1
|
Lé AM, Yilmaz O, Luz M, Torres T. Oral roflumilast for psoriasis: a real-world 24-week prospective cohort study. J DERMATOL TREAT 2025; 36:2464107. [PMID: 39933751 DOI: 10.1080/09546634.2025.2464107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
OBJECTIVE Psoriasis is a chronic inflammatory skin disease with significant physical and psychological burden, often associated with comorbidities such as obesity and cardiovascular disease. Current treatments include conventional systemic therapies and targeted biologic and non-biologic therapies, with several limitations related to safety, efficacy, and cost. Roflumilast, a selective PDE4 inhibitor, shows potential as an oral therapy for psoriasis due to its anti-inflammatory effects and favorable safety profile. This study aimed to evaluate the real-world effectiveness and safety of oral roflumilast in moderate-to-severe plaque psoriasis. METHODS Prospective cohort study at a single center in Portugal including adults with moderate-to-severe psoriasis treated with oral roflumilast 500 mcg once daily. RESULTS Among fifty-eight patients (baseline median PASI 13.7 ± 5.5), 63.0% achieved PASI < 5, 47.8% PASI < 3, and 21.7% PASI < 1 by week 24 (mNRI). Weight loss occurred in 53.4%, with a mean reduction of 6 kg ± 4.3. Mild gastrointestinal symptoms were common but rarely caused discontinuation. No serious adverse events were reported. CONCLUSION Roflumilast demonstrated real-world effectiveness and a favorable safety profile in moderate-to-severe plaque psoriasis. Additional benefits, including weight loss and no need for laboratory monitoring, make it a promising treatment option, particularly for patients with comorbidities or limited access to biologic therapies.
Collapse
Affiliation(s)
- Ana Maria Lé
- Department of Dermatology, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Orhan Yilmaz
- College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Martim Luz
- Department of Dermatology, Centro Hospitalar Universitário do Porto, Porto, Portugal
| | - Tiago Torres
- Department of Dermatology, Centro Hospitalar Universitário do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| |
Collapse
|
2
|
Augustin M, Barker J, Conrad C, Jullien D, Carrascosa JM, Reddy J, Amouzadeh H, Colgan S, Zou H, Mrowietz U. Efficacy and Safety of Apremilast Over 52 Weeks in Patients with Plaque Psoriasis in High-Impact Areas and Impaired Quality of Life. Dermatol Ther (Heidelb) 2025:10.1007/s13555-025-01389-z. [PMID: 40317400 DOI: 10.1007/s13555-025-01389-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/14/2025] [Indexed: 05/07/2025] Open
Abstract
INTRODUCTION In EMBRACE, significant benefits were seen with apremilast versus placebo on quality of life (QoL) and clinical outcomes after 16 weeks in patients with chronic plaque psoriasis in ≥ 1 high-impact area, limited skin involvement, and impaired QoL, in a European population previously unstudied with apremilast. Our objective was to evaluate the 52-week efficacy and safety of apremilast. METHODS EMBRACE was a phase 4, multicenter, randomized, placebo-controlled, double-blind study. Eligible patients had chronic plaque psoriasis for ≥ 6 months prior to baseline; involvement in ≥ 1 high-impact area; a Psoriasis Area and Severity Index score ≥ 3 to ≤ 10; and a Dermatology Life Quality Index score > 10. Patients were randomized (2:1) to receive apremilast 30 mg twice daily or placebo from weeks 0 to 16; after that, all patients continued with apremilast (apremilast/apremilast) or switched from placebo to apremilast (placebo/apremilast) until week 52. RESULTS Of 277 patients randomized (apremilast: 185, placebo: 92), 158 (apremilast/apremilast: 105; placebo/apremilast: 53) completed 52 weeks. Improvements in QoL, itch, skin discomfort/pain, patient-reported treatment benefits, and skin outcomes at week 16 were maintained over 52 weeks in patients continuing apremilast treatment. Similar improvements were seen at week 52 in patients who switched from placebo to apremilast at week 16. No new safety signals were observed. CONCLUSIONS Improvements in QoL and disease severity at week 16 were maintained through week 52 with continued apremilast treatment in EMBRACE, with no new safety signals. TRIAL REGISTRATION CLINICALTRIALS. GOV IDENTIFIER NCT03774875.
Collapse
Affiliation(s)
- Matthias Augustin
- Institute for Health Services Research in Dermatology and Nursing (IVDP), University Medical Center Hamburg-Eppendorf (UKE), Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| | - Jonathan Barker
- St John's Institute of Dermatology of King's College, London, UK
| | - Curdin Conrad
- Department of Dermatology, Lausanne University Hospital CHUV, Lausanne, Switzerland
| | - Denis Jullien
- Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Jose-Manuel Carrascosa
- Department of Dermatology, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma of Barcelona, IGTP, Badalona, Barcelona, Spain
| | | | | | | | | | - Ulrich Mrowietz
- Psoriasis-Center at the Department of Dermatology, University Medical Center Schleswig-Holstein, Campus Kiel, Kiel, Germany
| |
Collapse
|
3
|
Ren L, Zhao K, Wang B, Xiao S, Liu J, Tu C. Safety assessment of apremilast: real-world adverse event analysis from the FAERS database. Arch Dermatol Res 2025; 317:684. [PMID: 40195166 DOI: 10.1007/s00403-025-04193-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 02/17/2025] [Accepted: 03/21/2025] [Indexed: 04/09/2025]
Abstract
Apremilast, a unique oral immunomodulatory phosphodiesterase 4 inhibitor, has been approved by the U.S. Food and Drug Administration (FDA) for the treatment of psoriasis and psoriatic arthritis (PsA). But there is a lack of real-world studies on the safety of apremilast in large numbers of people. This research employs the FAERS database to assess the real-world safety profile of apremilast to evaluate the association between apremilast and adverse events, which can guide drug monitoring and clinical practice. Our study collected adverse event reports listing apremilast as the primary suspect drug from the first quarter of 2012 to the first quarter of 2024. We utilized multiple disproportionality analysis methods, including reporting odds ratio (ROR), proportional reporting ratio (PRR), Bayesian confidence propagation neural network (BCPNN), and multi-item gamma Poisson shrinker (MGPS), to evaluate adverse events associated with apremilast. A comprehensive analysis yielded 124,734 reports identifying apremilast as the primary suspect drug, encompassing 252,677 preferred terms (PT) for adverse reactions distributed across 27 system organ class (SOC) categories. Apart from the adverse reactions already mentioned in the drug label, this study identified some new clinically valuable potential adverse reactions, such as vomiting, depression, suicidal ideation, weight decreased, decreased appetite, nasopharyngitis, dyspepsia, abdominal pain, gastrooesophageal reflux disease, abdominal distension, migraine, insomnia, memory impairment, muscle spasms, musculoskeletal stiffness and so on. Our study provide new safety information for the clinical use of apremilast and establish a framework and insights for its further safety evaluation.
Collapse
Affiliation(s)
- Landong Ren
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Kaidi Zhao
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Bingqing Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Shengxiang Xiao
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Jiashu Liu
- Department of Dermatology, Xi'an Children's Hospital, Xi'an, 710000, China.
| | - Chen Tu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
4
|
Terui T, Murakami M, Okubo Y, Hayama K, Fujita H. Palmoplantar Pustulosis as an Immune-Mediated Inflammatory Disease with a Possible Relevance of Th17 Cell Plasticity: A Narrative Review. Dermatol Ther (Heidelb) 2025; 15:797-810. [PMID: 40088419 PMCID: PMC11971112 DOI: 10.1007/s13555-025-01382-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Palmoplantar pustulosis (PPP) is an immune-mediated inflammatory disease (IMID) of the skin that causes the formation of sterile pustules on the palms and soles. The clinical course of PPP is variable, with some patients having persistent skin lesions and others having lesions that wax and wane repeatedly. PPP has been treated with drugs such as those used for plaque psoriasis. The efficacy of biologics targeting the interleukin (IL)-23-helper T (Th)17 axis is not as conspicuous in PPP as in plaque psoriasis. Traditionally, CD4+ Th cell subsets have been defined by the expression of a small number of cytokines; however, recent advances in immunology have shown that some Th cell subsets can express cytokines of other Th subsets in response to changes in the environment. Recent studies have suggested the involvement of unique Th17 cells with Th2 cell characteristics in the pathogenesis of PPP. Thus, the insufficient efficacy of biologics targeting the IL-23-Th17 axis in PPP raises the question of whether Th17 cell plasticity is involved in the pathogenesis of PPP. In this review, we discuss the complexity of PPP pathogenesis with some speculation, compare the new knowledge obtained by other IMIDs or their mouse models with that of PPP elucidated thus far, and contribute to the development of future research.
Collapse
Affiliation(s)
- Tadashi Terui
- University Research Center, Nihon University, 30-1 Oyaguchi-Kamicho, Itabashi-ku, Tokyo, 173-8610, Japan
- Division of Cutaneous Science, Department of Dermatology, Nihon University School of Medicine, 30-1 Oyaguchi-Kamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Masamoto Murakami
- Department of Anatomy, Histochemistry and Cell Biology, Faculty of Medicine, Miyazaki University, 5600 Kihara, Kiyotake, Miyazaki, 889-1692, Japan
- Atsuta Skin Clinic, 2-44 Nakanohonnmachi, Nakagawa-ku, Nagoya, 454-0047, Japan
| | - Yukari Okubo
- Department of Dermatology, Tokyo Medical University, 6 Chome-7-1 Nishishinjuku, Shinjuku, Tokyo, 160-0023, Japan
| | - Koremasa Hayama
- Division of Cutaneous Science, Department of Dermatology, Nihon University School of Medicine, 30-1 Oyaguchi-Kamicho, Itabashi-ku, Tokyo, 173-8610, Japan
| | - Hideki Fujita
- Division of Cutaneous Science, Department of Dermatology, Nihon University School of Medicine, 30-1 Oyaguchi-Kamicho, Itabashi-ku, Tokyo, 173-8610, Japan.
| |
Collapse
|
5
|
Li W, Dong P, Zhang G, Hu J, Yang S. Emerging Therapeutic Innovations for Vitiligo Treatment. Curr Issues Mol Biol 2025; 47:191. [PMID: 40136446 PMCID: PMC11940846 DOI: 10.3390/cimb47030191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Vitiligo is a chronic autoimmune disorder with a multifactorial etiology, typically manifesting as localized or generalized hypopigmentation or depigmentation of the skin and mucous membranes. The pathogenesis of vitiligo is complex and significantly impacts patients' quality of life. Although traditional treatments such as hormone therapy, topical medications, and laser therapy can help control the disease to some extent, their outcomes remain unsatisfactory. Therefore, ongoing research is crucial to explore and develop novel treatment strategies while assessing their efficacy and safety. This review aims to classify and summarize various new candidate drugs for vitiligo currently undergoing clinical trials, providing a reference for clinical practice. Recent advancements in the understanding of the pathogenesis of vitiligo have facilitated the development of potential treatment strategies, such as Janus kinase inhibitors, cytokine blockers, and agents targeting tissue-resident memory or regulatory T cells. These emerging therapies offer hope to patients with vitiligo, though further investigation is needed to confirm their safety, efficacy, and optimal treatment regimens.
Collapse
Affiliation(s)
- Weiran Li
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei 230000, China; (P.D.); (G.Z.); (J.H.)
- Department of Dermatology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215128, China
- Key Laboratory of Dermatology, Ministry of Education of the People’s Republic of China, Hefei 230000, China
| | - Penghao Dong
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei 230000, China; (P.D.); (G.Z.); (J.H.)
- Key Laboratory of Dermatology, Ministry of Education of the People’s Republic of China, Hefei 230000, China
| | - Guiyuan Zhang
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei 230000, China; (P.D.); (G.Z.); (J.H.)
- Key Laboratory of Dermatology, Ministry of Education of the People’s Republic of China, Hefei 230000, China
| | - Junjie Hu
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei 230000, China; (P.D.); (G.Z.); (J.H.)
- Key Laboratory of Dermatology, Ministry of Education of the People’s Republic of China, Hefei 230000, China
| | - Sen Yang
- Institute of Dermatology and Department of Dermatology, The First Affiliated Hospital, Anhui Medical University, Hefei 230000, China; (P.D.); (G.Z.); (J.H.)
- Department of Dermatology, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215128, China
- Key Laboratory of Dermatology, Ministry of Education of the People’s Republic of China, Hefei 230000, China
| |
Collapse
|
6
|
Beltrami VA, Martins FRB, Martins DG, Queiroz-Junior CM, Félix FB, Resende LC, Santos FRDS, Lacerda LDSB, Costa VRDM, da Silva WN, Guimaraes PPG, Guimaraes G, Soriani FM, Teixeira MM, Costa VV, Pinho V. Selective phosphodiesterase 4 inhibitor roflumilast reduces inflammation and lung injury in models of betacoronavirus infection in mice. Inflamm Res 2025; 74:24. [PMID: 39862252 DOI: 10.1007/s00011-024-01985-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/03/2024] [Accepted: 10/16/2024] [Indexed: 01/27/2025] Open
Abstract
OBJECTIVE We aimed to understand the potential therapeutic and anti-inflammatory effects of the phosphodiesterase-4 (PDE4) inhibitor roflumilast in models of pulmonary infection caused by betacoronaviruses. METHODS Mice were infected intranasally with murine hepatitis virus (MHV-3) or severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Roflumilast was given to MHV-3-infected mice therapeutically at doses of 1 mg/kg or 10 mg/kg, or prophylactically at 10 mg/kg. In SARS-CoV-2-infected mice, roflumilast was given therapeutically at a dose of 10 mg/kg. Lung histopathology, chemokines (CXCL-1 and CCL2), cytokines (IL-1β, IL-6, TNF, IFN-γ, IL-10 and TGFβ), neutrophil immunohistochemical staining (Ly6G+ cells), macrophage immunofluorescence staining (F4/80+ cells), viral titration plaque assay, real-time PCR virus detection, and blood cell counts were examined. RESULTS Therapeutic treatment with roflumilast at 10 mg/kg reduced lung injury in SARS-CoV-2 or MHV-3-infected mice without compromising viral clearance. In MHV-3-infected mice, reduced lung injury was associated with decreased chemokines levels, prevention of neutrophil aggregates and reduced macrophage accumulation in the lung tissue. However, the prophylactic treatment strategy with roflumilast increased lung injury in MHV-3-infected mice. CONCLUSION Our findings indicate that therapeutic treatment with roflumilast reduced lung injury in MHV-3 and SARS-CoV-2 lung infections. Given the protection induced by roflumilast in inflammation, PDE4 targeting could be a promising therapeutic avenue worth exploring following severe viral infections of the lung.
Collapse
Affiliation(s)
- Vinícius Amorim Beltrami
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Flávia Rayssa Braga Martins
- Departamento Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Débora Gonzaga Martins
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Celso Martins Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Franciel Batista Félix
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Letícia Cassiano Resende
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Felipe Rocha da Silva Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Larisse de Souza Barbosa Lacerda
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Victor Rodrigues de Melo Costa
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Walison Nunes da Silva
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Pedro Pires Goulart Guimaraes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Goulart Guimaraes
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Frederico Marianetti Soriani
- Departamento Biologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Vivian Vasconcelos Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Vanessa Pinho
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| |
Collapse
|
7
|
Gosia M, Doshi G, Bagwe Parab S, Godad A. Innovative Approaches to Psoriasis: Small Molecules Targeting Key Signaling Pathways. Immunol Invest 2025:1-37. [PMID: 39819440 DOI: 10.1080/08820139.2025.2449960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
BACKGROUND Psoriasis (Pso) is a chronic, immune-mediated dermatological condition characterized by dysregulated inflammatory responses and the hyperproliferation of keratinocytes. Biologics, which target specific cytokines such as IL-17 and IL-23, have revolutionized the management by addressing key drivers of its pathophysiology. Despite their efficacy, biologics are not without limitations, including the need for intermittent administration and ongoing monitoring. In contrast, small molecules offer a promising alternative by selectively inhibiting key signaling pathways that modulate pro-inflammatory cytokines involved in the inflammatory cascade. METHODS AND RESULTS This review suggests a new therapeutic strategy for Pso treatment, emphasizing the intricate relationships between small molecules and important signaling pathways involved in the pathophysiology of skin conditions. Improving treatment outcomes and reducing the side effects associated with conventional medicines, this review aims to better understand how tailored small-molecule inhibitors might efficiently control these pathways. This creative approach promotes the creation of individualized treatment plans that can greatly enhance the quality of life of patients with Psoby utilizing the knowledge gathered from recent developments in signaling pathway research. CONCLUSION This review delves into the molecular mechanisms underlying Pso and explores how small molecules can be harnessed to enhance treatment outcomes, presenting a new paradigm for managing this chronic skin disorder.
Collapse
Affiliation(s)
- Meeral Gosia
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Siddhi Bagwe Parab
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Angel Godad
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| |
Collapse
|
8
|
Rizzo C, Grazzini S, Conticini E, Chinoy H, D'Alessandro R, Camarda F, Cantarini L, Frediani B, Guggino G, La Barbera L. Small molecules in idiopathic inflammatory myopathies: a systematic review and a multicenter case series about Janus kinase inhibitors and apremilast. Reumatismo 2025. [PMID: 39810571 DOI: 10.4081/reumatismo.2025.1718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/06/2024] [Indexed: 01/16/2025] Open
Abstract
OBJECTIVE Idiopathic inflammatory myopathies (IIM) are rare autoimmune diseases that primarily affect striated muscles; skin, joints, and lungs may be involved with different degrees of severity. Traditional treatment relies on high-dose glucocorticoids and conventional synthetic disease-modifying antirheumatic drugs. METHODS A growing amount of evidence is demonstrating the potential role of novel treatments in the management of IIM. We report our experience with Janus kinase inhibitors (JAKi) in these conditions and review the current evidence for the use of small molecules in real-life clinical practice. RESULTS A total of 41 papers were retrieved from PubMed, 37 papers concerning IIM and JAKi, and 4 papers concerning IIM and apremilast. CONCLUSIONS An overall good efficacy was evidenced in IIM-associated skin lesions, including rash, ulcers, and calcinosis. If present, muscle and joint involvement demonstrated a good response to therapy, while it was not possible to draw any conclusion about dysphagia. No life-threatening adverse events were reported.
Collapse
Affiliation(s)
- Chiara Rizzo
- Rheumatology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo
| | - Silvia Grazzini
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena
| | - Edoardo Conticini
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena
| | - Hector Chinoy
- Department of Rheumatology, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, Salford; Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester
| | - Roberto D'Alessandro
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena
| | - Federica Camarda
- Rheumatology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo
| | - Luca Cantarini
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena
| | - Bruno Frediani
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena
| | - Giuliana Guggino
- Rheumatology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo
| | - Lidia La Barbera
- Rheumatology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo
| |
Collapse
|
9
|
Mateescu LA, Savu AP, Mutu CC, Vaida CD, Șerban ED, Bucur Ș, Poenaru E, Nicolescu AC, Constantin MM. The Intersection of Psoriasis and Neoplasia: Risk Factors, Therapeutic Approaches, and Management Strategies. Cancers (Basel) 2024; 16:4224. [PMID: 39766123 PMCID: PMC11674284 DOI: 10.3390/cancers16244224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
The association between psoriasis and increased cancer risk is gaining recognition as studies reveal shared inflammatory and immune pathways. This review examines the relationship between psoriasis and neoplasia, focusing on cancer risk factors in psoriasis patients, the biological pathways underlying this connection, and the impact of various psoriasis treatments on cancer development. Psoriasis patients have a heightened incidence of certain cancers, such as lymphomas, skin cancers, and urological malignancies, potentially linked to immune dysregulation and chronic inflammation. Immunomodulatory treatments for psoriasis, including conventional systemic therapies and biologics, present varied cancer risks, with others, such as phototherapy, associated with an elevated risk of skin cancers. For oncologic patients with psoriasis, management necessitates a tailored approach, balancing effective psoriasis control with minimizing cancer progression risks. The emergence of IL-17 inhibitors, IL-23 inhibitors, and small-molecule therapies offers promising therapeutic alternatives with favorable safety profiles for these patients. This review underscores the need for interdisciplinary collaboration to optimize care for patients managing both psoriasis and malignancy.
Collapse
Affiliation(s)
- Larisa-Alexandra Mateescu
- 2nd Department of Dermatology, Colentina Clinical Hospital, 020125 Bucharest, Romania; (C.-C.M.); (C.-D.V.); (E.-D.Ș.); (Ș.B.); (M.-M.C.)
| | - Alexandra-Petruța Savu
- 2nd Department of Dermatology, Colentina Clinical Hospital, 020125 Bucharest, Romania; (C.-C.M.); (C.-D.V.); (E.-D.Ș.); (Ș.B.); (M.-M.C.)
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.P.); (A.-C.N.)
| | - Costina-Cristiana Mutu
- 2nd Department of Dermatology, Colentina Clinical Hospital, 020125 Bucharest, Romania; (C.-C.M.); (C.-D.V.); (E.-D.Ș.); (Ș.B.); (M.-M.C.)
| | - Cezara-Diana Vaida
- 2nd Department of Dermatology, Colentina Clinical Hospital, 020125 Bucharest, Romania; (C.-C.M.); (C.-D.V.); (E.-D.Ș.); (Ș.B.); (M.-M.C.)
| | - Elena-Daniela Șerban
- 2nd Department of Dermatology, Colentina Clinical Hospital, 020125 Bucharest, Romania; (C.-C.M.); (C.-D.V.); (E.-D.Ș.); (Ș.B.); (M.-M.C.)
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.P.); (A.-C.N.)
| | - Ștefana Bucur
- 2nd Department of Dermatology, Colentina Clinical Hospital, 020125 Bucharest, Romania; (C.-C.M.); (C.-D.V.); (E.-D.Ș.); (Ș.B.); (M.-M.C.)
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.P.); (A.-C.N.)
| | - Elena Poenaru
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.P.); (A.-C.N.)
| | - Alin-Codruț Nicolescu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.P.); (A.-C.N.)
- EgoClinic, District 1, 010235 Bucharest, Romania
| | - Maria-Magdalena Constantin
- 2nd Department of Dermatology, Colentina Clinical Hospital, 020125 Bucharest, Romania; (C.-C.M.); (C.-D.V.); (E.-D.Ș.); (Ș.B.); (M.-M.C.)
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (E.P.); (A.-C.N.)
| |
Collapse
|
10
|
Awasthi S, Nijhawan M, Mishra A, Gupta A. Comparing the efficacy of oral apremilast, intralesional corticosteroids, and their combination in patients with patchy alopecia areata: a randomized clinical controlled trial. Arch Dermatol Res 2024; 317:129. [PMID: 39673617 DOI: 10.1007/s00403-024-03642-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024]
Abstract
INTRODUCTION Alopecia areata (AA) is a chronic, immune-mediated inflammatory disorder characterized by nonscarring hair loss. The management of AA poses challenges due to its unpredictable course and variable response to treatment. In this comparative study, we evaluated the efficacy and safety of oral apremilast, intralesional corticosteroids (ILC) and a combination of both in patients with patchy AA. METHODS Sixty patients with patchy AA were randomly assigned to three treatment groups: oral apremilast (Group A), ILC (Group B), and a combination of both (Group C). The Severity of Alopecia Tool (SALT) score was used to assess the extent of hair loss before treatment, after 3 months, and at 6 months of follow-up. Adverse events and complications were also monitored. The changes in SALT score from baseline between the three groups were assessed by using non-parametric statistical tests. The statistical significance was judged at 5% level of significance. RESULTS Findings demonstrated significant higher reduction in median SALT scores after treatment i.e., 2.47 (1.76, 5.07), p < 0.001 as well as after six months follow up 5.08 (3.80, 7.53), p < 0.001 in patients treated with ILC compared to other two groups. Neither apremilast monotherapy nor its combination with ILC demonstrated statistically significant improvement, although individual responses were observed. Complications were minimal, with transient pain and burning sensation reported during ILC injections and a few cases of gastritis and relapse in the oral apremilast group. CONCLUSION These findings suggest that ILC remains an effective treatment option for patchy AA. This study did not demonstrate statistically significant efficacy of oral apremilast, either as monotherapy or in combination with ILC, though larger studies may be needed to evaluate potential benefits in specific patient subgroups. Further research with larger sample sizes and longer-term follow-up is needed to validate these findings and optimize treatment approaches for AA.
Collapse
Affiliation(s)
- Sankalp Awasthi
- Department of Dermatology, Venereology and Leprology, Mahatma Gandhi Medical College and Hospital, Jaipur, Rajasthan, India
| | - Manisha Nijhawan
- Department of Dermatology, Venereology and Leprology, Mahatma Gandhi Medical College and Hospital, Jaipur, Rajasthan, India
| | - Akash Mishra
- Centre of Biostatistics, (Department of Community Medicine), Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Anviti Gupta
- Department of Dermatology, Venereology and Leprology, Mahatma Gandhi Medical College and Hospital, Jaipur, Rajasthan, India.
| |
Collapse
|
11
|
Qiao X, Cao X, Xu S, Wang C, Guo R, Yao X, Zhang Q. Menisoxoisoaporphine A, a novel oxoisoaporphine alkaloid from Menispermi Rhizoma, inhibits inflammation by targeting PDE4B. Front Pharmacol 2024; 15:1505116. [PMID: 39691395 PMCID: PMC11649434 DOI: 10.3389/fphar.2024.1505116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
Background Dysregulated and excessive inflammatory reactions can lead to tissue damage, which is the underlying cause of most human diseases. Menisoxoisoaporphine A (MA), a novel oxoisoaporphine alkaloid, was obtained from Menispermi Rhizoma, a traditional Chinese medicinal herb used in the treatment of inflammatory conditions in clinical practice. This suggests that MA has very promising potential for the development of anti-inflammatory therapeutics. Hence, this study aims to investigate the anti-inflammatory effects and underlying mechanisms of MA. Method The anti-inflammatory effects of MA were evaluated in lipopolysaccharide (LPS)-induced mouse macrophage RAW264.7 cells. Its underlying mechanisms were explored through RNA sequencing and Western blotting. The binding modes and interactions sites between MA and phosphodiesterase 4B (PDE4B) were predicted using molecular docking and validated by molecular dynamics simulation. Results MA treatment significantly reduced LPS-induced morphological changes, inflammatory cytokine relesae, and proinflammatory genes expression in RAW264.7 cells compared to the LPS-induced controls. Transcriptome sequencing analysis suggested that PDE4B might be a key target for MA to exert its therapeutic effect. Mechanismly, MA directly acted on Tyr405 site of PDE4B, thus leading to a sustained elevation of the cyclic adenosine monophosphate (cAMP) levels, which subsequently inactivated NF-κB signaling pathway by phosphorylating protein kinase A (PKA). MA inhibited the NF-κB-mediated inflammatory response depending on PDE4B. Conclusion MA, a natural and novel compound, exerted anti-inflammatory effects in LPS-induced RAW264.7 macrophage cells. It demonstrated a strong binding ability to the Tyr405 sites of PDE4B, thereby inhibiting NF-κB signaling pathway by regulating the cAMP-PKA axis. Elucidating the interaction between MA and PDE4B holds significant potential for the advancement of innovative therapeutic strategies aimed at inflammatory diseases. By strategically modulating this interaction, it may be feasible to achieve more precise regulation of inflammatory responses, thereby offering promising therapeutic benefits for conditions such as rheumatoid arthritis, asthma, and inflammatory bowel disease.
Collapse
Affiliation(s)
- Xin Qiao
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan, China
| | - Xiaojuan Cao
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan, China
| | - Shuang Xu
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan, China
- Academy of Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Cunlin Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan, China
| | - Rui Guo
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan, China
| | - Xiaojuan Yao
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan, China
| | - Qiong Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
- Medicinal Basic Research Innovation Center of Chronic Kidney Disease, Ministry of Education, Shanxi Medical University, Taiyuan, China
- Shanxi Provincial Key Laboratory of Drug Synthesis and Novel Pharmaceutical Preparation Technology, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
12
|
Chen Q, Xia Y, Liu HN, Chi Y, Li X, Shan LS, Dai B, Zhu Y, Wang YT, Miao X, Sun Q. Synthetic approaches and clinical application of representative small-molecule inhibitors of phosphodiesterase. Eur J Med Chem 2024; 277:116769. [PMID: 39163778 DOI: 10.1016/j.ejmech.2024.116769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/25/2024] [Accepted: 08/11/2024] [Indexed: 08/22/2024]
Abstract
Phosphodiesterases (PDEs) constitute a family of enzymes that play a pivotal role in the regulation of intracellular levels of cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Dysregulation of PDE activity has been implicated in diverse pathological conditions encompassing cardiovascular disorders, pulmonary diseases, and neurological disorders. Small-molecule inhibitors targeting PDEs have emerged as promising therapeutic agents for the treatment of these ailments, some of which have been approved for their clinical use. Despite their success, challenges such as resistance mechanisms and off-target effects persist, urging continuous research for the development of next-generation PDE inhibitors. The objective of this review is to provide an overview of the synthesis and clinical application of representative approved small-molecule PDE inhibitors, with the aim of offering guidance for further advancements in the development of novel PDE inhibitors.
Collapse
Affiliation(s)
- Qingqing Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Xia
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - He-Nan Liu
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuan Chi
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Xun Li
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Li-Shen Shan
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Bing Dai
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Zhu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Ya-Tao Wang
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium.
| | - Xinxin Miao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Qian Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
13
|
Schmidt MF, Albuscheit N, Yazdi AS. [Phosphodiesterase 4 inhibitors in dermatology : Role in the treatment of skin diseases]. DERMATOLOGIE (HEIDELBERG, GERMANY) 2024; 75:791-797. [PMID: 39212723 DOI: 10.1007/s00105-024-05407-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Chronic inflammatory skin diseases are of great social and medical importance and require effective drug therapy. Phosphodiesterase 4 (PDE4) inhibitors represent a possible therapeutic option by regulating inflammatory processes. PDEs cause the release of proinflammatory cytokines by interfering with signaling pathways. The PDE4 inhibitors apremilast (treatment of psoriasis and Behçet's disease), roflumilast (treatment of chronic obstructive pulmonary disease), and crisaborole (treatment of atopic dermatitis) are currently approved in Europe. PSORIASIS Apremilast is used for second-line treatment of plaque psoriasis and psoriatic arthritis and has a favorable side effect profile. Topical PDE4 inhibitors are currently being researched and have not yet been approved by the European Medicines Agency (EMA). ATOPIC DERMATITIS The topical PDE4 inhibitor crisaborole was approved by the EMA in 2020 as a topical treatment alternative to glucocorticoids and calcineurin inhibitors. Although the substance has shown good tolerability in studies and also alleviates the accompanying itching, it did not find its way onto the German market. BEHçET'S DISEASE: Apremilast is approved for the treatment of Behçet's disease in adults with refractory, severe oral ulcers. OUTLOOK Case studies have also demonstrated the efficacy of systemic PDE4 inhibition in other skin diseases (including blistering autoimmune dermatoses, lichen planus, and acantholytic genodermatoses). The substances are also being researched and used to treat extracutaneous inflammatory diseases.
Collapse
Affiliation(s)
- Morna F Schmidt
- Uniklinik RWTH Aachen, Klinik für Dermatologie und Allergologie, Morillenhang 27, 52074, Aachen, Deutschland.
| | - Nicole Albuscheit
- Uniklinik RWTH Aachen, Klinik für Dermatologie und Allergologie, Morillenhang 27, 52074, Aachen, Deutschland
| | - Amir S Yazdi
- Uniklinik RWTH Aachen, Klinik für Dermatologie und Allergologie, Morillenhang 27, 52074, Aachen, Deutschland
| |
Collapse
|
14
|
Zhang Y, Yang Y, Liang H, Liang Y, Xiong G, Lu F, Yang K, Zou Q, Zhang X, Du G, Xu X, Hao J. Nobiletin, as a Novel PDE4B Inhibitor, Alleviates Asthma Symptoms by Activating the cAMP-PKA-CREB Signaling Pathway. Int J Mol Sci 2024; 25:10406. [PMID: 39408735 PMCID: PMC11477036 DOI: 10.3390/ijms251910406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Asthma is a chronic airway inflammation that is considered a serious public health concern worldwide. Nobiletin (5,6,7,8,3',4'-hexamethyl flavonoid), an important compound isolated from several traditional Chinese medicines, especially Citri Reticulatae Pericarpium, is widely used for a number of indications, including cancer, allergic diseases, and chronic inflammation. However, the mechanism by which nobiletin exerts its anti-asthmatic effect remains unclear. In this research, we comprehensively demonstrated the anti-asthmatic effects of nobiletin in an animal model of asthma. It was found that nobiletin significantly reduced the levels of inflammatory cells and cytokines in mice and alleviated airway hyperresponsiveness. To explore the target of nobiletin, we identified PDE4B as the target of nobiletin through pharmacophore modeling, molecular docking, molecular dynamics simulation, SPR, and enzyme activity assays. Subsequently, it was found that nobiletin could activate the cAMP-PKA-CREB signaling pathway downstream of PDE4B in mouse lung tissues. Additionally, we studied the anti-inflammatory and anti-airway remodeling effects of nobiletin in LPS-induced RAW264.7 cells and TGF-β1-induced ASM cells, confirming the activation of the cAMP-PKA-CREB signaling pathway by nobiletin. Further validation in PDE4B-deficient RAW264.7 cells confirmed that the increase in cAMP levels induced by nobiletin depended on the inhibition of PDE4B. In conclusion, nobiletin exerts anti-asthmatic activity by targeting PDE4B and activating the cAMP-PKA-CREB signaling pathway.
Collapse
Affiliation(s)
- Yan Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.Z.); (Y.Y.); (H.L.); (Y.L.); (G.X.); (F.L.); (K.Y.); (Q.Z.); (X.Z.)
| | - Yaping Yang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.Z.); (Y.Y.); (H.L.); (Y.L.); (G.X.); (F.L.); (K.Y.); (Q.Z.); (X.Z.)
| | - Huicong Liang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.Z.); (Y.Y.); (H.L.); (Y.L.); (G.X.); (F.L.); (K.Y.); (Q.Z.); (X.Z.)
| | - Yuerun Liang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.Z.); (Y.Y.); (H.L.); (Y.L.); (G.X.); (F.L.); (K.Y.); (Q.Z.); (X.Z.)
| | - Guixin Xiong
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.Z.); (Y.Y.); (H.L.); (Y.L.); (G.X.); (F.L.); (K.Y.); (Q.Z.); (X.Z.)
| | - Fang Lu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.Z.); (Y.Y.); (H.L.); (Y.L.); (G.X.); (F.L.); (K.Y.); (Q.Z.); (X.Z.)
| | - Kan Yang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.Z.); (Y.Y.); (H.L.); (Y.L.); (G.X.); (F.L.); (K.Y.); (Q.Z.); (X.Z.)
| | - Qi Zou
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.Z.); (Y.Y.); (H.L.); (Y.L.); (G.X.); (F.L.); (K.Y.); (Q.Z.); (X.Z.)
| | - Xiaomin Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.Z.); (Y.Y.); (H.L.); (Y.L.); (G.X.); (F.L.); (K.Y.); (Q.Z.); (X.Z.)
| | - Guanhua Du
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China;
| | - Ximing Xu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.Z.); (Y.Y.); (H.L.); (Y.L.); (G.X.); (F.L.); (K.Y.); (Q.Z.); (X.Z.)
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China;
- Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Jiejie Hao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.Z.); (Y.Y.); (H.L.); (Y.L.); (G.X.); (F.L.); (K.Y.); (Q.Z.); (X.Z.)
- Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China;
| |
Collapse
|
15
|
Zhang Q, Yu L, Wan L, Chen L, Chen J. Case report: Managing pemphigus foliaceus using apremilast without systemic glucocorticosteroids or immunosuppressive agents. Front Immunol 2024; 15:1408116. [PMID: 39139566 PMCID: PMC11319141 DOI: 10.3389/fimmu.2024.1408116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Pemphigus foliaceus (PF) is a superficial form of pemphigus. Treatment options for PF resemble pemphigus vulgaris, including glucocorticosteroids, immunosuppressive agents and rituximab et al. These treatment approaches can effectively improve the condition but may also be accompanied by high risks of side effects. Therefore, it is crucial to find a safe and effective treatment options for patients with PF. It will not only benefit/be necessary for patients who refuse glucocorticosteroids or immunosuppressive agents treatments, but also for patients who cannot be treated with glucocorticosteroids or immunosuppressive agents. Herein, we reported a case of PF that was treated with apremilast without systemic glucocorticosteroids or immunosuppressive agents. A 54-year-old woman presented with itchy erythema and erosions on the trunk for more than 1 month. The patient applied mometasonefuroate cream without improvement for a duration of two weeks. The past history of diabetes mellitus and atrophic gastritis was reported. Physical examination revealed scattered erythematous macules and erosions on the trunk. No mucosal involvement was observed. The condition was assessed by the pemphigus disease area index and numerical rating scale, with baseline scores of 7 and 8, respectively. Histopathological examination showed acantholysis and intraepithelial blister. Direct immunofluorescence revealed the presence of IgG and Complement 3 deposition between the acanthocytes with the reticular distribution. Based on enzyme-linked immunosorbent assay results, the levels of Dsg1 and Dsg3 antibodies were 28.18 and 0.26 kU/L respectively. The diagnosis of PF was made. This patient was successfully treated with apremilast without systemic glucocorticosteroids or immunosuppressive agents. The patient has continued with apremilast 30mg once daily for maintenance and no adverse events related to apremilast such as gastrointestinal side effects were observed during the 9-month follow-up period. In conclusion, apremilast therapy without systemic glucocorticosteroids nor immunosuppressive agents might provide an effective alternative to management of mild PF without obvious side effect.
Collapse
Affiliation(s)
- Quanhong Zhang
- Department of Dermatology, Wuhan No.1 Hospital, Wuhan, China
- Department of Dermatology, Traditional Chinese and Western Medicine Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lang Yu
- Department of Dermatology, Wuhan No.1 Hospital, Wuhan, China
- Jianghan University School of Medicine, Wuhan, China
| | - Li Wan
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Liuqing Chen
- Department of Dermatology, Wuhan No.1 Hospital, Wuhan, China
- Hubei Province & Key Laboratory of Skin Infection And Immunity, Wuhan, China
| | - Jinbo Chen
- Department of Dermatology, Wuhan No.1 Hospital, Wuhan, China
- Hubei Province & Key Laboratory of Skin Infection And Immunity, Wuhan, China
| |
Collapse
|
16
|
Campione E, Zarabian N, Cosio T, Borselli C, Artosi F, Cont R, Sorge R, Shumak RG, Costanza G, Rivieccio A, Gaziano R, Bianchi L. Apremilast as a Potential Targeted Therapy for Metabolic Syndrome in Patients with Psoriasis: An Observational Analysis. Pharmaceuticals (Basel) 2024; 17:989. [PMID: 39204094 PMCID: PMC11357209 DOI: 10.3390/ph17080989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 09/03/2024] Open
Abstract
Psoriasis (PsO) is a chronic inflammatory dermatosis that often presents with erythematous, sharply demarcated lesions. Although psoriasis is primarily a dermatological disease, its immune-mediated pathogenesis produces systemic effects and is closely associated with various comorbid conditions such as cardiovascular disease (CVD), metabolic syndrome (MetS), and diabetes mellitus type II (DMII). Apremilast, an oral phosphodiesterase 4 (PDE-4) inhibitor, has shown promise in treating moderate-to-severe psoriasis and is associated with potential cardiometabolic benefits. In a 12-month prospective observational study involving 137 patients with moderate-to-severe psoriasis, we assessed changes in psoriasis clinimetric scores and metabolic profiles from baseline (T0) to 52 weeks (T1) to evaluate the efficacy of apremilast. After 52 weeks of apremilast treatment, we documented a statistically significant decrease in low-density lipoprotein (LDL) and total cholesterol, triglycerides, and glucose levels. Our findings even suggest a potential synergistic effect among patients treated with apremilast, alongside concomitant statin and/or insulin therapy. Although the results of our study must be validated on a larger scale, the use of apremilast in the treatment of psoriatic patients with cardio-metabolic comorbidities yields promising results.
Collapse
Affiliation(s)
- Elena Campione
- Dermatology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy; (C.B.); (F.A.); (R.C.); (R.G.S.); (A.R.); (L.B.)
| | - Nikkia Zarabian
- School of Medicine and Health Sciences, George Washington University, 2300 I St NW, Washington, DC 20052, USA;
| | - Terenzio Cosio
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (T.C.); (R.G.)
- Dynamyc Research Team 7380, Université de Paris-Est-Créteil, 94000 Créteil, France
| | - Cristiana Borselli
- Dermatology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy; (C.B.); (F.A.); (R.C.); (R.G.S.); (A.R.); (L.B.)
| | - Fabio Artosi
- Dermatology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy; (C.B.); (F.A.); (R.C.); (R.G.S.); (A.R.); (L.B.)
| | - Riccardo Cont
- Dermatology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy; (C.B.); (F.A.); (R.C.); (R.G.S.); (A.R.); (L.B.)
| | - Roberto Sorge
- Laboratory of Biometry, Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Ruslana Gaeta Shumak
- Dermatology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy; (C.B.); (F.A.); (R.C.); (R.G.S.); (A.R.); (L.B.)
| | - Gaetana Costanza
- Unit of Virology, Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy;
| | - Antonia Rivieccio
- Dermatology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy; (C.B.); (F.A.); (R.C.); (R.G.S.); (A.R.); (L.B.)
| | - Roberta Gaziano
- Department of Experimental Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy; (T.C.); (R.G.)
| | - Luca Bianchi
- Dermatology Unit, Department of Systems Medicine, Tor Vergata University Hospital, 00133 Rome, Italy; (C.B.); (F.A.); (R.C.); (R.G.S.); (A.R.); (L.B.)
| |
Collapse
|
17
|
James J, Otto T, Gao J, Porter ML. Oral Psoriasis Therapies. Dermatol Clin 2024; 42:357-363. [PMID: 38796267 DOI: 10.1016/j.det.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2024]
Abstract
Oral psoriasis therapies include both older traditional immunosuppressants, such as methotrexate, cyclosporine, and acitretin, as well as newer, more targeted agents, such as apremilast, deucravacitinib, and oral interleukin-23 receptor antagonists. Patients may prefer oral therapies to injectable therapies based on the route of administration. Both older and newer oral psoriasis therapies can be utilized effectively in the treatment of psoriasis. Here, we will review oral agents used in the treatment of psoriasis as well as provide commentary on their role in our current, evolving psoriasis treatment paradigm.
Collapse
Affiliation(s)
- JaBreia James
- Harvard Combined Dermatology Residency Program, Department of Dermatology, Harvard Medical School, 55 Fruit Street, BH 616 Boston, MA 02114, USA
| | - Tracey Otto
- Department of Dermatology, Clinical Laboratory for Epidemiology and Applied Research in Skin (CLEARS), Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02115, USA
| | - Julia Gao
- Department of Dermatology, Clinical Laboratory for Epidemiology and Applied Research in Skin (CLEARS), Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02115, USA
| | - Martina L Porter
- Department of Dermatology, Clinical Laboratory for Epidemiology and Applied Research in Skin (CLEARS), Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA 02115, USA; Department of Dermatology, Harvard Medical School, 3 Blackfan Cir, Boston, MA 02115, USA.
| |
Collapse
|
18
|
Fukasawa T, Yoshizaki-Ogawa A, Enomoto A, Sato S, Yoshizaki A. Apremilast Decreased Proinflammatory Cytokines and Subsequently Increased Inhibitory ones in Psoriasis: A Prospective Cohort Study. Acta Derm Venereol 2024; 104:adv37555. [PMID: 38738774 PMCID: PMC11107833 DOI: 10.2340/actadv.v104.37555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/18/2024] [Indexed: 05/14/2024] Open
Abstract
Abstract is missing (Short communication)
Collapse
Affiliation(s)
- Takemichi Fukasawa
- Department of Dermatology, Psoriasis center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan; Department of Clinical Cannabinoid Research, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| | - Asako Yoshizaki-Ogawa
- Department of Dermatology, Psoriasis center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Atsushi Enomoto
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, Psoriasis center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, Psoriasis center, The University of Tokyo Graduate School of Medicine, Tokyo, Japan; Department of Clinical Cannabinoid Research, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
19
|
Bule P, Kadkanje P, Kshirsagar R, Puppala ER, Naidu VGM, Chella N. Formulation characterization of lecithin organogel as topical drug delivery system for psoriasis: In-vitro permeation and preclinical evaluation. Drug Dev Res 2024; 85:e22191. [PMID: 38685610 DOI: 10.1002/ddr.22191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 03/21/2024] [Accepted: 04/14/2024] [Indexed: 05/02/2024]
Abstract
Psoriasis is a chronic inflammatory and proliferative skin disease that causes pathological skin changes and has a substantial impact on the quality of patient life. Apremilast was approved by the US Food and Drug Administration as an oral medication for psoriasis and is beneficial in mild to moderate conditions for chronic usage. However, 5%-7% of withdrawals were reported due to severe side effects. To address the issue, a localized drug delivery strategy via the topical route may be a viable approach. However, poor physicochemical properties make it vulnerable to passing through the skin, requiring a specialized drug delivery system to demonstrate its full potential via a topical route like lecithin organogel. The formulation was optimized by screening the suitable lecithin type and non-polar solvents based on the gel formation ability of lecithin and the solubility of apremilast in the solvent. The pseudo-ternary diagram was used to optimize the water content required to form the gel. The optimized gel was found to be shear thinning characterized for rheological parameters, in-vitro diffusion studies, and in-vitro skin distribution studies. Preclinical studies in Imiquimod-induced mice showed a better reduction in severity index, cytokine levels, and epidermal hyperplasia from the lecithin organogel group compared to the apremilast oral administration and marketed standard topical gel group. Based on these results, lecithin organogel can be considered a promising approach to deliver molecules like apremilast by topical route in psoriatic-like conditions.
Collapse
Affiliation(s)
- Prajakta Bule
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Prashant Kadkanje
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Ravikiran Kshirsagar
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Eswara Rao Puppala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Vegi Ganga Modi Naidu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Naveen Chella
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| |
Collapse
|
20
|
Zhou Y, Xiao Y, Wang Y, Li W. Refractory atypical IgA pemphigus successfully treated with apremilast. J Dermatol 2024; 51:e86-e87. [PMID: 37864455 DOI: 10.1111/1346-8138.17007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/22/2023] [Accepted: 09/29/2023] [Indexed: 10/22/2023]
Affiliation(s)
- Yuxi Zhou
- Department of Dermatology & Venerology, Rare Diseases Center, Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yue Xiao
- Department of Dermatology & Venerology, Rare Diseases Center, Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yiyi Wang
- Department of Dermatology & Venerology, Rare Diseases Center, Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei Li
- Department of Dermatology & Venerology, Rare Diseases Center, Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
21
|
Li Y, Wang M, Su J, Zhong R, Yin S, Zhao Z, Sun Z. Hypersampsonone H attenuates ulcerative colitis via inhibition of PDE4 and regulation of cAMP/PKA/CREB signaling pathway. Int Immunopharmacol 2024; 128:111490. [PMID: 38218008 DOI: 10.1016/j.intimp.2024.111490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/15/2023] [Accepted: 01/02/2024] [Indexed: 01/15/2024]
Abstract
BACKGROUND AND OBJECTIVES Ulcerative colitis (UC) is a recurrent intestinal inflammatory disease which poses a serious threat to the life of patients. However, there are no specific drugs for UC yet. Hypericum sampsonii Hance (HS) is a Chinese herbal medicine traditionally used to treat enteritis and dysentery. Our previous studies have demonstrated that HS holds potential anti-UC effects, and a novel compound named Hypersampsonone H (HS-1) isolated from HS possesses significant anti-inflammatory activity. However, the beneficial effects of HS-1 on UC remain unclear. This study aimed to investigate the therapeutic effects of HS-1 on UC and its potential mechanisms, both in vitro and in vivo. METHODS The in vitro model was employed using LPS-induced RAW264.7 cells to investigate the anti-inflammatory effects of HS-1 and its possible mechanisms. Furthermore, the therapeutic efficacy and potential mechanisms of HS-1 against dextran sulfate sodium (DSS)-induced acute colitis were assessed through histopathological examination, biochemical analysis, and molecular docking. RESULTS In vitro, HS-1 significantly reduced LPS-induced inflammatory responses, as indicated by inhibiting NO production, down-regulating the overexpression of COX-2 and iNOS, as well as regulating the imbalanced levels of IL-6, TNF-α, and IL-10. Moreover, HS-1 also inhibited the expression of PDE4, elevated the intracellular cAMP level, and promoted the phosphorylation of CREB, thereby activating the PKA/CREB pathway in RAW264.7 cells. In vivo, HS-1 demonstrated therapeutic capacity against DSS-induced colitis by alleviating the symptoms of colitis mice, regulating the abnormal expression of inflammatory mediators, protecting the integrity of intestinal epithelial barrier, and reducing tissue fibrosis. Consistently, HS-1 was found to decrease the expression of PDE4 isoforms, subsequently activating the cAMP/PKA/CREB signaling pathway. Furthermore, the molecular docking results indicated that HS-1 exhibited a high affinity for PDE4, particularly PDE4D. Further mechanistic validation in vitro demonstrated that HS-1 possessed a synergistic effect on forskolin and an antagonistic effect on H-89 dihydrochloride, thereby exerting anti-inflammatory effects through the cAMP/PKA/CREB signaling pathway. CONCLUSION We disclose that HS-1 serves as a promising candidate drug for the treatment of UC by virtue of its ability to reduce DSS-induced colitis via the inhibition of PDE4 and the activation of cAMP/PKA/CREB signaling pathway.
Collapse
Affiliation(s)
- Yanzhen Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Mingqiang Wang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jianhui Su
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Ruimin Zhong
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan 512005, China
| | - Sheng Yin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhongxiang Zhao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Zhanghua Sun
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan 512005, China.
| |
Collapse
|
22
|
Tsiogkas SG, Karamitrou EK, Grammatikopoulou MG, Zafiriou E, Bogdanos DP. Efficacy of tyrosine-kinase-2 and phosphodiesterase-4 inhibitors for scalp psoriasis: a systematic review and meta-analysis. Curr Med Res Opin 2024; 40:155-163. [PMID: 37997745 DOI: 10.1080/03007995.2023.2288280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/22/2023] [Indexed: 11/25/2023]
Abstract
OBJECTIVES Psoriasis of the scalp is challenging to manage. The only approved oral tyrosine kinase 2 and phosphodiesterase 4 inhibitors for psoriasis are deucravacitinib and apremilast. The aim of this study was to explore their efficacy for scalp psoriasis utilizing data from randomized controlled trials. METHODS We searched Medline, Scopus, Web of Science, CENTRAL, and ClinicalTrials.gov up to August 4, 2023. To determine risk of bias, the revised Risk of Bias assessment tool 2.0 was used. Inverse variance random effects meta-analyses were executed. Heterogeneity was assessed utilizing Q and I2 statistics. Pre-determined outcomes included the proportion of participants with cleared scalp skin (Scalp Physician's Global Assessment [ScPGA] of 0/1), mean change in Psoriasis Scalp Severity Index (PSSI), and mean improvement in Dermatology Life Quality Index (DLQI). RESULTS Ten RCTs fulfilled inclusion criteria. Both apremilast (RR = 2.41, 95% CI = 2.08-2.79, Tau2 = 0, I2 = 0) and deucravacitinib (RR = 3.86, 95% CI = 3.02-4.94, Tau2 = 0, I2 = 0) were more effective in inducing ScPGA of 0/1 at 16 weeks compared to placebo. Furthermore, deucravacitinib was more effective than apremilast (RR = 1.70, 95% CI = 1.44-2.00, Tau2 = 0, I2 = 0). An analysis could not be executed for the rest of the outcomes. CONCLUSIONS Apremilast and deucravacitinib are effective for scalp psoriasis. Deucravacitinib may be more efficient in clearing the scalp.
Collapse
Affiliation(s)
- Sotirios G Tsiogkas
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | | | - Maria G Grammatikopoulou
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Efterpi Zafiriou
- Department of Dermatology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Dimitrios P Bogdanos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
23
|
Fogleman BM, McKinnon E, Nebeker S, Arukala KS. Severe Electrolyte Abnormalities and Distal Renal Tubular Acidosis in the Setting of Apremilast Use for Psoriatic Arthritis: A Case Report. Cureus 2024; 16:e53514. [PMID: 38440019 PMCID: PMC10910423 DOI: 10.7759/cureus.53514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2024] [Indexed: 03/06/2024] Open
Abstract
Renal tubular acidosis (RTA) involves dysfunction of the renal tubular system, which leads to electrolyte abnormalities and acid-base dysregulation. The case we present here discusses a patient with a past medical history of psoriatic arthritis who presented to the emergency department with progressive generalized weakness and anorexia in the preceding four weeks. She was found to have profound hypokalemia (1.2 mmol/L), hyperchloremic metabolic acidosis, and multiple other electrolyte abnormalities. Following an extensive workup, her principle problem was deemed to be distal (type 1) RTA. She was treated with sodium bicarbonate, spironolactone, and aggressive rehydration, which eventually led to the stabilization of her electrolytes alongside clinical improvement over the course of an eight-day hospitalization. The workup did not reveal a clear etiology for the RTA. One month prior to hospitalization, she was started on apremilast, a new medication for her psoriatic arthritis. Given the limited availability of alternative explanations and the temporality of clinical manifestations, our findings raise suspicion that apremilast might be associated with her clinical presentation.
Collapse
Affiliation(s)
- Brody M Fogleman
- Internal Medicine, Edward Via College of Osteopathic Medicine, Spartanburg, USA
| | - Emilie McKinnon
- Internal Medicine, Grand Strand Medical Center, Myrtle Beach, USA
| | - Schuyler Nebeker
- Internal Medicine, Grand Strand Medical Center, Myrtle Beach, USA
| | | |
Collapse
|
24
|
Chikhalkar SB, Gupta S, Chopade SU, Jadhav S, Kharkar V. Successful Remission with Combination of Diphenylcyclopropenone and Apremilast in Refractory Extensive Alopecia Areata. Int J Trichology 2024; 16:47-49. [PMID: 40309379 PMCID: PMC12039770 DOI: 10.4103/ijt.ijt_68_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/03/2022] [Accepted: 09/19/2022] [Indexed: 05/02/2025] Open
Abstract
Extensive alopecia areata including alopecia totalis and alopecia universalis is a therapeutic challenge in majority of the cases due to poor response to therapy, frequent relapses, minimal chances of spontaneous remission, and the associated psychological distress. Topical immunotherapy with diphenylcyclopropenone (DPCP) is one of the established therapeutic modalities for alopecia areata, although with a variable response rate. The use of systemic corticosteroids and other immunosuppressive drugs such as cyclosporine, azathioprine, and methotrexate is limited by their side effects. Apremilast, a phosphodiesterase-4 inhibitor, marked with a high safety profile and minimal monitoring, has been used off-label in many dermatological conditions including alopecia areata. We report three cases of extensive refractory alopecia areata with successful remission following combination therapy of DPCP and apremilast.
Collapse
Affiliation(s)
- Siddhi B Chikhalkar
- Department of Dermatology, Seth GS Medical College and KEM Hospital, Mumbai, Maharashtra, India
| | - Savera Gupta
- Department of Dermatology, Seth GS Medical College and KEM Hospital, Mumbai, Maharashtra, India
| | - Snehal Umesh Chopade
- Department of Dermatology, Seth GS Medical College and KEM Hospital, Mumbai, Maharashtra, India
| | - Sushanti Jadhav
- Department of Dermatology, Seth GS Medical College and KEM Hospital, Mumbai, Maharashtra, India
| | - Vidya Kharkar
- Department of Dermatology, Seth GS Medical College and KEM Hospital, Mumbai, Maharashtra, India
| |
Collapse
|
25
|
Crowley EL, Gooderham MJ. Phosphodiesterase-4 Inhibition in the Management of Psoriasis. Pharmaceutics 2023; 16:23. [PMID: 38258034 PMCID: PMC10819567 DOI: 10.3390/pharmaceutics16010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/16/2023] [Accepted: 12/17/2023] [Indexed: 01/24/2024] Open
Abstract
Psoriasis is a common chronic immune-mediated disease with many comorbidities and impacts on quality of life. Among the treatments for psoriasis, phosphodiesterase-4 (PDE4) inhibitors are emerging with expanding options. PDE4 inhibitors play a pivotal role in the inflammatory cascade by degrading cyclic adenosine monophosphate (cAMP), contributing to pro-inflammatory mediator production. Apremilast, an oral PDE4 inhibitor, is approved for psoriasis. While effective, its adverse effects can limit its utility. Roflumilast, a topical PDE4 inhibitor, was also recently approved for psoriasis and shows promise in clinical trials. Crisaborole, a PDE4 inhibitor approved for atopic dermatitis, has also been studied in psoriasis. This review summarizes evidence from randomized clinical trials regarding the efficacy and safety of PDE4 inhibitors in psoriasis treatment. By highlighting their potential benefits and limitations, this review provides valuable insights for clinicians and researchers aiming to optimize psoriasis management.
Collapse
Affiliation(s)
- Erika L. Crowley
- Faculty of Medicine, University of British Columbia Okanagan, 3333 University Way, Kelowna, BC V1V 1V7, Canada;
| | - Melinda J. Gooderham
- SKiN Centre for Dermatology, 775 Monaghan Rd, Peterborough, ON K9J 5K2, Canada
- Probity Medical Research, 139 Union St E, Waterloo, ON N2J 1C4, Canada
- Department of Medicine, Queen’s University, 99 University Ave, Kingston, ON K7L 3N6, Canada
| |
Collapse
|
26
|
Perez-Bootello J, Cova-Martin R, Naharro-Rodriguez J, Segurado-Miravalles G. Vitiligo: Pathogenesis and New and Emerging Treatments. Int J Mol Sci 2023; 24:17306. [PMID: 38139134 PMCID: PMC10743634 DOI: 10.3390/ijms242417306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/02/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Vitiligo is a complex disease with a multifactorial nature and a high impact on the quality of life of patients. Although there are multiple therapeutic alternatives, there is currently no fully effective treatment for this disease. In the current era, multiple drugs are being developed for the treatment of autoimmune diseases. This review assesses the available evidence on the pathogenesis of vitiligo, and a comprehensive review of treatments available for vitiligo now and in the near future is provided. This qualitative analysis spans 116 articles. We reviewed the mechanism of action, efficacy and safety data of phototherapy, afamelanotide, cyclosporine, phosphodiesterase 4 inhibitors, trichloroacetic acid, basic fibroblast growth factor, tumor necrosis factor (TNF) inhibitors, secukinumab, pseudocatalase and janus kinase (JAK) inhibitors. At the moment, there is no clearly outstanding option or fully satisfactory treatment for vitiligo, so it is necessary to keep up the development of new drugs as well as the publication of long-term effectiveness and safety data for existing treatments.
Collapse
|
27
|
Haddad A, Stein N, Lavi I, Shynkar L, Bergman I, Feldhamer I, Cohen AD, Saliba W, Zisman D. Treatment Persistence of Apremilast Among Patients with Psoriatic Arthritis. Biologics 2023; 17:129-136. [PMID: 37814674 PMCID: PMC10560465 DOI: 10.2147/btt.s425693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/27/2023] [Indexed: 10/11/2023]
Abstract
Introduction Persistence in drug therapy reflects treatment effectiveness and tolerability. We aim to estimate the persistence of apremilast prescribed to patients with psoriatic arthritis (PsA) and to identify characteristics associated with treatment discontinuation in a real-world setting. Methods Patients with PsA treated with apremilast from January 2016 were identified from a large health database and followed until medication stop date (using 3-months grace period), death or the end of observation period (June 2021). Demographic data, Charlson comorbidity index and concomitant and previous use of conventional and biologic DMARDs were extracted. The reasons for drug discontinuation were manually retrieved from patient charts. Time to discontinuation was estimated using survival analysis using Kaplan-Meier functions. Results Overall, 568 PsA patients treated with apremilast were identified. The mean age was 55.3±14.0 years, of whom 332 (58.5%) were females, 38.4% were obese (BMI>30), 75.2% had a Charlson comorbidity index>1, 24.1% were on concomitant treatment with methotrexate and 72.4% were biologic naïve. The median persistent period was 6.1,95% CI (5.2-6.9) months in which only 16.9% remained persistent on apremilast. No difference was found with regard to age, sex, socioeconomic status, ethnicity and obesity between patients who were persistent compared to patients who discontinued apremilast. Concomitant treatment with methotrexate and prior history of biologic therapy did not affect drug persistency (log rank P=0.957 and 0.082, respectively). Causes for treatment discontinuation were due to lack of skin efficacy in 19.4%, lack of joint efficacy in 33.3%, combined skin and joint inefficacy at 2.3% and due to side effects in 24.1%. Conclusion In this large observational retrospective cohort of patients treated with apremilast, a relatively low drug persistence was observed with 6-month and 1-year survival rates of 50.3% and 31.3%, respectively. Treatment discontinuation was mainly due to joint inefficacy, advocating for more studies for proper patient selection to assure treatment effectiveness and persistency.
Collapse
Affiliation(s)
- Amir Haddad
- Rheumatology Unit, Carmel Medical Center, Haifa, Israel
| | - Nili Stein
- Department of Epidemiology, Clalit Health Services, Haifa, Israel
| | - Idit Lavi
- Department of Epidemiology, Clalit Health Services, Haifa, Israel
| | - Lisa Shynkar
- Internal Medicine Department, Carmel Medical Centre, Haifa, Israel
| | - Irina Bergman
- Internal Medicine Department, Carmel Medical Centre, Haifa, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ilan Feldhamer
- Chief Physician’s Office, Central Headquarters, Clalit Health Services, Tel Aviv, Israel
| | - Arnon Dov Cohen
- Chief Physician’s Office, Central Headquarters, Clalit Health Services, Tel Aviv, Israel
- Siaal Research Center for Family Medicine and Primary Care, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheba, Israel
| | - Walid Saliba
- Department of Epidemiology, Clalit Health Services, Haifa, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Devy Zisman
- Rheumatology Unit, Carmel Medical Center, Haifa, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
28
|
Le Joncour A, Régnier P, Maciejewski-Duval A, Charles E, Barete S, Fouret P, Rosenzwajg M, Klatzmann D, Cacoub P, Saadoun D. Reduction of Neutrophil Activation by Phosphodiesterase 4 Blockade in Behçet's Disease. Arthritis Rheumatol 2023; 75:1628-1637. [PMID: 36862398 DOI: 10.1002/art.42486] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/17/2023] [Accepted: 02/15/2023] [Indexed: 03/03/2023]
Abstract
OBJECTIVE Behçet's disease (BD) is a systemic vasculitis with inflammatory lesions mediated by cytotoxic T cells and neutrophils. Apremilast, an orally available small-molecule drug that selectively inhibits phosphodiesterase 4 (PDE4), has been recently approved for the treatment of BD. We aimed to investigate the effect of PDE4 inhibition on neutrophil activation in BD. METHODS We studied surface markers and reactive oxygen species (ROS) production by flow cytometry, and neutrophil extracellular traps (NETs) production and molecular signature of neutrophils by transcriptome analysis before and after PDE4 inhibition. RESULTS Activation surface markers (CD64, CD66b, CD11b, and CD11c), ROS production, and NETosis were up-regulated in BD patient neutrophils compared to healthy donor neutrophils. Transcriptome analysis revealed 1,021 significantly dysregulated neutrophil genes between BD patients and healthy donors. Among dysregulated genes, we found a substantial enrichment for pathways linked to innate immunity, intracellular signaling, and chemotaxis in BD. Skin lesions of BD patients showed increased infiltration of neutrophils that colocalized with PDE4. Inhibition of PDE4 by apremilast strongly inhibited neutrophil surface activation markers as well as ROS production, NETosis, and genes and pathways related to innate immunity, intracellular signaling, and chemotaxis. CONCLUSION We highlight key biologic effects of apremilast on neutrophils in BD.
Collapse
Affiliation(s)
- Alexandre Le Joncour
- Sorbonne Université, INSERM, UMR S 959, Immunology-Immunopathology-Immunotherapy (I3), Laboratoire d'excellence TRANSIMMUNOM, Paris, and Biotherapy (CIC-BTi), Hôpital Pitié-Salpêtrière, Paris, and AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, Paris, France
| | - Paul Régnier
- Sorbonne Université, INSERM, UMR S 959, Immunology-Immunopathology-Immunotherapy (I3), Paris, and Laboratoire d'excellence TRANSIMMUNOM, Paris, and Biotherapy (CIC-BTi), Hôpital Pitié-Salpêtrière, Paris, France
| | - Anna Maciejewski-Duval
- Sorbonne Université, INSERM, UMR S 959, Immunology-Immunopathology-Immunotherapy (I3), Paris, and Laboratoire d'excellence TRANSIMMUNOM, Paris, and Biotherapy (CIC-BTi), Hôpital Pitié-Salpêtrière, Paris, France
| | - Erwan Charles
- Sorbonne Université, INSERM, UMR S 959, Immunology-Immunopathology-Immunotherapy (I3), Paris, and Laboratoire d'excellence TRANSIMMUNOM, Paris, and Biotherapy (CIC-BTi), Hôpital Pitié-Salpêtrière, Paris, France
| | - Stéphane Barete
- Sorbonne Université, INSERM, UMR S 959, Immunology-Immunopathology-Immunotherapy (I3), Paris, and Laboratoire d'excellence TRANSIMMUNOM, Paris, and AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, Paris, and AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Unit of Dermatology, Paris, France
| | - Pierre Fouret
- AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Anatomopathology, Paris, France
| | - Michelle Rosenzwajg
- Sorbonne Université, INSERM, UMR S 959, Immunology-Immunopathology-Immunotherapy (I3), Paris, and Laboratoire d'excellence TRANSIMMUNOM, Paris, and Biotherapy (CIC-BTi), Hôpital Pitié-Salpêtrière, Paris, France
| | - David Klatzmann
- Sorbonne Université, INSERM, UMR S 959, Immunology-Immunopathology-Immunotherapy (I3), Paris, and Laboratoire d'excellence TRANSIMMUNOM, Paris, and Biotherapy (CIC-BTi), Hôpital Pitié-Salpêtrière, Paris, France
| | - Patrice Cacoub
- Sorbonne Université, INSERM, UMR S 959, Immunology-Immunopathology-Immunotherapy (I3), Laboratoire d'excellence TRANSIMMUNOM, Paris, and Biotherapy (CIC-BTi), Hôpital Pitié-Salpêtrière, Paris, and AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, Paris, France
| | - David Saadoun
- Sorbonne Université, INSERM, UMR S 959, Immunology-Immunopathology-Immunotherapy (I3), Laboratoire d'excellence TRANSIMMUNOM, Paris, and Biotherapy (CIC-BTi), Hôpital Pitié-Salpêtrière, Paris, and AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Department of Internal Medicine and Clinical Immunology, Paris, France
| |
Collapse
|
29
|
Ayan G, Ribeiro A, Macit B, Proft F. Pharmacologic Treatment Strategies in Psoriatic Arthritis. Clin Ther 2023; 45:826-840. [PMID: 37455227 DOI: 10.1016/j.clinthera.2023.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 07/18/2023]
Abstract
PURPOSE The goal of this narrative review was to provide current data on psoriatic arthritis (PsA) therapeutic strategies, supporting treatment decisions with a domain-based approach. METHODS This narrative review of treatment strategies for PsA focused on several disease domains (ie, peripheral arthritis, enthesitis, axial disease, dactylitis, skin and nail disease), as well as the so-called "related conditions" of uveitis, Crohn's disease, and ulcerative colitis. We searched PubMed, EMBASE, international guidelines, and recent congress abstracts. FINDINGS Currently, multiple approved treatment options offer a wide range of options, such as tumor necrosis factor (TNF) inhibitors; inhibitors of interleukin-17 (IL-17), IL-12/23 (IL-12/23), IL-23 (IL-23), and Janus kinase; the phosphodiesterase 4 inhibitor apremilast; and the T-cell modulator abatacept. However, no treatment option shows clear superiority concerning efficacy on peripheral arthritis and dactylitis over the others, whereas limited evidence suggests that the IL-17 inhibitor ixekizumab and the IL-12/23 inhibitor ustekinumab may be superior to TNF inhibitors in treating enthesitis. Recent data on enthesitis have also shown promising results for methotrexate. Treatment of axial PsA is mostly derived from axial spondyloarthritis, and more data are needed focusing on this specific subgroup of PsA patients. Thus far, the most important finding from the only randomized controlled trial in this specific population is that the IL-17 inhibitor secukinumab was superior to placebo in terms of clinical and radiologic end-points in axial PsA. Regarding psoriatic skin involvement, head-to-head trials in PsA as well as skin psoriasis showed the superiority of IL-17, IL-23, and IL-12/23 inhibitors over TNF inhibitors. When treating PsA with concurrent uveitis, according to the existing data, monoclonal TNF inhibitor antibodies should be preferred. In PsA and concomitant inflammatory bowel disease, treatment decisions must include the consideration of which specific type of inflammatory bowel disease (Crohn's disease or ulcerative colitis) is present, as some of the agents either lack data or are ineffective in treating these 2 conditions. In both types, IL-17 inhibitors should be avoided. When determining treatment strategy, comorbidities should be carefully assessed, and the corresponding risk profile of the respective treatment modalities should be taken into consideration. IMPLICATIONS There are many approved therapeutic options for treating patients with PsA, and additional emerging treatment options are in the pipeline. Individualized treatment decisions for each patient, depending on the leading disease phenotype, underlying comorbidities, and patient preferences, should be made based on shared decision-making.
Collapse
Affiliation(s)
- G Ayan
- Hacettepe University, Department of Internal Medicine, Division of Rheumatology, Ankara, Turkey
| | - A Ribeiro
- Hospital de Clínicas de Porto Alegre, Department of Rheumatology, Porto Alegre, Brazil
| | - Betul Macit
- Department of Dermatology, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Fabian Proft
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
30
|
Bernard JW, Reddy S, Flowers RH. The successful use of oral apremilast for a case of dissecting cellulitis. JAAD Case Rep 2023; 39:122-124. [PMID: 37680571 PMCID: PMC10480467 DOI: 10.1016/j.jdcr.2023.06.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023] Open
Affiliation(s)
- Jate W. Bernard
- University of Kentucky College of Medicine, Lexington, Kentucky
| | - Soumya Reddy
- Department of Dermatology, University of Virginia, Charlottesville, Virginia
| | - R. Hal Flowers
- Department of Dermatology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
31
|
Kumar A, Smith PJ. Horizon scanning: new and future therapies in the management of inflammatory bowel disease. EGASTROENTEROLOGY 2023; 1:e100012. [PMID: 39944001 PMCID: PMC11731077 DOI: 10.1136/egastro-2023-100012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/15/2023] [Indexed: 01/02/2025]
Abstract
The current mainstay treatment modalities for inflammatory bowel disease (IBD) include immunomodulators (methotrexate and thiopurines), biologics (antitumour necrosis factor alpha (TNF-α) being the most commonly used) and other monoclonal antibodies such as the anti-integrins and anti-interleukins (IL-12/23). While ideally treatment should be initiated early in the disease process to avoid relapses and complications, the major recurring issue continues to be primary and secondary loss of response, with often 'diminishing returns' in terms of efficacy for the next line of therapies prescribed for patients with IBD. Additional concerns include the long-term risk factors such as malignancy and susceptibility to infections. Recently, there has been an influx of new and emerging medications entering the market that are showing promising efficacy results in patients with moderate-to-severe disease who have previously failed to respond to multiple drugs. This review will focus on these novel and emerging therapies-in essence, 'horizon scanning'-which includes the antiadhesion agents, cytokine inhibitors, Janus kinase inhibitors, phosphodiesterase inhibitors, sphingosine-1 phosphate receptor modulators and MicroRNA-124 (miR-124) upregulators.
Collapse
Affiliation(s)
- Aditi Kumar
- Department of Gastroenterology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Philip J Smith
- Department of Gastroenterology, Royal Liverpool Hospital, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
32
|
Mease PJ, Hatemi G, Paris M, Cheng S, Maes P, Zhang W, Shi R, Flower A, Picard H, Stein Gold L. Apremilast Long-Term Safety Up to 5 Years from 15 Pooled Randomized, Placebo-Controlled Studies of Psoriasis, Psoriatic Arthritis, and Behçet's Syndrome. Am J Clin Dermatol 2023; 24:809-820. [PMID: 37316690 PMCID: PMC10266699 DOI: 10.1007/s40257-023-00783-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Since US FDA approval in 2014, apremilast has consistently demonstrated a favorable benefit-risk profile in 706,585 patients (557,379 patient-years of exposure) worldwide across approved indications of plaque psoriasis, psoriatic arthritis, and Behçet's syndrome; however, long-term exposure across these indications has not been reported. OBJECTIVE The aim of this study was to conduct a pooled analysis of apremilast data from 15 clinical studies with open-label extension phases, focusing on long-term safety. METHODS We analyzed longer-term safety and tolerability of apremilast 30 mg twice daily across three indications for up to 5 years, focusing on adverse events of special interest, including thrombotic events, malignancies, major adverse cardiac events (MACE), serious infections, and depression. Data were pooled across 15 randomized, placebo-controlled studies and divided into placebo-controlled or all-apremilast-exposure groups. Treatment-emergent adverse events (TEAEs) were assessed. RESULTS Overall, 4183 patients were exposed to apremilast (6788 patient-years). Most TEAEs were mild to moderate in the placebo-controlled period (96.6%) and throughout all apremilast exposure (91.6%). TEAE rates of special interest were similar between treatment groups in the placebo-controlled period and remained low throughout all apremilast exposure. Exposure-adjusted incidence rates per 100 patient-years during all apremilast exposure were MACE, 0.30; thrombotic events, 0.10; malignancies, 1.0; serious infections, 1.10; serious opportunistic infections, 0.21; and depression, 1.78. Safety findings were consistent across indications and regions. No new safety signals were identified. CONCLUSIONS The incidence of serious TEAEs and TEAEs of special interest was low despite long-term exposure, further establishing apremilast as a safe oral option for long-term use across indications with a favorable benefit-risk profile. CLINICAL TRIAL REGISTRATION NCT00773734, NCT01194219, NCT01232283, NCT01690299, NCT01988103, NCT02425826, NCT03123471, NCT03721172, NCT01172938, NCT01212757, NCT01212770, NCT01307423, NCT01925768, NCT00866359, NCT02307513.
Collapse
Affiliation(s)
- Philip J. Mease
- Swedish Medical Center/Providence St, Joseph Health and University of Washington School of Medicine, Seattle, WA USA
| | - Gülen Hatemi
- School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Schmidt L, Wang CA, Patel V, Davidson D, Kalirai S, Panda A, Seigel L. Early Discontinuation of Apremilast in Patients with Psoriasis and Gastrointestinal Comorbidities: Rates and Associated Risk Factors. Dermatol Ther (Heidelb) 2023; 13:2019-2030. [PMID: 37517029 PMCID: PMC10442291 DOI: 10.1007/s13555-023-00975-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
INTRODUCTION Apremilast, the first oral targeted treatment for moderate to severe psoriasis, is associated with diarrhea, nausea, and vomiting, which have contributed to treatment discontinuation. This study describes early apremilast discontinuation rates in patients with psoriasis, including a cohort with gastrointestinal (GI) comorbidities, and associated characteristics. METHODS This retrospective cohort study used IBM® (now Merative™) MarketScan® commercial and Medicare claims data to identify adults with psoriasis who filled their first apremilast prescription between September 1, 2014 and March 31, 2020. Discontinuation was defined as a gap of > 30 days after exhausting the days' supply of a prescription fill. The GI comorbidity cohort included patients with ≥ 1 claim for inflammatory bowel disease (IBD), irritable bowel syndrome (IBS), or other GI comorbidity during the study period. RESULTS Discontinuation rates were high, regardless of previous biologic treatment or GI comorbidities. Among all patients, 25.5% discontinued within 60 days and 56.4% discontinued within 180 days. Patients who discontinued were more likely to be younger, female, and have IBD, Crohn's disease, or a mental health disorder. At 180 days, patients who used biologics previously were more likely to discontinue than biologic-naive patients. Patients with IBD discontinued at a greater rate than those without IBD at 60 days (30.3% vs 24.4%; P = 0.018) and 180 days (63.6% vs 57.2%; P = 0.026). Differences in discontinuation rates were minimal between GI comorbidity groups; patients with IBS discontinued at numerically higher rates than those without IBS. CONCLUSIONS High rates of early discontinuation were observed for patients with and without GI comorbidities. Early discontinuation, whether attributable to poor tolerability or effectiveness, suggests the need for additional oral treatment options.
Collapse
Affiliation(s)
| | | | - Vardhaman Patel
- Bristol Myers Squibb, Princeton, NJ, USA.
- Bristol Myers Squibb, 3410 Princeton Pike, Princeton, NJ, 08648, USA.
| | | | | | | | | |
Collapse
|
34
|
Gao W, Wang Z, Li W, Li Y, Liu M. Biomarkers and biologics related with psoriasis and psoriatic arthritis. Int Immunopharmacol 2023; 122:110646. [PMID: 37454633 DOI: 10.1016/j.intimp.2023.110646] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/06/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Over the past half century, psoriasis is considered as an immune-mediated inflammatory skin disease with the combined hallmarks of autoimmunity and autoinflammation, according to growing volumes of clinical and experimental findings. There is currently no cure for psoriasis, current treatment strategies focus on symptom control, disease minimization, and patient's quality of life enhancement. To meet these challenges, it keeps imperative to discover potential biomarkers, so that not only can they be used for the prediction and monitoring of psoriasis disease in clinic, but also can provide novel therapeutic targets or treatment strategies for psoriasis sufferers. This review systematically demonstrates the research progress of psoriasis-related biomarkers and elaborates their related mechanisms in the pathological development of psoriasis and psoriatic arthritis. In addition, we summarize the development of biologic therapies for psoriasis and psoriatic arthritis in order to drive the broader discussion of psoriasis as an autoimmune-mediated inflammatory skin disease.
Collapse
Affiliation(s)
- Weize Gao
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Zhan Wang
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Wenshuai Li
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yongxin Li
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Mingjun Liu
- Department of Clinical Laboratory, Key Laboratory of Laboratory Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
35
|
Dev PP, Verma P. A case of successful treatment of concurrent verrucous epidermal naevi and psoriasis with apremilast. Australas J Dermatol 2023; 64:e299-e301. [PMID: 37067296 DOI: 10.1111/ajd.14059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 04/18/2023]
Abstract
In a rare case of concurrent verrucous epidermal naevi (VEN) with psoriasis, previous treatments with oral methotrexate and acitretin showed minimal improvement. However, treatment with oral apremilast resulted in complete resolution of psoriasis and significant improvement in VEN lesions after 1 month. This is the first documented case of successful VEN treatment with apremilast, highlighting its potential efficacy in treating verrucous epidermal naevus. Further studies are needed to validate its effectiveness.
Collapse
Affiliation(s)
- Prabin P Dev
- Department of Dermatology & STD, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| | - Prashant Verma
- Department of Dermatology & STD, Vardhman Mahavir Medical College & Safdarjung Hospital, New Delhi, India
| |
Collapse
|
36
|
Tsai YF, Chen CY, Yang SC, Syu YT, Hwang TL. Apremilast ameliorates acute respiratory distress syndrome by inhibiting neutrophil-induced oxidative stress. Biomed J 2023; 46:100560. [PMID: 36103985 PMCID: PMC10345255 DOI: 10.1016/j.bj.2022.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND The pathogenesis of acute respiratory distress syndrome (ARDS) is attributed to the dysregulation of oxidative stress and neutrophil recruitment. We aimed to investigate the anti-inflammatory effects of apremilast on human neutrophils and assess its efficacy for treating ARDS. METHODS We analysed superoxide anion generation, integrin expression, and adhesion in activated human neutrophils using spectrophotometry, flow cytometry, and immunofluorescence microscopy. Phosphorylation of extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) was determined using immunoblotting. A murine lipopolysaccharide (LPS)-induced ARDS model was used to evaluate the therapeutic effects of apremilast. RESULTS Apremilast significantly decreased superoxide anion production, reactive oxygen species (ROS) generation, cluster of differentiation (CD)11 b expression, and neutrophil adhesion in formyl-l-methionyl-l-leucyl-l-phenylalanine activated human neutrophils. Apremilast elevated cyclic 3',5'-adenosine monophosphate (cAMP) and protein kinase A (PKA) activity in activated neutrophils. It reduced cellular cAMP-specific phosphodiesterase (PDE) activity and selectively inhibited enzymatic PDE4 activity. The activated cAMP/PKA pathway suppressed the phosphorylation of ERK and JNK as well as Ca2+ mobilization in activated neutrophils. All inhibitory effects of apremilast on activated neutrophils were reversed by a PKA inhibitor. In vivo examinations indicated that apremilast alleviated lung neutrophil infiltration, myeloperoxidase (MPO) activity, pulmonary oedema, and alveolar damage in LPS-induced ARDS. CONCLUSION Apremilast inhibits inflammatory responses after neutrophil activation via cAMP/PKA-dependent inhibition of ERK and JNK activation. Our study revealed apremilast suppresses oxidative stress and chemotaxis by selectively inhibiting PDE4 in neutrophils and thus protects against endotoxin-induced ARDS in mice. Apremilast can be used as an alternative off-label drug in treating acute lung damage.
Collapse
Affiliation(s)
- Yung-Fong Tsai
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Yu Chen
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shun-Chin Yang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan
| | - Yu-Ting Syu
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital at Taoyuan, Taoyuan, Taiwan; Research Center for Chinese Herbal Medicine and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei, Taiwan.
| |
Collapse
|
37
|
Patel HA, Revankar RR, Pedroza ST, Graham S, Feldman SR. The Genetic Susceptibility to Psoriasis and the Relationship of Linked Genes to Our Treatment Options. Int J Mol Sci 2023; 24:12310. [PMID: 37569685 PMCID: PMC10418823 DOI: 10.3390/ijms241512310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/12/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Understanding the factors creating genetic susceptibility in psoriasis may provide a basis for improving targeted treatment strategies. In this review, we discuss the genes linked to the pathogenesis of psoriasis and their relationship to the available treatment options. To identify the relevant genetic markers and treatments, we searched PubMed, Google Scholar, MEDLINE, and Web of Science with keywords, including genetic susceptibility to psoriasis, genetics and psoriasis, psoriasis treatments, and biologics treatments in psoriasis. The articles in English from database inception to 1/1/23 were included. Case reports and series were excluded. Gene variant forms commonly implicated in the pathogenesis of psoriasis include those encoding for interleukins, interferons, and other mediators involved in inflammatory pathways, such as JAK/STAT, and NF-κB. Several of the treatments for psoriasis (for example IL23 and TYK2 inhibitors) target the products of genes linked to psoriasis. Multiple genes are linked to the pathogenesis of psoriasis. This understanding may provide an avenue for the development of new psoriasis treatment strategies and for more effective, safer treatment outcomes.
Collapse
Affiliation(s)
- Heli A. Patel
- Center for Dermatology Research, Wake Forest School of Medicine, Winston-Salem, NC 27104, USA
| | | | | | - Shaveonte Graham
- Wright State University Boonshoft School of Medicine, Fairborn, OH 45435, USA
| | - Steven R. Feldman
- Center for Dermatology Research, Wake Forest School of Medicine, Winston-Salem, NC 27104, USA
- Department of Dermatology, Wake Forest School of Medicine, Winston-Salem, NC 27104, USA
| |
Collapse
|
38
|
Denis A, Sztejkowski C, Arnaud L, Becker G, Felten R. The 2023 pipeline of disease-modifying antirheumatic drugs (DMARDs) in clinical development for spondyloarthritis (including psoriatic arthritis): a systematic review of trials. RMD Open 2023; 9:e003279. [PMID: 37507210 PMCID: PMC10387652 DOI: 10.1136/rmdopen-2023-003279] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
OBJECTIVES The objective of this systematic review was to provide an overview of current developments and potentially available therapeutic options for spondyloarthritis (SpA) in the coming years. METHODS We conducted a systematic review of 17 national and international clinical trial databases for all disease-modifying antirheumatic drugs (DMARDs) for SpA that are already marketed, in clinical development or withdrawn. The search was performed on February 2023 with the keywords "spondyloarthritis", "ankylosing spondylitis" and "psoriatic arthritis". For each molecule, we only considered the study at the most advanced stage of clinical development. RESULTS Concerning axial SpA (axSpA), a total of 44 DMARDs were identified: 6 conventional synthetic DMARDs (csDMARDs), 27 biological DMARDs (bDMARDs) and 11 targeted synthetic DMARDs (tsDMARDs). Among the 18 targeted treatments (b+tsDMARDs) in current development, corresponding trials reached phase I (n=1), II (n=10) and III (n=7). Ten molecules are IL-17 inhibitors, two Janus kinase (JAK) inhibitors and two granulocyte-macrophage colony-stimulating factor inhibitors; four have another mode of action. Concerning psoriatic arthritis (PsA), 44 DMARDs were identified: 5 csDMARDs, 27 bDMARDs and 12 tsDMARDs. Among the 15 molecules in current development, corresponding trials reached phase II (n=8) and III (n=7). Six molecules are JAK inhibitors, six IL-17 inhibitors and one an IL-23 inhibitor; two have another mode of action. CONCLUSION This systematic review identified 18 and 15 molecules in clinical development for axSpA and PsA, respectively, which suggests a strengthening of the therapeutic arsenal in the coming years. However, with so many DMARDs but low target diversity, we will need to develop strategies or biomarkers to help clinicians make informed treatment decisions.
Collapse
Affiliation(s)
- Agathe Denis
- Service de Rhumatologie de Hautepierre, RESO, Centre de Référence des Maladies Autoimmunes Systémiques Rares Est Sud-Ouest, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Cédric Sztejkowski
- Service de Rhumatologie de Hautepierre, RESO, Centre de Référence des Maladies Autoimmunes Systémiques Rares Est Sud-Ouest, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Laurent Arnaud
- Service de Rhumatologie de Hautepierre, RESO, Centre de Référence des Maladies Autoimmunes Systémiques Rares Est Sud-Ouest, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Guillaume Becker
- Pôle Pharmacie-Pharmacologie, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Renaud Felten
- Service de Rhumatologie de Hautepierre, RESO, Centre de Référence des Maladies Autoimmunes Systémiques Rares Est Sud-Ouest, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- Centre d'Investigation Clinique, Inserm 1434, INSERM, Strasbourg, France
| |
Collapse
|
39
|
Drakos A, Vender R, Torres T. Topical roflumilast for the treatment of psoriasis. Expert Rev Clin Immunol 2023; 19:1053-1062. [PMID: 37243575 DOI: 10.1080/1744666x.2023.2219897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/13/2023] [Accepted: 05/26/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION New non-steroidal topical agents are needed for the treatment of psoriasis. Roflumilast cream 0.3% is a once daily phosphodiesterase-4 inhibitor that was recently approved by the FDA for the treatment of plaque psoriasis in adolescents and adults. It is indicated for use on all body surfaces including intertriginous areas. AREAS COVERED In this review, we summarize the current knowledge about roflumilast cream for the treatment of psoriasis, highlighting its efficacy and safety profile from published clinical trials. Roflumilast's mechanism of action and pharmacokinetic profile are also discussed. EXPERT OPINION Positive results were reported across trials with 48% of patients treated with roflumilast achieving an Investigator Global Assessment score of clear or almost clear at 8 weeks in phase III studies. Most adverse events were mild or moderate in severity and few application-site reactions were reported among participants. Unique advantages of the cream are its success in treating intertriginous areas and its ability to reduce symptoms of itch, results of which may significantly improve quality of life for patients. In the future, real-world data and active comparator trials with existing non-steroidal agents are needed to better understand roflumilast's place in the current treatment landscape.
Collapse
Affiliation(s)
| | - Ron Vender
- Dermatrials Research Inc. & Venderm Innovations in Psoriasis, Hamilton, ON, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Tiago Torres
- Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
- Department of Dermatology, Centro Hospitalar de Santo António, Porto, Portugal
| |
Collapse
|
40
|
Al-Harbi NO, Imam F, Al-Harbi MM, Qamar W, Aljerian K, Khalid Anwer M, Alharbi M, Almudimeegh S, Alhamed AS, Alshamrani AA. Effect of Apremilast on LPS-induced immunomodulation and inflammation via activation of Nrf2/HO-1 pathways in rat lungs. Saudi Pharm J 2023; 31:1327-1338. [PMID: 37323920 PMCID: PMC10267521 DOI: 10.1016/j.jsps.2023.05.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/23/2023] [Indexed: 06/17/2023] Open
Abstract
Lipopolysaccharides (LPS), the lipid component of gram-negative bacterial cell wall, is recognized as the key factor in acute lung inflammation and is found to exhibit severe immunologic reactions. Phosphodiesterase-4 (PDE-4) inhibitor: "apremilast (AP)" is an immune suppressant and anti-inflammatory drug which introduced to treat psoriatic arthritis. The contemporary experiment designed to study the protective influences of AP against LPS induced lung injury in rodents. Twenty-four (24) male experimental Wistar rats selected, acclimatized, and administered with normal saline, LPS, or AP + LPS respectively from 1 to 4 groups. The lung tissues were evaluated for biochemical parameters (MPO), Enzyme Linked Immunosorbent Assay (ELISA), flowcytometry assay, gene expressions, proteins expression and histopathological examination. AP ameliorates the lung injuries by attenuating immunomodulation and inflammation. LPS exposure upregulated IL-6, TNF-α, and MPO while downregulating IL-4 which were restored in AP pretreated rats. The changes in immunomodulation markers by LPS were reduced by AP treatment. Furthermore, results from the qPCR analysis represented an upregulation in IL-1β, MPO, TNF-α, and p38 whereas downregulated in IL-10 and p53 gene expressions in disease control animals while AP pretreated rats exhibited significant reversal in these expressions. Western blot analysis suggested an upregulation of MCP-1, and NOS-2, whereas HO-1, and Nrf-2 expression were suppressed in LPS exposed animals, while pretreatment with AP showed down regulation in the expression MCP-1, NOS-2, and upregulation of HO-1, and Nrf-2 expression of the mentioned intracellular proteins. Histological studies further affirmed the toxic influences of LPS on the pulmonary tissues. It is concluded that, LPS exposure causes pulmonary toxicities via up regulation of oxidative stress, inflammatory cytokines and stimulation of IL-1β, MPO, TNF-α, p38, MCP-1, and NOS-2 while downregulation of IL-4, IL-10, p53, HO-1, and Nrf-2 at different expression level. Pretreatment with AP controlled the toxic influences of LPS by modulating these signaling pathways.
Collapse
Affiliation(s)
- Naif O. Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Faisal Imam
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohammad Matar Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Wajhul Qamar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Khaldoon Aljerian
- Department of Pathology, College of Medicine, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Md. Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mohammed Alharbi
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Sultan Almudimeegh
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Abdullah S. Alhamed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Ali A Alshamrani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
41
|
Edwards A, Chandran V, Rahman P. Investigational monoclonal antibodies in early development for psoriatic arthritis: beyond the biosimilars. Expert Opin Investig Drugs 2023; 32:741-753. [PMID: 37655430 DOI: 10.1080/13543784.2023.2254684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/08/2023] [Accepted: 08/30/2023] [Indexed: 09/02/2023]
Abstract
INTRODUCTION Psoriatic Arthritis (PsA) is an inflammatory arthritis that is present in approximately 25% of psoriasis patients. Currently, several targeted therapies are available to manage PsA; however, many patients fail these therapies. Several new therapeutic options, with differing mechanisms of action, are currently being evaluated. AREAS COVERED This article reviews available results from phase I to phase III trials of several investigational monoclonal antibodies that the FDA has not yet approved for PsA. The proposed mechanisms of the new therapeutic agents and their relevance to the pathogenesis of PsA will be discussed. The investigational agents' efficacy and safety will be summarized, and their potential clinical applications for managing PsA will be contemplated. EXPERT OPINION Due to recent advances in understanding psoriatic arthritis, therapeutic agents are increasingly focused on inhibiting interleukin-17 and interleukin-23 pathways. Various strategies have been used to inhibit these cytokines, demonstrating favorable efficacy and acceptable safety profile.
Collapse
Affiliation(s)
- Anna Edwards
- Faculty of Pharmacy, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Vinod Chandran
- Department of Medicine, Division of Rheumatology, University of Toronto, Toronto, Ontario, Canada
| | - Proton Rahman
- Department of Medicine, Division of Rheumatology, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| |
Collapse
|
42
|
Xu Y, Li Z, Wu S, Guo L, Jiang X. Oral small-molecule tyrosine kinase 2 and phosphodiesterase 4 inhibitors in plaque psoriasis: a network meta-analysis. Front Immunol 2023; 14:1180170. [PMID: 37334353 PMCID: PMC10272578 DOI: 10.3389/fimmu.2023.1180170] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
Background Orally administered small-molecule drugs including tyrosine kinase 2 (TYK2) inhibitors and phosphodiesterase 4 (PDE4) inhibitors are new candidates for systemic therapy in plaque psoriasis. However, no previous articles evaluated the benefit and risk profile of TYK2 and PDE4 inhibitors in psoriasis. Objectives The objective of this study was to compare the efficacy and safety of oral small-molecule drugs, including TYK2 and PDE4 inhibitors, in treating moderate-to-severe plaque psoriasis. Methods PubMed, Embase, and Cochrane library were searched for eligible randomized clinical trials (RCTs). Response rates for a 75% reduction from baseline in Psoriasis Area and Severity Index (PASI-75) and Physician's Global Assessment score of 0 or 1 (PGA 0/1) were used for efficacy assessment. Safety was evaluated with the incidence of adverse events (AEs). A Bayesian multiple treatment network meta-analysis (NMA) was performed. Results In total, 13 RCTs (five for TYK2 inhibitors and eight for PDE4 inhibitors) involving 5274 patients were included. The study found that deucravacitinib at any dose (except for 3 mg QOD), ropsacitinib (200 and 400 mg QD), and apremilast (20 and 30 mg BID) had higher PASI and PGA response rates than placebo. In addition, deucravacitinib (3 mg BID, 6 mg QD, 6 mg BID, and 12 mg QD), and ropsacitinib (400 mg QD) showed superior efficacy than apremilast (30 mg BID). In terms of safety, deucravacitinib or ropsacitinib at any dose did not lead to a higher incidence of AEs than apremilast (30 mg BID). The ranking analysis of efficacy revealed that deucravacitinib 12 mg QD and deucravacitinib 3 mg BID had the highest chance of being the most effective oral treatment, followed by deucravacitinib 6 mg BID and ropsacitinib 400 mg QD. Conclusions Oral TYK2 inhibitors demonstrated satisfactory performance in treating psoriasis, surpassing apremilast at certain doses. More large-scale, long-term studies focusing on novel TYK2 inhibitors are needed. Systematic review registration PROSPERO (ID: CRD42022384859), available from: https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022384859, identifier CRD42022384859.
Collapse
Affiliation(s)
- Yuanyuan Xu
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Zhixuan Li
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Shuwei Wu
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Linghong Guo
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xian Jiang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Nikam RV, Gowtham M, More PS, Shinde AS. Current and emerging prospects in the psoriatic treatment. Int Immunopharmacol 2023; 120:110331. [PMID: 37210912 DOI: 10.1016/j.intimp.2023.110331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/23/2023]
Abstract
Psoriasis is an autoimmune chronic disorder that causes inflammation and a scaly epidermis. The exact pathogenesis of the disease is not known yet. According to the studies, psoriasis is considered an immune-mediated disease. Until now it is believed that genetic and environmental factors are responsible for the disease. There are many comorbidities associated with psoriasis which increases difficulties as patients in some cases get addicted to drugs, alcohol, and smoking which reduces their quality of life. The patient may face social ignorance or suicidal thoughts which may arise in the patient's mind. Due to the undefined trigger of the disease, the treatment is not fully established but by considering the severe impact of the disease researchers are focusing on novel approaches for successful treatment. which has succeeded to a large extent. Here we review pathogenesis, problems faced by psoriatic patients, the need for the development of new treatments over conventional therapies, and the history of psoriatic treatments. We thoroughly focus on emerging treatments like biologics, biosimilars, and small molecules which are now showing more efficacy and safety than conventional treatments. Also, this review article discusses novel approaches which are now in research such as drug repurposing, treatment by stimulation of the vagus nerve, regulation of microbiota, and autophagy for improving disease conditions.
Collapse
Affiliation(s)
- Rutuja Vilas Nikam
- Department of Pharmaceutics, Sanjivani College of Pharmaceutical Education and Research, At Sahajanandnagar, Post-Shinganapur, Tal-Kopargaon, Dist-Ahmednagar, Maharashtra 423603, India.
| | - M Gowtham
- Department of Pharmaceutics, Sanjivani College of Pharmaceutical Education and Research, At Sahajanandnagar, Post-Shinganapur, Tal-Kopargaon, Dist-Ahmednagar, Maharashtra 423603, India.
| | - Pratiksha Sanjay More
- Department of Pharmaceutics, Sanjivani College of Pharmaceutical Education and Research, At Sahajanandnagar, Post-Shinganapur, Tal-Kopargaon, Dist-Ahmednagar, Maharashtra 423603, India.
| | - Anuja Sanjay Shinde
- Department of Pharmaceutics, Sanjivani College of Pharmaceutical Education and Research, At Sahajanandnagar, Post-Shinganapur, Tal-Kopargaon, Dist-Ahmednagar, Maharashtra 423603, India.
| |
Collapse
|
44
|
Jullien D, Richard MA, Halioua B, Bessette C, Derancourt C, Bouloc A. The Needs of Patients with Psoriasis and Benefits of Apremilast in French Clinical Practice: Results from the Observational REALIZE Study. Dermatol Ther (Heidelb) 2023:10.1007/s13555-023-00933-z. [PMID: 37204608 DOI: 10.1007/s13555-023-00933-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/28/2023] [Indexed: 05/20/2023] Open
Abstract
INTRODUCTION Real-world data on the needs of patients with psoriasis and patient-perceived benefits of apremilast are limited. We report such data from France. METHODS The multicenter, observational REALIZE study was conducted in real-life clinical practice in France and enrolled patients with moderate-to-severe plaque psoriasis who had initiated apremilast per French reimbursement criteria in the 4 weeks preceding enrollment (September 2018-June 2020). Physician assessments and patient-reported outcomes (PROs) were collected at enrollment, 6 months, and 12 months. PROs included the Patient Benefit Index for skin diseases (PBI-S), Dermatology Life Quality Index (DLQI), and 9-item Treatment Satisfaction Questionnaire for Medication (TSQM-9). The primary outcome was PBI-S ≥ 1 (minimum clinically relevant benefit) at month 6. RESULTS Of 379 enrolled patients who received ≥ 1 dose of apremilast, most [n = 270 (71.2%)] remained on apremilast at 6 months and over half [n = 200 (52.8%)] persisted at 12 months. Patients reported the following treatment goals as most important (≥ 70% reported goal as "very important" in the Patient Needs Questionnaire): get better skin quickly, regain disease control, be healed of skin alterations, and have confidence in the therapy. Most patients persisting on apremilast achieved a PBI-S ≥ 1 at months 6 and 12 (91.6% and 93.8%, respectively). Mean (SD) DLQI decreased from 11.75 (6.69) at enrollment to 5.17 (5.35) and 4.18 (4.39) at months 6 and 12, respectively. Most patients (72.3%) reported moderate-to-severe pruritus at enrollment and no/mild pruritus at months 6 and 12 (78.8% and 85.9%, respectively). Mean (SD) TSQM-9 Global Satisfaction scores were 68.4 (23.3) and 71.7 (21.5) at months 6 and 12, respectively. Apremilast was well tolerated; no new safety signals were identified. CONCLUSIONS REALIZE provides insights regarding the needs of patients with psoriasis and the patient-perceived benefits of apremilast. Patients who persisted on apremilast reported improvements in quality of life, high treatment satisfaction, and clinically relevant benefits. TRIAL REGISTRATION NCT03757013.
Collapse
Affiliation(s)
- Denis Jullien
- Department of Dermatology, Hospices Civils de Lyon, Edouard Herriot Hospital, Hospital E. Herriot, HCL, 5 Pl. d' Arsonval, 69003, Lyon, France.
- Department of Dermatology, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France.
- INSERM U1111-CIRI, Lyon, France.
| | - Marie-Aleth Richard
- Department of Dermatology, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | | | | | | | | |
Collapse
|
45
|
Kopp KO, Greer ME, Glotfelty EJ, Hsueh SC, Tweedie D, Kim DS, Reale M, Vargesson N, Greig NH. A New Generation of IMiDs as Treatments for Neuroinflammatory and Neurodegenerative Disorders. Biomolecules 2023; 13:747. [PMID: 37238617 PMCID: PMC10216254 DOI: 10.3390/biom13050747] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/14/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
The immunomodulatory imide drug (IMiD) class, which includes the founding drug member thalidomide and later generation drugs, lenalidomide and pomalidomide, has dramatically improved the clinical treatment of specific cancers, such as multiple myeloma, and it combines potent anticancer and anti-inflammatory actions. These actions, in large part, are mediated by IMiD binding to the human protein cereblon that forms a critical component of the E3 ubiquitin ligase complex. This complex ubiquitinates and thereby regulates the levels of multiple endogenous proteins. However, IMiD-cereblon binding modifies cereblon's normal targeted protein degradation towards a new set of neosubstrates that underlies the favorable pharmacological action of classical IMiDs, but also their adverse actions-in particular, their teratogenicity. The ability of classical IMiDs to reduce the synthesis of key proinflammatory cytokines, especially TNF-α levels, makes them potentially valuable to reposition as drugs to mitigate inflammatory-associated conditions and, particularly, neurological disorders driven by an excessive neuroinflammatory element, as occurs in traumatic brain injury, Alzheimer's and Parkinson's diseases, and ischemic stroke. The teratogenic and anticancer actions of classical IMiDs are substantial liabilities for effective drugs in these disorders and can theoretically be dialed out of the drug class. We review a select series of novel IMiDs designed to avoid binding with human cereblon and/or evade degradation of downstream neosubstrates considered to underpin the adverse actions of thalidomide-like drugs. These novel non-classical IMiDs hold potential as new medications for erythema nodosum leprosum (ENL), a painful inflammatory skin condition associated with Hansen's disease for which thalidomide remains widely used, and, in particular, as a new treatment strategy for neurodegenerative disorders in which neuroinflammation is a key component.
Collapse
Affiliation(s)
- Katherine O. Kopp
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
| | - Margaret E. Greer
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
- Faculty of Medicine, Georgetown University School of Medicine, Washington, DC 20007, USA
| | - Elliot J. Glotfelty
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
- Department of Neuroscience, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Shih-Chang Hsueh
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
| | - Dong Seok Kim
- Aevisbio Inc., Gaithersburg, MD 20878, USA
- Aevis Bio Inc., Daejeon 34141, Republic of Korea
| | - Marcella Reale
- Department of Innovative Technologies in Medicine and Dentistry, G. d’Annunzio University of Chieti and Pescara, 66100 Chieti, Italy
| | - Neil Vargesson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Nigel H. Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program National Institute on Aging, Biomedical Research Center, 251 Bayview Blvd., NIH, Baltimore, MD 21224, USA
| |
Collapse
|
46
|
Kadagothy H, Nene S, Amulya E, Vambhurkar G, Rajalakshmi AN, Khatri DK, Singh SB, Srivastava S. Perspective insights of small molecules, phytoconstituents and biologics in the management of psoriasis: A focus on targeting major inflammatory cytokine pathways. Eur J Pharmacol 2023; 947:175668. [PMID: 36958476 DOI: 10.1016/j.ejphar.2023.175668] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 03/25/2023]
Abstract
Psoriasis is an enduring, pruritic and papulosquamous skin ailment that poses a significant burden on public health. It is mainly characterized by hyperkeratosis, acanthosis, parakeratosis, scaly and erythematous plaques. Biomarkers like interleukin-17, interleukin-12 and -23 and tumor necrosis factor-α serve as key drivers of psoriatic pathogenesis. Triggered release of pro-inflammatory cytokines from various up-regulated pathways leads to psoriatic inflammation. Several target moieties like biologics, small molecules and herbal moieties play a fundamental role in the repression of pathogenesis of psoriasis. Biologics and small molecules engaged in the management of psoriasis have been emphasized in detail. An insight into nano-carrier interventions on herbal moieties and clinical aspects of psoriasis are also highlighted. This review emphasizes various pathological targets involved in psoriasis.
Collapse
Affiliation(s)
- Husna Kadagothy
- Department of Pharmaceutics, Mother Theresa Post Graduate and Research Institute of Health Sciences, Puducherry, India
| | - Shweta Nene
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Etikala Amulya
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Ganesh Vambhurkar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - A N Rajalakshmi
- Department of Pharmaceutics, Mother Theresa Post Graduate and Research Institute of Health Sciences, Puducherry, India
| | - Dharmendra Kumar Khatri
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Shashi Bala Singh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
47
|
Oubaid EN, Abu-Raghif A, Al-Sudani IM. Ibudilast ameliorates experimentally induced colitis in rats via down-regulation of proinflammatory cytokines and myeloperoxidase enzyme activity. PHARMACIA 2023. [DOI: 10.3897/pharmacia.70.e98715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
Abstract
Objectives: This study was carried out to explore the possible anti-inflammatory effect of ibudilast on acetic acid-induced colitis in rats.
Methods: Fifty adult Wistar rats were separated into 5 groups, including the control group, acetic acid group, acetic acid + vehicle, acetic acid + sulfasalazine (100 mg/kg/day)group, and acetic acid + ibudilast (30 mg/kg/day) group. Colitis was induced in rats by the inter-rectal installation of 2 ml of 4% (v/v) acetic acid. Sulfasalazine and ibudilast were administered orally for ten days after 2 hours of induction.
Results: The treatment with ibudilast significantly reduced disease activity index (DAI), macroscopic colonic scores (MAC), and histopathological changes induced by acetic acid. Also, ibudilast markedly decreased the expression of proinflammatory markers (TNF-α and IL-1β) in colonic tissue. Moreover, ibudilast inhibited myeloperoxidase (MPO) enzyme activity that was increased by acetic acid.
Conclusion: Therefore, ibudilast may have a therapeutic effect in the management of ulcerative colitis.
Collapse
|
48
|
Azuaga AB, Ramírez J, Cañete JD. Psoriatic Arthritis: Pathogenesis and Targeted Therapies. Int J Mol Sci 2023; 24:4901. [PMID: 36902329 PMCID: PMC10003101 DOI: 10.3390/ijms24054901] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Psoriatic arthritis (PsA), a heterogeneous chronic inflammatory immune-mediated disease characterized by musculoskeletal inflammation (arthritis, enthesitis, spondylitis, and dactylitis), generally occurs in patients with psoriasis. PsA is also associated with uveitis and inflammatory bowel disease (Crohn's disease and ulcerative colitis). To capture these manifestations as well as the associated comorbidities, and to recognize their underlining common pathogenesis, the name of psoriatic disease was coined. The pathogenesis of PsA is complex and multifaceted, with an interplay of genetic predisposition, triggering environmental factors, and activation of the innate and adaptive immune system, although autoinflammation has also been implicated. Research has identified several immune-inflammatory pathways defined by cytokines (IL-23/IL-17, TNF), leading to the development of efficacious therapeutic targets. However, heterogeneous responses to these drugs occur in different patients and in the different tissues involved, resulting in a challenge to the global management of the disease. Therefore, more translational research is necessary in order to identify new targets and improve current disease outcomes. Hopefully, this may become a reality through the integration of different omics technologies that allow better understanding of the relevant cellular and molecular players of the different tissues and manifestations of the disease. In this narrative review, we aim to provide an updated overview of the pathophysiology, including the latest findings from multiomics studies, and to describe current targeted therapies.
Collapse
Affiliation(s)
- Ana Belén Azuaga
- Rheumatology Department, Hospital Clinic and IDIBAPS of Barcelona, 08036 Barcelona, Spain
| | | | - Juan D. Cañete
- Rheumatology Department, Hospital Clinic and IDIBAPS of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
49
|
Vander Does A, Ju T, Mohsin N, Chopra D, Yosipovitch G. How to get rid of itching. Pharmacol Ther 2023; 243:108355. [PMID: 36739914 DOI: 10.1016/j.pharmthera.2023.108355] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/01/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Itch is an unpleasant sensation arising from a variety of dermatologic, neuropathic, systemic, and psychogenic etiologies. Various itch pathways are implicated according to the underlying etiology. A variety of pruritogens, or itch mediators, as well as receptors have been identified and provide potential therapeutic targets. Recent research has primarily focused on targeting inflammatory cytokines and Janus kinase signaling, protease-activated receptors, substance P and neurokinin, transient receptor potential-vanilloid ion channels, Mas-related G-protein-coupled receptors (MRGPRX2 and MRGPRX4), the endogenous opioid and cannabinoid balance, and phosphodiesterase 4. Periostin, a newly identified pruritogen, should be further explored with clinical trials. Drugs targeting neural sensitization including the gabergic system and P2X3 are other potential drugs for chronic itch. There is a need for more targeted therapies to improve clinical outcomes and reduce side effects.
Collapse
Affiliation(s)
- Ashley Vander Does
- Dr Phillip Frost Department of Dermatology and Miami Itch Center, University of Miami, Miami, FL, USA
| | - Teresa Ju
- Dr Phillip Frost Department of Dermatology and Miami Itch Center, University of Miami, Miami, FL, USA
| | - Noreen Mohsin
- Dr Phillip Frost Department of Dermatology and Miami Itch Center, University of Miami, Miami, FL, USA
| | - Divya Chopra
- Dr Phillip Frost Department of Dermatology and Miami Itch Center, University of Miami, Miami, FL, USA
| | - Gil Yosipovitch
- Dr Phillip Frost Department of Dermatology and Miami Itch Center, University of Miami, Miami, FL, USA.
| |
Collapse
|
50
|
Liu CT, Yen JHJ, Brown DA, Song YC, Chu MY, Hung YH, Tang YH, Wu PY, Yen HR. Targeting Nrf2 with 3 H-1,2-dithiole-3-thione to moderate OXPHOS-driven oxidative stress attenuates IL-17A-induced psoriasis. Biomed Pharmacother 2023; 159:114294. [PMID: 36706632 DOI: 10.1016/j.biopha.2023.114294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Psoriasis, a chronic autoimmune disease characterized by the hyperproliferation of keratinocytes in the epidermis and parakeratosis, significantly impacts quality of life. Interleukin (IL)- 17A dominates the pathogenesis of psoriasis and facilitates reactive oxygen species (ROS) accumulation, which exacerbates local psoriatic lesions. Biologic treatment provides remarkable clinical efficacy, but its high cost and unignorable side effects limit its applications. 3 H-1,2-Dithiole-3-thione (D3T) possesses compelling antioxidative capacities against several diseases through the nuclear factor erythroid 2-related factor 2 (Nrf2) cascade. Hence, we aimed to evaluate the effect and mechanism of D3T in psoriasis. We found that D3T attenuates skin thickening and scaling by inhibiting IL-17A-secreting γδT cells in imiquimod (IMQ)-induced psoriatic mice. Interleukin-17A markedly enhanced IL-6 and IL-8 expression, lipid peroxidation, the contents of nitric oxide and hydrogen peroxide, oxidative phosphorylation and the MAPK/NF-κB pathways in keratinocytes. IL-17A also inhibited the Nrf2-NQO1-HO-1 axis and the activities of superoxide dismutase and glutathione peroxidase. D3T significantly reversed these parameters in IL-17A-treated keratinocytes. ML-385, a Nrf2 neutralizer, failed to improve D3T-induced anti-inflammatory and antioxidative effects in IL-17A-treated keratinocytes. We conclude that targeting Nrf2 with D3T to diminish oxidative and inflammatory damage in keratinocytes may attenuate psoriasis.
Collapse
Affiliation(s)
- Chuan-Teng Liu
- Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan; School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Jui-Hung Jimmy Yen
- Department of Microbiology and Immunology, Indiana University School of Medicine, Fort Wayne, IN, USA
| | - Dennis A Brown
- Manchester University College of Pharmacy, Natural and Health Sciences, Fort Wayne, IN, USA
| | - Ying-Chyi Song
- Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan; Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Mei-Yun Chu
- Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Yu-Hsiang Hung
- Department of Medical Education, China Medical University Hospital, Taichung, Taiwan
| | | | - Po-Yuan Wu
- Department of Dermatology, China Medical University Hospital, Taichung, Taiwan; School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan.
| | - Hung-Rong Yen
- Research Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan; School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, Taiwan; Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan; Department of Medical Biotechnology and Laboratory Science, Asia University, Taichung, Taiwan.
| |
Collapse
|