Efficacy and Safety of Apremilast for the Treatment of Japanese Patients with Palmoplantar Pustulosis: Results from a Phase 2, Randomized, Placebo-Controlled Study

Tadashi Terui, Yukari Okubo, Satomi Kobayashi, Shigetoshi Sano, Akimichi Morita, Shinichi Imafuku, Yayoi Tada, Masatoshi Abe, Masafumi Yaguchi, Natsuka Uehara, Takahiro Handa, Masayuki Tanaka, Wendy Zhang, Maria Paris, Masamoto Murakami, Tadashi Terui, Yukari Okubo, Satomi Kobayashi, Shigetoshi Sano, Akimichi Morita, Shinichi Imafuku, Yayoi Tada, Masatoshi Abe, Masafumi Yaguchi, Natsuka Uehara, Takahiro Handa, Masayuki Tanaka, Wendy Zhang, Maria Paris, Masamoto Murakami

Abstract

Background: Palmoplantar pustulosis (PPP) is a pruritic, painful, recurrent, and chronic dermatitis with limited therapeutic options.

Objective: To evaluate the efficacy and safety of apremilast for the treatment of Japanese patients with PPP and inadequate response to topical treatment.

Methods: This phase 2, randomized, double-blind, placebo-controlled study enrolled patients with Palmoplantar Pustulosis Area and Severity Index (PPPASI) total score ≥ 12 and moderate or severe pustules/vesicles on the palm or sole (PPPASI pustule/vesicle severity score ≥ 2) at screening and baseline with an inadequate response to topical treatment. Patients were randomized (1:1) to apremilast 30 mg twice daily or placebo for 16 weeks, followed by a 16-week extension phase during which all patients received apremilast. The primary endpoint was achievement of PPPASI-50 response (≥ 50% improvement from baseline in PPPASI). Key secondary endpoints included change from baseline in PPPASI total score, Palmoplantar Pustulosis Severity Index (PPSI), and patient's visual analog scale (VAS) for PPP symptoms (pruritus and discomfort/pain).

Results: A total of 90 patients were randomized (apremilast: 46; placebo: 44). A significantly greater proportion of patients achieved PPPASI-50 at week 16 with apremilast versus placebo (P = 0.0003). Patients receiving apremilast showed greater improvement in PPPASI at week 16 versus placebo (nominal P = 0.0013), as well as PPSI and patient-reported pruritus and discomfort/pain (nominal P ≤ 0.001 for all). Improvements were sustained through week 32 with apremilast treatment. The most common treatment-emergent adverse events included diarrhea, abdominal discomfort, headache, and nausea.

Conclusions: Apremilast treatment demonstrated greater improvements in disease severity and patient-reported symptoms versus placebo at week 16 in Japanese patients with PPP with sustained improvements through week 32. No new safety signals were observed.

Clinicaltrials: GOV: NCT04057937.

Conflict of interest statement

Tadashi Terui has received research grants, consulting fees, and/or speaker’s fees from AbbVie, Amgen, Boehringer Ingelheim, Eli Lilly, Eisai, Kyowa Hakko Kirin, LEO Pharma, Maruho, Mitsubishi Tanabe, Novartis, and Taiho Pharmaceutical. Yukari Okubo has received grants or contracts from AbbVie, Eisai, Jimro, Maruho, Shiseido, Sun Pharma, and Torii; consulting fees from AbbVie, Amgen, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Eisai, Eli Lilly, Janssen Pharma, Kyowa Kirin, LEO Pharma, Maruho, Pfizer, Sun Pharma, and UCB Pharma; and honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from AbbVie, Amgen, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Eisai, Eli Lilly, Janssen Pharma, JIMRO, Kyowa Kirin, LEO Pharma, Maruho, Novartis Pharma, Pfizer, Sanofi, Sun Pharma, Taiho, Tanabe-Mitsubishi, Torii and UCB Pharma. Satomi Kobayashi has received research grants from Kyowa Kirin; received honoraria from AbbVie, Eli Lilly, Janssen Pharma, Maruho Pharmaceutical, Novartis, and Taiho Pharmaceutical; and participated in clinical trials for AbbVie, Amgen, Boehringer Ingelheim, Celgene, Eli Lilly, Janssen Pharma, Kyowa Kirin, Maruho Pharmaceutical, and Pfizer. Shigetoshi Sano has received research grants from Kaken, Maruho, Nihon, Nippon Zoki, Sanofi, Taiho, and Torii; and honoraria from AbbVie, Eisai, Eli Lilly, Janssen Pharma, Kyowa Kirin, Maruho, Sun Pharma, Taiho, and UCB. Akimichi Morita has received research grants, consulting fees, and/or speaker’s fees from AbbVie, Amgen, Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Eli Lilly, Eisai, Janssen, Kyowa Hakko Kirin, LEO Pharma, Maruho, Mitsubishi Tanabe, Nichi-Iko, Nippon Kayaku, Novartis, Pfizer, Sun Pharma, Taiho Pharmaceutical, Torii Pharmaceutical, Ushio, and UCB Pharma. Shinichi Imafuku has received grants and/or personal fees from AbbVie, Amgen Inc., Boehringer Ingelheim, Bristol Myers Squibb, Daiichi-Sankyo, Eisai, Eli Lilly, GSK, Janssen, Kyowa Kirin, LEO Pharma, Maruho, Novartis, Sun Pharma, Taiho Pharmaceutical, Tanabe-Mitsubishi, Torii Pharmaceutical, and UCB Japan. Yayoi Tada has received honoraria and/or grants from AbbVie, Boehringer Ingelheim, Bristol Myers Squibb KK, Celgene, Eisai, Eli Lilly, Janssen, Kyowa Kirin, LEO Pharma, Maruho, Meiji Seika Pharma, Mitsubishi Tanabe Pharma, Novartis Pharma, Sun Pharma, Taiho Pharmaceutical, Torii Pharmaceutical, and UCB Pharma. Masatoshi Abe has received research grants, consulting fees, speaker fees, and/or participated in clinical trials for Celgene and Maruho. Masafumi Yaguchi, Natsuka Uehara, Takahiro Handa, and Masayuki Tanaka are employees of Amgen K.K. Wendy Zhang and Maria Paris are employees and stockholders of Amgen Inc. Masamoto Murakami has received research grants from AbbVie, ARISTEA Therapeutics, Eisai, Eli Lilly, Kyowa Kirin, and Novartis Pharma; honoraria from AbbVie, Amgen Inc., Boehringer Ingelheim, Celgene, Eisai, Eli Lilly, Janssen Pharma, Kyowa Kirin, Maruho, Novartis Pharma, Taiho Pharmaceutical, and Torii Pharmaceutical; and participated in clinical trials for AbbVie, Amgen Inc., Boehringer Ingelheim, Bristol Myers Squibb, Celgene, Eli Lilly, Janssen Pharma, Maruho, and Novartis Pharma.

© 2023. The Author(s).

Figures

Fig. 1
Fig. 1
Proportion of patients with PPPASI-50, PPPASI-75, and PPPASI-90 responses at Week 16. Intent-to-treat population. *Two-sided P-value is based on chi-square test. Missing data were imputed by nonresponder imputation. †Two-sided P-value (nominal) based on Cochran–Mantel–Haenszel test adjusting for baseline PPPASI score range and baseline focal infection status at baseline. BID twice daily, CI confidence interval, PPPASI Palmoplantar Pustulosis Area and Severity Index; PPPASI-50/75/90, ≥50%/75%/90% improvement from baseline in PPPASI total score
Fig. 2
Fig. 2
Change from Baseline in PPPASI total score and PPSI score at Week 16 after treatment with apremilast or placebo (ITT population). Error bars represent 95% CI. Intent-to-treat population. Differences are based on MMRM model for the change from baseline, with treatment group, visit, treatment-by-visit interaction, PPPASI total score range, and focal infection status at baseline as fixed effects and the baseline value as a covariate. An unstructured covariance matrix that is homogeneous across treatment groups was used. BID twice daily, CI confidence interval, MMRM mixed-effects model for repeated measures, PPPASI Palmoplantar Pustulosis Area and Severity Index, PPSI Palmoplantar Severity Index.
Fig. 3
Fig. 3
PGA response rate (score of 0 or 1 and ≥ 2 grade improvement) and change from Baseline in patient’s VAS for PPP symptoms at Week 16 after treatment with apremilast or placebo (ITT population). Data shown for week 16 are LS mean changes and for week 32 are mean changes from baseline. Error bars represent 95% CI. Intent-to-treat population. For PGA response, missing data were imputed by nonresponder imputation. Differences in patient’s VAS assessments are based on MMRM model for the change from baseline, with treatment group, visit, treatment-by-visit interaction, PPPASI total score range, and focal infection status at baseline as fixed effects and the baseline value as a covariate. An unstructured covariance matrix that is homogeneous across treatment groups was used. For patient’s VAS assessments, LS mean values are shown for week 16, and mean values are shown for week 32. APR apremilast 30 mg twice daily, BID twice daily, CI confidence interval, MMRM mixed-effects model for repeated measures, NRI nonresponder imputation, PBO placebo, PGA Physician Global Assessment, PPPASI Palmoplantar Pustulosis Area and Severity Index, VAS visual analog scale.
Fig. 4
Fig. 4
Change from Baseline in DLQI at Week 16 and Week 32 (exploratory endpoint). Error bars represent 95% CIs. Intent-to-treat population. Data are presented as observed. BID twice daily, CI confidence interval, DLQI Dermatology Life Quality Index.

References

    1. Murakami M, Terui T. Palmoplantar pustulosis: current understanding of disease definition and pathomechanism. J Dermatol Sci. 2020;98(1):13–19. doi: 10.1016/j.jdermsci.2020.03.003.
    1. Misiak-Galazka M, Zozula J, Rudnicka L. Palmoplantar pustulosis: recent advances in etiopathogenesis and emerging treatments. Am J Clin Dermatol. 2020;21(3):355–370. doi: 10.1007/s40257-020-00503-5.
    1. Masuda-Kuroki K, Murakami M, Kishibe M, Kobayashi S, Okubo Y, Yamamoto T, et al. Diagnostic histopathological features distinguishing palmoplantar pustulosis from pompholyx. J Dermatol. 2019;46(5):399–408. doi: 10.1111/1346-8138.14850.
    1. Murakami M, Hagforsen E, Morhenn V, Ishida-Yamamoto A, Iizuka H. Patients with palmoplantar pustulosis have increased IL-17 and IL-22 levels both in the lesion and serum. Exp Dermatol. 2011;20(10):845–847. doi: 10.1111/j.1600-0625.2011.01325.x.
    1. Liang Y, Xing X, Beamer MA, Swindell WR, Sarkar MK, Roberts LW, et al. Six-transmembrane epithelial antigens of the prostate comprise a novel inflammatory nexus in patients with pustular skin disorders. J Allergy Clin Immunol. 2017;139(4):1217–1227. doi: 10.1016/j.jaci.2016.10.021.
    1. Kubota K, Kamijima Y, Sato T, Ooba N, Koide D, Iizuka H, et al. Epidemiology of psoriasis and palmoplantar pustulosis: a nationwide study using the Japanese national claims database. BMJ Open. 2015;5(1):e006450. doi: 10.1136/bmjopen-2014-006450.
    1. Obeid G, Do G, Kirby L, Hughes C, Sbidian E, Le Cleach L. Interventions for chronic palmoplantar pustulosis. Cochrane Database Sys Rev. 2020 doi: 10.1002/14651858.CD011628.pub2.
    1. Benzian-Olsson N, Dand N, Chaloner C, Bata-Csorgo Z, Borroni R, Burden AD, et al. Association of clinical and demographic factors with the severity of palmoplantar pustulosis. JAMA Dermatol. 2020;156(11):1216–1222. doi: 10.1001/jamadermatol.2020.3275.
    1. Masuda-Kuroki K, Kawakami H, Abe N, Mori M, Tobita R, Fukushi R, et al. Nail lesions in palmoplantar pustulosis and pustulotic arthro-osteitis impairs patients' quality of life: suggesting new assessment tool of PPP nail lesions. J Dermatol Sci. 2022;106(1):29–36. doi: 10.1016/j.jdermsci.2022.03.001.
    1. Trattner H, Blüml S, Steiner I, Plut U, Radakovic S, Tanew A. Quality of life and comorbidities in palmoplantar pustulosis—a cross-sectional study on 102 patients. J Eur Acad Dermatol Venereol. 2017;31(10):1681–1685. doi: 10.1111/jdv.14187.
    1. Bissonnette R, Suárez-Fariñas M, Li X, Bonifacio KM, Brodmerkel C, Fuentes-Duculan J, et al. Based on molecular profiling of gene expression, palmoplantar pustulosis and palmoplantar pustular psoriasis are highly related diseases that appear to be distinct from psoriasis vulgaris. PLoS ONE. 2016;11(5):e0155215. doi: 10.1371/journal.pone.0155215.
    1. Yamamoto T. Similarity and difference between palmoplantar pustulosis and pustular psoriasis. J Dermatol. 2021;48(6):750–760. doi: 10.1111/1346-8138.15826.
    1. Twelves S, Mostafa A, Dand N, Burri E, Farkas K, Wilson R, et al. Clinical and genetic differences between pustular psoriasis subtypes. J Allergy Clin Immunol. 2019;143(3):1021–1026. doi: 10.1016/j.jaci.2018.06.038.
    1. Umezawa Y, Nakagawa H, Tamaki K. Phase III clinical study of maxacalcitol ointment in patients with palmoplantar pustulosis: a randomized, double-blind, placebo-controlled trial. J Dermatol. 2016;43(3):288–293. doi: 10.1111/1346-8138.13064.
    1. Muro M, Kawakami H, Matsumoto Y, Abe N, Tsuboi R, Okubo Y. Topical combination therapy with vitamin D3 and corticosteroid ointment for palmoplantar pustulosis: a prospective, randomized, left-right comparison study. J Dermatol Treat. 2016;27(1):51–53. doi: 10.3109/09546634.2015.1052036.
    1. Hayama K, Inadomi T, Fujisawa D, Terui T. A pilot study of medium-dose cyclosporine for the treatment of palmoplantar pustulosis complicated with pustulotic arthro-osteitis. Eur J Dermatol. 2010;20(6):758–762.
    1. Terui T, Kobayashi S, Okubo Y, Murakami M, Hirose K, Kubo H. Efficacy and safety of guselkumab, an anti-interleukin 23 monoclonal antibody, for palmoplantar pustulosis: a randomized clinical trial. JAMA Dermatol. 2018;154(3):309–316. doi: 10.1001/jamadermatol.2017.5937.
    1. Okubo Y, Morishima H, Zheng R, Terui T. Sustained efficacy and safety of guselkumab in patients with palmoplantar pustulosis through 1.5 years in a randomized phase 3 study. J Dermatol. 2021;48(12):1838–1853. doi: 10.1111/1346-8138.16132.
    1. Kawakami H, Nagaoka Y, Hirano H, Matsumoto Y, Abe N, Tsuboi R, et al. Evaluation of the efficacy of granulocyte and monocyte adsorption apheresis on skin manifestation and joint symptoms of patients with pustulotic arthro-osteitis. J Dermatol. 2019;46(2):144–148. doi: 10.1111/1346-8138.14717.
    1. Kavanaugh A, Mease PJ, Gomez-Reino JJ, Adebajo AO, Wollenhaupt J, Gladman DD, et al. Treatment of psoriatic arthritis in a phase 3 randomized, placebo-controlled trial with apremilast, an oral phosphodiesterase 4 inhibitor. Ann Rheum Dis. 2014;73(6):1020–1026. doi: 10.1136/annrheumdis-2013-205056.
    1. Cutolo M, Myerson GE, Fleischmann R, Liote F, Diaz-Gonzalez F, Van den Bosch F, et al. A phase III, randomized, controlled trial of apremilast in patients with psoriatic arthritis: results of the PALACE 2 trial. J Rheumatol. 2016;43(9):1724–1734. doi: 10.3899/jrheum.151376.
    1. Edwards CJ, Blanco FJ, Crowley J, Birbara CA, Jaworski J, Aelion J, et al. Apremilast, an oral phosphodiesterase 4 inhibitor, in patients with psoriatic arthritis and current skin involvement: a phase III, randomised, controlled trial (PALACE 3) Ann Rheum Dis. 2016;75(6):1065–1073. doi: 10.1136/annrheumdis-2015-207963.
    1. Wells AF, Edwards CJ, Kivitz AJ, Bird P, Nguyen D, Paris M, et al. Apremilast monotherapy in DMARD-naive psoriatic arthritis patients: results of the randomized, placebo-controlled PALACE 4 trial. Rheumatology. 2018;57(7):1253–1263. doi: 10.1093/rheumatology/key032.
    1. Papp K, Reich K, Leonardi CL, Kircik L, Chimenti S, Langley RG, et al. 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;73(1):37–49. doi: 10.1016/j.jaad.2015.03.049.
    1. Paul C, Cather J, Gooderham M, Poulin Y, Mrowietz U, Ferrandiz C, et al. 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;173(6):1387–1399. doi: 10.1111/bjd.14164.
    1. Stein Gold L, Bagel J, Lebwohl M, Jackson JM, Chen R, Goncalves J, et al. Efficacy and safety of apremilast in systemic- and biologic-naive patients with moderate plaque psoriasis: 52-week results of UNVEIL. J Drugs Dermatol. 2018;17(2):221–228.
    1. Strober B, Bagel J, Lebwohl M, Stein Gold L, Jackson JM, Chen R, et al. Efficacy and safety of apremilast in patients with moderate plaque psoriasis (UNVEIL phase IV study) [poster 4892]. Annual Meeting of the American Academy of Dermatology; March 3–7, 2017; Orlando, FL.
    1. Hatemi G, Mahr A, Ishigatsubo Y, Song YW, Takeno M, Kim D, et al. Trial of apremilast for oral ulcers of Behçet’s syndrome. N Engl J Med. 2019;381(20):1918–1928. doi: 10.1056/NEJMoa1816594.
    1. Schafer PH, Chen P, Fang L, Wang A, Chopra R. The pharmacodynamic impact of apremilast, an oral phosphodiesterase 4 inhibitor, on circulating levels of inflammatory biomarkers in patients with psoriatic arthritis: substudy results from a phase III, randomized, placebo-controlled trial (PALACE 1) J Immunol Res. 2015;2015:906349. doi: 10.1155/2015/906349.
    1. Hamzaoui K, Hamzaoui A, Guemira F, Bessioud M, Hamza M, Ayed K. Cytokine profile in Behçet's disease patients: relationship with disease activity. Scand J Rheumatol. 2002;31(4):205–210. doi: 10.1080/030097402320318387.
    1. Yamamoto T. Extra-palmoplantar lesions associated with palmoplantar pustulosis. J Eur Acad Dermatol Venereol. 2009;23(11):1227–1232. doi: 10.1111/j.1468-3083.2009.03296.x.
    1. Fujisawa T, Tawada C, Mizutani Y, Doi T, Yoshida S, Ogura S, et al. Efficacy of granulocyte and monocyte adsorption apheresis for treatment of palmoplantar pustulosis. Ther Apher Dial. 2014;18(3):238–243. doi: 10.1111/1744-9987.12210.
    1. Basra MK, Salek MS, Camilleri L, Sturkey R, Finlay AY. Determining the minimal clinically important difference and responsiveness of the Dermatology Life Quality Index (DLQI): further data. Dermatology. 2015;230(1):27–33. doi: 10.1159/000365390.
    1. Crocetti L, Floresta G, Cilibrizzi A, Giovannoni MP. An overview of PDE4 inhibitors in clinical trials: 2010 to early 2022. Molecules. 2022;27(15):4964. doi: 10.3390/molecules27154964.
    1. Strober B, Alikhan A, Lockshin B, Shi R, Cirulli J, Schafer P. Apremilast mechanism of efficacy in systemic-naive patients with moderate plaque psoriasis: pharmacodynamic results from the UNVEIL study. J Dermatol Sci. 2019 doi: 10.1016/j.jdermsci.2019.09.00.
    1. Garcet S, Nograles K, Correa da Rosa J, Schafer PH, Krueger JG. Synergistic cytokine effects as apremilast response predictors in patients with psoriasis. J Allergy Clin Immunol. 2018;142(3):1010–3.e6. doi: 10.1016/j.jaci.2018.05.039.
    1. Skov L, Beurskens FJ, Zachariae CO, Reitamo S, Teeling J, Satijn D, et al. IL-8 as antibody therapeutic target in inflammatory diseases: reduction of clinical activity in palmoplantar pustulosis. J Immunol. 2008;181(1):669–679. doi: 10.4049/jimmunol.181.1.669.
    1. Fukasawa T, Yoshizaki-Ogawa A, Enomoto A, Miyagawa K, Sato S, Yoshizaki A. Involvement of molecular mechanisms between T/B cells and IL-23: from palmoplantar pustulosis to autoimmune diseases. Int J Mol Sci. 2022;23(15):8261. doi: 10.3390/ijms23158261.
    1. Murakami M, Kaneko T, Nakatsuji T, Kameda K, Okazaki H, Dai X, et al. Vesicular LL-37 contributes to inflammation of the lesional skin of palmoplantar pustulosis. PLoS ONE. 2014;9(10):e110677. doi: 10.1371/journal.pone.0110677.
    1. Pincelli C, Schafer PH, French LE, Augustin M, Krueger JG. Mechanisms underlying the clinical effects of apremilast for psoriasis. J Drugs Dermatol. 2018;17(8):835–840.
    1. Mrowietz U, Bachelez H, Burden AD, Rissler M, Sieder C, Orsenigo R, et al. Secukinumab for moderate-to-severe palmoplantar pustular psoriasis: results of the 2PRECISE study. J Am Acad Dermatol. 2019;80(5):1344–1352. doi: 10.1016/j.jaad.2019.01.066.
    1. 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: a phase 3 randomized clinical trial. JAMA Dermatol. 2019;155(10):1153–1161. doi: 10.1001/jamadermatol.2019.1394.
    1. Van Zander J, Orlow SJ. Efficacy and safety of oral retinoids in psoriasis. Expert Opin Drug Saf. 2005;4(1):129–138. doi: 10.1517/14740338.4.1.129.
    1. Reitamo S, Erkko P, Remitz A, Lauerma AI, Montonen O, Harjula K. Cyclosporine in the treatment of palmoplantar pustulosis. A randomized, double-blind, placebo-controlled study. Arch Dermatol. 1993;129(10):1273–1279. doi: 10.1001/archderm.1993.01680310043006.
    1. Erkko P, Granlund H, Remitz A, Rosen K, Mobacken H, Lindelöf B, et al. Double-blind placebo-controlled study of long-term low-dose cyclosporin in the treatment of palmoplantar pustulosis. Br J Dermatol. 1998;139(6):997–1004. doi: 10.1046/j.1365-2133.1998.02555.x.
    1. Fujita H, Terui T, Hayama K, Akiyama M, Ikeda S, Mabuchi T, et al. Japanese guidelines for the management and treatment of generalized pustular psoriasis: the new pathogenesis and treatment of GPP. J Dermatol. 2018;45(11):1235–1270. doi: 10.1111/1346-8138.14523.
    1. Berth-Jones J, Exton LS, Ladoyanni E, Mohd Mustapa MF, Tebbs VM, Yesudian PD, et al. British Association of Dermatologists guidelines for the safe and effective prescribing of oral ciclosporin in dermatology 2018. Br J Dermatol. 2019;180(6):1312–1338. doi: 10.1111/bjd.17587.
    1. West J, Ogston S, Foerster J. Safety and efficacy of methotrexate in psoriasis: a meta-analysis of published trials. PLoS ONE. 2016;11(5):e0153740. doi: 10.1371/journal.pone.0153740.
    1. Kato N, Takama H, Ando Y, Yanagishita T, Ohshima Y, Ohashi W, et al. Immediate response to apremilast in patients with palmoplantar pustulosis: a retrospective pilot study. Int J Dermatol. 2021;60(5):570–578. doi: 10.1111/ijd.15382.
    1. Freitas E, Rodrigues MA, Torres T. Diagnosis, screening and treatment of patients with palmoplantar pustulosis (PPP): a review of current practices and recommendations. Clin Cosmet Investig Dermatol. 2020;13:561–578. doi: 10.2147/CCID.S240607.
    1. Wilsmann-Theis D, Kromer C, Gerdes S, Linker C, Magnolo N, Sabat R, et al. A multicentre open-label study of apremilast in palmoplantar pustulosis (APLANTUS) J Eur Acad Dermatol Venereol. 2021;35(10):2045–2050. doi: 10.1111/jdv.17441.

Source: PubMed

3
Abonner