,b/27a,c% 85.1a,c/68.9a,c/36.7a,c% 4.1 & 0.3% 57.2 & 18.5% 59.9 & 27% 79.6 & 36.7% – – – 80/73/53% p < 0.001 for the comparison with placebo p > 0.05 for the comparison with ustekinumab c p < 0.01 for the comparison with ustekinumab a b 21 Brodalumab treatment or to switch to placebo. Patients who experienced disease return with sPGA ≥ 3 after re-randomization to placebo were then re-started on their induction dose of brodalumab up to week 52. If patients had originally been randomized to the placebo group, or if they had sPGA ≥ 1 after week 12, they were started on brodalumab 210 mg every 2 weeks. By week 12, PASI 75 was achieved by 83.3% of the brodalumab 210 mg group, 60.3% of brodalumab 140 mg group, and in 2.7% of those receiving placebo [29]. In the 210 and 140 mg brodalumab arms, PASI 90 response was seen in 70.3 and 42.5% of patients respectively, compared to 0.9% in the placebo group. 41.9 and 23.3% of patients in the 210 and 140 mg arms achieved PASI 100 respectively, compared to 0.5% in the placebo group [29, 38, 42]. These significant differences in clinical response were sustained until week 52. sPGA scores of 0/1 were achieved by 75.7 and 53.9% of patients on brodalumab 210 mg and 140 mg, respectively, compared to placebo at 1.4%. Significant improvements on the Psoriasis Symptom Inventory (PSI) (which evaluates 8 psoriasis signs and symptoms; see Table 21.2) were also seen compared to placebo [43, 44]. Those who lost control of their psoriasis with withdrawal of brodalumab were able to recover their previous treatment sPGA score within 12 weeks of resuming therapy with brodalumab [45]. One hundred percent improvement in the Psoriasis Scalp Severity Index (PSSI) was achieved by 63.4 and 41% of patients in the 140 mg and 210 mg brodalumab groups, as comTable 21.2 Psoriasis symptom inventory items [44] Psoriasis Symptom Inventory (PSI)a,b • Itch • Redness • Scaling • Burning • Stinging • Cracking • Flaking • Pain Each item is scored on a scale of 0 (not at all severe) to 4 (very severe), with a total score from 0 to 32 b Response of PSI is defined as attaining a total score of ≤8 a 269 pared to 3.2% in the placebo group [46]. At week 12, the most frequently reported adverse events in the brodalumab cohorts were nasopharyngitis, upper respiratory tract infection, and headache. SAEs while on brodalumab 210 mg, 140 mg, and placebo occurred in 1.8, 2.7, and 1.4% of patients, respectively. The impact of brodalumab treatment on quality of life outcomes was also investigated in AMAGINE-1. At 12 weeks, in those receiving brodalumab, a significant proportion of patients reported PSI of 0, DLQI of 0, and satisfaction with treatment compared to placebo patients. Health-related quality-of-life measures also showed significantly greater improvement in patients treated with brodalumab compared with placebo. Over 60% of patients with moderate and severe anxiety or depression at baseline exhibited improvement to mild, moderate, or normal severity by week 12 on brodalumab [29]. In addition, psoriasis biomarkers such as mRNA expression of IL-17 cytokines, IL-23, and cell counts of epidermal Ki-67, dermal CD3, epidermal CD3, dermal CD8, and epidermal CD8, were all significantly reduced in psoriatic lesional skin at 12 and 52 weeks, compared to baseline in the patients treated with brodalumab [3]. AMAGINE-2 and AMAGINE-3 AMAGINE-2 and AMAGINE-3 were both large, double-blind, placebo-controlled, multinational studies in moderate-to-severe plaque psoriasis with identical designs [30]. The efficacy and safety of brodalumab 140 and 210 mg every 2 weeks was evaluated in comparison to both ustekinumab and placebo (in a 2:2:1:1 ratio). Ustekinumab is a human monoclonal antibody which acts against the p40 subunit common to IL-12 and IL-23. Ustekinumab works upstream compared to brodalumab which works by driving downstream signaling. Ustekinumab was approved for the treatment of moderate-to-severe plaque psoriasis in 2009 and for PsA in 2013. During the first 12 weeks the treatment arms of both AMAGINE-2 and -3 included: 140 or 210 mg brodalumab every 2 weeks; ustekinumab (45 mg if ≤100 kg, 90 mg if >100 kg), on day 1 and week 4; or placebo. Patients who were 270 receiving brodalumab were randomly reassigned to 1 of 4 maintenance schedules: 140 mg or 210 mg every 2 weeks, 140 mg every 4 weeks, or 140 mg every 8 weeks after the initial 12-week period. Those who were assigned to the ustekinumab arm from the beginning continued receiving ustekinumab every 12 weeks. Those who commenced on placebo were switched to brodalumab 210 mg every 2 weeks. After week 52 and completion of the maintenance phase, patients who were still continuing to receive ustekinumab were eligible to switch to brodalumab 210 mg every 2 weeks as part of the OLE. Two co-primary endpoints assessed the efficacy of both brodalumab arms in both studies compared to placebo: the percentage of patients who reached PASI 75 and sPGA 0/1 at week 12. The efficacy of 210 mg brodalumab was also compared to ustekinumab in both studies with PASI 100 as the primary endpoint at 12 weeks. Both AMAGINE-2 and -3 showed superiority of 210 mg brodalumab over ustekinumab and placebo. However, the patients in the brodalumab 140 mg arm had response rates significantly superior to ustekinumab only in AMAGINE-3 [30]. A pooled analysis of both trials showed similar efficacy of brodalumab in patients with prior biologic exposure, as well as those who were naïve. Non-neutralizing antibodies against brodalumab were found in AMAGINE-2 and AMAGINE-3 (in 1.8 and 2.3% of patients, respectively). No loss of efficacy or adverse events were associated with these antibodies. Neutralizing antibodies were not found in any patients [30]. The most common AEs included nasopharyngitis, upper respiratory tract infection, headache, and arthralgia, with the proportion of patients on brodalumab or ustekinumab reporting at least one AE greater than that of placebo patients. Transient, reversible, mostly mild cases of neutropenia also occurred more frequently in the brodalumab and ustekinumab groups as compared to placebo, but were not associated with serious infections. No clinically apparent differences were seen in the types of serious AEs among the study groups. Consistent with the role that IL-17A plays in mucocutaneous host A. S. Silfvast-Kaiser et al. defense, Candida infections occurred more commonly with brodalumab (1.2%) than with ustekinumab (0.5%) or placebo (0.5%) in the induction phase [47–49]. Over the 52-week trial periods, 6 deaths occurred in both the brodalumab and ustekinumab groups due to: stroke (1), cardiac arrest (3), pancreatic carcinoma (1), and accidental death after motor vehicle collision (1). In AMAGINE-2, one patient committed suicide 19 days after the last dose of brodalumab 210 mg. After the completion of the studies, an additional 3 deaths occurred: one due to suicide, another from hemophagocytic histiocytosis syndrome, and the third from cardiomyopathy. One additional suicide occurred during the OLE, after week 52 [30]. A pooled secondary data analysis of AMAGINE-1, -2, and -3, compared patients who achieved PASI 100 or sPGA 0 to those who achieved PASI 75 to <100 or an sPGA of 1, in order to evaluate the clinical meaningfulness of complete skin clearance based on PSI scores and DLQI. When comparing PASI groups, a significantly greater proportion of patients achieving PASI 100 reported a PSI score of 0 (45%) compared to those achieving PASI 75 to <100 (8%). A similarly significant trend was seen in these two groups as related to DLQI scores of 0/1 (80% of PASI 100 and 55% of PASI 75 to <100 patients). When comparisons were based on sPGA 0/1, similar results were also seen [50]. AMAGINE-2 In AMAGINE-2, brodalumab was superior to ustekinumab in achieving PASI 100 at both the 140 mg and 210 mg doses at week 12, but only significantly superior with the 210 mg group. Response rates for PASI 75/90/100 in the brodalumab 140 mg, brodalumab 210 mg, and ustekinumab groups were 67%/49.0%/25.7%, 86%/69.9%/44.4%, and 70%/47.0%/21.7%, respectively. Placebo response rates for PASI 75/90/100 were: 8.1%/1.9%/0.6%, respectively [30]. Rates of sPGA scores of 0/1 were also significantly higher with both doses of brodalumab (58 and 79%) as compared to placebo (4%). The most common AEs were the common cold, upper respiratory tract infection, headache, and joint pain [51]. 21 Brodalumab AMAGINE-3 In AMAGINE-3, brodalumab was also shown to be superior to ustekinumab and placebo with a primary endpoint of PASI 100. Week 12 response rates for PASI 75/90/100 in the brodalumab 140 mg and 210 mg arms were: 69%/52.0%/27.0% and 85%/68.9%/36.7%, respectively. Ustekinumab and placebo exhibited response rates of PASI 75/90/100 at 69%/47.9%/18.5% and 6%/2.9%/0.3%, respectively [30]. The secondary endpoints of PSI, PASI 100 and sPGA at week 12 were greater for brodalumab compared to ustekinumab and placebo; however, statistically significant results were not achieved against ustekinumab. Rates of sPGA scores of 0/1 were significantly higher with both doses of brodalumab (60 and 80%) when compared to placebo (4%) [30]. The most commonly reported AEs in the brodalumab groups were nasopharyngitis, upper respiratory tract infection, arthralgia, and headache [52]. At 52 weeks, PASI 75/90/100 was achieved by 80%/73%/53% of patients on 210 mg brodalumab, respectively, outperforming both the ustekinumab (69%/57%/19%) and placebo cohorts [42]. Significant clinical efficacy of brodalumab was maintained through 120 weeks in an OLE study [53]. A systematic review of the long-term efficacy of brodalumab compared to other novel biologic agents for psoriasis concluded that brodalumab showed superiority compared to secukinumab, ustekinumab, and etanercept in sustaining PASI response as well as 52-week complete clearance [54]. The majority of patients rescued with brodalumab after initial ustekinumab treatment achieved PASI 75 and sPGA 0/1 responses by week 12 [30]. By week 52, a significant number of these patients had also achieved PASI 100. Brodalumab showed greater effectiveness when compared to ustekinumab in non-naïve biologic patients, with 40% of brodalumab patients achieving PASI 100 compared to 17% of those on ustekinumab. Brodalumab exhibited more rapid onset of action as well, achieving PASI 75 at a median of 4.2 weeks versus 9.4 weeks with ustekinumab. 271 Clinical Efficacy in Psoriatic Arthritis Brodalumab has also been investigated as a potential treatment for psoriatic arthritis. Although early results from PsA trials showed some promise, further investigations are needed to evaluate its potential to benefit patients with PsA, as brodalumab does not yet carry an indication for the treatment of PsA. A phase II randomized, placebo-controlled trial showed that approximately 40% of 159 patients treated with brodalumab 140 or 280 mg every 2 weeks were able to achieve ACR20 (American College of Rheumatology response criteria) compared to 18% of placebo patients at week 12. At week 12, rates of ACR50 for brodalumab versus placebo were 14 and 4%, respectively. By 24 weeks, 51% patients in the 140 mg group and 64% in the 280 mg group achieved ACR20 response versus 44% for those who switched from placebo to open-label brodalumab. Response was sustained through week 52 [55]. It is important to recognize that in clinical trials for PsA, up to 50% of patients are maintained on their preexisting systemic therapies when commencing a biologic clinical trial. The most commonly reported AEs were similar to those of the plaque psoriasis trials. Rates of serious adverse events were similar as well (3 vs. 2% in brodalumab vs. placebo groups, respectively). No statistical difference in response was seen in non-naïve biologic patients [32]. In a Japanese phase II trial for moderate-to-­ severe plaque psoriasis, 20% improvement in ACR20 was seen in patients receiving brodalumab 70 mg (1 patient), 140 mg (2 patients), and 210 mg (4 patients) and none receiving placebo [40]. At week 52, 75% of the patients with PsA in the 210 mg group achieved ACR20 compared to 38% in the 140 mg group [41]. Brodalumab completed phase 3 testing for psoriatic arthritis in 2017 (AMVISION-2). Results have not been published. One phase 3 trial (AMVISION-1) was terminated in 2015, with a cited reason of “sponsor decision.” 272 linical Efficacy in Difficult-to-Treat C Psoriasis Subgroup analyses in psoriasis patients treated with brodalumab with nail and scalp involvement (both considered difficult-to-treat areas) showed improvement in nail psoriasis severity index (NAPSI) score as well as psoriasis scalp severity index (PSSI) from a baseline score of ≥6, and ≥15 with scalp surface area involvement of ≥30%, respectively. In patients with either of these two subtypes of psoriasis, significant improvements from baseline to week 12 were seen in both NAPSI and PSSI scores (p < 0.001). For the brodalumab 140 mg and 210 mg groups, 37.5 and 46.3% improvement of NAPSI was shown, respectively, compared to 11.6% for placebo patients at week 12. Of 661 patients enrolled in the AMAGINE-1 study, 9.5, 61.8, and 89% of the placebo, brodalumab 140 mg, and 210 mg groups, achieved PSSI 75 response at the end of the 12-week induction phase, respectively. PSSI 90 and 100 responses of 4.2%, 52.4%, 76.8% and 3.2%, 41%, and 63.4%, were seen in the same treatment groups, respectively. Significant improvement in both brodalumab groups was seen as early as week 2 and maintained until week 12 [46, 56]. Palmoplantar pustular psoriasis (PPPP), also known as palmoplantar pustulosis (PPP), is a form of psoriasis characterized by sterile yellow pustules of the palms and/or soles, sometimes in addition to the characteristic indurated, erythematous, scaly lesions of plaque psoriasis. Largely because of the location of involvement, this condition can be extremely debilitating due to pain and disease-related functional limitation. Recent studies have shown that the IL-17 cytokine plays an important role in the pustular forms of psoriasis, with detectable levels of IL-17A, -C, and -F in palmoplantar pustular lesions [57–59]. There are limited data available regarding the effectiveness of biologic treatment for this psoriatic variant. The few case reports regarding PPPP/PPP patients treated with brodalumab show inconsistent results. Formal clinical trials need to be performed to determine whether brodalumab could A. S. Silfvast-Kaiser et al. be an effective therapeutic option for pustular forms of psoriasis [60, 61]. Improvements in Quality of Life Health-related quality of life assessed by the Psoriasis Symptom Inventory (PSI) and the Dermatology Life Quality Index (DLQI) in all clinical trials with brodalumab exhibited significantly higher numbers of patients with improvement on both doses of brodalumab compared to placebo [29, 30]. At week 12 of a pooled analysis of AMAGINE-1, -2, and -3, significantly more brodalumab patients in the 210 mg every 2 weeks group had a DLQI score of 0/1 versus those on placebo (59 vs. 6% respectively, p < 0.001) [62]. A majority of patients (68–84%) receiving brodalumab also reported that psoriasis had no impact on their daily activities, leisure activities, personal relationships, and work/school as a result of their treatment [62]. In AMAGINE-2 and -3, brodalumab also improved productivity loss as indicated by the self-reported Work Limitations Questionnaire [63]. Depression and anxiety as assessed by the Hospital Anxiety and Depression Scale (HADS) were also improved. Among those patients with baseline moderate and severe anxiety or depression (n = 139), a majority improved to normal, mild, or moderate at week 12 [64]. Safety and Tolerability Common Adverse Events The most common adverse events relating to brodalumab therapy, reported through week 120 of an open label extension (OLE) study in patients receiving treatment with brodalumab can be found in Table 21.3 [32]. Most adverse events regarding brodalumab are related to its potential to increase the risk of infection due to its role as an immune modulator. Similar to other IL-17 inhibitors, brodalumab has demonstrated a small increased risk of neutrope- 21 Brodalumab Table 21.3 Most commonly reported adverse events of brodalumab through week 120 of open-label extension Most common adverse events with brodalumab therapy through 120 weeks • Nasopharyngitis (27%) • Upper respiratory tract infections (20%) • Arthralgia (16%) • Back pain (11%) • Gastroenteritis (10%) • Influenza (9%) • Oropharyngeal pain (9%) • Sinusitis (9%) nia and Candida infections, as well as potential exacerbation of inflammatory bowel disease [6, 32, 33]. In addition to the most common adverse events (seen in Table 21.2), mild to moderate oral candidiasis, injection site reactions, and serious adverse events occurred in 3, 8, and 15% of patients, respectively. Transient grade 2 absolute neutrophil abnormalities were seen in 2% of patients. All resolved on their own without treatment modification. Several cases of grade 3 neutropenias were also reported [32]. SAEs throughout the entirety of the study occurred at a rate of 8.3 in subjects treated with brodalumab, and at a rate of 13 with those on ustekinumab. Within AMAGINE-2 and AMAGINE-3 the rates were 7.9 and 4.0, respectively. Patients should be counseled to seek medical care should signs or symptoms of infection occur while on biologic therapies, including brodalumab. Inflammatory Bowel Disease There is a known association between psoriasis and inflammatory bowel disease (IBD). Patients with Crohn’s disease and ulcerative colitis (UC) have shown higher prevalence rates of psoriasis. Similarly, psoriasis patients have also shown higher rates of IBD. Crohn’s disease is genetically linked to psoriasis [28]. Brodalumab is contraindicated in patients with Crohn’s disease [28]. Clinical trials for brodalumab excluded patients with an established diagnosis of IBD as potential risk of exacerbation had already been observed in patients in the ixekizumab and secukinumab trials. The inci- 273 dence of new-onset Crohn’s disease in clinical trials with brodalumab was approximately less than 1:1000, with one patient on treatment with brodalumab experiencing new-onset Crohn’s disease during the maintenance phase. In general, providers should use caution when considering the use of IL-17 inhibitors in patients with IBD [28, 30, 56]. Serious Infections In the pivotal phase 3 clinical trials, serious infections and fungal infections were observed at a higher rate in patients on brodalumab as compared to placebo [30]. One patient had to discontinue therapy due to cryptococcal meningitis [31]. Immunizations The use of live vaccines in patients on brodalumab should be avoided [28]. If necessary, treatment may be interrupted for administration [65]. Pregnancy No specific contraindication exists for the use of brodalumab in pregnancy. However, risks and benefits should be carefully assessed prior to initiation of therapy as efficacy and safety of brodalumab use in pregnancy has not been investigated. It is unknown whether brodalumab is excreted in human milk. Elderly Brodalumab has not been associated with higher rates of adverse reactions in the elderly [66]. Suicidal Behavior During the phase III brodalumab clinical trials, 4 completed suicides (one case was ruled as indeterminate) were seen. These all occurred after the 274 A. S. Silfvast-Kaiser et al. first 12 weeks of the trials (after the placebo-­ Table 21.4 Screening guidelines prior to initiation of controlled portion), with 3 of 4 suicides occur- therapy with brodalumab ring in patients who had previously attained PASI Screening guidelines for brodalumab initiation 100 on brodalumab. All patients who completed – Complete blood count – Comprehensive metabolic panel suicide had underlying psychiatric disorders or – Hepatitis B/C stressors and had received brodalumab therapy at – HIV 210 mg every 2 weeks; however, none of the sui- – TB cides occurred during active treatment with bro- – Personal or family history of IBD or symptomatology consistent with IBD dalumab [67]. The suicides occurred 58, 27, and – Psychiatric history 19 days after each subject’s last dose [16, 68]. The FDA concluded there was no established drug-related risk of suicide or suicidal ideation; Contraindications and Cautions they also found no causal or temporal relationship. Although a causal or temporal relationship Brodalumab is contraindicated in patients with is lacking, brodalumab carries a black box warn- Crohn’s disease due to observed worsening. It ing for increased risk of suicidal ideation and should be used with caution in patients with behavior [69, 70]. As a result, it is only available IBD. Careful attention should also be paid to to registered providers to prescribe through the patients who present with chronic or recurrent Siliq® Risk Evaluation and Mitigation Strategy infections. Should a patient experience a non-­ (REMS) Program which requires providers to be resolving active infection, the medication should certified with the program in addition to having be discontinued. patients sign an agreement and receive a pocket-­ size guide explaining suicidal behavior and what behaviors would warrant immediate medical Initiation of Therapy evaluation [32, 68]. Oftentimes, the burden of psoriasis extends In addition to screening for any of the above conpast the skin’s surface. This includes significant traindications, most providers recommend estabeffects on patient quality of life and disease-­ lishing baseline health before initiating therapy related morbidity. Studies have shown that with biologics, including brodalumab. In general, patients with psoriasis are overall more prone to a patient’s health history, existence of comorbidiexperience psychological burden and to attempt ties, and baseline status prior to the initiation of suicide than patients without psoriasis [71–74]. biologic therapies such as IL-17 agents should be Patients with psoriasis experience higher rates of established by performing a thorough history and depression, anxiety, self-harm, and suicidality physical, along with the appropriate laboratory compared to the general population. assessments (Table 21.4). This includes recomUnsurprisingly then, most of the patients who mended tuberculosis (TB) testing [32]. completed suicide during the brodalumab clinical trials had a history of predisposing risk factors such as depression and/or suicidal behavior [67]. Conclusion Prior to prescribing brodalumab, providers should weigh the risks and benefits of therapy with Psoriasis is a complex and debilitating disease brodalumab in patients with a history of depres- which can impact both the physical and emosion, suicidal ideation, and/or behavior. Patients tional health of a patient. The scope of psoriasis and their caregivers should also be instructed to treatments has widened considerably within the seek medical attention immediately should the last decade. As the understanding of psoriasis and patient begin experiencing worsening or new its pathogenesis deepens, psoriasis therapies are depression, manifestations of suicidal ideation or becoming increasingly more effective and spebehavior, or other significant mood changes. cific. For many years, PASI 50 and later PASI 75, 21 Brodalumab were used as the standard end point in clinical trials for the evaluation of treatment efficacy. With newer biologic agents such as brodalumab, PASI 90 and PASI 100 are increasingly becoming the new standard as these are now realistic, achievable endpoints [18, 37, 75]. Brodalumab’s efficacy, rapid onset of action, and role in difficult-­to-treat psoriasis help distinguish it from other biologic agents [76]. Instead of blocking the individual IL-17 cytokines, brodalumab blocks the IL-17 receptor, which appears to yield additional clinical benefits for cutaneous disease. Its rapidity of action and its efficacy speak to the important role of the IL-17 receptor A in driving downstream signaling in keratinocytes, causing the expression of the pro-inflammatory molecules that contribute to psoriasis [24, 77, 78]. Further clinical trials regarding progression of joint disease and destruction in patients treated with brodalumab are needed as the efficacy results for psoriatic arthritis are less striking [38]. Despite its boxed label warning regarding suicidality, providers should consider brodalumab a realistic treatment option, especially for those who have experienced failure of other treatment options, or for those with a significant inflammatory flare of their psoriasis. Evaluation of the possible association of brodalumab with suicidality have failed to identify a link [69, 70]. It is important to remember that psoriasis patients overall are known to carry a significantly greater psychological burden (including higher incidences of both suicidal ideation and completed suicides) compared to patients without psoriasis [71–74]. Depression and anxiety symptoms have been shown to improve with treatment by biologics in these patients, including with treatment by brodalumab [79]. Compared to placebo and ustekinumab, brodalumab has shown significant superiority, with mean time to PASI 75 response nearly twice as rapid for brodalumab patients (210 mg every 2 weeks) compared to those treated with ustekinumab [80]. By targeting the Th17 inflammatory pathway which is specific and critical to the development of psoriatic plaques, IL-17 inhibitors, including brodalumab, have shown the potential to enhance and shorten 275 the duration to clinical improvement compared to other biologic agents available for the treatment of psoriasis. The efficacy and rapidity of improvement with brodalumab treatment speaks volumes regarding the exciting future of biologics for moderate-to-severe psoriasis. References 1. Takeshita J, Gelfand JM, Li P, et al. Psoriasis in the US Medicare population: prevalence, treatment, and factors associated with biologic use. J Invest Dermatol. 2015;135:2955–63. 2. Ouyang W. Distinct roles of IL-22 in human psoriasis and inflammatory bowel disease. Cytokine Growth Factor Rev. 2010;21(6):435–41. 3. Roostaeyan O, Kivelevitch D, Menter A. A review article on brodalumab in the treatment of moderate-­to-­severe plaque psoriasis. Immunotherapy. 2017;9(12):963–78. 4. Hugh JM, Weinberg JM. Update on the pathophysiology of psoriasis. Cutis. 2018;102(5S):6–12. 5. Cai Y, Fleming C, Yan J. New insights of T cells in the pathogenesis of psoriasis. Cell Mol Immunol. 2012;9(4):302–9. 6. Roman M, Chiu M. Spotlight on brodalumab in the treatment of moderate-to-severe plaque psoriasis: design, development, and potential place in therapy. Drug Des Devel Ther. 2017;11:2065–75. 7. Iwakura Y, Ishigame H, Saijo S, Nakae S. Functional specialization of the IL-17 family members. Immunity. 2011;34(2):149–62. 8. Numaski M, Fukushi J, Ono M, et al. Interleukin-17 promotes angiogenesis and tumor growth. Blood. 2003;101(7):2620–7. 9. Lynde CW, Poulin Y, Vender R, Bourcier M, Khalil S. Interleukin 17A: toward a new understanding of psoriasis pathogenesis. J Am Acad Dermatol. 2014;71(1):141–50. 10. Elder JT. Genome-wide association scan yields new insights into the immunopathogenesis of psoriasis. Genes Immun. 2009;10:201–9. 11. Ellinghaus E, Ellinghaus D, Stuart PE, et al. Genome-­ wide association study identifies a psoriasis susceptibility locus at TRAF3IP2. Nat Genet. 2010;42:991–5. 12. Georgescu SR, Tampa M, Caruntu C, Sarbu MI, et al. Advances in understanding the immunological pathways in psoriasis. Int J Mol Sci. 2019;20(3):739. 13. Patel D, Kuchroo VK. Th17 cell pathway in human immunity: lessons from genetics and therapeutic interventions. Immunity. 2015;43(6):1040–51. 14. Li J, Li D, Tan Z. The expression of interleukin-17, interferon-gamma, and macrophage inflammatory protein-3 alpha mRNA in patients with psoriasis vulgaris. J Huazhnog Univ Sci Technolog Med Sci. 2004;24:294–6. Biosimilars for Psoriasis 22 Sarah Lonowski, Nirali Patel, Nika Cyrus, and Paul S. Yamauchi Abstract Biologics have revolutionized the treatment of numerous conditions. While the therapeutic benefit of biologics has been dramatic, the development of these agents has been accompanied by significant increases in healthcare costs. Recently there has been growing interest in the development of biosimilar agents, which have the potential to decrease costs and improve access to this powerful class of medications. In this chapter, we discuss the legisla- S. Lonowski Department of Medicine, Division of Dermatology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA e-mail: slonowski@mednet.ucla.edu N. Patel David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA e-mail: niralipatel@mednet.ucla.edu N. Cyrus Division of Dermatology, Kaiser Permanente Orange County, Irvine, CA, USA P. S. Yamauchi (*) Department of Medicine, Division of Dermatology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA Dermatology Institute & Skin Care Center, Inc., Clinical Science Institute, Santa Monica, CA, USA © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_22 tive, regulatory, and scientific framework for the development and approval of biosimilars. We also review the current state of the biosimilars market as it relates to psoriasis, and examine current barriers to widespread utilization of biosimilar agents in clinical practice. Key Learning Objectives 1. To understand the development pathway for biosimilars 2. To understand the approval process for biosimilars 3. To review the use of biosimilars for process Background Biologics are highly complex pharmaceutical agents isolated or synthesized from natural sources. These may take the form of antibodies, recombinant proteins, vaccines, blood, and plasma products [1–3]. Biologics are distinguished from chemically-synthesized drugs by their much larger size (100–1000×) and complex molecular structures [4]. Biologics have revolutionized the treatment of numerous dermatologic diseases, chronic inflammatory diseases, and certain malignancies. Biologics are the fastest-­growing segment of the 279 S. Lonowski et al. 280 Table 22.1 Available patent expiration dates for psoriasis biologics in the US and EU [2, 6] Brand name Humira Remicade Stelara Cimzia Enbrel Cosentyx Taltz Generic name Adalimumab Infliximab Ustekinumab Certolizumab Etanercept Secukinumab Ixekizumab Expiration year (US) 2016 2018 2023 2024 2028 2028 2036 pharmaceutical industry, accounting for 22% of the drugs approved by the United States Food and Drug Administration (FDA) between 2008 and 2017 [5]. Within the field of dermatology, the wave of biologics has undoubtedly had the greatest impact on the treatment of psoriasis, with 11 biologic agents currently FDA-approved for this indication and 5 new agents since 2017. As many biologics reach the end of their patent protection (Table 22.1), there has been growing interest in the development of biosimilar agents [7]. Biosimilars are reproductions of biologics which are highly similar to the original biologic agent (called the reference product) in terms of structure, mechanism of action, safety, and efficacy and must be given in the same form and dosage as the reference product [2, 8–10]. The FDA defines a biosimilar as a biologic product that is “highly similar to a US licensed reference biological product notwithstanding minor differences in clinically inactive components, and for which there are no clinically meaningful differences between the biological product and the reference product in terms of the safety, purity, and potency of the product” [11]. The European Medicines Agency (EMA) defines a biosimilar as “a biological medicinal product that contains a version of the active substance of an already authorized original biological medicinal product” [10]. While the therapeutic benefit of biologics has been dramatic, the development of these complex agents has been accompanied by significant increases in healthcare costs. Biologics are expensive, with an average cost of $10,000–30,000 per year [12]. Biologics accounted for nearly a quarter (24%) of worldwide pharmaceutical spending in 2015 and 40% of drug spending in the United Expiration year (EU) 2018 2015 2024 2021 2015 2030 Unavailable States (US) in 2017 [3]. Tumor necrosis factoralpha inhibitors adalimumab, etanercept, and infliximab were 3 of the top 5 most expensive drugs in terms of overall expenditures in 2016 [13]. The economic burden of psoriasis alone in the US is substantial, with estimates ranging from $35.2 to $112 billion [14, 15]. The Biologics Price Competition and Innovation Act (BCPI) of 2009 was approved as part of the Patient Protection and Affordable Care Act and provided a legislative framework for an abbreviated process for the approval of biosimilars. The stated goal of the BPCI was to improve patient access to biologic therapies by decreasing costs [2]. In addition to lowering drug development costs, the BCPI was expected to lead to increased competition among manufacturers [16]. The BPCI enabled the FDA to approve biosimilar medications following a review of the “totality of evidence” from extensive analytical, nonclinical, and clinical studies for these agents [4, 11, 17]. The first biosimilar agent was not approved in the US until 2015, however the speed of development has increased dramatically in the past few years. As of June 27, 2019 there are 21 biosimilars approved by the FDA, though not all of these are commercially available [18]. The potential for future growth is substantial, with over 1000 agents currently in the biosimilar pipeline [19]. The global biosimilars market size is expected to reach a value of USD $61.47 billion by 2025 [20]. Generics Versus Biosimilars While conceptually similar to generic small molecule medications, biosimilars differ from generics in several important ways. Generic drugs are, 22 Biosimilars for Psoriasis by definition, identical to their brand name equivalents, whereas biosimilars are not exact copies of their reference products [17]. Biosimilars are much larger products and have complex, dynamic structures that are highly vulnerable to alterations in the manufacturing process. Even slight changes in temperature, purification, and storage can lead to chemical modifications, such as misfolding or glycosylation, which affect the ultimate form and potentially the functionality of the drug [3, 17, 21]. For this reason, biologics cannot be exactly reproduced. Biosimilars are therefore considered “unique but related” to their reference product [22]. It is important to note that manufacturing of reference biologic products is also intricate, and changes in manufacturing processes, handling, and storage have the potential to cause slight modifications in the end-product, a phenomenon known as batch variability [23]. Biosimilars take longer and are more expensive to produce than generics. Biosimilars take on average 5–10 years and cost $100–250 million to develop, compared with 2 years and $1–10 million for most generics [3]. Another important distinction between generics and biosimilars is the potential for immunogenicity. Biologics, and therefore biosimilars, have the potential to elicit an immune response resulting in the production of anti-drug antibodies. Therefore, the development of biosimilars requires a thorough assessment of immunogenic potential and immunologic adverse events, which is not required for generic agents [24]. Naming of Biosimilars In January 2017 the FDA released guidelines which require a four-letter suffix to be appended to the original nonproprietary biologic name. For instance, Amjevita, a biosimilar to adalimumab, is designated as adalimumab-atto. The stated purpose of the guideline is to prevent confusion between products and to facilitate ongoing pharmacovigilance monitoring [25]. FDA requires that biosimilar package inserts contain the same information as the reference biologic regarding safety and efficacy, in a man- 281 ner similar to the labeling of generic medications [2]. Notably, these labels may not include information on clinical data demonstrating no clinically meaningful differences between the biosimilar and its reference product [2, 17]. Development of Biosimilars The process of biosimilar development centers on reproduction of the essential aspects of the reference product following a thorough examination of its structure and function. Biosimilar manufacturers must first study the reference product based on publicly available information, then use “reverse engineering” to independently design a process that will reproduce a highly similar biological agent [3, 4]. Because of the large and dynamic structure of these agents, the goal of this undertaking is not to create an exact copy of the originator biologic, but to create a bioequivalent product, with only minor variations in clinically insignificant components of the structure. Initially, detailed characterization studies are performed in order to identify the critical quality attributes (CQAs) of the reference biologic agent [11]. These CQAs determine the biologic function and therefore the resulting clinical effect [11]. Once a biosimilar manufacturer has produced a product that is highly similar to the reference product in terms of structure, mechanism of action, and CQAs, the product can undergo approval through appropriate regulatory bodies. Approval Process The approval process for biosimilars focuses on establishing biosimilarity between the proposed product and the reference biologic agent rather than demonstrating independent safety and efficacy, as is the case for most novel pharmaceutical agents. The manufacturer of the biosimilar product submits comparative data to the reference agent, which is subsequently analyzed by the regulatory bodies to determine whether the proposed product will be approved as a biosimilar. S. Lonowski et al. 282 Characterization studies Nonclinical studies (Animal) Clinical pharmacologic studies • Analysis of structure and function • Animal PK, PD, and toxicity profile • Pharmacokinetic (PK) assays • Pharmacodynamic (PD) assays • Immunogenicity assessment Comparative clinical study • Confirmatory safety and efficacy studies • Phase III clinical trial for at least one specific indication Fig. 22.1 Totality of evidence approach in approval of biosimilars by regulatory agencies The FDA has an abbreviated pathway for the approval of biosimilars which was established through the BCPI in 2009 [26]. Initially, manufacturers submit a biologics license application. In this 351(k) application, the biosimilar is tested across an array of different analytical assays, which must demonstrate by standard criteria that the activity of the new product is equivalent to the original agent, utilizes the same mechanism of action for the proposed condition(s) of use, and has the same route of administration, dosage form, and strength [26]. In addition, they must present data showing absence of clinically meaningful differences. The approval of biosimilars through the FDA and the EMA uses a “totality of evidence” approach. This involves a stepwise investigational process which includes analytical studies, animal studies, clinical pharmacokinetic (PK) and pharmacodynamic (PD) studies, i­mmunogenicity assessments, and, in some cases, additional clinical studies (Fig. 22.1) [11]. Analysis of Structure and Function First, comparative analytical characterization of the structure and function of the biosimilar is performed in order to demonstrate that the biological product is “highly similar” to the reference agent. Structural analyses ensure that the biosimilar encodes the same primary amino acid sequence as the original biologic agent [1]. The primary structure of the protein, its arrangement, size variants, level of glycosylation, and molecular charge are evaluated to characterize the quality attributes of the proposed biosimilar and to ­identify any potential differences that could impact clinical performance [27]. Minor differences in areas of the compound that are not clinically active, such as N- or C- terminal truncations, are acceptable [1]. Next, functional assays are performed to assess the pharmacologic activity of the biosimilar. In vitro functional assays must demonstrate similarity between the biosimilar and the reference product in terms of mechanism of action, receptor binding, effector cell response, chemokine production, cytotoxicity, and other measures [8]. In other words, these studies must prove the biosimilar replicates the effect of the originator product. Nonclinical (Animal) Studies In vivo (i.e. animal) studies are not required for biosimilar approval, but may be undertaken in some instances. Animal studies can provide more data in support of biosimilarity; in particular, animal toxicity studies may be utilized to demonstrate a similar safety profile compared to the reference product. Animal studies may be considered unnecessary if available structural and functional data already strongly support a high degree of similarity between the biosimilar and the originator biologic [2]. Clinical Studies After completion of the analytical/functional and nonclinical studies, clinical pharmacologic studies are designed as the ultimate comparative assessment to establish bioequivalence. These studies 22 Biosimilars for Psoriasis are typically conducted in healthy subjects, and must be statistically powered to determine differences between the biosimilar agent and the reference product. The purpose of pharmacokinetic (PK) assays is to determine how the human body affects the drug in terms of absorption, distribution, metabolism, and elimination [7]. Phase I human PK studies are required for all biosimilar candidates, and must demonstrate pharmacologic bioequivalence in certain parameters such as serum concentration of drug over time [17]. Human pharmacodynamic (PD) studies are not always required, but may be conducted to supplement PK data. For a human or humanized monoclonal antibody drug it can be difficult to determine how much of the drug is present in the serum at any given time. Through an assessment of the PK and PD data available for the biosimilar and the reference product, exposure response profiles can be compared. Clinical immunogenicity assessments are also undertaken to evaluate for any differences in the incidence and severity of immune responses between the reference product and the biosimilar. Such studies are important because alterations in immune response elicited by the biosimilar agent may promote the development of anti-drug antibodies and/or lead to acute clinical responses such as anaphylaxis [11, 17]. At least one clinical study comparing the immunogenicity of the proposed biosimilar to the reference product is required by the FDA [11]. The formation of anti-­drug antibodies is compared through titer changes, time to development of antibodies, and impact on pharmacokinetics [27]. Following PK, PD, and immunogenicity studies, biosimilar candidates must undergo at least one phase III clinical trial for a specific indication per FDA guidelines. Typically designed as an equivalence trial, the objective is not to establish efficacy of the biosimilar agent per se but rather to demonstrate non-inferiority to its reference product in terms of efficacy and safety [2, 11]. The biosimilar sponsor may choose which indication is tested. Importantly, biosimilar agents may eventually be approved for non-tested indications based on the concept of extrapolation. Extrapolation refers 283 to the approval of a given biosimilar medication for additional clinical indications, outside of those tested in a clinical trial. According to the FDA, this is allowable if there is “sufficient scientific justification for extrapolating clinical data to support a determination of biosimilarity for each condition of use for which licensure is sought” [11]. For example the biosimilar Inflectra (infliximab-dyyb) underwent clinical trials in patients with rheumatoid arthritis and ankylosing spondylitis, but has now been FDA-approved for all the same indications as the reference product, including plaque psoriasis and psoriatic arthritis [28, 29]. In fact, all infliximab and some adalimumab and etanercept biosimilars were approved for psoriasis via extrapolation [4]. Manufacturers must provide justification for extrapolation via explanation of the drug’s mechanism of action, pharmacokinetics, biodistribution, and immunogenicity [24]. Biosimilars may not be approved for indications that are protected by regulatory exclusivity, such as adalimumab’s exclusive indication for adults with moderate to severe hidradenitis suppurativa [2, 24]. Extrapolation decreases biosimilar development costs by decreasing the extent of clinical trials that must be performed. However, the lack of clear clinical trial data may be a point of hesitation for some clinicians considering prescribing biosimilars for extrapolated indications. Pharmacovigilance Studies The clinical studies required for FDA approval are of limited duration and are conducted in a limited population, and therefore may not identify all potential adverse effects. Post-approval safety monitoring is recommended by the WHO, FDA, and the EMA, but the specific requirements for this monitoring varies by agency [4]. The EMA requires pharmacovigilance plans as part of the initial biosimilar approval process, however the FDA does not [30]. The FDA instead recommends that manufacturers “consult with appropriate FDA divisions to discuss the sponsor’s proposed approach to post-marketing safety monitoring” on a case-by-case basis [11]. S. Lonowski et al. 284 In response to concerns regarding lack of safety data, the International Psoriasis Council has suggested the establishment of registries of all patients treated with biosimilars to enable monitoring of adverse treatment events over time [25]. biosimilar does not have any impact on safety, immunogenicity, or effectiveness. A biosimilar with an interchangeable designation may be substituted for an originator biologic without active intervention (such as a change in prescription) by a prescribing provider, though individual state substitution laws will impact this practice [33]. In some states, a product that is designated as interchangeable may be exchanged for the reference product by a pharmacist without notifying the ordering physician [33]. The NPF has issued a position statement on interchangeable biosimilar substitution, detailing the minimum requirements that should be met in order for biosimilar substitution to occur [34]. At the time of this publication there are no FDA-approved interchangeable biosimilars in the US. This may not be true for long, however. Boehringer Ingelheim is currently pursuing an interchangeability designation for BI 695501, its adalimumab biosimilar [35]. It is likely that other pharmaceutical companies will follow suit in the coming months and years as the market continues to grow and additional agents obtain regulatory approval (Fig. 22.2). Interchangeability Biosimilars can obtain a special classification as an “interchangeable biosimilar” through an additional approval process by the FDA, which was finalized in May 2019 [31]. Interchangeability is a specific designation given to biosimilars that are expected to achieve the same clinical outcome as the biologic product in any given patient, without any change in the safety or efficacy profile between the new product and original agent [32]. The approval process for these agents requires additional information from the manufacturer to ensure that the biologic agent can be effectively deemed interchangeable. Importantly, the application for interchangeability must include “switching studies,” which demonstrate that switching between the reference product and the Biologic manufacturer Payment for product Payment for product Drug wholesaler Dispensing pharmacy Drug ent m burse Formulary rebate r Presc reim iption Payer reimbursement Pharmacy benefits manager (PBM) Drug Payer/ Health Plan Insurance premiums % Pass through of rebate Legend Payments andProduct cash flow Premium payment Patient Employer Fig. 22.2 US Pharmaceutical Distribution and Reimbursement System (for self-administered drugs)—simple version [36] 22 Biosimilars for Psoriasis 285 Of note, several interchangeable biosimilars have been approved outside the US. Other countries have been studying the interchangeability of biosimilars in their respective markets. The NOR-SWITCH study was a randomized controlled study initiated by the Norwegian government to study the safety and viability of switching and interchanging infliximab and its biosimilar agents in several diseases including psoriasis, RA, spondyloarthritis, psoriatic arthritis, ulcerative colitis, and Crohn’s disease [37]. The study found that there were no differences or clinical worsening in any of these conditions when patients were switched between original products and their biosimilars. They also found no difference in the development of anti-drug antibodies [37]. Additional studies regarding multiple switch designs, including VOLTAIRE-X, will provide additional information about the effect of switching between biosimilars and innovator products [38]. Biosimilars in Dermatology Current FDA-approved biosimilars for the treatment of moderate to severe psoriasis are shown in Table 22.2. Amjevita (adalimumab-atto), Cyltezo (adalimumab-adbm), and Hyrimoz (adalimumab-­adaz) are biosimilar medicines to Table 22.2 Currently approved biosimilars for psoriasis [18] Brand name Erelzi Eticovo Amjevita Cyltezo Hyrimoz Inflectra Renflexis Ixifi Generic name Etanercept-­szzs Etanercept-­ykro Adalimumab-­ atto Adalimumab-­ adbm Adalimumab-­ adaz Infliximab-­dyyb Infliximab-­abda Infliximab-­qbtx Phase III trial for FDA approval date psoriasis August Yes 2016 April 2019 No September Yes 2016 August No 2017 October Yes 2018 April 2016 No April 2017 No No December 2017 adalimumab. Erelzi (etanercept-szzs) and Eticovo (Etanercept-­ ykro) are biosimilar medicines to etanercept. Inflectra (infliximab-dyyb), Renflexis (infliximab-­abda), and Ixifi (infliximab-qbtx) are biosimilar medicines to infliximab. While these biologic agents have all been approved for use in patients with moderate to severe psoriasis, only three (Amjevita, Erelzi, and Hyrimoz) have undergone clinical trials specifically for the indication of psoriasis [4]. The remaining biosimilars were approved via extrapolation. There are several other biosimilars in phase III clinical trials for the treatment of moderate to severe plaque psoriasis. These include BCD457, CHS-1420, GP2017, M923, MSB11022, and Myl-1401A, which are all biosimilars of adalimumab. Biosimilars of etanercept ONS3010 and CHS-0214 are also undergoing clinical trials for psoriasis [4]. There are also several biosimilars currently being studied for rheumatoid arthritis that may be approved for psoriasis via extrapolation if approved by the regulatory agencies their respective countries. These include FKB327 and LBALM (adalimumab biosimilars), PF-06410293 and LBEC0101 (etanercept biosimilars), and ABP 710, BCD-055, and N1-071 (infliximab biosimilars), all currently in phase III clinical trials [4]. Data regarding biosimilar use among dermatologists in the United States is limited, due to multiple factors including limited access to biosimilars in the current market and prescriber unfamiliarity with these medications [38]. Biosimilars are more widely used in Europe resulting in accumulation of significant data supporting the use of biosimilars for psoriasis. A 10-year study from Danish registries, for example, found no significant differences in safety or drug survival between reference and biosimilar versions of etanercept and infliximab [39]. In some European countries, national legislation has influenced the use of biosimilars as part of efforts to decrease costs and increase the availability of these drugs. In May 2015, for instance, Denmark instituted a national policy that patients should be started on or switched to the cheapest version of a given biologic (i.e. the biosimilar version) [39]. S. Lonowski et al. 286 Economic Advantage of Biosimilars In general, cost savings with biosimilars are less than those seen with generic small molecule agents. While cost reductions associated with small molecule generics can be 80% or higher, estimated biosimilar acquisition cost savings in the US have ranged from 10 to 40% [5, 40–43]. According to a 2017 estimate, the average cost for an 8-week supply of an infliximab biosimilar was 18% less than its reference product [5]. Outside the US, cost savings on biosimilars are variable, ranging from as high as 70% in Norway (due to discounted government pricing) to 30% in India [25]. Europe has approved 23 biosimilars, with an average price reduction of 20–30% [40]. While estimated US cost savings may seem modest, they remain significant when viewed in the context of the overall market size for biologic agents. In 2014, the Rand Corporation estimated a direct cost savings potential of $44.2 billion between 2014 and 2024, accounting for about 4% of total biologic spending over that period [17, 44]. As the biosimilar market expands, competition grows, and regulatory changes allow for streamlining of the developmental process for biosimilars, greater cost decreases may be seen. arriers to Use and Future B Directions Despite the passage of the BCPI nearly a decade ago, market uptake of biosimilars in the US has been slow. Reports from 2017 indicated that less than 10% of total biologic spending ($11.5 billion) was subject to competition from biosimilars [45]. There are several factors at play which ­continue to limit access to and acceptance of biosimilars, some of which will be reviewed here. atent Disputes and Extended P Patents for Branded Drugs As of July 2019, 20 biosimilars have received FDA approval but only 7 are commercially ­available [46]. Most have been withheld from the market due to ongoing patent disputes between the reference product manufacturer and the biosimilar manufacturer [12]. Additionally, while many original drug patents for biologics are approaching their expiration dates, some have additional patents related to manufacturing processes which provide ongoing protection; for example two additional patents for etanercept (Enbrel) can extend patent protection until 2029 [3, 21]. In many instances, patent litigation has had the effect of postponing market entry for biosimilar products. Recent litigation settlements involving Amgen and Sandoz, for instance, have resulted in delayed introduction of adalimumab biosimilars until 2023 [5]. Prescriber Considerations Physician awareness and comfort level with prescribing biosimilars is another barrier to widespread use of these medications. Incomplete understanding of what biosimilars are, apprehension about the significance of “minor differences” between reference products and biosimilars, and lack of long-term safety data are factors that may limit physician willingness to prescribe biosimilars. According to a recent online survey of US dermatologists, only 25% stated they “will definitely or are highly likely to prescribe biosimilars” [47]. Frequently cited concerns included safety (66%), efficacy (71%), immunogenicity (63%) and the potential for patients to be switched from a biologic to a biosimilar without clinician awareness [47]. Since no biosimilars have yet been given an interchangeable designation in the US, some clinicians remain wary of the potential for adverse immunologic effects if patients switch between originator biologics and biosimilars. It is worthy of note, however, that switching studies completed to date suggest that immunologic risk is low [5, 48] (Fig. 22.3). Prescriber acceptance of biosimilars hinges on knowledge and comfort level with these agents. A 2017 cross-sectional survey of dermatologists found that physicians who were “fairly to very 22 Biosimilars for Psoriasis 287 familiar” with biosimilars were more comfortable prescribing biosimilars for psoriasis, even if they had not been clinically studied for this ­indication [49]. Patient Factors While the economic benefits of biosimilars are easy to appreciate from a systems level perspective, this benefit may not be immediately apparent to the end user, i.e. the patient. The decreased acquisition cost of a biosimilar is most relevant to patients who have high deductible plans or who have co-insurance plans with expenses calculated as a percentage of a given drug’s list price [24]. Patients with fixed copays are not incentivized to seek out medications with a lower acquisition cost, since this cost is not directly passed on to them. In fact, such patients may be likely to prefer brand name reference products over biosimilars in these instances, given greater name recognition and overall familiarity. Patient Employer Insurance Coverage Premium Payment Dispenses Prescriptions Coinsurance Contracts and Services Medication Purchase (Contracted Price) In summary, biosimilars are a rapidly growing sector of the pharmaceutical industry, both in the US and globally. Despite the slow adoption of biosimilars in the US to date, there is potential for greater use of biosimilars in the future; the adoption of generic agents in the United States was similarly slow, but now generics account for the vast majority of prescribed medications [50]. At present, legal, regulatory, and financial constructs, lack of commercial availability, and low provider comfort level with the use of these agents remain barriers to large-scale adoption of biosimilars. Numerous biosimilar agents are currently in the pipeline as multiple commonly prescribed biologics are approaching their patent expiration dates. In the future, as utilization of these novel agents becomes more widespread, biosimilars have the potential to decrease healthcare costs globally and increase patient access to additional valuable diseasemodifying therapies. Premium Payment Legend Payments and Cash Flow Conclusion Prescription Reimbursement (Contracted Rate) Dispensing Pharmacy Health Plan/ Medical Benefit Network Agreement Wholesale Agreement Prescription Benefit Pharmacy Benefit Manager (PBM) Pharmaceutical Wholesaler Management Fees Distribution Agreement Medication Purchase (Contracted Price) Rebate Pharmaceutical Manufacturer Formulary Placement Fig. 22.3 US Pharmaceutical Distribution and Reimbursement System (for self-administered drugs)—complex ­version [36] Research Pipeline I: Oral Therapeutics for Psoriasis 23 D. Grand, K. Navrazhina, J. W. Frew, and J. E. Hawkes Abstract Over the last two decades, tremendous advances have been made in the treatment of psoriatic disease. Our understanding of the complex immunopathogenesis of this chronic inflammatory condition has led to the development of highly effective, safe, targeted monoclonal antibodies that result in clearance in the vast majority of treated patients. However, these recently approved therapies have multiple drawbacks and limitations that necessitate the development and testing of additional treatment modalities. To address this need, scientists and the pharmaceutical industry have pursued the identification and testing of several oral small molecules as novel treatments for plaque and psoriatic arthritis. These molecules in testing include JAK/ TYK2 inhibitors, phosphodiesterase inhibitors, RORγt inhibitors, fumarate esters, sphingosine 1 phosphate antagonists, neurokinin-­1 receptor D. Grand · K. Navrazhina · J. W. Frew Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, USA e-mail: dgrand@rockefeller.edu; knavrazhin@rockefeller.edu; jfrew@rockefeller.edu J. E. Hawkes (*) Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, USA Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA e-mail: jhawkes@rockefeller.edu © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_23 antagonists, adenosine A3 receptor agonists, H4 receptor antagonists, PRINS inhibitors, and spleen tyrosine kinase inhibitors. Key Learning Objectives 1. To understand the mechanism of action of novel oral agents in development for the treatment of psoriasis 2. To understand the appropriate use and efficacy of novel oral agents in development for the treatment of psoriasis 3. To understand the safety issues and appropriate monitoring of novel oral agents in development for the treatment of psoriasis Introduction Psoriasis is chronic inflammatory skin disease with a complex etiology. Current research demonstrates that interleukin-23 (IL-23) and IL-17-­producing T (T17) cells are central to the pathogenesis of this condition [1]. The effectiveness of current anti-psoriatic therapies is largely correlated with the ability of the treatment to disrupt the dysregulated IL-23/T17 signaling axis in the skin. With the recent approval of multiple selective monoclonal antibodies against IL-23 291 D. Grand et al. 292 Table 23.1 List of novel therapies currently in clinical trials for the treatment of psoriasis Drug name Baricitinib Upadacitinib Drug class JAK1/2 inhibitor JAK1 inhibitor Phase 2b 3 Company Eli Lilly AbbVie Filgotinib JAK1 inhibitor 2 Galapagos ASP015K BMS-986165 JAK3 inhibitor Tyk2 inhibitor 2 3 Janssen BMS LEO32731 PDE4 inhibitor 2 LEO VTP-43742 RORγt inhibitor 2 Vitae Pharma LAS41008 Dimethyl fumarate 3 Almirall FP187 Fumaric acid 2/3 Forward Pharma XP23829 Ponesimod (ACT-128800) Amiselimod (MT-1303) Serlopitant (VPD-737) CF101 Fumaric acid S1P receptor antagonist 2 2 Xenoport Actelion S1P receptor antagonist 2 Mitsubishi Tanabe Pharma Clinical trials NCT01490632 NCT03104374 NCT03104400 NCT03320876 NCT03101670 NCT03926195 NCT01096862 NCT04036435 NCT03624127 NCT02888236 NCT03231124 NCT03724292 NCT02555709 NCT02955693 NCT01726933 NCT01230138 NCT01815723 NCT02475304 NCT02173301 NCT01208090 NCT00852670 NCT01987843 Neurokinin-1 receptor antagonist Adenosine A3 receptor agonist H4 receptor antagonist IL-20 and PRINS inhibitor 3 Menlo Therapeutics NCT03343639 3 Can-Fite Biopharma NCT03168256 2 2 Ziarco Pharma CellCeutix NCT02618616 NCT02101216 NCT02494479 NCT02949388 ZPL-389 Prurisol and IL-17, we are witnessing unprecedented advances in the treatment of patients with psoriasis. Nevertheless, monoclonal antibodies have limitations including their high cost, insurance accessibility, loss of efficacy over time potentially due to anti-drug antibodies, and the need for intravenous or subcutaneous administration. The potential advantage of novel oral small molecules for the treatment of psoriasis include a decreased cost, avoidance of proteinrelated immune mechanisms leading to loss of efficacy with time, and ease of administration. Accordingly, many companies are testing the efficacy of a variety of small molecules for the potential treatment of plaque and psoriatic psoriasis. The purpose of this chapter is to provide an overview of the research pipeline for novel, oral therapeutics being studied for the potential treatment of psoriatic disease (Table 23.1). JAK Inhibitors The Janus kinase (JAK) family is composed of four tyrosine kinases (JAK1, JAK2, JAK3, and TYK2) that have essential roles as signal transducers downstream of activated cytokine receptors. In contrast to c-KIT and insulin, cytokine receptors lack intrinsic protein kinase domains and, therefore, rely upon constitutively active JAK tyrosine kinases (TYK) to convey immune signals [2]. Following JAK activation, one of the six STAT family members are consequently activated (STAT1–6) leading to its function as a transcrip- 23 Research Pipeline I: Oral Therapeutics for Psoriasis H4 Receptor Pathway SYK Pathway 293 GPCR Pathway (S1P, A3AR, NK1) NF-kB Pathway JAK-STAT Pathway CF101 ponesimod amiselimod ZPL-399 JAK/ TYK2 SYK cAMP fostamatinib apremilast LEO32731 Protein Kinase A baricitinib upadacitinib filgotinib ASP015k BMS-986165 MyD88 Protein Kinase C NF-kB JNK/ERK MEK/ERK STAT LAS41008 FP187 XP23829 CYTOPLASM VTP-43742 RORy abacavir hydroxyacetate NUCLEUS PRINS Fig. 23.1 Schematic of a T lymphocyte and the proposed mechanisms of action for several small molecules being tested in clinical trials for the treatment of psoriatic disease tion factor. Significant overlap between the various JAK and STAT isoforms have been demonstrated in mouse knockout models, thus highlighting the need to carefully study the impact of combination versus more selective JAK blockade [3]. Novel small molecules target one or more JAK isoforms (Fig. 23.1). JAK1 and JAK2 are involved in interferon-gamma signaling, whereas JAK3 is involved in signaling from type I cytokine receptors that use the common gamma chain (γc) including IL-2, IL-4, IL-7, IL-9, IL-15 and IL-21 [4, 5]. Although the JAK-STAT pathway does not directly suppress IL-17, it works indirectly through other STAT-dependent cytokines acting upstream (e.g. IL-23) [6, 7]. Novel therapies targeting the JAK-STAT pathway must balance suppression of pro-inflammatory signaling versus the over-suppression and disruption of vital developmental functions, such as neuronal development, protection against infections, and hematopoiesis [8]. Some of the differences between JAK isoforms can be inferred from the effects observed in individuals living with JAK deficiencies as 294 D. Grand et al. well as the clinical results from human studies. [13, 14]. SELECT-PsA 1 is an active phase III JAK3 deficiency (severe combined immunodefi- study with 640 participants who have had an ciency or SCID) is associated with decreased T inadequate response to at least one non-biologic and NK cell numbers, but unaffected B cells [9]. disease-modifying anti-rheumatic medication TYK2 deficiency is associated with susceptibility (e.g. methotrexate). SELECT-PsA 2 is currently to viral, fungal, and mycobacterial infection [10]. recruiting participants who have had an inadActivating somatic mutations in JAK1, JAK2, equate response to at least one biologic disease-­ and JAK3 have also been identified in myelopro- modifying anti-rheumatic drug with estimated liferative disease, leukemia, and lymphoma [11] enrollment of 1640 participants. Both trials raising the concern for a potential increase in wills compare two daily doses of upadacitinib malignancy risk with long-term treatment. versus placebo and adalimumab and will meaIn May 2012, the FDA approved tofacitinib, an sure ACR20 at week 12 as the primary endpoint oral pan-JAK inhibitor, for the treatment of rheu- [13, 14]. No clinical trials testing this compound matoid arthritis. This was the first approval of a for the treatment of plaque psoriasis have been medication in this specific drug class. Subsequent reported. FDA approval of tofacitinib for the treatment of The EQUATOR trial evaluated the efficacy psoriatic arthritis occurred in December 2017. and safety of JAK1-selective inhibitor, filgotinib, Other first-generation JAK Inhibitors include in patients with psoriatic arthritis [15]. 131 subruxolitinib, baricitinib, and oclacitinib. jects were randomly assigned to daily filgotinib A phase 2b trial for a JAK1/2-selective inhibi- 200 mg versus placebo. 80% of the filgotinib tor, baricitinib, randomized 271 participants to group achieved ACR20 at week 16 compared to placebo and four daily doses of baricitinib (2, 4, 33% of the placebo group (P value < 0.0001). 8, or 10 mg) for 12 weeks [12]. Based on treat- A greater percentage of participations in the filment response at week 12, doses were either con- gotinib group achieved PASI75 than the placebo tinued or increased for the next 12 weeks. 75% group (45.2 vs. 15%, P value = 0.0034). The improvement in the Psoriasis Area and Severity incidence of adverse events was similar between Index (PASI75) at week 12 was achieved in filgotinib and placebo with nasopharyngitis 42.9% of the 8 mg group and 54.1% of the and headache being the most common reported 10 mg group. Both proportions were statisti- adverse events [15]. An open-label extension cally significant compared to the placebo group study is ongoing and will provide long-term data (17%). Greater than 81% of treatment respond- on filgotinib therapy [16]. No clinical trials testers at week 12 maintained a PASI75 at week ing this compound for the treatment of plaque 24. With dose escalation, 48% of those who had psoriasis have been reported. not achieved PASI75 at week 12 achieved it by A phase 2a trial evaluating the efficacy and week 24. The most common adverse events of the safety of JAK3-selective inhibitor, ASP015K, placebo and baricitinib groups were infection-­ randomized 124 participants to four twice-daily related (e.g. nasopharyngitis). Cytopenia was doses (10, 25, 60, or 100 mg) or one daily dose observed in 4.6% of patients receiving baricitinib (50 mg) versus placebo for 6 weeks [17]. A compared to 0% in placebo; higher cytopenia greater change in PASI score was observed in a rates were documented in the 8 and 10 mg treat- dose-dependent manner in those receiving active ment groups. Rates of drug discontinuation due treatment with ASP015K compared to control to adverse events was highest in the 8 mg (6.3%) (P value < 0.001). Histological studies of biopand 10 mg (5.8%) treatment groups [12]. Phase sied lesional skin demonstrated active treatment III clinical trials for baricitinib in the treatment of dose-­ dependent decreases in epidermal thickplaque psoriasis are currently underway. ness, number of proliferating epidermal cells There are two ongoing phase 3 trials study- (measured by Ki67 staining), number of dermal ing the efficacy of a JAK1-selective compound, CD3+ T cells, and number of CD11c+ dendritic upadacitinib, in participants with psoriatic arthritis cells. A greater percentage of ASP015K subjects 23 Research Pipeline I: Oral Therapeutics for Psoriasis (46.3%) experienced treatment-related adverse events compared to the placebo group (37.9%), but the number of drug-related adverse events were similar among the two groups. No serious adverse events were reported [17]. No clinical trials testing this compound for the treatment of psoriatic arthritis have been reported. Tyk2 Inhibitor BMS-986165 is a selective Tyk2 inhibitor. A phase 2 trial randomized 267 participants to five doses of BMS-986165 (3 mg every other day, 3 mg daily, 3 mg twice daily, 6 mg twice daily, or 12 mg daily) versus placebo for 12 weeks followed by a 30-day follow-up period [18]. PASI75 at 12 weeks was observed in 9, 39, 69, 67, and 75% of the treatment doses, respectively, versus 7% of the placebo group. PASI100 was observed in 18% of the 6 mg twice daily group and 25% of the 12 mg daily group compared to 0% of the placebo group. Treatment effects were evident as early as day 15 and persisted after the 30-day follow-up period. Nasopharyngitis, headache, diarrhea, nausea, and upper respiratory infections were the most common reported adverse effects. A greater proportion of patients in the active treatment group experienced mild-to-moderate acne, possibly due to a change in the microbiome in response to the altered cytokine profile following treatment. However, no cases of pathologic agents such as herpes zoster, tuberculosis, or opportunistic infections, were documented [18]. Two phase 3 trials (POETYK-PSO-1 and 2) further testing BMS-986165 are currently underway and recruiting participants for a multicenter study in subjects with moderate-to-severe plaque psoriasis [19, 20]. An estimated 600 and 1000 participants will be randomized to BMS-986165 versus apremilast or placebo. The proportion of patients who achieved a Static Physicians Global Assessment (sPGA) score of 0/1 and PASI75 at week 16 of treatment will be the primary measure of treatment efficacy [19, 20]. No clinical trials testing this compound for the treatment of psoriatic arthritis have been reported. 295 PDE4 Inhibitors Phosphodiesterases (PDEs) are comprised of 11 families of molecules responsible for the degradation of cyclic nucleotides on adenosine and guanine monophosphates (cAMP and cGMP, respectively). PDE4 is a specific isoform found to be highly expressed in keratinocytes and inflammatory leukocytes (T cells, monocytes, dendritic cells, and neutrophils), as well as brain, cardiovascular, and smooth muscle tissues [21]. Suppression of PDE4 leads to elevation in cAMP and activation of protein kinase A (Fig. 23.1), as well as other downstream mediators such as Epac1/2. Several PDE4 inhibitors have been developed based upon the subtypes of PDE4 elevated in the primary tissues involved in specific inflammatory diseases. In psoriasis and psoriatic arthritis, PDE4B and PDE4D were identified as differentially expressed in psoriasis peripheral blood mononuclear cells. Accordingly, apremilast, which has high specificity for PDE4, was developed as an oral therapy for the treatment of psoriasis and psoriatic arthritis [22, 23]. The downstream effects of apremilast in psoriasis include the downregulation of interferon-γ and TNF-α with resultant Th17 pathway suppression and skin clearance [24]. Apremilast was approved by the FDA for the treatment of moderate-to-­ severe plaque psoriasis in September 2014. It is also approved for the treatment of psoriatic arthritis and Behcet’s disease. Since the FDA’s approval of apremilast, another PDE4 inhibitor, LEO32731, is currently being studied in human trials. A phase 2 single-­ center study of 36 participants with moderateto-­severe psoriasis compared twice-daily 30 mg LEO32731 to placebo. Endpoints included PASI score and the Itch Numeric Rating Scale score between baseline and week 16 of treatment. The study was completed in June 2017, but results have not yet been published [25]. No clinical trials testing this compound for the treatment of psoriatic arthritis have been reported. However, several other topical PDE4 inhibitors are currently under development, including pefcalcitol and HFP034. 296 RORγt Inhibitors D. Grand et al. New oral formulations of fumarates with greater gastrointestinal tolerability are being RAR-related orphan receptor gamma t (RORγt developed, including FP187 and XP23829. A or RORγ2) is a DNA-binding transcription factor dose-ranging phase 2 clinical trial at two study (Fig. 23.1) expressed in CD4+ T cells leading to locations in Germany investigated the efficacy for Th17 differentiation. It is essential for IL-17 of FP187 on moderate-to-severe psoriasis [33]. production and is, therefore, an attractive target Participants were randomized to three-times daily for small molecule inhibition in psoriatic disease FP187 (250 mg), twice-daily FP187 (375 mg), [26]. Murine models have shown that RORγt twice-daily and FP187 (250 mg) versus plaknockdown suppressed the Th17 response cebo. The primary endpoint was the proportion induced by intradermal IL-23 injection. IL-17A/ of patients achieving PASI75 at 20 weeks. This C/F, CCL20, and CCR6 are relevant genes known study was completed in January 2012, but results to be regulated by RORγt and RORα [27]. have not yet been published [33]. Additional A phase 1 single ascending dose study ran- phase 2 and 3 clinical trials involving FP187 for domized 53 healthy volunteers to a single dose of the treatment of psoriatic arthritis and plaque VTP-43742 (ranging from 30 to 2000 mg) versus psoriasis, respectively, were initiated but subseplacebo [28]. A phase 1 multiple ascending dose quently withdrawn. A multicenter, dose-­ranging, study randomized 40 healthy volunteers to VTP-­ phase 2 clinical trial randomized plaque psoria43742 (ranging from 100 to 1400 mg/dL) versus sis participants to three XP23829 doses (400 mg placebo [29]. In both studies, VTP-43742 was daily, 800 mg daily, or 400 mg twice daily) vershown to be safe and well-tolerated at all doses, sus placebo [34]. Efficacy was assessed as the and ex vivo whole blood assays showed a dose-­ percent change in PASI score at 12 weeks. This dependent decrease in IL-17A secretion following study was completed in May 2015, but results treatment. In the multiple ascending dose study, all have not yet been published [34]. No clinical but the lowest dose resulted in a >90% inhibition of trials testing this compound for the treatment of RORγt-dependent IL-17A secretion for 24 h [28– psoriatic arthritis have been reported. 30]. No clinical trials testing this compound for the The BRIDGE trial was a phase 3 noninferiority treatment of psoriatic arthritis have been reported. trial where participants were randomly assigned into three groups: LAS41008 (dimethyl fumarate), fumaderm (dimethyl fumarate combined Fumarate Esters with monomethyl fumarate salts), or placebo [35]. Doses of each were escalated based upon Oral fumarates are a longstanding treatment clinical response with a maximum daily dimethyl for moderate-to-severe psoriasis, particularly fumarate dose limited to 720 mg. 37.5% of subin Europe. However, the exact mechanism of jects receiving LAS41008 at week 16 achieved action by which this class of drugs act is incom- PASI75 making it superior to placebo (15.3%, pletely understood. The esters of fumaric acid, p < 0.001) and noninferior to fumaderm (40.3%, monomethyl fumarate and dimethyl fumarate, p < 0.001). LAS41008 demonstrated superiorare the active fumarates responsible for the anti-­ ity to placebo upon assessment with sPGA 0/1 inflammatory properties of this therapeutic class at week 16 (33 vs. 13%, p < 0.001). Rebound [31]. Multiple mechanisms of action have been was observed in 1.1% of LAS41008 subjects at proposed, including lowering glutathione levels, 2 months off treatment compared to 9.3% of plaactivation of Nrf2 antioxidant pathways, inhibi- cebo subjects. Lymphopenia is a known adverse tion of NF-κB activity (Fig. 23.1), shifting Th1/ effect of dimethyl fumarate-­ containing comTh17 to a Th2 dominant phenotype, binding of pounds. In this study, lymphocyte counts lower monomethyl fumarate to GPR109A (implicated than 0.7 × 109 cells/L were observed in a greater in the flushing response of patients on fuma- percentage of LAS41008 (7.9%) and fumaderm rates), and modulation of hypoxia inducible fac- (7.4%) subjects compared to placebo (0.7%). tor and JAK-STAT signaling pathways [32]. However, the lymphopenia resolved upon treatment 23 Research Pipeline I: Oral Therapeutics for Psoriasis discontinuation. Most adverse effects were mild in severity and included gastrointestinal disorders and flushing [35]. No clinical trials testing this compound for the treatment of psoriatic arthritis have been reported. 297 ease, ­ macular degeneration, ulcerative colitis, pyoderma gangrenosum, and uveitis. Two S1P modulators, amiselimod and ponesimod, have undergone Phase 2 trials for chronic plaque psoriasis. A phase 2a study of ponesimod randomized 66 participants with moderate-to-severe chronic plaque psoriasis to either ponesimod 20 mg daily S1P Receptor Antagonists or placebo [40]. The primary endpoint was percentage change in PASI score at week 6. Results Sphingosine 1 phosphate (S1P) is a bioac- have not yet been published. In 2012, a phase 2 tive lipid molecule first identified as a stimula- trial randomized 326 participants to two daily tor of fibroblast proliferation. It was originally doses of ponesimod (20 or 40 mg) versus pladescribed as an intracellular second messenger of cebo [41]. A greater proportion of participants in G protein coupled receptors (GPCR) mediating the 20 and 40 mg groups (46 and 48.1%, respecintracellular calcium levels secondary to platelet-­ tively) achieved PASI75 at week 16 compared derived growth factor (PDGF) and IgE signaling to placebo (13.4%, P value < 0.0001 for both (Fig. 23.1). However, the discovery of high affin- groups). Additionally, sPGA 0/1 was noted in ity cell surface receptors (S1P1–5) and the role 27.8 and 32.3% of the ponesimod 20 and 40 mg of these receptors in angiogenesis, tissue remod- groups, respectively, compared to only 4.5% of eling, and inflammatory mediator synthesis have the placebo group. At week 16, those who had accelerated the development of target molecules received ponesimod and achieved a greater than to modulate these pathways [36]. The fungus 50% decrease in PASI score either continued ponIsaria sinclairii, used in traditional Chinese med- esimod at the same dose or switched to placebo. icine, contains Myriocin from which synthetic At week 28, a greater proportion of those who S1P modulators have subsequently been devel- remained on ponesimod achieved PASI75 comoped for clinical use and testing [37]. pared to those who switched to placebo. In addiFingolimod is a non-selective S1P agonist and tion, those that switched to placebo had a greater functional antagonist of S1P receptors 1, 3, 4 and likelihood of disease relapse. Laboratory analy5 [38]. The binding of phosphorylated fingoli- sis revealed dose-dependent lymphopenia in the mod results in intracellular signaling via GPCRs ponesimod groups (56 and 65% mean decrease in (Fig. 23.1). The overall effect of fingolimod is lymphocyte count, respectively) compared to 2% immune modulation via trapping of lymphocytes of the control group. The incidence of infection in secondary lymphoid tissues, thus causing T was similar across all groups. The most common and B cells to accumulate rather than migrate into reported adverse events associated with ponesithe blood stream. Sustained lymphopenia is the mod were dyspnea, elevated liver enzymes, and suspected mechanism of action responsible for dizziness [41]. the efficacy of this drug in the treatment of relapsA phase 2a dose-ranging study of amiselimod ing-remitting multiple sclerosis. Lymphopenia recruited 142 subjects with moderate-to-severe associated with fingolimod resolves in approxi- psoriasis and randomized them to low, medium, mately 7 days after cessation of the drug [39]. and high doses of amiselimod versus placebo The mechanism of action of fingolimod and [42]. The proportion of participants achieving other S1P modulators makes this class of drug an a PASI75 at week 16 was the primary measure attractive potential therapy for multiple autoim- of treatment efficacy. This study was completed mune or inflammatory conditions. in September 2014, but study results have not Other S1P modulators are in various stages yet been published. No clinical trials testing of development for the treatment of psoriasis, ­ponesimod or amiselimod for the treatment of SLE, dermatomyositis, graft versus host dis- psoriatic arthritis have been reported. 298 Neurokinin-1 Receptor Antagonists Neurokinin-1 (NK1) is the preferred receptor for substance P, the binding of which results in rapid downstream activation of NF-κB, ERK, and p21 associated pathways (Fig. 23.1) [43, 44]. Substance P has a vital role in the stimulation of pro-inflammatory leukocytes, as well as the innate response to viral, parasitic, and bacterial infections through enhanced production of IL-1, IL-6, TNF-α, MIP-1B and IFN-γ [45]. NK1 pathways are also implicated in the modulation of itch, stress, sleep, anxiety, and nausea via the central nervous system. This has led to the development of NK1 receptor (NK1R) antagonists for pruritus as well as chemotherapy-associated nausea and vomiting [46]. NK1R antagonists have also been tested in psoriasis given the associated pruritus and the contribution of pro-inflammatory cytokines to the keratinocyte mediated feed-­forward pathway in psoriasis [47]. Additionally, localized TGFβ elevation in psoriatic skin is associated with delayed internalization of the NK1R complex, which leads to increased elevation of IL-1, IL-6, and TNFα levels [43]. A phase 2 study randomized participants to a serlopitant 5 mg daily group or placebo group. The primary endpoint WI-NRS 4-point responder rate at 8 weeks was achieved in 33.29% of the serlopitant group versus 21.07% of the placebo group [48]. Serlopitant was not associated with any serious adverse events. Diarrhea, nasopharyngitis, and headache were the most common adverse events reported. An open-label extension study is ongoing and will provide long-term data on serlopitant therapy for the treatment of psoriasis [49]. No clinical trials testing this compound for the treatment of psoriatic arthritis have been reported. Adenosine A3 Receptor Agonists Adenosine A3 receptors (A3AR) are GPCRs (Fig. 23.1) associated with regulation of local tissue function in the absence of an adequate energy supply. They are specifically overexpressed in inflamed and malignant tissues leading to the D. Grand et al. identification of this receptor as a potential therapeutic target [50]. The binding of adenosine to A3AR constitutively inhibits adenylyl cyclase and mitogen-activated protein kinase pathways. Chronic elevation of adenosine in the setting of pro-inflammatory cytokines leads to increased levels of membrane bound A3AR, which has been identified in malignancy and chronic inflammatory disorders [51]. This chronic elevation of adenosine results in delayed internalization of A3AR and upregulation of downstream NF-κB (Fig. 23.1), and β-catenin signaling resulting in transcriptional alterations leading to a pro-­ inflammatory environment. Therefore, A3AR agonists are being evaluated for the treatment of rheumatoid arthritis, inflammatory bowel disease, and uveitis, as well as melanoma, prostate, colon, breast, and hepatocellular carcinomas [52]. A phase 2 dose-ranging clinical trial randomized 75 participants to twice-daily doses of CF101 (1 mg, 2 mg, or 4 mg) versus placebo [53]. Efficacy, as measured by percent change in PASI score, showed a bell-shaped response in which the 2 mg group had the greatest change in PASI score at weeks 4, 8, and 12. Adverse events were similar among the treatment and placebo groups. However, the results of this study are limited due to its small sample size [53]. Another phase 2/3 clinical trial randomized 293 patients with moderate-tosevere plaque psoriasis to either twice daily 2 mg CF101 versus placebo [54]. 8.5% of participants in the CF101 group achieved the primary endpoint (PASI75) at week 16 compared to 6.9% of placebo group. 31% of the CF101 group achieved the secondary endpoint (PASI50 or 75) at week 16 compared to 17% in the placebo group [54]. An ongoing phase 3 clinical trial will assess CF101 therapy in those with moderate-tosevere plaque psoriasis [55]. An estimated 407 ­participants will be randomized to twice daily regimens of CF101 2 mg, CF101 3 mg, or apremilast 30 mg versus placebo. The primary endpoint will be the proportion of subjects who achieve PASI75. This trial is currently recruiting participants [55]. No clinical trials testing this compound for the treatment of psoriatic arthritis have been reported. 23 Research Pipeline I: Oral Therapeutics for Psoriasis H4 Receptor Antagonists Histamine, as a ligand for the H1-H4 receptors, is classically released by mast cells in response to immunological and non-immunological stimuli. The localization of histamine receptor subtypes differs between tissues with H4 receptors predominately expressed on the surface of hematopoietic cells, including eosinophils, T cells, neutrophils, and other leukocytes (Fig. 23.1) [56]. Aside from its well-documented effect on eosinophils migration, H4 receptor antagonists also inhibit CD4+ T cell activation. While the primary T cell population affected by H4 receptor blockade are Th2 lymphocytes, modulation of the Th17 cell population with a subsequent decrease in the production of IL-17 production has also been noted [57–59]. This suggests that blockade of the H4 receptor in psoriasis may represent a novel therapeutic strategy in psoriatic disease. A phase 2 trial for ZPL389, a novel H4 receptor antagonist, involving 129 participants with moderate-to-severe plaque psoriasis was completed in December 2016 [60]. Participants were randomized to either ZPL389 (30 mg daily) or placebo, and the primary endpoint was change in PASI score at week 12. Results have not yet been published for this trial. No clinical trials testing this compound for the treatment of psoriatic arthritis have been reported. IL-20/PRINS Inhibitor Psoriasis-associated non-protein coding RNA induced by stress (PRINS) are cytosolic RNA found in non-lesional psoriatic skin and are purported to oppose chronic inflammation caused by cytosolic DNA fragments [61]. PRINS expression is modified by UVB irradiation, hypoxia, and microbial stimulation and interacts with the innate immune response of keratinocytes via IL-6 and CCL-5/RANTES [61]. Compounds that directly inhibit PRINS are attractive as potential novel therapies in inflammatory conditions like psoriasis due to their modulation of the inflammatory and immune response. 299 Abacavir hydroxyacetate, also known as Prurisol, is a chemical known to inhibit PRINS (Fig. 23.1). Two phase 2 trials investigating the efficacy of prurisol in chronic plaque psoriasis have been completed. One trial randomized 115 participants with mild-to-moderate psoriasis into four daily treatment arms: prurisol 50 mg, prurisol 100 mg, and prurisol 200 mg versus placebo [62]. The primary endpoint was percentage of participants with a ≥2-point improvement in IGA rating at day 84. Another trial randomized 199 participants with moderate-to-severe psoriasis to twice daily prurisol 150 or 200 mg versus placebo [63]. The primary endpoint was the proportion of participants achieving PASI75 at week 12. These studies were completed in April 2016 and June 2017, respectively, but results of these studies have not yet been published. No clinical trials testing this compound for the treatment of psoriatic arthritis have been reported. SYK Inhibitors Spleen tyrosine kinase (SYK) is a cytoplasmic tyrosine kinase (Fig. 23.1) mainly expressed in hematopoietic cells. It has crucial T cell, macrophage, neutrophil, and mast cell functions via the IgG and IgE receptors, as well as involvement in the TLR9-mediated B cell response [64]. It also demonstrates other diverse functions including cell adhesion, innate immune recognition, osteoclast maturation, and vascular remodeling [65]. Zap70 is a SYK tyrosine kinase essential to T cell development, the absence of which results in SCID [66]. SYK inhibition using fostamatinib (a prodrug of tamatinib) has been demonstrated as effective in rheumatoid arthritis as well as in murine models of psoriasis [67, 68]. Interestingly, tamatinib demonstrates inadequate SYK specificity compared to fostamatinib. The effect of fostamatinib is mediated through suppression of SYK expression in dendritic cells leading to downstream Th17 pathway downregulation [69]. SYK inhibition has been suggested as a potential therapeutic option for lupus, autoimmune ­thrombocytopenic 300 purpura, and B cell malignancies [70–72]. However, no clinical trials testing this compound for the treatment of plaque psoriasis or psoriatic arthritis have been reported. Clinical studies involving the treatment of inflammatory conditions with SYK inhibitors will be of interest to the field and those studying this class of molecules. Conclusion Despite the tremendous scientific and clinical advances witnessed in the field of psoriatic disease, our understanding of this complex disease remains incomplete. A noteworthy proportion of patients are inadequately treated, remain resistant to single or combination therapy, or experience loss of clinical response to previously effective therapies. This is particularly true for patients with psoriatic arthritis. Therefore, there is a continued need for new treatment modalities that overcome the limitations of first-generation and subsequent novel monoclonal antibodies. Several classes of small molecules currently in early and late phase clinical testing represent an exciting new frontier for the treatment of psoriatic disease. While promising, the clinical impact of these novel molecules remains undetermined and will depend on the results of ongoing, larger clinical trials and their implementation in realworld clinical scenarios. References 1. Hawkes JE, et al. Discovery of the IL-23/IL-17 signaling pathway and the treatment of psoriasis. J Immunol. 2018;201(6):1605–13. 2. Babon JJ, et al. The molecular regulation of Janus kinase (JAK) activation. Biochem J. 2014;462(1):1–13. 3. Welsch K, et al. Targeting JAK/STAT signalling in inflammatory skin diseases with small molecule inhibitors. Eur J Immunol. 2017;47(7):1096–107. 4. Igarashi K, et al. Interferon-gamma induces tyrosine phosphorylation of interferon-gamma receptor and regulated association of protein tyrosine kinases, Jak1 and Jak2, with its receptor. J Biol Chem. 1994;269(20):14333–6. 5. Zhou YJ, et al. Hierarchy of protein tyrosine kinases in interleukin-2 (IL-2) signaling: activation of syk D. Grand et al. depends on Jak3; however, neither Syk nor Lck is required for IL-2-mediated STAT activation. Mol Cell Biol. 2000;20(12):4371–80. 6. Damsky W, King BA. JAK inhibitors in dermatology: the promise of a new drug class. J Am Acad Dermatol. 2017;76(4):736–44. 7. Works MG, et al. Inhibition of TYK2 and JAK1 ameliorates imiquimod-induced psoriasis-like dermatitis by inhibiting IL-22 and the IL-23/IL-17 axis. J Immunol. 2014;193(7):3278–87. 8. Nicolas CS, et al. The role of JAK-STAT signaling within the CNS. JAKSTAT. 2013;2(1):e22925. 9. Notarangelo LD, et al. Mutations in severe combined immune deficiency (SCID) due to JAK3 deficiency. Hum Mutat. 2001;18(4):255–63. 10. Kreins AY, et al. Human TYK2 deficiency: mycobacterial and viral infections without hyper-IgE syndrome. J Exp Med. 2015;212(10):1641–62. 11. Sonbol MB, et al. Comprehensive review of JAK inhibitors in myeloproliferative neoplasms. Ther Adv Hematol. 2013;4(1):15–35. 12. Papp KA, et al. A randomized phase 2b trial of baricitinib, an oral Janus kinase (JAK) 1/JAK2 inhibitor, in patients with moderate-to-severe psoriasis. Br J Dermatol. 2016;174(6):1266–76. 13. A study comparing Upadacitinib (ABT-494) to placebo in participants with active psoriatic arthritis who have a history of inadequate response to at least one biologic disease modifying anti-rheumatic drug (SELECT—PsA 2) (NCT03104374). https://clinicaltrials.gov/ct2/show/NCT03104374. Updated April 30, 2019. Accessed June 25 2019. 14. A study comparing Upadacitinib (ABT-494) to placebo in participants with active psoriatic arthritis who have an inadequate response to at least one non-biologic disease modifying anti-rheumatic drug (SELECT—PsA 1) (NCT03104400). https://clinicaltrials.gov/ct2/show/NCT03104400. Updated June 28, 2019. Accessed June 25 2019. 15. Mease P, et al. Efficacy and safety of filgotinib, a selective Janus kinase 1 inhibitor, in patients with active psoriatic arthritis (EQUATOR): results from a randomised, placebo-controlled, phase 2 trial. Lancet. 2018;392(10162):2367–77. 16. An open-label, long-term extension study with filgotinib in active psoriatic arthritis (NCT03320876). https://clinicaltrials.gov/ct2/show/NCT03320876. Updated April 16, 2019. Accessed June 25 2019. 17. Papp K, et al. A phase 2a randomized, double-blind, placebo-controlled, sequential dose-escalation study to evaluate the efficacy and safety of ASP015K, a novel Janus kinase inhibitor, in patients with moderate-to-­ severe psoriasis. Br J Dermatol. 2015;173(3):767–76. 18. Papp K, et al. Phase 2 trial of selective tyrosine kinase 2 inhibition in psoriasis. N Engl J Med. 2018;379(14):1313–21. 19. An investigational study to evaluate experimental medication BMS-986165 compared to placebo and a currently available treatment in participants with moderate to severe plaque psoriasis (POETYK-­ Research Pipeline II: Upcoming Biologic Therapies 24 Ahuva D. Cices and Jeffrey M. Weinberg Abstract Psoriasis is a chronic inflammatory condition mediated by activated T-cells. Cutaneous manifestations of psoriasis have been shown to have a tremendous impact on quality of life, particularly in patients with moderate to severe disease. Given the high morbidity associated with psoriasis, disease control is paramount. Systemic treatments are indicated for effective management of cutaneous disease and the comorbidities that result from chronic inflammation. Prior systemic treatments were non-specific and associated with significant side effect profiles. With the development of biologics, treatments have become increasingly targeted and efficacious. This chapter will review biologic drugs currently in development that may eventually add to the growing armamentarium of psoriasis treatments. Key Learning Objectives 1. To understand the mechanism of action of novel biologic agents in development for the treatment of psoriasis A. D. Cices · J. M. Weinberg (*) Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA e-mail: jmw27@columbia.edu © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_24 2. To understand the appropriate use and efficacy of novel biologic agents in development for the treatment of psoriasis 3. To understand the safety issues and appropriate monitoring of novel biologic agents in development for the treatment of psoriasis Introduction Psoriasis is a chronic, inflammatory, multisystem disease mediated by activated T-cells with an estimated prevalence of 3.2% of the adult US population [1, 2]. Comorbidities of psoriasis include psoriatic arthritis, metabolic syndrome, cardiovascular disease, depression, and impaired quality of life. While mild to moderate disease can typically be controlled with topical medications or phototherapy, traditional treatments are often insufficient for moderate to severe disease. Topical treatments and phototherapy are limited by logistic issues related to application and do not target systemic disease manifestations. In patients with psoriatic arthritis, systemic treatment preventing irreversible joint destruction is critical. Prior to the advent of biologics, systemic therapies were non-specific immunosuppressive 303 304 agents with toxic side effects. Cyclosporine, methotrexate, and acitretin were the cornerstones of treatment until our understanding of the underlying aberrant pathways allowed for development of targeted therapies. Biologics are complex protein based molecules produced by living organisms that are utilized for their ability to target specific genes or proteins [3]. The relevant proteins in psoriasis are interleukins, cytokines involved in cell-to-cell signaling related to immune cell differentiation, or their receptors. Binding of biologics to these targets allows for specific modulation of the immune response. Overview of Pathogenesis Advances in our understanding of psoriasis at the molecular level have paved the way for the development of targeted and safe biologic therapies. Studying the complex interactions and signaling between T-cells, dendritic cells, and neutrophils with keratinocytes has been integral to the development of novel effective psoriasis therapies. Psoriatic disease is initiated by the interaction of self-nucleotides and innate antimicrobial peptides with toll-like receptors of plasmacytoid dendritic cells resulting in production of type I interferons, which then stimulate myeloid dendritic cells to secrete cytokines including tumor necrosis factor alpha (TNFα), interleukin 12 (IL-­ 12), and interleukin 23 (IL-23) [4, 5]. Dendritic cells bridge the innate and adaptive immune system by promoting T-helper 1-cell and T-helper 17-cell (Th17) differentiation through activation of innate pathways. TNFα, IL-23, and interleukin 17 (IL-17) promote inflammatory pathways and amplification of these responses. IL-23 stimulates hyperproliferation that is a hallmark of psoriasis by inducing differentiation and survival of Th17 cells, maintaining a favorable cytokine milieu. Clinical studies supporting the integral role of these inflammatory mediators include increased expression of IL-23 subunits p19 and A. D. Cices and J. M. Weinberg p40 in psoriatic lesions, and elevated levels of serum IL-17 and TNFα in patients with psoriasis [6, 7]. Granulocyte-macrophage colony-stimulating factor (GM-CSF) has been implicated in maintaining positive feedback loops involved in activated T-cell populations in patients with autoimmune disease, particularly related to Il-17A [8]. Programmed cell death- protein 1 (PD-1) or CD279 is an immune checkpoint that promotes self-tolerance and has been hypothesized to play a role in autoimmune disease including psoriasis as demonstrated in a study showing decreased levels of PD-1 expression on T-cells of psoriasis patients compared with healthy controls as well as case series describing psoriasis flares in patients treated with PD-1 inhibitors [9, 10]. Overview of Biologic Treatments TNFα antagonists are the first generation of biologics widely used in the treatment of psoriasis. While anti-TNFα therapies are effective treatments for psoriasis, they are not without risk. Due to the broad activity of TNFα throughout the immune system, TNFα inhibitors have significant side effects including increased risk for TB reactivation, infections, and certain malignancies [4]. Meta analysis of rheumatoid arthritis patients treated with anti-TNFα therapies showed increased risk of serious infections and a dose-­ depended increase in malignancy; however, meta analysis of psoriasis patients treated with anti-­ TNFα therapies showed a small increased risk of infection without increased risk of serious infections or malignancy, suggesting that some of the risks associated with TNFα antagonists may be specific to rheumatoid arthritis populations [11, 12]. Second generation biologics for psoriasis that target IL-17 and IL-23 have improved efficacy and safety profiles with increased risk of some non-serious infections, but importantly without increased risk of malignancy or serious infections [1]. 24 Research Pipeline II: Upcoming Biologic Therapies Clinical Trial Background Clinical trials consist of organized, prospective, exposures of patients to interventions [13]. In order for a new drug to be marketed in the United States, the U.S. Food and Drug Administration (FDA) must approve the drug after careful review of pre-clinical and clinical tests, including Phase I-III studies. Phase I studies are initial human studies designed to document initial safety data. In these studies, investigational drug is given to a small number of people, typically 20–80 patients or healthy volunteers, to evaluate preliminary safety and dosing. Phase II studies evaluate the effectiveness of a drug for a specific medical indication. Typically these studies consist of several hundred patients and look at the effect of treatment on clinical outcomes as well as side effects. Ideally, phase II studies are double blind and placebo controlled. Phase III studies are conducted for experimental drugs with evidence of efficacy in previously completed phase II trials in order to gather additional evidence of efficacy and safety. These are randomized clinical trials conducted in large populations (often >1000 patients) to further evaluate efficacy and safety. Typically, at least two successful phase III clinical trials demonstrating adequate efficacy and safety are required by the FDA prior to approval. Standardized measurements are utilized in clinical trials to allow for consistent comparison of data across studies. The Psoriasis Area and Severity Index (PASI) serves as the gold standard for measuring disease activity. PASI score takes into account the affected body surface area (BSA) as well as severity of lesions. Lesions are assessed separately over four body regions: the head and neck, upper extremities, trunk, and lower extremities. Average intensity of erythema, thickness, and scale is graded in each of the four locations from 0 (none) to 4 (very severe). Within each region, the severity sum is multiplied by an area score based on the affected BSA and adjusted by 305 a fixed variable correlated to the BSA of the representative region. The sum of these scores from each region yields the PASI score, which can range from 0 (no active disease) to 72. Clinical trials for biologics tested in moderate to severe populations typically include inclusion criteria of BSA ≥10% and PASI ≥12. In clinical trials percent of improvement in PASI score is used to assess efficacy, with many of the newer drugs achieving high rates of PASI 90 (90% improvement in PASI score at a given time point from baseline) and even PASI 100 (no residual disease). Physician’s global assessment (PGA) or investigator’s global assessment (IGA) is a useful tool for measuring cutaneous psoriatic disease activity in clinical trials. It allows for a quick, but subjective method of stratification based on overall severity without taking into account the affected BSA. There are multiple versions of the scale which utilize variations in the criteria for each score [14]. The IGA mod 2011 scale is a 5 point scale commonly used in psoriasis clinical trials, the scores are designated as follows: 0 (clear), 1 (almost clear), 2 (mild), 3 (moderate), and 4 (severe). Inclusion criteria for psoriasis studies evaluating patients with moderate to severe psoriasis require a PGA/IGA score of 3 or 4 and treatment success is defined by the FDA as a PGA/IGA score of 0 or 1 with at least 2 grade improvement from baseline. American College of Rheumatology (ACR) score is an instrument specifically developed for rheumatoid arthritis and extended for use in psoriatic arthritis, which is utilized in rheumatologic clinical trials as a measure of disease activity in inflammatory arthritis. ACR measurement incorporates number of tender or swollen joints, physician global assessment of disease severity, patient global assessment of disease severity, patient reported functional ability, patient reported visual analogue pain scale, and acute phase reactants, namely erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP). Improvement is measured by the percent 306 improvement in number of tender or swollen joints as well as at least 3 out of 5 of the above criteria. Previous biologics were considered successful with achievement of ACR 20, while many newer medications are evaluating treatment success with ACR 50. A. D. Cices and J. M. Weinberg cebo dosed every 4 weeks [17]. Primary endpoint of PASI 90 at week 12 was achieved in significantly more patients in all bimekizumab treated groups (46.2–79.1%) compared with placebo (0%) (p < 0.0001) [17]. Also seen across all doses, were significant improvements (p ≤ 0.0003) in all secondary endpoints compared to placebo, which included PASI 90 at week 8, PASI 75 at week 12, Psoriasis Biologics Pipeline PASI 100 at week 12, and IGA 0/1 at weeks 8 and 12. Three serious TEAEs were reported in 2 IL-17 patients, however they were not considered related to study treatment (viral meningitis in a patient on Bimekizumab/UCB4940 is a humanized mono- placebo and a large intestinal polyp/colon cancer clonal IgG1 antibody targeting IL-17A and in a subject 15 days after initiating treatment with IL-17F being studied as a treatment for chronic bimekizumab 480 mg). There were no unexpected plaque psoriasis, psoriatic arthritis, ankylosing safety signals: common TEAEs occurring in >5% spondylitis, non-radiographic axial spondyloar- of bimekizumab treated subjects included nasothritis, hidradenitis suppurativa, and rheumatoid pharyngitis, upper respiratory infections, arthralarthritis. A phase I placebo-controlled, dose-­ gia, elevated GGT, tonsillitis, oral candidiasis, escalating study (NCT02529956) in 39 subjects headache, leukopenia, vomiting, and neutropenia. with mild to moderate psoriasis showed efficacy No subjects in the placebo group had neutropenia, of bimekizumab compared to placebo in treat- while five subjects receiving bimekizumab had ment of mild to moderate psoriasis with dose-­ transient, non-serious neutropenia that resolved dependent improvements in PASI and PGA spontaneously and did not interrupt treatment. following a single dose of intravenous bimeki- Maintenance of response through week 60 was zumab at doses ranging from 8 to 640 mg [15]. seen in the phase II extension study BE ABLE 2 Statistically significant clinical improvement was (NCT03010527) with PASI 90 in 80–100% and seen as early as week 2, with maintenance of near PASI 100 in 70–83% of study subjects [18]. No new maximal response through weeks 12–20 (depend- safety signals were identified, and consistent with ing on endpoint) following a single IV dose previous data oral candidiasis and nasopharyngi≥160 mg. No unexpected safety signals and simi- tis were the most commonly occurring treatment lar numbers of treatment emergent adverse events emergent adverse events. The phase II psoriatic (TEAE) were observed in bimekizumab and pla- arthritis BE ACTIVE study (NCT02969525) rancebo treated groups. PA007, a phase I placebo-­ domized 206 adults with active psoriatic arthricontrolled study (NCT02141763) in 53 subjects tis 1:1:1:1:1 to one of four bimekizumab doses with psoriatic arthritis, that compared five dos- (16 mg, 160 mg, 160 mg with 320 mg loading, or ages of bimekizumab (240 mg/160 mg/160 mg; 320 mg) or placebo administered every 4 weeks. 80 mg/40 mg/40 mg; 160 mg/80 mg/80 mg and After week 12, subjects administered placebo and 560 mg/320 mg/320 mg) administered at weeks 0, the lowest bimekizumab dose were re-randomized 3 and 6 demonstrated efficacy in treatment of and maintained through week 48. The primary arthritis and skin lesions with an ACR 20 in 80% outcome of ACR 50 at week 12 was achieved in and PASI 100 in 86.7% of subjects in the top 3 significantly more subjects treated with 16 mg doses at week 8 compared to 16.7 and 0% respec- bimekizumab (OR 4.2, p = 0.032), 160 mg bimetively in placebo treated subjects [16]. kizumab (OR 8.1, p = 0.012), and 160 mg bimeIn the 12 week, phase II BE ABLE 1 trial kizumab with loading dose (OR 9.7, p = 0.0004) (NCT02905006) subjects were randomized compared to placebo. 1:1:1:1:1:1 to 64 mg, 160 mg, 160 mg with Multiple phase III studies for bimekizumab in 320 mg loading dose, 320 mg, 480 mg, or pla- the treatment of moderate to severe chronic 24 Research Pipeline II: Upcoming Biologic Therapies 307 plaque psoriasis are underway and have shown promising preliminary results with all three studies achieving the co-primary endpoints of PASI 90 and IGA 0/1 with at least two-grade improvement and with a safety prolife similar to that demonstrated in the BE ABLE studies [19]. The BE VIVID 52 week study (NCT03370133) randomized 570 subjects to bimekizumab, low dose ustekinumab if weight ≤100 kg, high dose ustekinumab if weight >100 kg, or placebo, with subjects randomized to placebo crossed over to bimekizumab after week 16. Bimekizumab was superior to ustekinumab and placebo in achieving PASI 90, PASI 100, IGA 0, and IGA 1. The 56-week BE READY study (NCT03410992) randomized 435 subjects to bimekizumab or placebo for an initial 16-week treatment period followed by a randomized withdrawal period through week 56. All ranked secondary endpoints were met notably PASI 100 at week 16, patient reported itch, pain and scaling, and scalp IGA 0/1 superior to placebo. The BE SURE 56-week study (NCT03412747) randomized 480 subjects to one of two bimekizumab regimens or adalimumab for 24 weeks, followed by a dose-blind maintenance through week 56. All ranked secondary endpoints were achieved, including statistically significant superior PASI 75 at week 4 and PASI 100 at weeks 16 and 24 compared to adalimumab. Given the positive results from these phase III studies, plans are underway for bimekizumab to be submitted for FDA approval in mid-2020. Additional continuing studies evaluating bimekizumab for the treatment of moderate to severe psoriasis include the following: BE RADIANT (NCT03536884), comparing two bimekizumab doses and secukinumab with a primary endpoint of PASI 100 at week 16, a phase III long-term extension study (NCT03598790) comparing two bimekizumab doses through week 160, and a phase III study (NCT03766685) comparing bimekizumab administered by auto injector or syringe. Ongoing studies for bimekizumab in the treatment of psoriatic arthritis include the phase II long term extension study BE ACTIVE 2 (NCT03347110), a phase III, placebo-­controlled study BE VITAL (NCT03896581), and another phase III study (NCT03895203) comparing bimekizumab to adalimumab with placebo control though week 16. Netakimab/BCD-085 is a humanized monoclonal antibody to IL-17 developed and approved in Russia for treatment of moderate to severe plaque psoriasis based on favorable results in the phase II BCD-085-2 study (NCT02762994) with long-term extension and phase III BCD-085-7/ PLANETA study (NCT03390101) conducted in Russia. Results have not been published, but a press release from the company show PASI 75/90/100 in 83%/92%/59% of subjects at week 12 respectively [20, 21]. Netakimab is also being evaluated for the treatment of ankylosing spondylitis, psoriatic arthritis, and primary biliary cholangitis. Vunakizumab/SHR-1314 is a humanized IL-17A inhibitor under development for psoriasis and axial spondyloarthritis. Preliminary safety was established in healthy adults in a phase I study (NCT02934412) with doses ranging from 20 to 240 mg. A second phase I study (NCT03710681) is underway, comparing 160 and 240 mg administered every 2 weeks for 168 days in subjects with moderate to severe plaque psoriasis. A multiple dose, escalating design, combined phase I and phase II study (NCT03463187) is ongoing for moderate to severe plaque psoriasis and an upcoming phase II study (NCT04121143) will compare low dose short interval, high dose long interval, high dose short interval, and placebo for 16 weeks in subjects with moderate to severe psoriasis. M1095/ALX-0761/MSB0010841 is a trivalent monomeric nanobody specific for IL-17A, IL-17F, and human serum albumin allowing for increased circulation time in the bloodstream [22]. Data from a recently completed phase I placebo-controlled, dose escalation 6 week study (NCT02156466) showed promising results [22]. 44 subjects with moderate to severe psoriasis were randomized 4:1 to 30 mg, 60 mg, 120 mg or 240 mg M1095 or placebo administered subcutaneously bi-weekly. A dose response relationship was observed. In the 240 mg cohort, all subjects achieved PASI 90 and 56% of subjects achieved PASI 100 at day 85. The most common TEAEs were pruritus and headaches. 2 patients withdrew A. D. Cices and J. M. Weinberg 308 due to adverse events, one due to an injection site reaction and another due to transaminitis. An upcoming placebo-controlled, active comparator (secukinumab) phase II study (NCT03384745) will compare 30 mg every 4 weeks, 60 mg every 4 weeks, 120 mg every 8 weeks, and 120 mg every 4 weeks subsequent to a loading dose. ABY-035/AFO2 is a novel multivalent and specific molecule targeting both IL-17A subunits as well as albumin. Favorable safety and tolerability was established across multiple doses and dosing regimens in healthy volunteers and psoriasis subjects in an initial phase I/II study (NCT02690142) [23]. A two cohort phase I trial (NCT03580278) evaluating 75 mg daily for 14 days or 150 mg daily for up to 28 days in psoriasis subjects is ongoing. Encouraging interim results have been reported in the AFFIRM-35 phase II trial (NCT03591887), in which subjects with moderate to severe psoriasis were randomized 1:1:1:1:1 to 2 mg, 20 mg, 80 mg, 160 mg or placebo administered subcutaneously every 2 weeks for 12 weeks followed by dose adjustments and re-randomization after week 12 [23]. IL-12/23 Promising results have been shown with mirikizumab, a humanized IgG4 monoclonal antibody to the p19 subunit of IL-23, which is being tested for treatment of moderate to severe psoriasis as well as inflammatory bowel disease. In AMAF, a placebo controlled phase II trial (NCT02899988) evaluating single doses of subcutaneous mirikizumab in subjects with moderate to severe psoriasis at weeks 0 and 8, the percentage of patients meeting the primary objective of PASI 90 at week 16 compared to placebo was clinically significant in all tested doses with rates of 0, 29, 59 and 67% for subjects treated with placebo, 30, 100, and 300 mg respectively [24]. Risk of adverse events was not elevated in subjects treated with mirikizumab compared to placebo. Pivotal phase III studies OASIS-1/AMAK (NCT03482011) and OASIS-2/AMAJ (NCT03535194), which compared mirikizumab to placebo and secukinumab or placebo respectively, have been completed, with results expected in the near future. OASIS-3 (NCT03556202) is an open label extension study currently underway comparing two different mirikizumab doses for 208 weeks. Miscellaneous CC-90006 is a PD-1 agonist antibody in the initial stages of development. A phase I study (NCT03337022) for treatment of mild to moderate plaque psoriasis was recently completed. The study consists of 40 subjects divided into 4 cohorts of 10 patients with escalating doses administered at weeks 0, 2, and 4. Data has not yet been released, but continued development is underway [25]. Namilumab/MT203/AMG203 is an IgG1 kappa monoclonal antibody targeting GM-CSF being pursued as a novel treatment for psoriasis, rheumatoid arthritis, and axial spondyloarthritis. Safety was established in a phase I, placebo controlled study (NCT02354599) in healthy Japanese and Caucasian adult men. The NEPTUNE phase II placebo controlled, proof of concept study (NCT02129777) randomized 122 subjects with moderate to severe psoriasis to 20, 50, 80, or 150 mg subcutaneous namilumab [26]. The number of subjects in all groups achieving the primary endpoint of PASI 75 at week 12 was low in all cohorts, with no clinical significance between groups and similar results for other assessments. Given the results of the phase II study, which suggest that namilumab inhibition does not inhibit pathogenic inflammatory processes of psoriasis Namilumab is no longer being studied in psoriasis. Summary Psoriasis is a multi-system disorder that is not skin-limited and often has significant effects on patient quality of life. Recent advances in immunology have vastly expanded our comprehension of psoriasis. Identifying key mediators of cell signaling pathways associated with pathogenic subsets of T-cells paved the way for the development of novel therapies, namely biologics. These 24 Research Pipeline II: Upcoming Biologic Therapies 309 medications are remarkably effective and safe due to their specificity. New therapeutic targets will continue to emerge as our understanding of psoriasis continues to increase, and may include innate immune pathways or genetic predispositions. The development of novel psoriasis treatments will continue to produce highly effective and safe medications. infections and malignancies: systematic review and meta-analysis of rare harmful effects in randomized controlled trials. JAMA. 2006;295(19):2275–85. 12. The safety of tumor necrosis factor antagonists in patients with psoriatic disease: a systematic review and metaanalysis of randomized controlled trials. J Am Acad Dermatol. 2011;64(2):AB8. 13. Junod SW. FDA and clinical drug trials: a short history FDA. Available from https://www.fda.gov/ media/110437/download. 14. Langley RG, Feldman SR, Nyirady J, van de Kerkhof P, Papavassilis C. The 5-point Investigator’s Global Assessment (IGA) Scale: a modified tool for evaluating plaque psoriasis severity in clinical trials. J Dermatolog Treat. 2015;26(1):23–31. 15. Glatt S, Helmer E, Haier B, Strimenopoulou F, Price G, Vajjah P, et al. First-in-human randomized study of bimekizumab, a humanized monoclonal antibody and selective dual inhibitor of IL-17A and IL-17F, in mild psoriasis. Br J Clin Pharmacol. 2017;83(5):991–1001. 16. Glatt S, Baeten D, Baker T, Griffiths M, Ionescu L, Lawson ADG, et al. Dual IL-17A and IL-17F neutralisation by bimekizumab in psoriatic arthritis: evidence from preclinical experiments and a randomised placebo-controlled clinical trial that IL-17F contributes to human chronic tissue inflammation. Ann Rheum Dis. 2018;77(4):523–32. 17. Papp KA, Merola JF, Gottlieb AB, Griffiths CEM, Cross N, Peterson L, et al. Dual neutralization of both interleukin 17A and interleukin 17F with bimekizumab in patients with psoriasis: results from BE ABLE 1, a 12-week randomized, double-blinded, placebo-­ controlled phase 2b trial. J Am Acad Dermatol. 2018;79(2):277–86.e10. 18. Blauvelt A, Papp KA, Merola JF, Gottlieb AB, Cross N, Madden C, et al. AB0738 dual neutralisation of interleukin (IL)-17A and IL-17F with bimekizumab in moderate-to-severe plaque psoriasis: 60-week results from a randomised, double-blinded, phase 2B extension study. Ann Rheum Dis. 2019;78(Suppl 2):1834. 19. Available from: https://www.ucb.com/stories-media/ Press-Releases/article/Bimekizumab-PositiveResults-Confirmed-in-Second-Phase-3-PsoriasisStudy. 20. BIOCAD registered the first Russian original therapeutic monoclonal antibody 2020. Available from: https://biocadglobal.com/post/Russian_original_ therapeutic_monoclonal_antibody. 21. Bakulev AS, Kubanov AA, Khairutdinov V, Kokhan M, Artemyeva A, Derbin S, Chernyaeva E, Ivanov R. Long-term efficacy and safety of netakimab in patients with moderate-to-severe psoriasis. Results of phase II open-label extension clinical study BCD-085-­ 2-ext. Vestn Dermatol Venerol. 2019;95(3):54–64. 22. Svecova D, Lubell MW, Casset-Semanaz F, Mackenzie H, Grenningloh R, Krueger JG. A randomized, double-blind, placebo-controlled phase 1 study of multiple ascending doses of subcutaneous M1095, an anti-interleukin 17A/F nanobody, in References 1. Menter A, Strober BE, Kaplan DH, Kivelevitch D, Prater EF, Stoff B, et al. Joint AAD-NPF guidelines of care for the management and treatment of psoriasis with biologics. J Am Acad Dermatol. 2019;80(4):1029–72. 2. Armstrong AW, Siegel MP, Bagel J, Boh EE, Buell M, Cooper KD, et al. From the Medical Board of the National Psoriasis Foundation: treatment targets for plaque psoriasis. J Am Acad Dermatol. 2017;76(2):290–8. 3. Morrow T, Felcone LH. Defining the difference: what makes biologics unique. Biotechnol Healthc. 2004;1(4):24–9. 4. Greb JE, Goldminz AM, Elder JT, Lebwohl MG, Gladman DD, Wu JJ, et al. Psoriasis. Nat Rev Dis Primers. 2016;2:16082. 5. Georgescu SR, Tampa M, Caruntu C, Sarbu MI, Mitran CI, Mitran MI, et al. Advances in understanding the immunological pathways in psoriasis. Int J Mol Sci. 2019;20(3):739. 6. Lee E, Trepicchio WL, Oestreicher JL, Pittman D, Wang F, Chamian F, et al. Increased expression of interleukin 23 p19 and p40 in lesional skin of patients with psoriasis vulgaris. J Exp Med. 2004;199(1):125–30. 7. Arican O, Aral M, Sasmaz S, Ciragil P. Serum levels of TNF-alpha, IFN-gamma, IL-6, IL-8, IL-12, IL-17, and IL-18 in patients with active psoriasis and correlation with disease severity. Mediat Inflamm. 2005;2005(5):273–9. 8. Hamilton JA. GM-CSF as a target in inflammatory/ autoimmune disease: current evidence and future therapeutic potential. Expert Rev Clin Immunol. 2015;11(4):457–65. 9. 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(2):190–7. 10. Shin D, Kim DS, Kim SH, Je JH, Kim HJ, Young Kim D, et al. Decreased PD-1 positive blood follicular helper T cells in patients with psoriasis. Arch Dermatol Res. 2016;308(8):593–9. 11. Bongartz T, Sutton AJ, Sweeting MJ, Buchan I, Matteson EL, Montori V. Anti-TNF antibody therapy in rheumatoid arthritis and the risk of serious Pediatric Psoriasis 25 Starling Tolliver, Amber N. Pepper, Salma Pothiawala, and Nanette B. Silverberg Abstract Psoriasis is a chronic multi-system inflammatory condition with an autoimmune component. One-third to one-half of cases have onset in childhood. There are a variety of pediatric variant types of psoriasis, including generalized disease, that is similar to adult psoriasis, and types distinctive to childhood, including diaper involvement and pityriasis amiantacea. Pediatric psoriasis is associated with infectious triggers and exacerbation by the Koebner phenomenon. Children with psoriasis may have more of a tendency toward obesity as reflected by a large waist circumference. More extensive disease in childhood is associated with a poor quality of life, anxiety, and depression. Therapeutics of pediatric psoriasis gen- erally requires a global approach including identifying infectious triggers, addressing health risks including obesity, and psychological support. Prescription care includes application of mid-potency topical corticosteroids and/or calcipotriene, phototherapy with Narrowband UVB or excimer laser. In severe cases, cyclic prescribing of systemic agents for 6–12 months including methotrexate, cyclosporine, etanercept, adalimumab, and recently approved agents ixekizumab and ustekinumab can aid in disease clearance with minimization of side effects. Emerging studies for the efficacious and safe use of biologics in children broaden treatment options for managing severe psoriatic disease early. Key Learning Objectives S. Tolliver · N. B. Silverberg (*) Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA e-mail: starling.tolliver@osumc.edu; nanette.silverberg@mountsinai.org A. N. Pepper Department of Internal Medicine, USF Health Morsani College of Medicine, Tampa, FL, USA e-mail: apepper1@health.usf.edu 1. To understand the clinical presentation of psoriasis in the pediatric population 2. To understand the therapeutic management of psoriasis in the pediatric population 3. To understand the specific safety concerns in the management of psoriasis in the pediatric population S. Pothiawala Department of Dermatology and Cutaneous Surgery, University of South Florida, Tampa, FL, USA e-mail: salma.pothiawala@post.harvard.edu © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_25 311 312 Introduction Psoriasis is a chronic inflammatory disorder, believed to have an autoimmune basis. It is characterized by aberrant T cell activity causing hyperproliferation of epidermal keratinocytes, with development of erythematous skin plaques covered by a silvery or micaceous scale [1, 2]. Pediatric psoriasis differs from adult onset psoriasis in types of environmental triggers, with trauma, stress, and bacterial infection being the most common pediatric disease triggers [1, 3], while drug-reactions, smoking, alcohol use, and underlying HIV infection are triggers more common in adulthood [4]. Recently, obesity and the metabolic syndrome have been linked to both pediatric and adult psoriasis [5]. Treatment options are similar for adult and pediatric psoriasis patients, although therapy selection for pediatric psoriasis varies according to patient age and is limited by the fact that systemic treatments for pediatric psoriasis are off-label. In the United States, high-potency topical corticosteroids were the most frequently prescribed outpatient treatment for pediatric patients overall from 1979 to 2007 [6]. Few therapies for psoriasis are approved by the FDA for the pediatric age group, and drug dosages must be altered based on weight and/or age in children. However, with emerging data on the safety and efficacy of these therapies for pediatric patients, this is changing. This chapter will explore the epidemiology, pathogenesis, diagnosis, and therapeutic management options of pediatric psoriasis, with a special focus on aspects of the disease specific to the pediatric patient. Epidemiology (Table 25.1) Psoriasis vulgaris is a common dermatologic disorder, affecting 3.15% of the United States population and 1.5% of the population in the United Kingdom [14, 15]. Onset occurs before 16 years of age in approximately one third of all cases [7, 16], making psoriasis a significant dermatologi- S. Tolliver et al. cal problem in the pediatric population. These data likely represent under reported incidences secondary to potential disparities in access to care, mild undertreated disease, misdiagnoses, and racial disparities in patient reporting [17]. The peak age of onset varies in the literature, but usually ranges anywhere between 2 and 11 years of age [1, 3, 8, 9]. The mean age at diagnosis has been reported to be between 10 and 11 years of age by a recent large populationbased study [10]. Onset may occur slightly earlier for females [18], although studies have shown nearly equal male to female prevalence ratios [8, 9, 11]. Onset may also occur earlier in those with a positive family history of psoriasis [18]. Table 25.1 shows a review of worldwide presentation of pediatric psoriasis. The annual incidence of psoriasis has been shown to be increasing, nearly doubling (from 29.6 cases per 100,000 to 62.7 cases per 100,000) from 1970 to 2000 in both pediatric and adult patients [10, 19]. Recent studies in the United States have reported a prevalence of mild and moderate-to-severe pediatric psoriasis to be 128 per 100,000 and 16 per 100,000 respectively [20]. Plaque psoriasis (Figs. 25.1 and 25.2) is the most common variant, occurring in 34–74% of children, with the majority of lesions localized to the extensor surfaces of elbows and knees, scalp, face (Fig. 25.3), trunk, and posterior auricular region (Table 25.1) [8–11, 22]. Guttate psoriasis is more common in pediatric patients, occurring in approximately 6–33% of cases of pediatric psoriasis (Fig. 25.4) [1, 18]. Pustular disease, is a relatively uncommon presentation in children, however, a Turkish group reported 13% of their pediatric patients had pustular lesions [1, 8]. A single study from Greece sited 8% of children as having erythroderma [24] however, most studies demonstrate that <1% of children present this way [3, 8, 16, 18]. Glossitis and mucosal diseases are uncommon in childhood [1, 3, 8, 16, 18]. In infants and toddlers (less than 2 years old), psoriatic diaper rash is the most common presentation sometimes involving the intertriginous 223 India USA China Kumar et al. 2004 [3] Raychaudhuri and Gross 2000 [7] Fan 2007 [8] 277 419 Country Turkey Author, year Seyhan et al. 2006 [1] Number of kids 61 1:1.1 47; 53 1:1.3 44; 56 1.1:1 52; 48 Sex distribution (M:F and %M; %F) 1:1.7 38; 62 Table 25.1 Worldwide demographics of pediatric psoriasis Not discussed (Chinese) Not discussed Not discussed (Indian) Race and or ethnicity Not discussed (Turkish) Median age of onset 10 Range: 9 months to 15 years Not discussed Onset mean 9.1 overall (M) 8.1 ± 2.1 (F) 9.3 ± 2.3 Range 4–14 Ages of kids (range, median, mean) Onset mean 6.9 ± 4.1 At time of study 10.0 ± 4.1 Plaque 69% Guttate 29% Pustular 1% Erythroderma 1% Not discussed Plaque 61% Guttate 10% Pustular <1% Inverse <1% Erythrodermic 1% Palmoplantar 6% Plantar 13% Nails 3% Types of psoriasis (%) Plaque 54% Guttate 33% Pustular 13% Inverse 7% Erythrodermic 3% Palmoplantar 2% (At onset) Scalp 57% Trunk 14% Extremities 52% Face 9% Nails 13% (At onset) Scalp 47% Face 20% Trunk 43% Arms 51% Legs 66% Nail 6% Palmoplantar 37% Areas of involvement (At onset) Trunk 44% Scalp 36% Extremities 54% Diaper rash 5% Nails 21% (At onset) Trunk 8% Extremities 35% Scalp 21% Soles 19% Palms 3% Not discussed 4.5% Only recalled by 7% of participants Trauma 14 pts Throat infection ten patients Psych 3 pts Drug rxn 1 pt Trauma 50% Stress 50% Pharyngitis 28% (continued) 8% (7 1st, 1 2nd) 68% Family history 23% Triggers URTI 15% +Group B strep culture 21% 25 Pediatric Psoriasis 313 Country Korea USA Australia Author, year Kwon 2011 [9] Tollefson 2010 [10] Morris 2001 [11] Table 25.1 (continued) 1262 357 Number of kids 358 1:1.1 47;53 1:1.1 48;52 Sex distribution (M:F and %M; %F) 1.1:1 51; 49 Not discussed Mostly Caucasian but not specified Race and or ethnicity Not discussed No median or mean described Range: 1 month to 15 years Median age of onset 10.6 Range 6.8–14.4 Ages of kids (range, median, mean) Mean age onset: 10.5 ± 4.3 Plaque 34% Scalp 12% Diaper rash 13% Guttate 6% Face only 4% Pustular <1% Nail <1% Erythrodermic <1% Types of psoriasis (%) Plaque 67% Guttate 18% Generalized pustular pustulosis 7% Palmoplantar 4% Erythroderma 1% Plaque 74% Guttate 14% Sebo-psoriasis 8% Pustular 1% Areas of involvement Trunk 70% Legs 65% Arms 48% Face 46% Palms/soles 22% Scalp 47% Extremities 60% Palms/soles 5% Trunk 35% Face 18% Genital/groin 9% Nails 17% Face 38% Otherwise not discussed Family history 32% Not discussed 71% (based on 61% who responded) Triggers Not discussed Not discussed Not discussed 314 S. Tolliver et al. USA USA Greece Vogel 2012 [6] Wu 2011 [12] Stefanaki 2011 [13] 125 1361 3.8 million outpt VISITS for ped psoriasis 1.4:1 59; 41 1:1.2 45; 55 1:1 50;50 Race White 93% Black 3% Asian/ Pacific Islander 3% American Indian/ Eskimo/ Aleut 0.5% Ethnicity 85% non-­ Hispanic 8% Hispanic 7% other Non-Hispanic white 38% Hispanic white 38% Black 3% Asian or Pacific Islander 7% Others 3% 81% Greek 16% Not discussed Extremities 56% Scalp 48% Trunk 47% Nails 10% Plaque 57% Scalp? 34% Guttate 12% Flexural 10% Erythrodermic 8% Nails only 5% Peak age of onset 9–10 grp Range: <1–13 years No mean or median given Not discussed Not discussed Not discussed Not discussed Not discussed 11.8 ± 4.4 in M 12.5 ± 4.4 in F Not discussed Not discussed Not discussed (NB: article discusses outpatient visits and treatment choices, not individual patients) Mean age at visit 11.3 25 Pediatric Psoriasis 315 316 S. Tolliver et al. Fig. 25.3 Facial psoriasis can be a challenging condition due to the combination of disfigurement, psychological distress and limitations of therapy on the thin-skinned face Fig. 25.1 Untreated plaque psoriasis affecting the trunk with large circinate plaques with overlying micaceous scale (Reprinted with permission from Silverberg [21]) Fig. 25.2 Close-up of plaque with micaceous scale demonstrates areas of Auspitz sign, pinpoint bleeding due to manual removal of scale neck and axillary areas (Fig. 25.5) [11, 22]. Psoriasis presenting in infancy has a better prognosis for long-term remission than childhood psoriasis [24]. Involvement of the folds, i.e. inverse psoriasis, is an uncommon appearance in older children. Inverse psoriasis in any age group can be exacerbated by concurrent cutaneous overgrowth of streptococcal, staphylococcal and candidal species. Fig. 25.4 Guttate psoriasis of the trunk in a child with psoriasis flared by an upper respiratory infection (Taken from Silverberg [23] with permission) Nail psoriasis can arise in the setting of plaque-type psoriasis, psoriatic arthritis, or with 25 Pediatric Psoriasis Fig. 25.5 Inverse psoriasis in an infant (Taken from Silverberg [23] with permission) isolated nail disease. This manifestation can be found in as many 16–32% of pediatric patients [25, 26]. It is associated with male sex and higher disease burden at onset as well as follow-up [25, 26]. Nail disease may also suggest a more morbid disease course for patients [25]. Common features include nail pitting on the fingernails while onycholysis and pachyonychia were found on the toenails [25]. Psoriatic arthritis with active joint involvement is less common in childhood than adulthood, but should remain in the differential for children with arthritis [3]. In the majority of children, dermatologic manifestations precede the onset of psoriatic arthritis [27]. There is a bimodal age of psoriatic arthritis onset with peaks during the first few years of life and in early adolescence [28]. Skin manifestations are often established before joint involvement [22]. The early onset form of psoriatic arthritis is most likely to be polyarticular, ANA (antinuclear antibody) positive, and favors the female sex [29, 30]. The adolescent-onset form of psoriatic arthritis has a higher incidence of axial joint involvement and favors the male sex. The proximal and distal interphalangeal joints of the hand, knees, and ankles are most commonly involved initially. Children often present with oligoarthritis, which may progress to polyarthritis; dactylitis can be associated. Psoriatic arthritis can be differentiated from idiopathic juvenile rheumatoid arthritis by the presence of blue discoloration of the joints, nail pitting and/ or nail dystrophy [27]. Predominant involvement of the wrists and small joints of the hands and feet also 317 point toward a psoriatic etiology of arthritis in affected children [30]. As many as 25% of patients with psoriatic arthritis have joint damage by 5 years after symptoms begin, thus early screening and treatment is important [31]. Pediatric psoriasis has been shown to negatively affect quality of life. The psychological impact of skin disease is becoming an important area of research with some studies reporting prevalence rates as much as 70% [32]. Studies have shown that cutaneous diseases such as, atopic dermatitis, psoriasis and urticaria have a direct impact on psychological well-being [33]. Pediatric patients with psoriasis are increasingly being found with higher rates of depression, anxiety, and even bipolar disorder in some studies [34]. One study showed a 9- and 6-fold risk for anxiety and depression respectively verses healthy controls [32]. The psychological stress caused by psoriasis may even be significantly higher than other chronic cutaneous diseases. Multiple studies have determined depression to be higher in patients with psoriasis compared to atopic dermatitis [33, 35]. This may be more pronounced in younger children who have less developed coping mechanisms [36]. Mounting pressures within their social circles and decreased levels of maturity may increase the risk of lower quality of life [32]. Children with moderate-tosevere psoriasis have similar health-related quality of life as compared to children with arthritis or asthma, but worse than children with diabetes [37]. Adult females with psoriatic arthritis suffer greater disability than their male counterparts, but no gender differences have been reported in children thus far [32, 38]. Impairment in quality of life is also described in families with children who have psoriasis [39]. This phenomenom is likely multifactorial with emotional well-being and burden of care being the factors with the most impact [39, 40]. There is current debate on the role psychiatric disorders play in the development of psoriasis. Some studies have shown distress as an important trigger in the onset as well as exacerbating factor in leading to psoriatic lesions [32, 41]. One paper described an association between other autoimmune diseases and schizophrenia suggest- 318 ing potential shared genetic risk factors in the development of both diseases [42]. The gene implicated in this pathogenesis is EPHB4, a transmembrane receptor that has activity in regulating cell migration with multiple roles in immune cells and neuronal pathways [42]. Lower psoriatic incidence has been noted in Inuit people; the single most prominent ethnic variation that has been noted. This information suggests that diets high in omega-3 fatty acids may be protective against the development of psoriasis [43]. New studies on ethnic variations have suggested Asians and Hispanics are more likely to have pustular and erythrodermic psoriasis as compared to their Caucasian counter parts [44]. The authors suggested these differences are associated with socioeconomic disparities between these ethnic groups which may allow for more severe presentations of disease [44]. One study of children in southern California found the highest prevalence of pediatric psoriasis to be in non-Hispanic whites and the lowest but not negligible incidence in blacks. Also, of note, a lower overall prevalence was noted than historically would be seen in other areas of the country. The authors speculated that increased sunlight exposure and a lower population-based proportion of non-Hispanic whites may have led to this observation [12]. Pathogenesis Although the exact pathogenesis of psoriasis has not yet been revealed, it is clear that both genetic and environmental factors play a role in the development of psoriasis. A positive family history is present in 23–71% of pediatric psoriatic cases [1, 11, 45]. There is a higher rate of concordant psoriasis in identical as compared to fraternal twins (65–72 vs. 15–30% respectively) [46, 47]. Psoriasis vulgaris cannot be linked to a single gene. It appears that several genes may play a role in the genetic susceptibility of psoriasis, most notably HLA-Cw6 [46]. HLA-Cw6 may S. Tolliver et al. interact with the susceptibility alleles, cyclindromatosis gene, CYLD; and transglutaminase 5, TGM5 [48]. Other genes that have been implicated include IL12-B9 (1p31.3); IL-13 (5q31.1); IL-23R (1p31.3); HLABW6; PSORS6, signal transducer and activator of transcription gene 2 STAT2 and IL-23A (12q13.2); tumor necrosis factor α-induced protein 3, TNFAIP3 (6q23.3); and TNFAIP3 interacting protein 1, TNIP1 (5q33.1) [46]. The gene PSORS1 within HLA-C and PSORS2 (17q24-q25) have also been implicated as increasing the risk for psoriasis development [49, 50]. Yet another gene, PSORS6 (19p13), has been shown to interact with PSORS1 [49]. SCL12A8, which belongs to the solute carrier gene family, has also been named as a susceptibility gene [48]. The role of these genes is regulation of Th2 and Th17 lymphocyte activity as well as the NF-κB signaling pathway, which indicates both Th2 and Th17 cells may play a role in the pathogenesis of psoriatic disease. Collections of Th17, Th2 and Th1 cells have been found in psoriatic skin lesions [46, 51]. It is theorized interactions among these cells, along with dendritic cells and neutrophils lead to the release of cytokines, like IL-17 and IL-23, which causes hyperproliferation of keratinocytes and the release of proinflammatory factors within the skin [52]. New susceptibility genes are continually being discovered. Additionally, it appears that susceptibility genes for psoriasis may overlap with other autoimmune disorders. For example, the genes IL-12B and IL-23R are shared with Crohn’s disease and IL-23R is shared with ulcerative colitis. Patients with Crohn’s disease are five times more likely to develop psoriasis as compared to the general population, a fact that may be explained, as least in part, by shared susceptibility genes [46]. Decreased expression of the CD18 gene, mutations in which result in leukocyte adhesion deficiency type I, may also be low in certain patients with psoriasis [53]. Another example includes the gene CARD14 which involves the NF-κB signaling pathway. This aberrant gene is evident in chronic condi- 25 Pediatric Psoriasis tions such as psoriasis vulgaris, pityriasis rubra pilaris, and pustular psoriasis [54, 55]. These diseases, especially pityrisiasis rubra pilaris and psoriasis vulgarius, share similar clinical manifestations that are distinguishable histologically [54]. Conversely, a gain of function mutation in CARD14 is associated with developing psoriasis while a loss of function mutation in the same gene is associated with developing atopic dermatitis [56]. Although no studies have shown a difference in the genetics of pediatric-onset psoriasis as compared to adult-onset psoriasis, preliminary studies have shown differences in inflammatory composition in adults and pediatric patients [57]. Additional genes such as IL36RN in generalized pustular psoriasis is a growing area of research. This gene is an inhibitor of IL-36a, IL-35B, and IL-36y via competitive binding to the IL1RL2 receptor which leads to downstream inhibitor effects in the NF-KB and MAPK signaling pathways [58]. New pathogenic mechanisms are being considered in the literature. Emerging studies on serum levels of IL-22 in adult patients suggest other potential pathogenic mechanisms for future therapeutic use. Some studies report higher levels of IL-22 in psoriatic patients compared to controls and increase with disease burden [59]. Other studies have linked an IL-22 variant that was associated with disease manifestation at an earlier age [59]. Psoriasis is increasingly linked to several autoimmune diseases. Diseases like celiac disease, IBD, uveitis and non-alcoholic fatty liver diseases, which involve the gastrointestinal tract, may share a pathogenic link [60]. Some studies are researching the differences in composition of the gut microbiome in psoriasis patients [61]. Conflicting studies demonstrate both increased and decreased bacterial diversity in their participants necessitating further studies in this area [61]. Table 25.2 gives a list of potential laboratory or diagnostic evaluations for children with psoriasis. The most common precipitating environmental factors in pediatric psoriasis appear to be stress and upper respiratory infections 319 Table 25.2 Suggested work-up for children with psoriasis (Wolverton [62]) At onset: Throat culture or ASO (especially in guttate psoriasis) Biopsy in atypical cases Annually: Joint evaluation (referral to rheumatology for specific symptoms) Cardiac markers: weight, height, body mass index, blood pressure, lipid profile (cholesterol, triglyceride) Optional evaluations: Cultures of macerated or crusted lesions for diagnosis of superinfection (bacterial culture, fungal culture) Celiac panel for children with stomach symptoms, difficulty with clearance or severe disease Thyroid screen for co-morbid arthritis or vitiligo and in symptomatic cases Screening for medications (co-manage with rheumatology when arthritis suspected): Cyclosporine: blood pressure, complete blood count, complete metabolic profile, urinalysis, magnesium, fasting lipid profile (q1–2 weeks for 2 months or for dosage elevations then monthly); urine pregnancy screening at baseline in girls of child bearing age Methotrexate: PPD (baseline and annually), complete blood count with platelets, liver function testing and renal function testing (every few weeks for dosage escalations and at initiation of therapy, then monthly), Hepatitis A, B and C screening, HIV testing for individuals at risk (at baseline); urine pregnancy screening at baseline in girls of child bearing age Liver biopsies are rarely performed in children at this time. Co-management with rheumatology and/or gastroenterology may be helpful in prolonged usage Acitretin. Isotretinoin: complete blood count, liver function tests, cholesterol, triglycerides, urine pregnancy in girls of child bearing age (monthly for isotretinoin due to the iPLEDGE program for the duration of therapy; for acitretin after 6 months can change to quarterly labs); spiral x-rays for suspected DISH; ophthalmology with prolonged usage Etanercept/adalimumab/infliximab/ixekizumab/ ustekinumab: PPD at baseline and annually; screening for prior hepatitis; periodic evaluation for lymph node enlargement; periodic laboratory re-evaluation Referrals: Endocrinology for comorbid endocrinopathy, obesity management, work-up for HPA axis suppression in chronic steroid users Rheumatology for joint pains, limited mobility, co-management of methotrexate or biologics (optional, but suggested for cases with arthritis or suspicion of) (continued) 320 Table 25.2 (continued) Gastroenterology for management/work-up of suspected comorbid inflammatory bowel disease, celiac disease; co-management of methotrexate (optional) Nutritionists/weight down programs Ophthalmology screening for issues arising from oral retinoids (Table 25.1), especially with group A β-hemolytic Streptococcus (Streptococcus pyogenes). Upper respiratory tract infections have been reported to be an inciting factor in 14.8– 28% of pediatric psoriasis cases, while 21.3% of asymptomatic children have tested positive for group A ß-hemolytic streptococcus on throat culture prior to developing psoriasis [1]. Streptococcal pharyngitis may trigger up to 2/3 of all cases of guttate psoriasis [63] as well as exacerbate existing plaque psoriasis [2]. Children with psoriasis induced by Streptococcal infections frequently experience remission [17, 22]. It appears that patients with psoriasis have a unique host-specific response to the streptococcal antigens [63, 64]. Enterotoxin-producing Staphylococcus aureus, Candida albicans, and Human papillomavirus DNA has also been isolated from patients with psoriasis, suggesting that these entities and the corresponding immune response to them may also play a role in the pathogenesis of psoriasis [65–67]. Environmental factors, such as exposure to cigarette smoke, as well as obesity, have also recently been associated with the development of pediatric psoriasis [45]. Psoriasis is an autoimmune disease which, by definition, means it is characterized by the immune system mounting a response against self-antigens. Thus, it is of no surprise that psoriasis is associated with other autoimmune conditions, most frequently of the skin. Patients with morphea have been found to have a higher prevalence of psoriasis than the general population [68]. There have also been case reports of familial associations between psoriasis and vitiligo [69, 70]. Individuals with celiac disease are at increased risk for the development of psoriasis and this association is most pronounced in pedi- S. Tolliver et al. atric patients, meriting celiac disease screening in children with severe psoriasis [71]. An uncertain relationship remains between psoriasis and autoimmune thyroid diseases. In adults, psoriatic arthritis has been linked to a higher incidence of autoimmune thyroiditis than the general population, especially in patients with concomitant rheumatoid arthritis [72]. However no such correlation has been shown with psoriasis localized to the skin and without associated arthritis, as thyroiditis markers are statistically similar between psoriatic adults and age-matched controls [73]. Pediatric studies investigating a link between psoriasis and autoimmune thyroid diseases have yet to be reported. Since the data in the adult population is inconsistent and no studies with children participants exist to be used as guidelines, routine annual screening for thyroid disorders is not recommended in pediatric patients with psoriasis. Screening for thyroid disorders may be warranted in children with psoriatic arthritis or in psoriatic pediatric patients with concomitant vitiligo, or another autoimmune diseases that are more closely associated with thyroid abnormalities [74]. A suggested work-up for children with psoriasis is included in Table 25.2. Obesity and Metabolic Syndrome Recently, an association between psoriasis and the various characteristics of the metabolic syndrome has surfaced in the literature. A growing body of evidence points to an increased risk of cardiovascular mortality in adult patients with severe psoriatic disease. New research is showing similar cardiac risk factors present even pediatric patients [22, 34, 75, 76]. Increased rates of hypertension, hyperlipidemia, diabetes mellitus, and obesity are seen in children and adolescents with psoriasis, with the former three comorbidities occurring twice as often in pediatric psoriatic patients as compared to healthy controls [77]. Furthermore, there is an association with increasing weight and psoriasis severity with some studies showing that adiposity may precede psoriatic lesions by 2 years in a majority of patients [17, 25 Pediatric Psoriasis 22]. Waist to height ratio, a tool commonly used to determine central adiposity, was found to be more common in patients with moderate to severe psoriasis, suggesting increased risk for cardiovascular disease [78]. Even in patients with both normal and overweight, central adiposity was associated with psoriasis which also suggest that this chronic condition effects the body even before lesions are seen [79]. Newly established screening protocol guidelines for comorbidities associated with pediatric psoriasis have been created to aid clinicians in early management and prevention strategies [80]. Adolescents with psoriasis have been found to elevated plasma lipids irrespective of body mass index, suggesting psoriasis itself may lead to metabolic abnormalities [5, 81]. Some studies have suggested an association with higher blood pressures in patients with psoriasis irregardless of weight [82, 83] while others have found no association with hypertension and psoriasis [34]. Regardless, this evidence suggests a need for early screening and management of these co-morbidities in children [75, 84]. Recent studies suggest obesity may even be an independent risk factor in the development of psoriasis [85]. Pubescent females (aged 12–13) with an elevated BMI appear to be at an increased risk for the development of severe psoriasis later in adolescence [86]. As for obesity in psoriatic arthritis, no meaningful differences in weight were found in patients with psoriasis and patients with psoriatic arthritis [87]. Interestingly, this study did not show an increased prevalence of obesity in psoriatic arthritis compared to healthy controls. Whether this was secondary to unmeasured confounding factors or accounting for a patients BMI only at the time of the study remains unclear. Nevertheless, future studies are needed to understand the association between obesity and psoriatic arthritis in pediatric patients [87]. In adults with psoriasis, unhealthy lifestyle habits such as cigarette smoking, excess alcohol intake, and poor dietary choices have been linked to an increased risk of cardiovascular comorbidities [88]. This highlights the need for prevention through the early development of healthy habits in pediatric patients with psoriasis. Referrals to nutritionists, endocrinology, and weight-down 321 programs are advisable interventions in obese adolescents with psoriasis. Diagnosis and Clinical Characteristics (Tables 25.1 and 25.3) The various clinical variants of psoriasis are listed in Tables 25.1 and 25.3. The most common presentation in children is plaque type, with erythematous plaques with overlying silvery scale localized to the extensor surfaces and scalp (Table 25.3). In children, psoriatic lesions are generally thinner, possess less scale and are more pruritic as compared to adults [22, 89]. Certain physical findings are characteristic of psoriasis, including pediatric cases. These findings include: (1) The Koebner phenomenon, (2) post-inflammatory pigmentary alteration, especially in children of color, (3) the presence of punctate bleeding spots when scales are removed or the Auspitz sign, and (4) nail pitting [90]. The scalp is the leading site of psoriatic lesions in all age groups with koebnerization as a common initial insult in some studies [22, 91]. Scalp psoriasis was found to be associated with more extensive disease process and suggests that it may indicate scalp involvement as an early clinical marker for more aggressive treatment [91]. Typical presentation include thick erythematous plaques of the scalp, extending onto the forehead, over the ears and onto the nape of the neck. The forehead is an especially important site in childhood psoriasis due to the fact that facial disease is more prevalent in childhood (Fig. 25.2). It is important to differentiate scalp psoriasis from tinea capitis [92]. A specific scalp finding that is associated with psoriasis, but may be noted even in the absence of psoriasis, is pityriasis amiantaceia, presenting as thick hyperkeratotic concretions attached to the hairs accompanied by scalp erythema. Children in Scandinavia with pityriasis amiantaceia have a stronger family history and personal tendency towards psoriasis than children in the general population [93]. Some authors believe pityriasis amiantaceia is de facto psoriasis, often scalp limited, and usually of childhood. Staphylococcal overgrowth is noted in Inverse Guttate Type of psoriasis Plaque-type Erythematous, sometimes macerated thick plaques of the intertriginous skin, including axillae and groin Can be associated with plaque type psoriasis in other sites Although clinical diagnosis is often possible, similarity to other diseases may require biopsy for differentiation Secondary infection with Candida and/or Streptococcus may require cutaneous culture for diagnosis and prescription of topical anti-infectives Diagnostic features and tests Co-morbidities Check for recent Usually a clinical diagnosis Nail pitting can be noted as a pharyngitis, (Group A beta hemolytic clue to the diagnosis of Streptococcus) psoriasis in children Rule out secondary Biopsy (when needed) infection with S. features aureus Thickened epidermis, neutrophils in the horny layer, Consider comorbid autoimmunity (e.g. the spongiform pustules of thyroid disease, celiac Kogoj and the subcorneal disease) microabscess of Munro (collections of neutrophils in In the setting of obesity, disease the epidermis) Pharyngeal bacterial culture or severity can be a marker of ASO testing cardiovascular risk Recent pharyngitis, Clinical diagnosis is often Small, annular localized (Group A beta erythematous to salmon colored possible hemolytic Biopsy is similar to that of plaques with mild Streptococcus) plaque-type psoriasis hyperkeratosis, sometimes Check for recent pharyngitis, micacous Commonly noted on the trunk, (Group A beta hemolytic Streptococcus), by Pharyngeal abdomen and back bacterial culture or ASO testing Clinical appearance Erythematous plaques with micaceous scale Typical areas: Scalp extending to forehead, nuchal, postauricular, elbows, knees, umbilical and buttocks Table 25.3 Clinical variants of pediatric psoriasis [22, 30] Topical therapy similar to plaque-type psoriasis Oral antibiotics are often used initially due to presumptive infectious precipitating factors and for anti-­ inflammatory capabilities (erythromycin, azithromycin cephalosporins) Systemic agents and phototherapy may be needed in moderate to severe disease unresponsive to oral antibiotics Topical medications should be non-steroidal or low-potency topical corticosteroids to avoid atrophy of the occluded skin Oral or topical anti-infectives should be added where appropriate Nummular dermatitis Lichen planus Secondary syphilis Pityriasis rosea ID reaction Tinea corporis Pityriasis rubra pilaris Pityriasis lichenoides Intertrigo Erythrasma Tinea corporis Langerhans cell histiocytosis Candidiasis Darier’s/Hailey-Hailey disease Contact dermatitis Treatment Treatments must be tailored based upon: (1) the age of the patient, (2) quality of life issues, (3) surface area and (4) sites affected Topical therapeutic regimens are prescribed including keratolytic agents, topical anti-inflammatory compounds (e.g. corticosteroids and calcineurin inhibitors) and topical vitamin A and D analogues Systemic agents and phototherapy may be needed in moderate to severe disease Differential diagnosis Nummular dermatitis Seborrheic dermatitis Tinea capitis ID reaction Pityriasis rubra pilaris Lichen planopilaris Tinea corporis Atopic dermatitis (overlap can rarely be seen) 322 S. Tolliver et al. Macerated, well-demarcated shiny erythema of the groin region including the folds and the genital skin Napkin or diaper Psoriasis (“nappy”) Erythroderma Generalized erythema and thickening of the skin, sometimes with hyperkeratosis Pitting, onycholysis, onychodystrophy, splinter hemorrhages oil spots, subungual hyperkeratosis trachyonychia: (i.e. Extensive pitting and subungual hyperkeratosis) Nail Onychomycosis Secondary infection 1. Fungal culture and nail Alopecia areata plate biopsy to rule out tinea with Lichen planus Candida and/or infection or secondary Pityriasis rubra pilaris candidal infection of the nail Streptococcus bed is needed when subungual And/or dermatophytes Trauma hyperkeratosis is present 2. Avoid trauma orexcess manipulation (e.g. aggressive manicures) Secondary infections Irritant contact dermatitis Biopsy may need to be or perianal strep Diaper dermatitis performed Candidal diaper dermatitis Bacterial and fungal cultures Allergic contact dermatitis may be needed for suspected secondary infections or perianal strep Atopic dermatitis Fever, chills and Two biopsies are generally Congenital nonbullous required from separate sites to malaise can ichthyosiform accompany differentiate psoriatic erythroderma, making Atopic dermatitis erythroderma from other bacteremia a possible Lichen planus causes of erythroderma in Pityriasis rubra pilaris co-morbidity which childhood Seborrheic dermatitis should be ruled out conclusively ivia blood Langerhans cell histiocytosis cultures Pityriasis rubra pilaris Mycosis fungoides Staph scalded skin syndrome (continued) Topical anti-inflammatory agents can be used similar to the therapy of extensive plaque-type psoriasis; however, control of extensive disease is difficult with topical agents and systemic antiinflammatory agents and/ or phototherapy are often required to control severe and extensive disease NB: Special care must be taken to acoid systemic corticosteroids which can precipitate a pustular flare 1. After treatment of any fungal super-infection, topical corticosteroids with tazarotene or calcipotriene can be applied to the paronychial skin. Intralesional kenalog can also be used in the same region to reduce the subungual inflammation 2. May require usage of topical anti-infectives Mild topical corticosteroids with or without topical anti-candidal agents can be helpful Barrier therapy with zinc oxide pastes reduces secondary irritant reactions 25 Pediatric Psoriasis 323 Source: Modified from Silverberg [24] Co-morbidities Bacterial or fungal infections should be ruled out through culture Generally, a clinical diagnosis, Rarely has any however, biopsy is similar to morbidity that of pustular psoriasis Clinical appearance Diagnostic features and tests Erythroderma accompanied by 1. Biopsy is often needed 2. Bacterial culture of the sterile pustule formation, sometimes localized to the distal pustules 3. Fungal culture of the extremities, sometimes generalized. Prior history of oral pustules 4. ASO titers can be drawn to steroid usage may be noted rule out streptococcal precipitant Mucosal/oral Annular plaques on the tongue may be noted in patients with psoriasis Type of psoriasis Pustular Table 25.3 (continued) Differential diagnosis Blistering distal dactylitis Acute generalized exanthamatous pustulosis Staphylococcal scalded skin syndrome Subcorneal pustular dermatosis Sweets syndrome Infected dyshidrotic dermatitis Tinea infection with Tinea mentagrophytes Herpetic whitlow Hand foot mouth disease Aphthosis Lichen planus White sponge nevus Lichen sclerosis Lichen simplex Acrodermatitis enteropathica Contact dermatitis No therapy is usually needed, however topical medicaments in an oral base can be used when needed Treatment Topical therapy (see plaque-type psoriasis) are often ineffective and systemic agents (especially cyclosporine, acitretin) or topical PUVA may be needed 324 S. Tolliver et al. 25 Pediatric Psoriasis a majority of cases of pityriasis amiantaceia and may play a causative role in disease [92]. Several case studies in the literature have reported oral mucosal changes in adult patients with psoriasis and there is debate over whether these changes are part of the spectrum of psoriatic disease. Due to the transient nature of these lesions clinical detection maybe difficult and may be underdiagnosed [94]. Lesions of all areas of the oral cavity, including the tongue, lips, buccal mucosa, gingivae and palate have been reported. However, the overall incidence among patients with psoriasis is low [95]. One study found a 7.7% prevalence of this manifestation in their cohort [94]. Fissured tongue and geographic tongue (also called benign migratory glossitis) are the most commonly reported oral lesions [95, 96], and the incidence of geographic tongue increases as the severity (as measured by the PASI score) of psoriasis increases [96]. Cases of geographic tongue in children with psoriasis have been also reported [97]. In a systematic review of the literature the authors found no clear association with of tongue psoriasis with plaque psoriasis [94]. The histopathology of geographic tongue resembles that of pustular psoriasis, leading some experts in the field to believe that geographic 325 tongue and psoriatic skin lesions may be two manifestations of the same disease process [98]. Although this manifestation has been difficult to confirm it as a discrete clinical entity, it is now commonly accepted in the community [94, 95]. Other potential causes such as candidiasis, lichen planus, leukoplakia, and other autoimmune mucosal manifestations must be ruled out before the diagnosis can be confirmed [94]. The PASI score was devised to give objective measurement to the severity of psoriasis, especially in regard to clinical research, however the score has not been validated in childhood and may not adequately reflect severity of childhood disease especially as it lacks a pruritus or quality of life component. It is a calculation based upon the severity of psoriatic lesions (determined by erythema, induration, and scale), location of lesions, and total body surface area involved. This calculation is outlined in Table 25.4, ranges from 0 to 72, and can be used in older children and adults [4]. A website is also available to simplify the PASI calculation [99]. Other methods of psoriasis scoring base the overall severity of psoriasis on total body surface area involved, quality of life impairment, and the presence or absence of psoriatic arthritis. Still Table 25.4 Psoriasis area severity index (PASI) calculation [4] Body locations Head Trunk Upper extremity Lower extremity Severity of psoriatic lesions 0 = none; 1 = slight; 2 = moderate; 3 = severe; 4 = very severe Erythema 0–4 0–4 0–4 0–4 Induration 0–4 0–4 0–4 0–4 Scaling 0–4 0–4 0–4 0–4 Totals Head severity Trunk severity UE severity LE severity total total total total Surface area (SA) of psoriatic involvement 0 = none; 1 = 10% or less; 2 = 10–29%; 3 = 30–49%; 4 = 50–69%; 5 = 70–89%; 6 = 90–100% Degree of involvement 0–6 0–6 0–6 0–6 Totals Head SA total Trunk SA total UE SA total LE SA total Multiply severity total × SA total Head combined Trunk combined UE combined LE combined total total total total Correction factor (based on area of 0.10 0.30 0.20 0.40 involvement) Multiply combined totals × correction Head corrected Trunk corrected UE corrected LE corrected factor total total total total Add together Corrected Totals for each Body Location to obtain final PASI score Source: Van de Kerkhof and Schalkwijk [4] S. Tolliver et al. 326 others are based solely upon the total body surface area involved, with <3% being mild disease, 3–10% being moderate disease, and >10% being severe disease. However, the latter method may be oversimplified, as quality of life may be significantly impaired with minimal total body surface area involvement [100, 101]. One such example is psoriatic lesions localized to the face, which is much more common in children (affecting 38%) than their adult counterparts [11]. Alterations of PASI with extra weighting for head and neck disease in children may more adequately reflect body surface area distribution in smaller children. Facial disease in our experience is the leading cause of patients and parents seeking systemic therapy. Differential Diagnosis (Table 25.5) The differential diagnosis of pediatric psoriasis includes other papulosquamous diseases of childhood, including psoriaform id reactions, pityriasis rosea, and pityriasis rubra pilaris (PRP), as well as lichen planus, dermatomyositis and lupus erythematosus; as well, pustular disease and nail Table 25.5 Differential diagnosis of pediatric psoriasis Differential diagnosis of psoriasis Differential of generalized psoriasis Clues to diagnosis Lichen planus Purple, polygonal, planar papules over the extensor surfaces Hypertrophic skin lesions in individuals of color Oral white erosive plaques Nail changes include longitudinal ridging and pterygium formation Pityriasis rubra Small follicular papules, pilaris disseminated as well as notably over anterior shins Disseminated yellowish-pink to salmon scaling patches including scalp Palmoplantar hyperkeratosis Dermatomyositis Poikiloderma, atrophy, heliotrope sign, nailfold changes Prominent hyperpigmentation in patients of color Table 25.5 (continued) Differential diagnosis of psoriasis Differential of generalized psoriasis Clues to diagnosis Cutaneous lupus Does not typically involve erythematosus extensor surfaces Discoid lupus with follicular hyperkeratosis, including ear comedones and scarring SCLE usually more annular and in sun-exposed areas Photoexacerbation noted Nasal bridge and cheeks more commonly involved than forehead/ nuchal Malar rash/ photosensitivity associated with systemic disease Pityriasis rosea Herald patch may be present Disease comes up over 6 weeks and resolves over 6 weeks Typically oval lesions with a collarette of scale that follow skin cleavage lines of Langer Differential of scalp psoriasis Seborrheic Greasy and yellowish scale dermatitis Tinea capitis Positive potassium hydroxide stain and fungal culture (for dermatophyte) Broken-off stumps of hairs, often with pustules, alopecia and cervical lymph nodes Differential of guttate psoriasis Small plaque Variably erythematous (but often parapsoriasis less intense than psoriasis) Covered with a fine scale Red–brown and finely scaly Pityriasis papules lichenoides chronica Indolent course and recurrent crops Pityriasis rosea See above Differential of inverse psoriasis Tinea Annular appearance with central clearing Positive potassium hydroxide prep and fungal culture Candidiasis Beefy red macerated plaques Satellite pustules Erythrasma Coral red fluorescence on Wood’s lamp evaluation Contact dermatitis Usually follows the pattern of contactant exposure Generally spares inguinal folds (continued) 25 Pediatric Psoriasis 327 Table 25.5 (continued) hyperkeratosis is distinguished from psoriasis by the scales, which in psoriasis are silvery, light and overlapping as well as by the papules with extend peripherally to form patches in PRP [4]. Only about 1–4% of cases of lichen planus are seen in children. Lichen planus mainly affects the flexor surfaces of the wrists and ankles with purple to violaceous, pruritic papules Nail changes in lichen planus include longitudinal ridging and pterygium formation. In addition, chronic plaque psoriasis may need to be distinguished from mycosis fungoides. As both can have scaling and fissures, palmoplantar plaque psoriasis mimics keratotic eczema of the palms and soles. Examination of the rest of the skin for evidence of psoriasis as well as margination of the lesions, which favors psoriasis, can provide clues to the diagnosis. Further, chronic lesions of dermatitis may look clinically and histologically similar to partially treated psoriasis. Histological examination via biopsy is usually the best method to differentiate these skin disorders from true psoriasis [4]. In the case of guttate psoriasis, the differential diagnosis includes small plaque parapsoriasis, pityriasis lichenoides chronica (PLC), and pityriasis rosea. Small plaque parapsoriasis ­typically occurs in the middle aged and elderly, but can occur in childhood. However, the lesions of guttate psoriasis do not typically affect the palms or soles and are often more erythematous than those of parapsoriasis. In PLC, the papules are erythematous to red–brown and scaly. There is no Auspitz sign when the scale is lifted off. The lesions of PLC come up in successive crops every 6–8 weeks and regress over weeks to months, often with post-inflammatory hypopigmentation in individuals of color. Tinea corporis is also on the differential of guttate psoriasis, when the lesions are more limited number or are annular. Psoriasis of the flexures (inverse psoriasis) is one cause of intertrigo. Other etiologies include tinea corporis, cutaneous candidiasis, erythrasma, and contact dermatitis. Candidiasis will usually present with a beefy red color and satellite pustules. Contact dermatitis (usually irritant in infants) typically spares the inguinal folds, unlike psoriasis. Erythrasma can be differentiated by Differential diagnosis of psoriasis Differential of generalized psoriasis Clues to diagnosis Langerhans cell Typically also have scalp scaling histiocytosis and crust Adenopathy may accompany lesions Differential of palmoplantar psoriasis Atopic dermatitis Intensely pruritic vesicles and/or bullae May have other classic areas of involvement (flexural surfaces) Juvenile plantar Balls of feet toe pads, and hands dermatosis symmetrically tender and reddened, dry with shiny appearance, may have scale and painful cracks and fissures Palmoplantar Hereditary vs Acquired thick, keratoderma yellow hyperkeratosis, involves the lateral aspects of the hands and feet transgredient vs non-transgredient Reiter’s syndrome Urethritis, arthritis, ocular findings, and oral ulcers in addition to psoriasis form skin lesions Lesions on the plantar surface with thick yellow scale and often pustular (keratoderma blennorrhagicum) changes can be mimicked by cutaneous infections. Like psoriasis, the distribution of dermatomyositis can include extensor surfaces especially elbows and knees. However, patients with dermatomyositis have skin changes that show poikiloderma, atrophy, a heliotrope sign, and nailfold changes (on dermoscopy or capillaroscopy), none of which are typically seen in psoriasis [4]. Lupus erythematosus, however, usually lacks involvement of extensor surfaces. Furthermore, psoriasis is generally improved by, not exacerbated by UV exposure [4]. The eruption of pityriasis rosea is typically located on the upper arms, trunk, and thighs with a duration of a few weeks. A herald patch is frequently noted, and lesions are typically oval and follow skin cleavage lines. Individual lesions only have a collarette of scale and have crinkling of the epidermis [4]. PRP with its small follicular papules, disseminated yellowish-pink scaling patches, and palmoplantar S. Tolliver et al. 328 using a Wood’s lamp, demonstrating coral red fluorescence. Tinea corporis typically shows an annular border. In infants especially, the possibility of Langerhans cell histiocytosis needs to be considered. In these patients, there may also be scalp involvement with scaling and crust, lymph node enlargement and internal organ involvement. On the scalp, tinea capitis, and seborrheic dermatitis often mimic psoriasis. Distinguishing feature of tinea capitis are broken-off stumps of hairs, often in patches in which there are crusts and pustule. When examined, the broken-off hairs are loose and found to be surrounded by or contain the fungus. Seborrheic dermatitis often co-exists with psoriasis, although it’s uncommon past infancy and prior to puberty. Although typically the scales in psoriasis are dry, shiny and white, versus those of seborrheic dermatitis which are greasy and yellowish, distinguishing the two requires fungal culture. Biopsy and Histology Psoriatic lesions contain characteristic histologic features when biopsied that can help differentiate psoriasis from other skin diseases if the clinical picture is unclear. A mixed inflammatory perivascular infiltrate is commonly seen in the dermis. The epidermis appears acanthotic with focal areas of spongiosis containing inflammatory cells. There is thinning of the suprapapillary plate, and the stratum granulosum may be absent. Parakeratosis, the persistence of nucleated keratinocytes in the stratum corneum, is common [22]. Psoriatic plaques with dotted vessels can be seen on dermoscopy [22]. There are two possible pathognomonic findings in psoriasis histology. The first is the spongiform pustule of Kogoj, characterized by a spongiotic pustule filled with neutrophils in the stratum spinosum. The second is the microabscess of Munro, characterized by the presence of neutrophils in the stratum corneum. As the lesion progresses, the rete ridges elongate and become blunted, and the hyperproliferation of the epidermal keratinocytes becomes more apparent histo- logically. In pustular psoriasis, there are more marked accumulations of neutrophils in the epidermis, similar to the spongiform pustules of Kogoj and microabscesses of Munro [4]. Lesser known pathognomonic findings such as sandwich sign depict neutrophils and parakeratosis alternating in the statum corneaum [102]. Therapeutic Management (Table 25.2 and Fig. 25.6) [62] Topical Therapies Topical therapies are the initial treatment of choice for pediatric patients with limited psoriasis of the skin (i.e. in the absence of joint disease). In general, thicker vehicles, such as ointments, are more effective, but the location of [62] involvement and patient preference will ultimately affect the decision of which vehicle will be used. Thinner preparations are commonly used on the scalp and face while thicker ones are used on the extremities [103]. Keratolytics, such as urea, salicylic acid, and α-hydroxy acids, are the most simplified of the topical therapies. The mechanism of action of these agents is through removal of the characteristic superficial hyperkeratosis of psoriatic lesions, allowing for better penetration of other topical therapies [21, 103]. Salicylic acid application can lead to percutaneous salicylism in infants and is therefore avoided in this age group [103]. One of the earliest topical therapies developed, and still in use today in some areas of the world, is the Goeckermann regimen. It involves topical application of coal tar, or a modified version with addition of a cream called liquor carbons detergents, followed by exposure to UV light. It is proven to be effective in children, with 85% achieving >80% clearance in psoriatic skin lesions [104]. The mechanism of action remains unclear, but antimitotic effects, inhibition of DNA synthesis, and enzyme inactivation may play a role [103]. Despite its effectiveness, the 25 Pediatric Psoriasis 329 Diagnosis of psoriasis in a child Small BSA involved Topical agents Adjuncts Corticosteroids Topical exfoliants Goeckermann regimen Omega-3 fatty acids (oral) Anthalin Vitamin D3 derivatives Calcineurin inhibitors None Excimer laser Oral antibiotic therapy + topical agents; tonsillectomy in recurrent streptococcal pharyngitis induced flares Larger BSA involved; decreased quality of life Perform screening tests before prescribing systemic agents Prior TB or HBV infection, prior malignancy/lymphoma, immunosuppressed state Localized phototherapy PUVA or narrowband UVB Evidence of streptococcal infection or rapid-onset cutaneous disease with pharyngitis CBC, liver function tests, lipid panel, blood pressure, UA, pregnancy test Prior TB, HBV, cancer risk, or immunosuppressed TNF-a Inhibitors to Biologic Agents Topical agents Enteracept Generalized phototherapy Adalimumab infliximab PUVA or narrowband UVB Rotational therapy (6–12 mo alternating) Methotrexate Cyclosporin A Acitretin Ixekizumab Ustekinumab BSA = body surface area, TB = tuberculosis, HBV= hepatitis B virus, CBC = complete blood count, UA = urinalysis, TNF-α = tumor necrosis factor alpha PUVA = psoralen plus UVA Fig. 25.6 Schema of psoriasis therapy in childhood, based on Silverberg [21] Goeckermann treatment has fallen out of favor in industrialized nations due to concern over its long-term consequences. The coal tar contains polycyclic hydrocarbons and, when exposed to UV irradiation, this combination has been shown to cause increased chromosomal abnormalities in peripheral lymphocytes and an elevated Heat Shock protein (specifically Hsp70) response in children [105]. However, there is no concrete evidence demonstrating that treatment with coal tar or its derivatives leads to an increase in skin cancer risk as compared to the general population [103] A milder variation on this theme is to use dilute tar bath additives the night before narrowband UVB therapy (1–2 caps tar added to tub and soak 10–15 min). Unfortunately, there are no Food and Drug Administration (FDA) (United States) topical therapies specifically approved for the treatment of psoriasis in children under 12 years of age, but there have been several studies demonstrating efficacy of off-label agents, including topical corticosteroids, vitamin D2 and D3 derivatives, and immunosuppressants (e.g. calcineurin inhibitors) [106–108]. The strength and formulation of therapies should be based on the patient’s age, PASI score, and impact on quality of life. A recent systematic review of 64 studies from The Netherlands proposed an algorithm of beginning with topical synthetic vitamin D3 derivatives with or without topical corticosteroids, followed by anthralin cream [106]. However, in the United States, topi- 330 cal corticosteroids are usually the initial treatment of choice [103]. Topical corticosteroids are frequently employed by practitioners for the treatment of pediatric psoriasis. The choice of which topical steroids to use depends upon the skin areas involved, and parallels the treatment algorithms for atopic dermatitis in children. Low potency steroids (classes 5–7) are used for more sensitive areas such as the head, neck, and intertriginous regions. Moderate potency (classes 2–4) steroids are generally used on the scalp and extremities. Higher potency (class 1) steroids are reserved for persistent, thickened lesions that do not respond to lower potency steroids [21, 103]. Two class 1 corticosteroids, clobetasol and halobetasol, have been studied and found to be effective in children. Clobetasol (in some specific preparations) is approved for children >12 years of age for 2 weeks or less, while halobetasol is not currently FDA-approved for use in children. For scalp psoriasis, the combination of calcipotriene 0.005% and betamethasone dipropionate 0.064%, or taclonex, has been FDA-approved for patients 12 years or older [17]. Treatment duration with class 1 steroids is often limited to 2 weeks in children, as these agents have an extensive side effect profile with long-term use, including skin atrophy, striae, and potential hypothalamic-pituitaryadrenal (HPA) axis dysfunction [106, 107, 109]. All topical corticosteroids if used prolongedly and especially over large surface areas, including classes 2–7, are theoretically associated with these adverse effects as well. The risk of systemic absorption and HPA dysfunction increases with the body surface area to which the steroid is applied increases. This is of particular concern in children, as they have usually not yet reached their growth potential [21]. A recent systematic review of the use of topical corticosteroids showed no systemic and minimal topical adverse effects in the studies included. The incidence of adverse events can be decreased by rotating the use of topical corticosteroids with other topical treatments listed below [103]. Anthralin 1% cream, or dithranol, is a topical therapy that can be used for localized areas of psoriasis. It is an anti-proliferative and immuno- S. Tolliver et al. suppressive agent which inhibits T-lymphocytes through an unclear mechanism of action. Adverse effects are minimal due to limited systemic absorption and include irritation and staining of clothing or hair [103, 106]. Short contact therapy for several minutes a day minimizes staining and irritation of the skin [103]. Vitamin D3 derivatives, such as calcipotriol/ calcipotriene or calcitriol, have also been proven to be effective in children with psoriasis. Side effects include local reactions such as erythema, irritation and pruritis. Mechanistically, they inhibit keratinocyte proliferation which allows them to work synergistically with corticosteroids [22]. There is also a theoretical risk of systemic absorption leading to elevated serum calcium levels, especially when applied to a large body surface area [106, 110, 111]. Doses of calcipotriene up to 45 g/m2/week have been shown to have no significant effect on calcium homeostasis in children [103]. A head-to-­head comparison of calcipotriol and anthralin showed similar efficacy in psoriatic children [112]. Clinical trials determining the long-term safety and efficacy of calcitriol ointment, vectical, are underway and may lead to the first FDA-approved topical treatment for pediatric patients under 12 years old. Tazarotene, a topical retinoid, is also effective for limited psoriatic skin disease and nail psoriasis [103]. Topical calcineurin inhibitors, such as tacrolimus and pimecrolimus, have also been shown to promote the clearance of psoriatic lesions in children [106, 113]. These agents are immunosuppressants that inhibit cytokine production (IL-2 in particular) by T lymphocytes and thus limit their proliferation. Tacrolimus (0.03% for children ages 2–15 years and 0.1% ointment for ages 16 years and over) and pimecrolimus 1% cream are approved for the treatment of atopic dermatitis in children >2 years of age, but its use is off-label for psoriasis [103, 113]. The advantage of these agents is the lack of potential skin atrophy, allowing their use in more sensitive areas such as the face, neck, and intertriginous areas [103, 113]. However, use of calcineurin inhibitors on plaque psoriasis how been shown to be less effective likely secondary to poor absorption rates in the skin [111]. Side effects most fre- 25 Pediatric Psoriasis quently noted include local irritation and pruritis [106]. This class of topical medication also carries a black box warning in the US due to a theoretical increased risk of skin cancer and lymphoma. Sun protection is therefore advisable concurrent with calcineurin inhibitor products [114]. 331 adults. Therefore, the use of systemic antibiotics for the treatment of pediatric psoriasis remains controversial [106, 116, 119]. Tonsillectomy has been recommended by some as a treatment for recurrent guttate psoriasis [117], but this has also not been proven to be effective in an evidencebased manner [106, 119]. Methotrexate was the original therapy available for extensive psoriasis. It is generally safe in Systemic Therapies (Table 25.2 children and is a well-established treatment for moderate to severe psoriatic disease. Methotrexate and Fig. 25.6) is also the drug of choice for patients with psoriSystemic therapies for the treatment of pediatric atic arthritis in addition to skin disease [120]. psoriasis are usually reserved for those patients Methotrexate is associated with a >75% improvewho have failed topical treatments, have exten- ment in PASI score [121]. Dosages used range sive body surface area involvement, have signifi- from 0.2 to 0.7 mg/kg per week [120–122]. The cant impairment in quality of life and/or most common side effect in children is nausea psychological health, or have concomitant psori- and vomiting. Although routine monitoring of atic arthritis. Like all systemic medications, the blood counts and liver function tests are recomagents available for use in pediatric psoriasis mended, bone marrow suppression, such as have side effects. For this reason, many physi- microcytic anemia and pancytopenia, and liver cians employ a cyclic approach for systemic ther- toxicity are rare adverse drug events. apy, alternating between different available Liver toxicity appears to be relatively rare in treatments every 6–12 months to minimize long- children, although obesity and associated fatty term adverse events. Systemic therapies include liver changes are associated with an increased oral antibiotics, methotrexate, cyclosporine A, risk of this adverse reaction [106, 120, 122]. retinoids, TNF-α inhibitors, and biologics. Concomitant folic acid supplementation (1 mg/ Oral antibiotics are considered the safest of day) is protective against bone marrow suppresthe systemic agents due to their superior side sion and liver enzyme abnormalities [123]. effect profile. Benefits with oral antibiotics are Cyclosporine, another immunosuppressant most impactful in the setting of severe, rapid-­ which inhibits cytokine signaling by lymphoonset cutaneous disease following streptococcal cytes, may be effective for psoriatic skin disease pharyngitis or perianal streptococcal disease, and in children. In doses ranging from 2 to 4 mg/kg/ in pustular and guttate psoriasis [115–117]. A day, cyclosporine can improve the overall severrecent controlled trial of 50 patients (ages ity and appearance of skin lesions, but studies 13–63 years) found that a 48-week course of investigating its effectiveness in children are limazithromycin therapy (4 days of azithromycin ited [124, 106, 125, 126]. Recent studies from 500 mg tab once-daily, followed by 10 days off India followed 8 children’s use of cyclosporine therapy, with repeat of the cycle every 2 weeks) and found the drug to have few side effects, resulted in a PASI 75 of 80% at 48 weeks in the abdominal pain and increased creatinine, with treatment group as compared to no significant significant improvement in psoriatic lesions. difference in the control group. Only 12 patients These patients were treated until achieving a had a positive ASLO test for streptococcal infec- PASI 75 then subsequently tapered 50 mg every 2 tion [118]. Despite this and other studies demon- weeks [126]. Because of its rapid onset this medstrating an association between streptococcal ication can be utilized as a “rescue” treatment infection and the onset of psoriasis, it remains and then switched to a safer drug for maintenance unclear whether antibiotic therapy improves therapy [126, 127]. In addition, its use is hindered PASI scores or disease clearance in children or by the side effects of nephrotoxicity, secondary 332 hypertension, hyperlipidemia, and immunosuppression as well as the requirement for close monitoring of blood pressure and renal functioning, which is more likely to become permanently impaired in individuals on therapy beyond 6 months (Table 25.2). Cyclosporine is of particular benefit for the therapy of pustular psoriasis (Poster SPD) [127]. Reports of an increased cancer risk secondary to immunosuppression have also been reported, but the low doses, diligent monitoring, short courses and rotational therapies used in treatment of skin-limited psoriasis decrease these risks [120]. Retinoids, such as acitretin or etretinate, are other treatment options for children with pustular, erythrodermic, or plaque psoriasis, although they are not as extensively studied or as safe as methotrexate [106, 128]. Adverse effects include teratogenicity, elevation of liver enzymes, impaired lipid profile, alterations in blood counts, and bony abnormalities. Due to the ability of retinoids to cause severe birth defects, oral contraceptives are recommended for 1 month before initiating therapy and for 3 years after the cessation of therapy in girls of childbearing age [22]. Isotretinoin may be a preferred choice for those at risk of pregnancy. Good pregnancy prevention (2 methods of birth control) and monitoring for pregnancy are needed in those of child bearing age. Close monitoring of liver function tests and lipid profiles during therapy are necessary as well. High-dose systemic retinoids used for greater than 2.5 years have been associated with premature epiphyseal closure in children. Hyperostosis and osteoporosis are other potential bony side effects [129]. However, cyclic shortterm therapy for 6–12 months, as indicated in rotational therapy, decreases the risk for bony abnormalities. Longer courses of treatment in children require periodic skeletal surveys [120]. TNF-α inhibitors, such as etanercept, adalimumab, and infliximab, have been used to treat pediatric psoriasis, but only entercept is approved for this purpose. This recent change comes after a recent randomized double-blinded, placebo-controlled study showed promising long-­term safety data with beneficial effects on quality of life in pediatric patients [17, 130]. With the paucity of S. Tolliver et al. literature determining long term safety data in pediatric patients treated specifically for psoriasis, there is much debate on this use of biologics in this population. Early studies determining long term safety profiles for these drugs are being conducted. One study followed 12 patients over 8 years and found similar side effects, infections and injection-site reactions, previously reported by other investigations [131]. Furthermore, they found that up to 50% of patients initially treated with etanercept needed to switch to another biologic. Whether this is due to the changing immunologic make-up in pediatric patients remains to be seen. However, this suggests a potential treatment strategy for failed biologic therapies [124, 131–134]. TNF-α inhibitors have been used to treat rheumatoid arthritis, tumor necrosis factor 1-­associated fever, juvenile idiopathic arthritis, ulcerative colitis and Crohn’s disease in children for more than a decade [21]. The most extensive studies of the safety of TNF-α inhibitors in children come from research investigating their use in juvenile rheumatoid arthritis, with up to 8 years of published data on this subject. Side effects include reactivation of latent tuberculosis or hepatitis infections as well as increased risks for malignancies including lymphoma, opportunistic infections, and demyelinating disease. The FDA has recently placed a black box warning on Enbrel for pediatric psoriasis stating, “Lymphoma and other malignancies, some fatal, have been reported in children and adolescent patients treated with TNF blockers, including Enbrel.” A recent retrospective cohort study comparing 9045 pediatric psoriasis patients and 77, 206 healthy controls found that there was no overall increased cancer risk when taking TNF-a inhibitors. However, lymphoma the primary cancer reported in these patients [135]. In one study following pediatric patients with juvenile rheumatoid arthritis treated with etanercept, no cases of tuberculosis, opportunistic infections, malignancies, lymphomas, lupus, demyelinating disorders, or deaths were reported after 8 years [136]. The most commonly reported side effects with etanercept specifically are injection site irritation and non-­opportunistic infection 25 Pediatric Psoriasis [106]. Interestingly, there have studies showing that TNF-α inhibitors are associated with outbreaks of psoriasis and other cutaneous manifestations in children and adults being treated for other autoimmune disease processes [124, 132, 133, 137, 138]. One study suggests that this potential is a result from the loss of inhibition of TNF-α on dermal plasmacytoid dendritic cells which leads to increases in IFN-γ [132, 133]. Emerging data regarding this phenomenon is especially seen in children with IBD with one study demonstrating 8.8 percent of their patients treated with TNF-α inhibitors developing psoriasis [132]. Another study based on patients with JIA found a 5.4% prevalence of psoriasis when using TNF-α inhibitors, with female predominance and low incidence of personal or family history of psoriasis [132]. As for which TNF-α drug is most likely to cause this adverse effect remains to be seen, some studies showed equal prevalence of psoriasiform lesions when using both adalimumab, infliximab and etanercept while other studies implicate infliximab as the culprit [132–134, 139, 140]. More studies on this subject are needed to understand this paradoxical side effect of TNF-α inhibitors. The literature as well as clinical practice are trending toward increased use of biologics with as much as 25% of children with psoriasis being treated with these medications [141]. When determining which patients will benefit from biologic therapy consider children and adolescents with greater than 10% BSA, DLQI >10, failed standard therapies or other contraindications [142] with special attention to educating patients on the risks and benefits of using these medications. Some studies suggest using systemic therapies earlier in mild psoriasis and patients with lower DLQI scores. Allowing patients to advance to systemic treatment may improve quality of life [135]. Vaccinations are another special consideration for patients starting biologic therapy. Live vaccines are absolute contraindications to biologic therapy [142, 143]. Biologics, over other systemic treatments like methotrexate and cyclosporine, require less stringent treatment schedules, have fewer adverse effects, decreased need for monitoring, and greater efficacy [144]. 333 However, the safety profile of these medications are still being elucidated [141]. Biologic agents initially approved in adults have been examined and approved in childhood and adolescent psoriasis [145]. Etanercept, which is the most extensively studied of the TNF-α inhibitors in children, has recently been approved for use in children 6 years and older with ulcerative colitis and Crohn’s disease, and is now approved to treat psoriatic arthritis and plaque psoriasis in adults and pediatric patients 4 years and older [130, 146]. Etanercept at doses of 0.8 mg/kg weekly has been shown to achieve PASI 90 in 27% and PASI 75 in 57% of treated patients as compared to 11 and 7% of placebo-treated patients after 12 weeks of treatment respectively [147]. This benefit appears to be maintained in the majority of patients after 96 weeks [148]. Etanercept has also been shown to improve overall and disease-specific quality of life in children with moderate to severe plaque-type psoriasis [149]. It has also been shown to improve depression in adults, irrespective of disease clearance. This suggests that the inflammatory response itself, rather than the psychological effects of an undesirable physical appearance, may contribute to depressed mood in psoriatic patients [150, 151]. Other TNF-α inhibitors have not been extensively studied in children with psoriasis [106]. Adalimumab is currently not FDA-approved in the USA. However, with increasing understanding of the chronicity of psoriasis and associated co-morbidities, long-term treatment with safe and effective drugs are an important area of research. Recent results from Phase 3 clinical trials comparing the efficacy and safety of adalimumab and methotrexate are promising [152]. Patients were able to maintain resolution of disease with 0.8 mg/kg every other week in this study for 52 weeks. Furthermore, patient’s refractory to methotrexate were able to achieve improvement when switched to adalimumab every other week. Patients were found to have a similar safety profile as adults with nasopharyngitis and upper respiratory infections being the most common adverse effects [152]. The safety of adalimumab in pediatric patients was further studied in patients who were being evaluated for its use in JIA, pediatric enthesitis-related arthritis, psoriasis, and crohns disease [153]. In 577 patients who partici- S. Tolliver et al. 334 pated in clinical trials dating back to September 2002 the most common side effects reported were infections with no malignancies reported at all. This study adds to the amounting evidence that adalimumab is safe in pediatric patients with similar adverse events as in adults [153]. Ustekinumab or Stelara, is emerging as safe and efficacious treatment modality for improvement of moderate to severe psoriasis in adolescents. Ustekinumab is an Il-12 and IL-23 inhibitor that is FDA approved for patients 6 years or older [140]. Its dosing schedule recommends starter doses at week 0 and week 4 then once every 12 weeks thereafter. This less involved dosing schedule is ideal for adolescents with many time sensitive activities [128]. It has been shown to have a favorable side effect profile and efficacy comparable to adult populations being treated for psoriasis [140]. The CADMUS Jr trial addressed efficacy of ustekinumab in 6-12 year old children with moderate to severe psoriasis, with 84% of children achieving PASI 75 and 64% PASI 90 by 12 weeks, with no new safety signals [154]. Approval in children 6 years of age and older has been granted in the United States. The IXORA-PEDS study addressed the highaffinity IL-17a monoclonal antibody ixekizumab in pediatric psoriasis. PASI-75 was 89% at week 12. Approval in children 6 years of age and older has been granted in the United States [155]. Other potential treatments are being considered for cases refractory to the TNF-a inhibitors, ixekizumab and ustekinumab such as guselkumab, apremilast, secukinumab. Guselkumab is an anti-­ interleukin-­ 23 monoclonal antibody. There is a paucity of literature for the use of guselkumab in even adult patients. However, the recent NAVIGATE clinical trial demonstrates the efficacy of guselkumab in ustekinumab refractory patients [156]. In this study guselkumab was shown to have a greater efficacy when compared to ustekinumab in patients unable to achieve 0 or 1 investigator assment scores. The safety profile of guselkumab and ustekinumab were similar with guselkumab having a slightly higher incidence of adverse events, infection being reported most often [156]. As for pediatric patients, there is one case report of guselkumab use in a patient refractory to high potency topical steroids, phototherapy, and systemic treatments such as methotrexate, adalimumab, and ustekinumab [157]. In this patient guselkumab was added to the dosage of methotrexate and the patient’s psoriatic plaques thinned by week 4 with full resolution by 5 months. No adverse effects were reported. This case report demonstrates a potential future treatment modality in which patient’s refractory to other biologics and systemic agents can achieve remission of their disease [157]. There is one case report in the literature that details the use of apremilast an oral phosphodiesterase-4 inhibitor, in a 14-year-­old boy with guttate psoriasis [102]. Previous therapies used include topical therapies, natural light exposure, antibiotics. Apremilast was prescribed at the adult dosing based on the patient’s weight. After 6 months of therapy the patient had improved in both his skin disease and quality of life [102]. There have been reports of psychological distress in adults taking apremilast, depression and suicidal ideation, thus warranting monitoring [158]. To date there are no case report detailing the use of secukinumab, and IL-17 inhibitor, treating plaque psoriasis in pediatric patients. There have been case reports demonstrating its use in patients with recalcitrant nails disease which suggests other potential indications for pediatric patients [143]. Several clinical trials are underway to determine the safety profile and efficacy of secukinumab in patients’ children and adolescents with moderate to severe psoriasis as well as dosing strategies. Phototherapy UV light therapy is used to treat a variety of skin diseases in children, including atopic dermatitis, vitiligo, acne vulgaris and psoriasis [159]. When determining what patients are good candidates for phototherapy a thorough assessment of history, former unsuccessful treatments, availability and transportation should be completed [111]. Indications for use include older pediatric patients with extensive disease, contraindications to systemic treatments, guttate psoriasis, and thinner plaques. Phototherapy is available in the form of 25 Pediatric Psoriasis broadband UVB, narrowband UVB or UVA in combination with psoralen (PUVA). Both UVB and UVA have been shown to be efficacious when be used in combination with topical steroids, topical retinoids, emollients, coal tar or vitamin D analogs [111]. Narrowband UVB (NB UVB) and PUVA therapy have shown the most success in treating children with psoriasis [21, 103, 159]. However, NBUVB (311 nm) is the treatment of choice, as PUVA therapy has been associated with an increased risk of carcinogenesis in adult patients with psoriasis and development of cataracts in pediatric patients with psoriasis [111, 159]. NB UVB is also highly effective and safe, with a response rate approaching 80% in pediatric patients [111, 160]. Topical PUVA therapy is therefore recommended for refractory palmoplantar psoriasis based on its side effect profile [111]. If PUVA therapy is used, topical psoralen is preferred, due to the necessity for 24 h of protective eyewear following treatment with oral psoralen. For this reason and the increased risk of other systemic adverse reactions, oral psoralen should be avoided children less than 12 years of age. Absolute contraindications include diseases characteristically exacerbated by UV light, such as lupus erythematosus, dermatomyositis, and xeroderma pigmentosum [111]. Common adverse events of phototherapy include local irritation and erythema. There is also the possibility of reactivation of a latent herpesvirus infection, photoaging, and the long-term risk of carcinogenesis [106, 159–162]. Phototherapy maybe administered in the form of a handheld laser, phototherapy booth, or hand/ foot units. If a phototherapy booth is employed, the child must be old enough to remain still for the length of each therapeutic session [21]. Compliance issues in younger children may restrict treatment, thus phototherapy is often more useful in adolescents [21, 159]. Although there are no strict guidelines on the age in which phototherapy can be initiated, school age children are a reasonable starting point, however younger patients, if able, can still receive therapy [111]. Special considerations for teenagers using phototherapy include tanning bed use. This presents an increased risk for adverse reactions such 335 as burning, redness, and pain because of the increased amount of ultraviolet exposure [111]. Therapy is required once or twice per week for a 1–3 months before psoriatic lesions clear and a maintenance phase can be reached [21]. This can be difficult for many families with busy schedules and varying temperaments in young children. A potential option for this barrier is to consider home treatment using either 10–15 min of natural sunlight or at home light boxes. As for natural sunlight, this option can be considered during the spring and summer months and is cost effective [111]. Potential barriers include schedules during day light hours that may make it difficult to be outside like school in adolescent children. As for home light therapy, this option is for children in need of long-term therapy and has the benefit of it less intimidating that a doctor’s office. As a tradeoff, these treatments are usually longer because of lower fluences and necessitate direct parental supervision [111]. Excimer laser, a narrowband UVB 308 nm laser light source has been described to be effective for the therapy of localized psoriasis. Scalp, palms and soles and areas on the face and body can be treated. Some data on usage in childhood shows benefit for children with localized psoriasis, and children may experience superior benefit with excimer usage. Therapy has some ultraviolet light induced side effects, but is generally well tolerated [163]. Natural Supplements Various natural supplements have been investigated in the treatment of pediatric psoriasis and this is a common topic of inquiry among parents. No natural remedies have found to be curative, but a few may help alter the disease progress. Omega-3 fatty acids, such as those found in salmon and other fatty fish, may lead to slight improvements in PASI score. As previously mentioned, this finding is supported by the observation that Inuit populations, which consume diets high in omega-3 fatty acids, have a low incidence of psoriasis. The benefits of omega-3 fatty acid supplementation are superior to omega-6 fatty acid supplementation. The mecha- S. Tolliver et al. 336 nism of action appears to be alterations in the level of eicosapentanoic acid and arachidonic acid, leading to decreased production of pro-inflammatory metabolites [43]. The benefits of omega-3 fatty acids are minimal or possibly non-existent, but this supplement appears to be essentially harmless, and thus safe for use in children. There may be a more important role for dietary changes in pediatric patients with metabolic syndrome and psoriasis, due to the recent association between these two disease entities. Diets low in fat and high in fiber have been found to beneficial in treating metabolic syndrome in children, and along with exercise, can even cure patients of this condition with lasting benefits into adulthood [164, 165]. Indigo naturalis, a traditional Chinese medicine, has been reported to aid in clearance of pediatric psoriatic lesions after 8 weeks of topical application in one anecdotal study [166]. Other traditional Chinese medicines and natural supplements may ultimately cause more harm than benefit and it is important for dermatologists to be aware of the potential side effects of these therapies [167]. The role of this medication in children is unknown at this time. Dietary Recommendations The role of diet and supplementation in the management of psoriasis is controversial. Anecdotally, some patients report improvement of symptoms with changes in diet. Healthier food choices lead to reductions in heart disease, diabetes, and obesity, however, the effect of healthier food choices on the disease burden of psoriasis remains unclear. The National Psoriasis Foundation overall recommends meals nutritionally balanced with lean proteins, whole grains, fresh produce, and healthy fats, and is sometimes referred to as the “anti-­ inflammatory” or Mediterranean diet [168, 169]. Foods that promote inflammation include fatty red meats, processed foods, refined sugars, and dairy products [168]. Although there is limited evidence to justify changing eating habits there is a well-established linked between metabolic syndrome, obesity and psoriasis [169]. Body fat has pro inflammatory mediators that may exacerbate psoriatic lesions [84]. Thus, the benefits of a healthy balanced diet remains an important factor in overall management of psoriatic patients. Furthermore, adiposity has been noted to precede onset of psoriasis, making diet a potentially important preventive agent [170, 171] Dietary modifications that eliminate foods and the role of large supplements in children are concerning for risk of side effects and potential malnutrition. Recent studies investigating dietary recommendations for adults with psoriasis or psoriatic arthritis have shown that hypocaloric diets leading to weight loss in overweight and obese adults has a positive impact on patients regarding psoriasis severity and dermatologic quality of life [169]. Hypocaloric diets refer to ranges from 800 to 1400 kcal per day. Diet alone does not maintain psoriasis remission and adjunctive biologic therapy is recommended [169]. Whether the type of diet or weight reduction leads to the reduced severity needs further investigation. The benefit of calorie restriction in children is unknown and therefore, it is best to work with the parents, children, nutritionists and the pediatrician to find the best strategy for healthier weight achievement and maintenance. Gluten-free diets in psoriatic with celiac disease were highly recommended. Psoriatic patients with positive serologic markers for gluten sensitivity were recommended to undergo a 3-month trial of gluten-free diet with adjunctive medical therapy [169]. As for adult patients with psoriatic arthritis, it was highly recommended that they undergo a trial of vitamin D supplementation with 0.5 μg alfacalcidol or 0.5– 2.0 μg of calcitriol daily [169]. Usage of oral vitamin D supplements in children with psoriasis and/ or psoriatic arthritis has not been adequately explored at this time. For other supplements, such as omega-3 fatty acids, selenium, Vitamin B12, and micronutrients evidence was limited and therefore no recommendations were made. Conclusions Psoriasis of childhood is a common and complex disorder that can occur from birth up through adolescence. Consideration for the child’s emo- 25 Pediatric Psoriasis tional well-being and general health, including risk factors for the metabolic syndrome and possible side effects of therapy, have to be given when choosing therapy for this disease. Recent advances in phototherapy and biologic therapy have expanded the armamentarium of this disease. Greater research is needed to assess the benefits of therapy on long-term disease manifestations and the risks of lymphoma and other potential side effects of therapy. References 1. Seyhan M, Coşkun BK, Sağlam H, Ozcan H, et al. Psoriasis in childhood and adolescence: evaluation of demographic and clinical features. Pediatr Int. 2006;48(6):525–30. 2. Gudjonsson JE, Thorarinsson AM, Sigurgeirsson B, Kristinsson KG, et al. Streptococcal throat infections and exacerbation of chronic plaque psoriasis: a prospective study. Br J Dermatol. 2003;149(3):530–4. 3. Kumar B, Jain R, Sandhu K, Kaur I, et al. Epidemiology of childhood psoriasis: a study of 419 patients from northern India. Int J Dermatol. 2004;43(9):654–8. 4. Van de Kerkhof PCM, Schalkwijk J. Psoriasis. In: Bolognia JL, Jorizzo JL, Rapini RP, editors. Dermatology. 2nd ed. Philadelphia: Mosby Elsevier; 2008. 5. Koebnick C, Black MH, Smith N, Der-Sarkissian JK, et al. The association of psoriasis and elevated blood lipids in overweight and obese children. J Pediatr. 2011;159(4):577–83. 6. Vogel SA, Yentzer B, Davis SA, Feldman SR, et al. Trends in pediatric psoriasis outpatient health care delivery in the United States. Arch Dermatol. 2012;148(1):66–71. 7. Raychaudhuri SP, Gross J. Comparative study of pediatric onset psoriasis with adult onset psoriasis. Pediatr Dermatol. 2000;17:174–8. 8. Fan X, Xiao FL, Yang S, Liu JB, et al. Childhood psoriasis: a study of 277 patients from China. J Eur Acad Dermatol Venereol. 2007;21(6):762–5. 9. Kwon HH, Na SJ, Jo SJ, Youn JI. Epidemiology and clinical features of pediatric psoriasis in tertiary referral psoriasis clinic. J Dermatol. 2012;39:260–4. 10. Tollefson MM, Crowson CS, McEvoy MT, Maradit KH. Incidence of psoriasis in children: a population-­ based study. J Am Acad Dermatol. 2010;62(6):979–87. 11. Morris A, Rogers M, Fischer G, Williams K. Childhood psoriasis: a clinical review of 1262 cases. Pediatr Dermatol. 2001;18(3):188–98. 12. Wu JJ, Black MH, Smith N, Porter AH, et al. Low prevalence of psoriasis among children and adolescents in a large multiethnic cohort in southern California. J Am Acad Dermatol. 2011;65(5):957–64. 337 13. Stefanaki C, Lagogianni E, Kontochristopoulos G, et al. Psoriasis in children: a retrospective analysis. J Eur Acad Dermatol Venereol. 2011;25(4):417–21. 14. Kurd SK, Gelfand JM. The prevalence of previously diagnosed and undiagnosed psoriasis in US adults: results from NHANES 2003–2004. J Am Acad Dermatol. 2009;60:218–24. 15. Gelfand JM, Weinstein R, Porter SB, Neimann AL, et al. Prevalence and treatment of psoriasis in the United Kingdom: a population-based study. Arch Dermatol. 2005;141(12):1537–41. 16. Radtke MA, Fölster-Holst R, Beikert F, Herberger K, et al. Juvenile psoriasis: rewarding endeavours in contemporary dermatology and pediatrics. G Ital Dermatol Venereol. 2011;146(1):31–45. 17. Eichenfield LF, Paller AS, Tom WL, Sugarman J, Hebert AA, Friedlander SF, Siegfried E, Silverberg N, Cordoro KM. Pediatric psoriasis: evolving perspectives. Pediatr Dermatol. 2018;35(2):170–81. 18. Zhang X, Wang H, Te-Shao H, Yang S, et al. The genetic epidemiology of psoriasis vulgaris in Chinese Han. Int J Dermatol. 2002;41(10):663–9. 19. Icen M, Crowson CS, McEvoy MT, Dann FJ, et al. Trends in incidence of adult-onset psoriasis over three decades: a population-based study. J Am Acad Dermatol. 2009;60(3):394–401. 20. Paller AS, Singh R, Cloutier M, Gauthier-Loiselle M, Emond B, Guérin A, Ganguli A. Prevalence of psoriasis in children and adolescents in the United States: a claims-based analysis. J Drugs Dermatol. 2018;17(2):187–94. 21. Silverberg NB. Update on pediatric psoriasis, part 2: theurapeutic management. Cutis. 2010;86(4):172–6. 22. Smith R. Pediatric psoriasis treated with apremilast. JAAD Case Rep. 2016;2(1):89–91. 23. Silverberg NB. Atlas of pediatric cutaneous biodiversity. New York: Springer; 2012. 24. Silverberg NB. Update on pediatric psoriasis, part 1: clinical features and demographics. Cutis. 2010;86(3):118–24. 25. Schwartz G, Paller AS. Targeted therapies for pediatric psoriasis. Semin Cutan Med Surg. 2018;37(3):167–72. 26. Pourchot D, Bodemer C, Phan A, Bursztejn AC. et all. Nail psoriasis: a systematic evaluation in 313 children with psoriasis. Pediatr Dermatol. 2017;34(1):58–63. 27. Lewkowicz D, Gottlieb AB. Pediatric psoriasis and psoriatic arthritis. Dermatol Ther. 2004;17(4):364–75. 28. Stoll ML, Nigrovic PA. Subpopulations within juvenile psoriatic arthritis: a review of the literature. Clin Dev Immunol. 2006;13(2–4):377–80. 29. Stoll ML, Punaro M. Psoriatic juvenile idiopathic arthritis: a tale of two subgroups. Curr Opin Rheumatol. 2011;23(5):437–43. 30. Stoll ML, Nigrovic PA, Gotte AC, Punaro M. Clinical comparison of early-onset psoriatic and non-­psoriatic oligoarticular juvenile idiopathic arthritis. Clin Exp Rheumatol. 2011;29(3):582–8. 31. Kaushik SB, Lebwohl MG. Psoriasis: which therapy for which patient: focus on special popula- Challenges in Psoriasis Treatment: Nail, Scalp, and Palmoplantar Involvement 26 Jeffrey J. Crowley Abstract Psoriasis can affect many areas of the body and treatments should target the areas of involvement. Nail disease is difficult to treat with topical therapy as the vehicle must be optimized to penetrate the nail and surrounding tissues. Some of the inflammation in nail disease is deep in the nail matrix and thus difficult for topical therapies to access. Scalp disease is difficult to treat with topical and phototherapy due to the presence of hair, bathing habits, and convenience issues. The palms and soles often have particularly thick plaques of psoriasis which may prevent absorption of topical therapy and resist phototherapy. Patients with primarily palmoplantar disease often do not respond to multiple therapies, and many require combination therapy for disease control. Collectively, nail, scalp, and palmoplantar psoriasis are considered “tough to treat” and often do not respond as well as plaque psoriasis elsewhere on the body. This chapter will critically address the challenges posed by each condition and evaluate available treatment options. J. J. Crowley (*) Bakersfield Dermatology and Skin Cancer Medical Group, Bakersfield, CA, USA © Springer Nature Switzerland AG 2021 J. M. Weinberg, M. Lebwohl (eds.), Advances in Psoriasis, https://doi.org/10.1007/978-3-030-54859-9_26 Key Learning Objectives 1. To understand the diagnosis and management of nail psoriasis 2. To understand the diagnosis and management of scalp psoriasis 3. To understand the diagnosis and management of palmoplantar psoriasis Nail Psoriasis Psoriasis is a chronic systemic inflammatory disease involving the skin, nails, and joints. Approximately 50% of psoriasis patients have some nail involvement and the lifetime incidence of nail disease is estimated at 80–90%. Nail disease may rarely be the only manifestation of psoriasis [1]. Nail psoriasis is associated with higher overall disease severity and male gender. Importantly, nail psoriasis can cause significant pain, discomfort, and embarrasment, leading to impairment in quality of life (QoL) and work function [2, 3]. There is a strong correlation of nail psoriasis and psoriatic arthritis [4]. Psoriatic arthritis patients have rates of nail disease as high as 70% and there is evidence that nail psoriasis may be a predictor of joint disease developing later in life [5]. Psoriatic arthritis may involve the distal interphalangeal joints and the anatomic link 343 J. J. Crowley 344 between these joints and the nail unit may result in nail changes. In fact, nail disease is one of the components of the CASPAR criteria, which is used to aid in the diagnosis of psoriatic arthritis [6]. Fingernail psoriasis, due to its visibility and impacts on function, is more problematic for many patients compared with toenail disease. Additionally, psoriatic toenails are often secondarily infected with dermatophytes which may complicate treatment assessment [7]. Treatment with immuosuppressive medications may even contribute to the development of onychomycosis in patients with toenail psoriasis. For these ­reasons, most studies evaluate fingernail psoriasis alone [8]. Clinically, nail disease has many different presentations which depend on the location of the infllammatory process. Nail pitting, the most common finding in nail psoriasis, nail dystrophy, and leukonychia (white discoloration of nails) are due to nail matrix involvement [9] (Fig. 26.1). Nail bed psoriasis is characterized by onycholysis (lifting of the distal nail from the nail bed), oil drop patches (yellow discolorations below the nail), subungal hyperkeratosis (thickening of the nail), and splinter hemorrhages (linear streaks of dried blood in the nail) [1] (Fig. 26.2a, b). One recent study demonstrated that nail clippings of clinically uninvolved nails from patients with psoriasis may show abnormalities, thus subclinical nail disease exists [10]. a The methods of reporting changes in nail disease within clinical trials have not been standardized. The nail psoriasis and severity index (NAPSI) was developed to measure changes in nail disease over time [11]. When utilizing this measure each nail is divided into 4 quadrants and then assessed for the presence or absence of signs of both nail matrix and nail bed disease. Each quadrant of nail with disease present is given a score of “1” for signs of matrix disease and “1” for signs of nail bed disease. A normal nail is scored “0” and the maximum value for each nail is 8, 4 for matrix involvemet and 4 for nail bed disease. Thus, the maximum NAPSI value for a study measuring fingernail disease is 80 and the maxi- Fig. 26.1 Nail matrix psoriasis—note the extensive pitting in this nail. Some of the pits are linear and a splinter hemorrhage is also present. This patient has concomitant psoriatic arthritis and inflammatory bowel disease b Fig. 26.2 (a) Nail bed psoriasis. Note the hyperkeratosis, onycholysis, oil droplet formation in the nail bed, and destruction of the distal nail. (b) Nail bed psoriasis. Note the onycholysis, oil droplet formation, and distal hyperkeratosis 26 Challenges in Psoriasis Treatment: Nail, Scalp, and Palmoplantar Involvement mum for a target nail is 8. NAPSI measures the extent of nail disease but not the severity of nail involvement, so a modified NAPSI (mNAPSI) has also been used as a clinical endpoint in some studies [12]. The mNAPSI has a maximum score of 13 for each nail. Some studies utilize a single target nail or an overall nail severity score. Other scales have been used to measure nail involvement but most of the studies reviewed herein utilize some form of the NAPSI. The wide variety of objective measures used in psoriasis studies to measure nail disease and the variety of time points when the measurements are made, make comparison of treatment outcomes for various interventions difficult. The goal of treating nail psoriasis is total clearance of the nail and given the current highly efficacious therapies, this goal is attainable for some patients. Treatment of Nail Psoriasis An array of treatment options are available for nail psoriasis including topical products, procedural interventions, and oral systemic and biologic agents. The challenges to treating nail disease are many; poor penetration of topical therapy into the nail and surrounding tissue, pain associated with intralesional injections, side effects and monitoring of systemic therapies and patient adherence to therapy [13]. A Cochrane review has recently been published which reviewed the published literature on randomized double-blind placebo controlled trials (RDBPCT) of nail psoriasis [14]. Unfortunately, many of the treatments most commonly used to treat psoriasis do not have published RDBPCT specifically addressing nail disease. Others have reviewed nail psoriasis treatments and the results of a Delphi consensus conference have also been published [15–17]. Topical agents are often the first-line option for treating patients with nail disease. These products are readily available, may have cost advantages, and rarely require laboratory monitoring. Although data are limited and placebo controlled trials rare, there are data to 345 support the efficacy of topical cyclosporine, topical tacrolimus, clobetasol nail lacquer, calcipotriol, calcipotriol plus betamethasone, tazarotene, and indigo naturalis extract [18– 25]. Keratolytics such as urea and salicylic acid are also used in patients with nail psoriasis and, in particular, for nail debridement [26]. Topical preparations, in particular, take several months to show efficacy and adherence to therapy over months is challenging. In a study comparing calcipotriol twice daily with calcipotriol/betamethasone once daily, adherence to the twice daily regimen was only 26% [21]. Therefore, topical therapy may not only be limited by penetration into the nail unit but also by adherence to lengthy treatment regimens lasting weeks or months. In a Cochrane review of topical agents for nail psoriasis the strongest evidence was for the two compound products (calcipotriol and betamethasone dipropionate) although this product is only marginally superior to betamethasone dipropionate alone [27]. Procedures have also been studied in the treatment of nail psoriasis. Phototherapy, a common treatment for skin disease, in the absence of psoralen or a retinoid, is not likely to improve nail disease and is therefore not a viable option for nail psoriasis. There is limited efficacy data showing improvement of NAPSI scores with psoralen plus UVA (PUVA), acitretin plus UVA (Re-PUVA) and acitretin plus UVB (Re-UVB) [28]. In a 1999 study of Grenz ray therapy (superficial x-ray therapy), 22 patients with nail psoriasis had one hand treated with weekly radiation and the other hand serve as an internal control. The treated hand showed significant but moderate improvement but the effect proved more limited with hyperkeratotic nails [29]. Limited access to Grenz ray therapy, a mostly historical therapy in the United States, may further inhibit the use of this modality. Several studies evaluated pulsed dye laser (PDL) in nail psoriasis. The studies noted that therapy is limited by pain, experienced by all patients, and an incidence of both petechiae and hyperpigmentation in one third of patients [30, 31]. A combination of PDL and tazarotene cream was studied in two groups J. J. Crowley 346 of nail psoriasis patients; those on stable doses (10 mg/ml) versus methotrexate (25 mg/ml) of systemic medication and those on no systemic versus cyclosporine (50 mg/ml) showed ≥75% medication. The study showed efficacy in both improvement in over half of the nails injected groups but did not show any difference between with triamcinolone and methotrexate but a low the groups [31]. Another study evaluated PDL response to cyclosporine [35]. both with and without the addition of methyl-­ The systemic agents acitretin, methotrexaminolevulinic acid (photodynamic therapy, ate and cyclosporine, which are effective in PDT) and found no additional benefit of PDT plaque psoriasis, are also effective in nail disover PDL [32]. Intralesional injection of corti- ease. Apremilast, a newer oral agent for psoriasis costeroid is an accepted clinical treatment for which targets phosphodiesterase 4, also has data localized nail psoriasis. Even though this tech- showing efficacy in nail disease. Additionally, nique has been used for many decades, pub- tofacitinib, approved for psoriatic arthritis but lished data to support the safety or efficacy of not for psoriasis, shows efficacy in nail disease. intralesional injections is extremely limited. Appropriate monitoring of both the patient and Injection techniques vary but generally involve the laboratory values pertinent to the systemic injection of 0.1–0.2 ml of 5–10 mg/ml triamcin- therapy should be performed. olone acetonide suspension into the lateral nail Supporting evidence for the efficacy of oral folds [33]. Nerve blocks and/or topical anesthet- systemic therapies is detailed in Table 26.1. ics, to ease the pain associated with injection, Cyclosporine and methotrexate, in a comparamay be performed prior to steroid injection [34]. tor trial, showed a 43.3 and 37.2% mean reducInjections are repeated at various intervals. Side tion in NAPSI, respectively, over a 24 week effects from intralesional steroids include pain on period [38]. The most robust efficacy data with injection, skin atrophy, depigmentation, second- methotrexate is from a randomized double blind ary infection, cyst formation, subungual hemor- trial of methotrexate and the experimental anti-­ rhage, and tendon rupture [34]. Nonetheless, for interleukin-­12/23 antibody briakinumab (a drug a patient with one or two isolated psoriatic nails, no longer being studied). Methotrexate showed a intralesional injection after ring block anesthe- 48% improvement in target nail NAPSI at 1 year sia has been a useful method in the author’s prac- [36]. In a 6 month open label trial of acitretin for tice. A trial comparing intra-matrix triamcinolone nail psoriasis there was a 41% mean reduction Table 26.1 Efficacy of systemic therapies in nail psoriasis Agent Methotrexate Dose 15–20 mg/ weeks Length 52 weeks Patients (n) 317 Acitretin 24 weeks 36 Cyclosporine 0.2–0.3 mg/ kg/day 5.0 mg/kg/day 24 weeks 37 Apremilast 30 mg bid 52 weeks 558 Tofacitinib 5 mg bid 10 mg bid 16 weeks 487 476 Trial type Randomized, double blind, controlled trial (no placebo) Open-label Comparator (vs. methotrexate) Randomized, double blind, placebo controlled phase III trials, subanalysis of patients with baseline nail disease Randomized, double blind, placebo controlled phase III trials, subanalysis of patients with baseline nail disease NAPSI improvement 48% References [36] 41% [37] 43.3% [38] 60.2% (ESTEEM 1) 59.7% (ESTEEM 2) [39] 16.9% (NAPSI-75) 28.1% (NAPSI-75) [40] 26 Challenges in Psoriasis Treatment: Nail, Scalp, and Palmoplantar Involvement in NAPSI [37]. Apremilast, FDA approved for psoriasis and psoriatic arthritis, has been studied in nail disease. In the phase III ESTEEM 1 and 2 studies, 66.1 and 64.7% of patients had nail psoriasis (NAPSI ≥ 1). Mean percent change in target NAPSI score from baseline was −22.5 and −29% at week 16 and −43.6 and −60% at week 32 for ESTEEM 1 and 2 respectively [41]. In a post-hoc analysis of two phase III clinical trials of tofacitinib in psoriasis, NAPSI was evaluated for patients with nail disease. NAPSI75 was achieved by 16.9, 28.1, and 8.8% at 16 weeks in the 5 mg bid, 10 mg bid and placebo groups respectively. Continued tofacitinib use to 52 weeks demonstrated additional improvement in nail disease [40]. These agents may be an excellent option for a patient who is otherwise a candidate for systemic therapy for psoriatic ­disease. Combination treatment for nail psoriasis, though widely used, is little studied. A 2004 study of oral cyclosporine and topical calcipotriol showed that at 12 weeks the combination group showed 79% of patients with improvement compared with 47% of patients on cyclosporine alone [42]. Combination therapy with topical agents and systemic therapy is frequently employed in clinical practice and is likely safe and may be more effective than systemic therapy alone. Table 26.2 reviews the data on biologics in nail psoriasis. In a rare trial of a biologic, designed specifically to target patients with nail psoriasis, adalimumab showed a modified NAPSI 75 response at 26 weeks in patients with and without psoriatic arthritis of 61.5 and 40.9% respectively (placebo 0.5 and 4.6%) [50]. Adalimumab, in a RPCDBT of patients with psoriasis involving the hands and/or feet, demonstrated 50% improvement in NAPSI at 16 weeks compared with 8% for placebo [43]. In a study evaluating both fingernails and toenails, open label adalimumab showed improvements at 6 months in NAPSI of 85 and 72% in fingernails and toenails respectively [51]. An open label study of etanercept in nail psoriasis compared patients randomized to 2 dosing regimens; 50 mg twice weekly for 12 weeks followed by 50 mg weekly for 12 weeks, or 50 mg weekly for 24 weeks. A 50% improvement in NAPSI was demonstrated in 347 58.1 and 82.3% of patients in the twice weekly/ weekly group and 50.5 and 80.7% in the weekly group at 12 and 24 weeks respectively [44]. In a RDBPCT patients with psoriatic arthritis were treated with placebo, golimumab 50 mg every 4 weeks, or golimumab 100 mg every 4 weeks for 24 weeks. Median percent change in NAPSI at weeks 14 and 24 was 0%, 25%, 43%, and 0%, 33%, and 54% for the placebo, golimumab 50 mg, and golimumab 100 mg groups respectively. Patients in this trial for psoriatic arthritis were allowed to use stable doses of methotrexate and prednisone during the study [45]. Currently golimumab is FDA approved for psoriatic arthritis but not for psoriasis. In a RDBPCT of infliximab, patients were randomized to either placebo or infliximab 5 mg/kg at weeks 0, 2, 6, and every 8 weeks through week 46, with placebo crossover at week 24. Mean percentage improvement in target nail NAPSI at week 10 and 24 was 26.8 and 57.2%, compared with −7.7 and −4.1% for infliximab and placebo respectively [52]. In a retrospective analysis of patients from this same infliximab study, mean NAPSI improvement was 28.3, 61.4, and 67.8%, at weeks 10, 24 and 50 respectively [46]. No controlled comparisons of anti-TNF agents in nail psoriasis have been performed. However, an open label prospective study showed that infliximab was superior to adalimumab and etanercept at 14 weeks [53]. A retrospective comparison of these agents also showed high efficacy of all the TNF blocking agents in nail psoriasis with some greater improvement with infliximab [54]. The differences in efficacy of these agents in nail psoriasis may parallel the differences seen in skin response. Infliximab may also have an advantage in the speed to which clearance is achieved. However, after 4 months or more, all anti-TNF therapies are successful in improving nail disease by at least 50%, as measured by NAPSI. Robust data is also available for IL-12/23 blockade in nail psoriasis. Data from a large phase III trial of ustekinumab showed significant improvement in NAPSI compared with placebo at 12 weeks. Additionally, the ustekinumab 45 and 90 mg groups showed 46.5 and 48.7% NAPSI J. J. Crowley 348 Table 26.2 Efficacy of biologic therapies in nail psoriasis Agent Adalimumab Dosing 80 mg/40 mg eow Etanercept 50 mg biw/50 mg qw 100 mg q4 weeks Golimumab Patients (n) Trial type 72 RPCDBT in hand/ foot psoriasis 711 405 Randomized dose comparison RPCDBT in psoriatic arthritis Infliximab 5 mg/kg weeks 0,2, and 6 then q8 weeks 378 RPCDBT, data from the open label extension Ustekinumab 45 or 90 mg at weeks 0, 4, 16 545 RDBPCT Secukinumab 150 or 300 mg at weeks 1–5 then q4 weeks 198 Ixekizumab 80 mg q2 or q4 weeks for 12 weeks then 80 mg q4 weeks 1346 RDBPCT of patients with nail disease, data from the open label extension RDBPCT, retrospective analysis of open label period Outcomes (note different primary outcome References Weeks measures) [43] 16 50% reduction in NAPSI (8% for placebo) 24 82.3% NAPSI 50 [44] response [45] 24 54% reduction in NAPSI (0% for placebo) [46] 50 67.8% reduction in NAPSI (49.2% with complete nail clearance) 24 46.5% (45 mg), 48.7% [47] (90 mg) reduction in NAPSI [48] 32 52.6% (150 mg) and 63.2% (300 mg) reduction in NAPSI 24 34 and 30% with no nail involvement (NAPSI = 0) on q2 and q4 doses, respectively [49] eow every other week, NAPSI nail area psoriasis severity index, RDBPCT randomized double-blind placebo controlled trial improvement, respectively, at 24 weeks [47]. Long-term data with 68 weeks of ustekinumab in a Japanese study showed a 56.6 and 67.8% improvement from baseline NAPSI in patients treated with 45 or 90 mg of ustekinumab respectively [55]. Antibodies to interleukin 17-A and an interleukin 17 receptor antagonist are currently approved for psoriasis. Data from a phase II dose finding trial of the anti-interleukin 17 antibody ixekizumab showed improvement of 57.1 and 49.3% in NAPSI scores at the 75 and 150 mg doses respectively at 16 weeks [56]. Further data with ixekizumab from the UNCOVER 3 phase III clinical trial has also been published [49]. At week 12, patients with baseline nail involvement showed reduction in NAPSI of 39, 40, 28, and −4.7% in the ixekizumab every 2 weeks, ­ixekizumab every 4 weeks, etanercept 50 mg twice weekly, and placebo groups, respectively. Additionally, at 24 weeks, 34 and 30% of patients had no nail involvement on ixekizumab q2weeks/q4 weeks, and ixekizumab q4 weeks/q4 weeks, respectively. At 60 weeks, half of the patients with initial nail disease had a NAPSI = 0 while on continuous ixekizumab. Secukinumab, another antibody to IL-17A, was studied in a clinical trial specifically designed to study patients with nail psoriasis. In the TRANSFIGURE study, mean improvement of NAPSI from baseline was 10.8, 37.9, and 45.3 at week 16 for placebo, secukinumab 150 mg, and secukinumab 300 mg, respectively. At week 32, in the open label period, a 52.6 and 63.2% improvement in mean NAPSI was seen for secukinumab 150 and 300 mg respectively [48]. Currently in development for psoriasis and psoriatic arthritis is bimekizumab which targets both IL-17A and IL-17F and data on efficacy in nail disease will be forthcoming [57]. The rapid onset of action and high levels of efficacy make these anti-IL-17 agents particularly attractive for patients with extensive nail disease. 26 Challenges in Psoriasis Treatment: Nail, Scalp, and Palmoplantar Involvement Treatment Approach to Nail Disease A treatment algorithm for nail psoriasis, based on published data for treatment of nail psoriasis and expert opinion where data is lacking, has been previously published by the Medical Board of the National Psoriasis Foundation [58] and is summarized here. All psoriasis patients should be evaluated for nail disease and the extent to which their nail disease contributes to overall disease burden. Nail disease should be classified as mild if it has minimal impact on QoL and poses no functional impact for the patient. Significant or extensive nail disease has real impact on daily activities, may be disfiguring, and may be associated with significant pain. Patients with significant nail disease need therapies to address their nail psoriasis. Patients with moderate to severe psoriasis and significant nail disease should be treated with appropriate therapy which addresses skin and nail disease. Previous consensus guidelines for the treatment of moderate to severe psoriasis also apply to the patient with nail disease [59]. Options include cyclosporine, methotrexate, acitretin, apremilast, and the biologics. Patients with significant nail disease and psoriatic arthritis (PsA) should be treated with an appropriate systemic treatment for PsA that has efficacy in nails. Data support the following options in this patient group; methotrexate, apremilast, the TNF inhibitors, IL-17 inhibitors and the IL-12/23 inhibitor. For the rare patient with nail disease as the primary manifestation of their psoriatic disease, a 3–6 month trial of topical therapy and/or intralesional injections may be appropriate. For patients with severe nail disease affecting QoL, systemic and biologic therapies may be needed. Patients who have their skin and/ or joint disease well controlled but still have significant nail involvement may need combination topical therapy or intralesional injections. If this fails, a change in the systemic or biologic therapy may be warranted. Improvements in nail psoriasis often trail the improvements in skin and joint disease. Fingernails grow at a rate of 3–4 mm/month, thus it takes 5–7 months for a nail to grow from matrix to distal fingertip [60]. Therefore, the 349 length of a clinical trial must reflect this delay. Few studies, with the exception of the anti-IL 17 inhibitors and infliximab, demonstrate any significant improvement before 12 weeks and several studies with adalimumab, etanercept, infliximab, ustekinumab, ixekizumab, and scukinumab demonstrate continued improvement up to and even beyond 6 months [44, 46, 53, 61]. One of the problems in reviewing data on nail psoriasis is the lack of consistent outcome reporting among the studies. Even when NAPSI data is presented, it may be presented in different ways. Target nail NAPSI, mean improvement in NAPSI, percent of patients with no nail disease (NAPSI = 0), and modified NAPSI, are some of the many outcomes reported. The dermatology life quality index (DLQI) contains some questions pertinent to nail signs and symptoms but does not specifically account for the influence of nail disease on QoL. Some researchers have used QoL measures validated in onychomycosis and applied these to psoriasis [62]. A validated measure specific to nail psoriasis, the NPQ10, has been published but not extensively utilized [63]. Additionally, the Nail Assessment in Psoriasis and Psoriatic Arthritis (NAPPA) may prove useful in evaluating response to therapy in nail disease [64]. Clearly there is a need for more uniform reporting of nail outcomes and adoption of a valid and easily performed QoL metric that is specific to nail disease. Scalp Psoriasis Is scalp psoriasis difficult to treat? Indeed, it may respond faster than other body regions to some biologic therapies but it remains a challenge to obtain good results with both topical and phototherapy. While the head and neck represent about 10% of the body surface area, the impact of psoriasis in this region may be disproportionate to the area, and may have social and emotional impacts on affected individuals. Most scalp psoriasis is treated with topical therapies including shampoos, oils, foams, liquids, and gels. Over the counter remedies, containing tar, salicylic acid, J. J. Crowley 350 zinc, and others are relatively inexpensive and widely available. One of the reasons that scalp psoriasis has been labeled “tough to treat” is the difficulty with using and complying with topical medications. Prescription topical therapy mainly involves the use of mid to high potency topical steroids and topical vitamin D analogues. Many larger trials for the biologics have had measures of scalp psoriasis as secondary endpoints. A few trials have been designed specifically to assess scalp disease and these will be highlighted in this chapter. Indeed, scalp psoriasis provides challenges to both practitioner and patient. The incidence of scalp psoriasis in patients with psoriasis is estimated between 40 and 90 percent. In up to half of patients, psoriasis may initially present on the scalp [65, 66]. Some patients only have psoriasis on the scalp. The scalp is characterized by the presence of the prominent pilosebaceous unit and has a microbiome that differs from other skin sites. Both yeast and bacteria colonize the scalp [67]. Indeed Malessazia furfur, M. globosa, candida, and other commensal organisms are found on the scalp in significant numbers and may influence scalp diseases such as seborrheic dermatitis and psoriasis [68, 69]. The role of these organisms in psoriasis is not well established but they likely play a role. Treatments targeting these organisms have shown some efficacy in seborrheic dermatitis [70, 71]. Friction and trauma to the scalp from scratching and hair grooming my also contribute to scalp disease severity [72, 73]. Recent studies have shown that the hair in patients with scalp psoriasis may also be modified by the presence of psoriatic inflammation. Indeed, hair shafts evaluated in patients with psoriasis reveal pits, thought to be analogous to the pitting seen in nails [74]. Additionally, recent studies evaluating the transcriptome of both scalp and body psoriasis suggest there may be differences in gene activation between the scalp and body [75]. ­ ell-­defined plaques with silvery scale and an w erythematous border (Fig. 26.3). Classic scalp lesions are asymmetric, sharply demarcated, covered with silvery-white or gray scale, and may extend beyond scalp margins to affect the forehead, ear, and neck [76]. Most patients with psoriasis of the face also have scalp disease [77]. Multiple surveys have cited itching and scaling as the most disturbing aspects of scalp psoriasis [78, 79]. This itch may be so severe that it can interfere with sleep and lead to actual hair loss due to hair breakage and trauma. Scalp psoriasis is usually not associated with significant hair loss, however, some patients may have alopecia due to the trauma of chronic itching leading to hair breakage. In an analysis of 47 patients with psoriatic alopecia followed for several years, many patients only had alopecia in some of the scalp plaques and indeed, as the psoriasis cleared, the hair regrew [80]. In some cases the differentiation of scalp psoriasis and seborrheic dermatitis can be difficult. Seborrheic dermatitis is characterized by diffuse thin scale that is localized to the hair bearing scalp and may involve the central face and chest. Psoriasis, on the other hand, may extend beyond the scalp, is sharply marginated, and may occur elsewhere on the body [81]. Measuring Scalp Disease A variety of measures have been developed to assess the extent and burden of scalp psoriasis linical Presentation of Scalp C Psoriasis Scalp lesions may vary from mild, with minimal erythema and scaling, to severe with thick Fig. 26.3 Scalp psoriasis 26 Challenges in Psoriasis Treatment: Nail, Scalp, and Palmoplantar Involvement 351 Table 26.3 Measures of scalp psoriasis Measure Psoriasis Scalp Severity Index (PSSI) Scalp-modified PASI (S-mPASI) Parameters measured Extent of involvement, erythema, induration, and desquamation of scalp Extent of involvement, erythema, induration, desquamation of scalp Head and Neck PASI (HN-PASI) Extent of involvement, erythema, induration, desquamation of head and neck Erythema, induration, desquamation of scalp Overall scalp disease, as judged by the patient, as compared to baseline involvement Overall scalp disease measured by investigator Measure of symptoms, functioning, and emotions on scalp disease (23 items) Total Severity Score (TSS) Scalp-specific Patient’s Global Assessment (SPaGA) Global Severity Score (GSS) Scalpdex Max. score Description 72 Sum of 3 parameter scores (0–4) multiplied by a score for the area of involvement (1–6) 7.2 Sum of 3 parameter scores (0–4) multiplied by a score for the area of involvement (1–6) times a constant (0.1) 7.2 Sum of 3 parameter scores (0–4) multiplied by a score for the area of involvement (1–6) times a constant (0.1) 9 Sum of scores (0–3) for erythema, induration, and desquamation −2 to +2 Current scalp disease severity compared with baseline visit on a 5-point scale; “much worse” (−2) through “much improved” (+2) 5 Assessment of scalp disease from “none” (0) to “very severe” (5) 0–100 Measures may be combined to form subscores for symptoms, functioning, and emotions References [82] [83] [84] [85] [83] [85] [78] (Table 26.3). The Psoriasis Scalp Severity Index issue for many scalp psoriasis patients, may be (PSSI) and the Scalp-modified PASI (S-mPASI) measured by a visual analogue scale [87] of itch. are modifications of the standard PASI measure- Scalpdex is a validated measure of quality of life ment of psoriasis [82, 83]. Both the PSSI and the for scalp conditions (not specific to psoriasis) Sm-PASI measure erythema, induration, and des- that utilizes 23 questions that address symptoms, quamation of the plaques of psoriasis on the scalp emotions, and functions surrounding the scalp only in contrast to PASI where the entire body is condition [78]. evaluated. Some studies, including one on the IL-17 inhibitor secukinumab, have utilized the head and neck portion of the PASI score Management of Scalp Psoriasis: (HN-PASI) as a proxy for scalp involvement Topical Therapies [84]. Other measures have also been used for evaluation of scalp psoriasis including the Total Topical therapies are the foundation in the manSeverity Scale (TSS), the Scalp-specific agement of scalp psoriasis and utilized by Physicians Global Assessment (S-PGA), and the approximately 60% of patients [66]. These theraGlobal Severity Scale (GSS) [85]. A recent study pies have been formulated into shampoos, gels, of 102 patients with predominately scalp psoria- foams, oils, and solutions. Some of these prodsis treated with secukinumab or placebo utilized ucts have been evaluated in large clinical trials. a 90% improvement of PSSI and a 2 point High costs limit the use of many of these propriimprovement in Investigators Global Assessment etary products. Calcipotriol is a synthetic derivaas the two primary endpoints to the study [86]. tive of calcitriol (Vitamin D3); calcipotriol binds Patient assessment tools have also been devel- to the vitamin D3 receptor, but unlike calcitriol, oped to evaluate scalp psoriasis. The Scalp-­ is a poor regulator of calcium metabolism. The specific Patient’s Global Assessment (S-PaGA) efficacy and safety of calcipotriol solution suphas been utilized to gauge patient assessment of port its use as a first-line therapy as well as its use disease change from baseline. Pruritus, a major as maintenance therapy [88, 89]. This treatment, J. J. Crowley 352 however, can be associated with some burning, redness, dryness, and itching in some patients. The combination of calcipotriol and topical steroids decreases some of the irritation of calcipotriol and is certainly more efficacious than calcipotriol monotherapy. There may also be an advantage to this combination due to the potential protective effect of calcipotriol on corticosteroid induced skin atrophy. Shampoos and foams containing high potency topical steroids such as clobetasol propionate 0.05% have demonstrated improvement in scalp psoriasis in as little as 2 weeks. In these short, 2–4 week studies, skin atrophy, folliculitis and telangiectasia were not observed [90, 91]. Clobetasol propionate 0.05% shampoo was superior in efficacy and tolerability compared with calcipotriol 0.005% solution [85]. In a study comparing 1% tar blend shampoo with clobetasol shampoo, the clobetasol product was cosmetically more acceptable to patients [91]. Betamethasone valerate 0.12% in a mousse (foam) vehicle was compared to topical corticosteroid solution and calcipotriol lotion and showed superior efficacy over 4 weeks. Most of these patients preferred the foam formulation [88]. An open label study of the combination product (betamethasone/calcipotriene) in patients 12–17 years of age showed efficacy over 8 weeks with one patient demonstrating mild adrenal suppression and no patients demonstrating hypercalcemia [92]. The authors concluded that this treatment was safe and efficacious in adolescents with scalp psoriasis. A 2016 Cochrane review of topical therapies reviewed 59 randomized controlled clinical trials in scalp psoriasis [93]. They concluded that a corticosteroid of high or very high potency or a combination of a corticosteroid and vitamin D (calcipotriol) was superior to vitamin D alone. The two-compound combination also led to fewer withdrawals from treatment compared to the other treatments. Further the authors concluded “Given the comparable safety profile and only slim benefit of the two-compound combination over the corticosteroid alone, monotherapy with generic topical steroids of high and very high potency may be fully acceptable for short term therapy.” However, do patients adhere to treatment with topical therapy? There is some evidence that optimizing the vehicle may improve adherence to scalp psoriasis treatments, however adherence to topical therapy for psoriasis is low [94]. Even with interventions to assist and remind patients to apply topical therapies, adherence is less than 50% after several weeks [95]. In clinical practice, the use of anti-dandruff shampoos is generally encouraged by dermatologists to help remove the scale from the plaques of psoriasis. The use of high potency topical steroids is usually limited to a few weeks but patients may continue to use these products intermittently for months and even years. The use of topical vitamin D products, when available and when well tolerated, is also a reasonable approach to maintenance therapy. Many practitioners will utilize a high potency topical steroid 1 or 2 days per week and a topical vitamin D analogue on the other days in an effort to minimize potential side effects of long term topical steroid use [96]. Procedural Therapies for Scalp Psoriasis Intralesional injection of corticosteroids has also been used by dermatologists to address localized treatment resistant plaques of psoriasis with some success. Triamcinolone acetonide (5–10 mg/ml) is injected directly into the psoriatic plaques in small (0.1–0.2 ml) aliquots. The aim is to deliver the steroid directly into the area of inflammation in the dermis. This technique, however, has not been studied in a rigorous fashion [34]. Phototherapy for scalp psoriasis is limited due to hair blocking the penetration of the light. Devices have been designed and are available to help deliver phototherapy to the scalp. These devices use fiber-optics that penetrate through the hair and deliver the phototherapy, broadband or narrow-band UVB, directly to the scalp [97]. Additionally, the 308-nM laser can be used to treat scalp psoriasis. In one study, 17 of 35 patients had >95% clearance after a mean of 21 treatments [98]. In this study the hair was manually parted to gain access to the psoriatic plaques. 26 Challenges in Psoriasis Treatment: Nail, Scalp, and Palmoplantar Involvement Notably, all patients demonstrated erythema and some blistering to treated sites. In another study, patients treated with the laser administered with a blow dryer to part the hair, had significant improvement after twice weekly treatments for up to 15 weeks [99]. Grenz ray therapy has also been efficacious in scalp psoriasis. The lack of availability of Grenz ray therapy units and the potential for skin cancer formation in the treated area significantly limit this modality [100, 101]. Systemic Therapy for Scalp Psoriasis There are few studies that specifically address the efficacy of methotrexate, cyclosporine, and acitretin in scalp psoriasis. Nonetheless, these therapies may be effective in treating scalp disease. Appropriate monitoring of liver function (methotrexate, acitretin), creatinine (cyclosporine, methotrexate), complete blood counts (methotrexate), triglycerides (acitretin), and blood pressure (cyclosporine) should be maintained during treatment with these agents. 353 Apremilast, an oral phosphodiesterase 4 inhibitor, is approved for psoriasis and psoriatic arthritis. Scalp disease was evaluated as a secondary endpoint in the two phase III psoriasis programs for apremilast. Scalp disease was moderate or greater in about two thirds of patients in these trials. Approximately half of those with baseline moderate scalp involvement achieved a clear or almost clear PGA at 16 weeks. Most patients who continued in trial maintained this response out to 1 year [39, 41]. Apremilast may be a reasonable option for patients with significant scalp disease. Biologic Therapy for Scalp Psoriasis The treatment of scalp psoriasis with biologics is summarized in Table 26.4. There have been some studies specifically designed to evaluate scalp psoriasis. Etanercept, a TNF-fusion protein that binds TNF-alpha, was used in a study specifically addressing patients with scalp disease [102]. In this placebo-controlled trial, PSSI at week 12 Table 26.4 Efficacy of systemic therapies in scalp psoriasis Agent Apremilast Dosing 30 mg biw Etanercept 50 mg biw or placebo Adalimumab 80 mg weeks. 0, then 40 mg eow Secukinumab 300 mg weeks. 0–4, then 300 mg q4 weeks 80 mg weeks. 0, then 80 mg q2 or q4 weeks, also comparator arm with etanercept biw Ixekizumab Patients (n) Trial type 558 RPCDBT, subanalysis of patients with moderate scalp involvement 124 Placebo controlled study in moderate scalp disease 730 RPCDBT, subanalysis of patients with baseline scalp disease 102 RPCDBT in patients with scalp psoriasis 3866 RDBPCT with etanercept comparator arm Outcomes (note different primary outcome References Weeks measures) [41] 16 Scalp PGA of clear or almost clear (0 or 1) of 46.5% (ESTEEM 1) and 40.9% (ESTEEM 2) [102] 12 Improvement in PSSI from baseline 86.8% (vs. 20.4% for placebo) 16 77.2% improvement in [103] PSSI 12 12 52.9% PSSI improvement (2% for placebo) PSSI 100 response of 74.6, 68.8, 48.1, and 6.7% in Ixe q2w, Ixe q4 wk, etanercept, and placebo respectively [86] [104] biw twice weekly, eow every other week, RPCDBT randomized placebo-controlled double blind clinical trial, PGA physician’s global assessment, PSSI psoriasis scalp severity index J. J. Crowley 354 was improved by 86.8 and 20.4% in the etanercept 50 mg biw group and placebo group respectively. Improvement continued in the open label period to week 24. In a subanalysis of a larger trial of adalimumab in psoriasis, PSSI was improved by an median of 100% (mean 77.2%) at 16 weeks [103]. Blockade of interleukin-17 has also been studied in scalp psoriasis. In a placebo controlled study designed specifically for scalp disease patients, secukinumab 300 mg achieved 90% improvement in PSSI in 52.9% of patients compared with 2% on placebo at 12 weeks. Investigators global assessment of clear or almost clear was achieved by 56.9% on secukinumab and 5.9% on placebo at 12 weeks [105]. In a subanalysis of larger phase III trials for ixekizumab, scalp psoriasis was evaluated [104]. Of patients with baseline scalp psoriasis, PSSI 90 and PSSI 100 at 12 weeks were obtained by 81.7 and 74.6, 75.6 and 68.8, 55.5 and 48.1, and 7.6 and 6.7, in the ixekizumab q2 weeks, ixekizumab q4 weeks, etanercept 50 mg biw, and placebo groups, respectively. Thus, more than half of patients on etanercept, and over three quarters of patients on ixekizumab had complete clearance of scalp psoriasis at 12 weeks. reatment Algorithm for Scalp T Psoriasis For limited scalp disease, treatment with potent topical steroids with or without vitamin D derivatives is a reasonable initial treatment. This may be augmented by OTC shampoos to remove scale. For patients who fail to respond to a trial of topical therapy, phototherapy (if available), and oral treatment with methotrexate or apremilast may be appropriate. For patients who prove recalcitrant to these therapies or for patients with significant psoriasis elsewhere, biologic therapies that target TNF or interleukin-17 are recommended. TNF Induced Psoriasis Paradoxically, treatment with anti-TNF agents may induce scalp psoriasis in patients who have not had a previous history of psoriasis [106, 107]. These cases have been reported with all the anti-­ TNF agents and the patients are often being treated for rheumatoid arthritis and inflammatory bowel disease. The scalp, palms, soles, and other body areas may be involved. Occasionally, these patients may have lesions consistent with pustular psoriasis [107]. Some patients can be treated with topical or phototherapy and remain on the TNF agent while others may require discontinuation of the TNF inhibitor to control the eruption. The incidence of this eruption is between 1 and 2% of patients treated with anti-TNF agents [108]. The cause of this phenomenon is not well elucidated but may involve compensatory increases in interferon in patients treated with anti-TNF’s [109]. Palmoplantar Psoriasis Psoriasis of the hands and feet may be one of the most difficult therapeutic dilemmas for the dermatologist. Palmoplantar psoriasis affects up to 5% of patients with psoriasis [110]. First, however, we need to define the spectrum of psoriatic disease that can affect the palms and soles. There are several distinct presentations of psoriasis on the palms and soles: • Plaque psoriasis of the hands and feet with significant psoriasis elsewhere (Fig. 26.4) • Plaque psoriasis of the hands and feet with little psoriasis elsewhere (palmoplantar psoriasis, PPP, Fig. 26.5) • Pustular psoriasis of the hands and feet (palmoplantar pustulosis, PPPP, Fig. 26.6). This may also be called Acrodermatitis Continua of Hallopeau, particularly when it involves bony resorption of the distal phalanx [111]. • Psoriasiform dermatitis of the hands and feet—with features of hand dermatitis and psoriasis These presentations represent distinct patient populations that may respond differently to various therapies and may have differing genetic and environmental factors involved in their disease 26 Challenges in Psoriasis Treatment: Nail, Scalp, and Palmoplantar Involvement Fig. 26.4 Palm psoriasis in a patient with chronic plaque psoriasis Fig. 26.5 Severe palmoplantar plaque psoriasis in a patient with minimal psoriasis on body Fig. 26.6 Palmar pustular psoriasis. Note resorption of distal digits and extensive pustules. These are features of a variant of pustular psoriasis termed acrodermatitis continua of Hallopeau development. This review will focus on plaque psoriasis of the hands and feet (PPP) and pustular psoriasis of the hands and feet (PPPP). 355 Recently, pustular psoriasis (particularly the generalized form) has been associated with mutations in the IL-36 receptor [112]. There is also a small clinical trial demonstrating that an antibody that targets the IL-36 receptor is effective in treating generalized pustular psoriasis [113]. Some PPPP patients have mutations in the IL-36 receptor but other genes are also implicated including CARD14 and AP1S3. Notably, patients with PPPP do not have mutations in PSORS1 which is common in patients with plaque psoriasis [114]. These genetic associations may provide therapeutic targets to address PPPP in the future. A key factor in palmoplantar disease is disability. The use of the hands and feet may be significantly compromised by disease. The scale, fissures, cracking, and hyperkeratosis can lead to pain with movement. Chung et al. evaluated the quality of life (QoL) of patients with PPP and compared them with plaque psoriasis [115]. These patients were receiving treatment with systemic or phototherapy. The palmoplantar psoriasis patients had significantly more impairment in QoL characterized by limitations of mobility, self-care, and pursuing usual activities. Additionally, palmoplantar patients were more likely to be taking multiple topical and oral therapies for their disease. Others have reported on physical disability and discomfort associated with PPP [116]. Smoking appears to play a role in palmoplantar disease and there is some data suggesting that pustular disease severity may improve with smoking cessation [117]. There are few controlled clinical trials addressing treatments for palmoplantar disease. Most treatment has been based on case series, small clinical trials, and anecdotal clinical evidence. If you were to ask 10 dermatologists for their top five treatments for hand and foot psoriatic disease, you would get dramatically different answers. Reviews of the literature have also yielded differing recommendations for treatment of palmoplantar psoriasis. Seravin et al. concluded in 2013 “Phototherapy, cyclosporine and topical corticosteroids seem to be able to control PPPP. However, the standard of care for PPPP remains an issue and there is a strong need for reliable RCTs to better define treatment strategies for 356 PPPP” [118]. The National Psoriasis Foundation concluded in a 2012 review of treatments for pustular psoriasis that “acitretin, cyclosporine, methotrexate, and infliximab are considered to be first line therapies in generalized pustular disease” [119]. All sources conclude that better data is needed to guide clinical decisions regarding treatment. Topical steroids, Vitamin D analogues, tar and anthralin preparations, either by themselves or in combination, and under occlusion have all been reported to improve psoriasis and are regularly used in palmoplantar disease. In a comparative trial, in PPP, of clobetasol plus tar compared with topical psoralen plus UVA, both treatments were effective [120]. Phototherapy with psoralen plus UVA (PUVA), and narrowband UVB, have shown some efficacy in hand and foot disease [121]. Multiple small studies and case series have demonstrated the efficacy of the 308 nM laser in the treatment of palmoplantar disease [122–124]. A recent comparison of paint PUVA (psoralen not ingested but instead painted on the skin) and broad band UVB showed improvement with both treatments but more complete and longer remissions with paint PUVA [125]. Another study used a split hand design to treat one side with narrowband-UVB and the other with topical PUVA. The PUVA side fared better than the hand treated with narrowband UVB [126]. Thus, topical therapies and phototherapy may be beneficial in treating palmoplantar psoriasis. Systemic treatments for palmoplantar disease have also been studied. Methotrexate (0.4 mg/ kg weekly) and acitretin (0.5 mg/kg daily) were studied in an active comparator trial of 111 PPP patients [127]. Both treatments showed improvement but methotrexate significantly improved more patients. Wald et al. published a series of 48 hand foot psoriasis patients treated with methotrexate, most in combination with other therapies, and concluded that methotrexate was effective in PPP [128]. In a study of cyclosporine in PPPP, most patients improved on relatively low doses (1–2 mg/kg/day) of cyclosporine [129]. In a pooled analysis of phase II and two phase III trials of apremilast, a phosphodiesterase 4 inhibitor, in psoriasis, patients with baseline palmoplantar physicians global assessment (PP-PGA) of moderate or greater were evaluated during J. J. Crowley the 16 week placebo controlled period. At week 16, 46 and 25% of patients showed a PP-PGA of clear or almost clear for the apremilast and placebo groups, respectively [130]. Thus, acitretin, cyclosporine, and apremilast all demonstrate some efficacy in PPP and/or PPPP. However, there are no randomized placebo controlled studies specifically addressing these systemic agents in PPP or PPPP. The biologics have also been studied in PPP and PPPP. Adalimumab was studied in patients with at least moderate plaque psoriasis of the hands and feet and moderate to severe plaque psoriasis elsewhere [43]. In this placebo c­ ontrolled trial, 31% of patients in the adalimumab group were able to achieve a hand/foot PGA of clear or almost clear at week 16, compared to only 4% in the placebo group. In a study of 24 palmoplantar psoriasis patients randomized to placebo or infliximab infusions, more patients reached 75% improvement in palmoplantar PASI in the infliximab group (33.3 vs. 8.3%), but this did not reach statistical significance [131]. Success with treating PPP and PPPP has also been demonstrated in case reports with etanercept [132, 133]. The TNF inhibitors do appear to show some efficacy in treating PPP, but this efficacy is significantly attenuated compared with overall plaque psoriasis results. Ustekinumab, in an open label study of patients with PPP and PPPP, cleared 35% (7/20), however the 90 mg dose was superior to 45 mg [134]. The interleukin-17 inhibitors have been studied in both PPP and PPPP. In a subanalysis of the large phase III trials of ixekizumab in psoriasis, patients with significant palmoplantar disease (PP-PASI of ≥8) at baseline were evaluated [135]. This analysis involved 105 patients (total study n = 1224) and revealed PP-PASI 75 and PP-PASI-100 results of 16.7, 44.1, 81.8, 74.2 and 5.6, 29.4, 45.5, and 51.6 in the placebo, etanercept 50 mg biw, ixekizumab q2 weeks, and ixekizumab q4 weeks, respectively. Thus, roughly half of the ixekizumab treated patients achieved total clearance of their plaque psoriasis of the hands and feet. Secukinumab was studied in a placebo controlled trial of patients with moderate to severe palmoplantar psoriasis [136]. Many of these patients would not have had 26 Challenges in Psoriasis Treatment: Nail, Scalp, and Palmoplantar Involvement enough ­psoriasis to meet enrollment criteria for typical phase III psoriasis trials and thus may better represent the vexing patients with primarily palmoplantar disease. 205 patients were randomized 1:1:1 to secukinumab 300 mg, secukinumab 150 mg, and placebo. In this study, palmoplantar IGA of clear or almost clear at week 16 was achieved by 33.3, 22.1, and 1.5% of patients treated with secukinumab 300 mg, secukinumab 150 mg, and placebo respectively. These results are impressive but are not as robust as seen in the larger phase III trials of plaque psoriasis. An additional study of secukinumab in PPPP was also performed [137]. In this study 237 patients with palmoplantar pustulosis (PPPP) were randomized to placebo, secukinumab 150 mg, or secukinumab 300 mg. Forty percent of these patients did not have plaque psoriasis elsewhere. Change in PP-PASI was measured at week 16 and 29.7, 30.2, and 42.3% improvement was noted in the placebo, secukinumab 150 mg, and secukinumab 300 mg, respectively. Thus, there was minimal response of PPPP to IL-17 inhibition. More studies are needed to confirm this finding but it does appear that IL-17 is not a key target for PPPP. PPP remains a difficult disease to treat. Multiple therapies and often necessary, and a trial of many treatments is often needed in order to achieve adequate control. There are some data to support topical therapy, phototherapy (especially PUVA), cyclosporine, methotrexate, acitretin, and apremilast. The anti-TNF antibodies may also improve some patients. The anti-IL-17 antibodies may offer some additional efficacy with PPP. Treatment options for PPPP are even more dismal, with few studies to guide treatment. The inflammatory cascade in PPPP is different from plaque psoriasis and this disease will likely need different targets to control disease. Cyclosporine is likely the best oral option for pustular disease. References 1. Jiaravuthisan MM, Sasseville D, Vender RB, Murphy F, Muhn CY. Psoriasis of the nail: anatomy, pathology, clinical presentation, and a review of the literature on therapy. J Am Acad Dermatol. 2007;57(1):1–27. 357 2. de Jong EM, Seegers BA, Gulinck MK, Boezeman JB, van de Kerkhof PC. Psoriasis of the nails associated with disability in a large number of patients: results of a recent interview with 1,728 patients. Dermatology. 1996;193(4):300–3. 3. van der Velden HM, Klaassen KM, van de Kerkhof PC, Pasch MC. The impact of fingernail psoriasis on patients’ health-related and disease-specific quality of life. Dermatology. 2014;229(2):76–82. 4. Langenbruch A, Radtke MA, Krensel M, Jacobi A, Reich K, Augustin M. Nail involvement as a predictor of concomitant psoriatic arthritis in patients with psoriasis. Br J Dermatol. 2014;171(5):1123–8. 5. Armesto S, Esteve A, Coto-Segura P, Drake M, Galache C, Martinez-Borra J, et al. Nail psoriasis in individuals with psoriasis vulgaris: a study of 661 patients. Actas Dermosifiliogr. 2011;102(5):365–72. 6. Chandran V, Schentag CT, Gladman DD. Sensitivity and specificity of the CASPAR criteria for psoriatic arthritis in a family medicine clinic setting. J Rheumatol. 2008;35(10):2069–70. author reply 70 7. Gupta AK, Lynde CW, Jain HC, Sibbald RG, Elewski BE, Daniel CR 3rd, et al. A higher prevalence of onychomycosis in psoriatics compared with non-psoriatics: a multicentre study. Br J Dermatol. 1997;136(5):786–9. 8. Al-Mutairi N, Nour T, Al-Rqobah D. Onychomycosis in patients of nail psoriasis on biologic therapy: a randomized, prospective open label study comparing Etanercept, Infliximab and Adalimumab. Expert Opin Biol Ther. 2013;13(5):625–9. 9. Halprin KM. Afflictions of a vestigial appendage. II. Inflammatory processes affecting the nail matrix. (trauma, eczema, psoriasis, lichen planus, alopecia areata). JAMA. 1967;202(11):1045. 10. Werner B, Fonseca GP, Seidel G. Microscopic nail clipping findings in patients with psoriasis. Am J Dermatopathol. 2015;37(6):429–39. 11. Rich P, Scher RK. Nail Psoriasis Severity Index: a useful tool for evaluation of nail psoriasis. J Am Acad Dermatol. 2003;49(2):206–12. 12. Cassell SE, Bieber JD, Rich P, Tutuncu ZN, Lee SJ, Kalunian KC, et al. The modified Nail Psoriasis Severity Index: validation of an instrument to assess psoriatic nail involvement in patients with psoriatic arthritis. J Rheumatol. 2007;34(1):123–9. 13. Radtke MA, Beikert FC, Augustin M. Nail psoriasis—a treatment challenge. J Dtsch Dermatol Ges. 2013;11(3):203–19. quiz 20 14. de Vries AC, Bogaards NA, Hooft L, Velema M, Pasch M, Lebwohl M, et al. Interventions for nail psoriasis. Cochrane Database Syst Rev. 2013;1:CD007633. 15. Oram Y, Akkaya AD. Treatment of nail psoriasis: common concepts and new trends. Dermatol Res Pract. 2013;2013:180496. 16. Langley RG, Saurat JH, Reich K, Na