51
|
Yanase T, Tsuruta N, Yamaguchi K, Ohata C, Ohyama B, Katayama E, Sugita K, Kuwashiro M, Hashimoto A, Yonekura K, Higashi Y, Murota H, Koike Y, Matsuzaka Y, Kikuchi S, Hatano Y, Saito K, Takahashi K, Miyagi T, Kaneko S, Ota M, Harada K, Morizane S, Ikeda K, Furue M, Nakahara T, Okazaki F, Sasaki N, Okada E, Yoshida Y, Ito K, Imafuku S. Survival rates of systemic interventions for psoriasis in the Western Japan Psoriasis Registry: A multicenter retrospective study. J Dermatol 2023. [PMID: 36786158 DOI: 10.1111/1346-8138.16737] [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: 01/27/2022] [Revised: 12/27/2022] [Accepted: 01/15/2023] [Indexed: 02/15/2023]
Abstract
Psoriasis affects approximately 0.3% of the Japanese population. Recently, various effective systemic drugs have become available, and the continuation of a given treatment has become critical because of the chronic nature of psoriasis. Factors affecting drug survival (the time until treatment discontinuation) in psoriasis treatment include efficacy, safety, ease of use, and patient preference. In the present study, the authors retrospectively surveyed a multifacility patient registry to determine the real-world evidence of the survival rate of systemic interventions for psoriasis treatment. Patients with psoriasis who visited 20 facilities in the Western Japan area between January 2019 and May 2020 and gave written consent were registered as study participants, and their medical history of systemic interventions for psoriasis (starting from 2010) was retrospectively collected and analyzed. The drugs investigated were adalimumab, infliximab, ustekinumab, secukinumab, ixekizumab, brodalumab, guselkumab, risankizumab, cyclosporine, and apremilast. When drugs were discontinued, the reasons were also recorded. A total of 1003 patients with psoriasis including 268 with psoriatic arthritis (PsA) were enrolled. In biologics, more recently released drugs such as interleukin 17 inhibitors showed a numerically higher survival rate in the overall (post-2010) analysis. However, in the subset of patients who began treatment after 2017, the difference in the survival rate among the drugs was smaller. The reasons for discontinuing drugs varied, but a loss of efficacy against dermatological or joint symptoms were relatively frequently seen with some biologics and cyclosporine. The stratification of drug survival rates based on patient characteristics such as bio-naive or experienced, normal weight or obese, and with or without PsA, revealed that bio-experienced, obese, and PsA groups had poorer survival rates for most drugs. No notable safety issues were identified in this study. Overall, the present study revealed that the biologics show differences in their tendency to develop a loss of efficacy, and the factors that negatively impact the survival rate of biologics include the previous use of biologics, obesity, and PsA.
Collapse
Affiliation(s)
- Tetsuji Yanase
- Department of Dermatology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Noriko Tsuruta
- Department of Dermatology, Fukuoka University Faculty of Medicine, Fukuoka, Japan.,Department of Dermatology, Kitakyuhsu City Yahata Hospital, Kitakyushu, Japan
| | - Kazuki Yamaguchi
- Department of Dermatology, Fukuoka University Faculty of Medicine, Fukuoka, Japan.,Department of Dermatology, Saiseikai Futsukaichi Hospital, Fukuoka, Japan
| | - Chika Ohata
- Department of Dermatology, Kurume University School of Medicine, Kurume, Japan.,Department of Dermatology, Osaka General Medical Center, Osaka, Japan
| | - Bungo Ohyama
- Department of Dermatology, Kurume University School of Medicine, Kurume, Japan.,Ohyama Dermatology Clinic, Kumamoto, Japan
| | - Eri Katayama
- Department of Dermatology, Kurume University School of Medicine, Kurume, Japan
| | - Kazunari Sugita
- Division of Dermatology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan.,Division of Dermatology, Department of Medicine of Sensory and Motor Organs, Tottori University Faculty of Medicine, Yonago, Japan
| | - Maki Kuwashiro
- Division of Dermatology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Aki Hashimoto
- Division of Dermatology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | - Kentaro Yonekura
- Department of Dermatology, Imamura General Hospital, Kagoshima, Japan
| | - Yuko Higashi
- Department of Dermatology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hiroyuki Murota
- Department of Dermatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yuta Koike
- Department of Dermatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Yuki Matsuzaka
- Department of Dermatology, Onomichi General Hospital, Onomichi, Japan
| | - Satoko Kikuchi
- Department of Dermatology, Kyushu Central Hospital, Fukuoka, Japan
| | - Yutaka Hatano
- Department of Dermatology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kanami Saito
- Department of Dermatology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Kenzo Takahashi
- Department of Dermatology, Graduate School of Medicine University of the Ryukyus, Nishihara, Japan
| | - Takuya Miyagi
- Department of Dermatology, Graduate School of Medicine University of the Ryukyus, Nishihara, Japan
| | - Sakae Kaneko
- Department of Dermatology, Shimane University Faculty of Medicine, Izumo, Japan.,Department of Dermatology, Masuda Red Cross Hospital, Masuda, Japan
| | - Masataka Ota
- Department of Dermatology, Shimane University Faculty of Medicine, Izumo, Japan
| | - Kayo Harada
- Department of Dermatology and Allergy, National Hospital Organization, Kyushu Medical Center, Fukuoka, Japan
| | - Shin Morizane
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kenta Ikeda
- Department of Dermatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masutaka Furue
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fusako Okazaki
- Department of Dermatology, Okayama City General Medicine Center, Okayama, Japan
| | - Natsuko Sasaki
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Etsuko Okada
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yuichi Yoshida
- Division of Dermatology, Department of Medicine of Sensory and Motor Organs, Tottori University Faculty of Medicine, Yonago, Japan
| | - Kotaro Ito
- Department of Dermatology, Fukuoka University Faculty of Medicine, Fukuoka, Japan.,Ito Medical Clinic, Dermatology, Kitsuki, Japan
| | - Shinichi Imafuku
- Department of Dermatology, Fukuoka University Faculty of Medicine, Fukuoka, Japan
| |
Collapse
|
52
|
Spencer RK, Elhage KG, Jin JQ, Davis MS, Hakimi M, Bhutani T, Liao W. Apremilast in Palmoplantar Psoriasis and Palmoplantar Pustulosis: A Systematic Review and Meta-analysis. Dermatol Ther (Heidelb) 2023; 13:437-451. [PMID: 36609960 PMCID: PMC9884725 DOI: 10.1007/s13555-022-00877-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/07/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND This review's goals were to investigate apremilast's efficacy versus placebo in palmoplantar psoriasis (PP) and palmoplantar pustulosis (PPP), and apremilast's efficacy versus methotrexate in PP. METHODS A literature search was conducted in PubMed, clinicaltrials.gov, and Embase in July 2022. Publications investigating subjects with PP or PPP, treated with apremilast, which reported palmoplantar-specific outcomes were used. Exclusion criteria included cases of drug-induced PP/PPP, case studies, non-English texts, omission of palmoplantar-specific outcomes, and incomplete publications. Studies were assessed for risk of bias using Cochrane Review Manager application and CASP checklist. Primary endpoints were a 50% improvement of the Palmoplantar Psoriasis/Pustulosis Area and Severity Index (PPPASI 50) and improvement of the Palmoplantar Physician Global Assessment (PPPGA) to 0 or 1 in patients with baseline PPPGA ≥ 3. RESULTS Seventeen original studies including five placebo-controlled randomized clinical trials (RCTs), one phase II clinical trial, two randomized methotrexate comparative trials, six cohort studies, and three case series were analyzed, totaling 1117 participants. Meta-analysis of four placebo-controlled RCTs investigating PP found apremilast treatment to be superior to placebo in achieving a PPPGA of 0/1 (baseline PPPGA of ≥ 3) after 16 weeks of treatment (n = 244; OR = 2.69 [1.39-5.22]). Apremilast was superior to placebo in achieving PPPASI 50 at week 16 in the only placebo-controlled RCT of PPP (78.3 vs. 40.9%) [P = 0.0003]. Apremilast was comparable to methotrexate in achieving PPPASI 50 at week 16 in PP (59.5 vs. 64.3%; n = 84; [P = 0.65]). Non-randomized studies generally showed marked improvement in PPPASI, PPPGA, and DLQI scores following apremilast treatment. DISCUSSION Apremilast treatment in PP and PPP resulted in significant improvement in objective, palmoplantar-specific clinical parameters versus placebo, and comparable efficacy with methotrexate in PP. Limitations in interpreting these results include variations in palmoplantar-specific metrics used and risk of bias of included studies.
Collapse
Affiliation(s)
- Riley K Spencer
- Department of Dermatology, University of California San Francisco, 515 Spruce Street, San Francisco, CA, 94118, USA.
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, USA.
| | - Kareem G Elhage
- Department of Dermatology, University of California San Francisco, 515 Spruce Street, San Francisco, CA, 94118, USA
| | - Joy Q Jin
- Department of Dermatology, University of California San Francisco, 515 Spruce Street, San Francisco, CA, 94118, USA
- School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Mitchell S Davis
- Department of Dermatology, University of California San Francisco, 515 Spruce Street, San Francisco, CA, 94118, USA
| | - Marwa Hakimi
- Department of Dermatology, University of California San Francisco, 515 Spruce Street, San Francisco, CA, 94118, USA
| | - Tina Bhutani
- Department of Dermatology, University of California San Francisco, 515 Spruce Street, San Francisco, CA, 94118, USA
| | - Wilson Liao
- Department of Dermatology, University of California San Francisco, 515 Spruce Street, San Francisco, CA, 94118, USA
| |
Collapse
|
53
|
Mao P, Huang C, Li Y, Zhao Y, Zhou S, Zhao Z, Mu Y, Wang L, Li F, Zhao AZ. Pharmacological targeting of type phosphodiesterase 4 inhibits the development of acute myeloid leukemia by impairing mitochondrial function through the Wnt/β-catenin pathway. Biomed Pharmacother 2023; 157:114027. [PMID: 36436494 DOI: 10.1016/j.biopha.2022.114027] [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: 08/18/2022] [Revised: 11/09/2022] [Accepted: 11/17/2022] [Indexed: 11/27/2022] Open
Abstract
Acute myeloid leukemia (AML) is prone to drug-resistant relapse with a low 5-year survival rate. New therapeutic modalities are sorely needed to provide hope for AML relapse patients. Herein, we demonstrated a specific inhibitor of type 4 phosphodiesterase (PDE4), Zl-n-91, could significantly reduce the proliferation of AML cells, block DNA replication process, and increase AML cell death. Zl-n-91 also impeded the growth of subcutaneous xenograft and prolonged the survival of the MLL-AF9-driven AML model. Bioinformatic analysis revealed that elevated mitochondrial gene signatures inversely correlate with the survival of AML patients; and importantly, Zl-n-91 strongly suppressed the function of mitochondria. In addition, this PDE4 inhibitor induced alterations in multiple signaling pathways, including the reduction of β-catenin activity. Stimulation of the Wnt/β-catenin pathway could attenuate the inhibitory effect of Zl-n-91 on AML cell proliferation as well as mitochondrial function. Taken together, we revealed for the first time that targeting PDE4 activity could attenuate mitochondrial function through a Wnt/β-catenin pathway, which in turn would block the growth of AML cells. Specific PDE4 inhibitors can potentially serve as a new treatment modality for AML patients.
Collapse
Affiliation(s)
- Ping Mao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Changhao Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Yuyu Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Yuanyi Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Sujin Zhou
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Zhenggang Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Yunping Mu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Lina Wang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China
| | - Fanghong Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China.
| | - Allan Z Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Xiaoguwei Street, Panyu District, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
54
|
Expert recommendations for the use of apremilast in psoriatic arthritis. REUMATOLOGIA CLINICA 2023; 19:34-44. [PMID: 35537933 DOI: 10.1016/j.reumae.2022.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 01/07/2022] [Indexed: 01/10/2023]
Abstract
INTRODUCTION AND OBJECTIVES Despite the evidence, there are doubts about the positioning of apremilast in the psoriatic arthritis (PsA) treatment algorithm. The objective of this project was to collect the scientific evidence and the experience of a group of rheumatologists who are experts in the management of PsA with apremilast in clinical practice in Spain. MATERIAL AND METHODS A scientific committee made up of 6 experts proposed 5 clinical scenarios where the evidence on the use of apremilast in PsA was controversial: (i) Efficacy in peripheral PsA; (ii) Efficacy in enthesitis and dactylitis; (iii) Efficacy in PsA with skin involvement; (iv) Comorbidities; and (v) Apremilast safety. After this, a panel of 17 rheumatologists with expertise in PsA management discussed these scenarios and generated a questionnaire with 50 questions and 156 items following the Delphi methodology. This questionnaire was anonymously answered by the panel. RESULTS After 2 voting rounds, the panel of experts reached consensus in 93 of the 156 items raised (59.6%) (67 in agreement and 26 in disagreement). The degree of consensus was 53.3% in the area of "Efficacy in peripheral PsA"; 60.0% in "Efficacy in enthesitis and dactylitis"; 50.0% in "Efficacy in PsA with skin involvement"; 57.1% in "Management of comorbidities in patients with PsA"; and 67.3% in "Implications of safety in the use of apremilast". CONCLUSIONS The structured opinion of the experts complements the available evidence and contributes to the establishment of consensual guidelines for the use of apremilast in PsA.
Collapse
|
55
|
Gelfand JM, Shin DB, Armstrong AW, Tyring SK, Blauvelt A, Gottlieb S, Lockshin BN, Kalb RE, Fitzsimmons R, Rodante J, Parel P, Manyak GA, Mendelsohn L, Noe MH, Papadopoulos M, Syed MN, Werner TJ, Wan J, Playford MP, Alavi A, Mehta NN. Association of Apremilast With Vascular Inflammation and Cardiometabolic Function in Patients With Psoriasis: The VIP-A Phase 4, Open-label, Nonrandomized Clinical Trial. JAMA Dermatol 2022; 158:1394-1403. [PMID: 36129688 PMCID: PMC9494263 DOI: 10.1001/jamadermatol.2022.3862] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 07/24/2022] [Indexed: 01/13/2023]
Abstract
Importance Psoriasis is an inflammatory condition associated with metabolic and cardiovascular disease. Apremilast, a phosphodiesterase 4 inhibitor, is commonly used for psoriasis and can cause weight loss. Objective To determine the association between apremilast and aortic vascular inflammation as assessed by 18F-fluorodeoxyglucose positron emission tomography/computed tomography (FDG-PET/CT), cardiometabolic markers (primary outcomes at week 16), and abdominal fat composition. Design, Setting, and Participants A single-arm, open-label, interventional, nonrandomized clinical trial in which the imaging and laboratory outcomes were measured by an investigator who was blinded to time was conducted between April 11, 2017, and August 17, 2021, at 7 dermatology sites in the United States. A total of 101 patients with moderate to severe psoriasis were screened, 70 enrolled, 60 completed week 16, and 39 completed week 52. Intervention Apremilast, 30 mg, twice daily. Main Outcomes and Measures Aortic vascular inflammation (measured by FDG-PET/CT), 68 cardiometabolic biomarkers, and abdominal fat composition (measured by CT) at week 16 and week 52 compared with baseline. Results The mean (SD) age of the 70 patients was 47.5 (14.6) years, 54 were male (77.1%), 4 were Black (5.7%), and 58 were White (82.9%). There was no change in aortic vascular inflammation at week 16 (target to background ratio, -0.02; 95% CI, -0.08 to 0.05; P = .61) or week 52 (target to background ratio, -0.07; 95% CI, -0.15 to 0.01; P = .09) compared with baseline. At week 16, potentially beneficial decreases in interleukin 1b, valine, leucine, isoleucine, fetuin A, and branched-chain amino acids were observed. At week 52 compared with baseline, potentially beneficial decreases in ferritin, β-hydroxybutyrate, acetone, and ketone bodies, with an increase in apolipoprotein A-1, were observed, but there was a reduction in cholesterol efflux. There was an approximately 5% to 6% reduction in subcutaneous and visceral adiposity at week 16 that was maintained at week 52. Conclusions and Relevance The findings of this nonrandomized clinical trial suggest that apremilast has a neutral association with aortic vascular inflammation, variable but generally beneficial associations with a subset of cardiometabolic biomarkers, and associations with reductions in visceral and subcutaneous fat, indicating that the drug may have an overall benefit for patients with cardiometabolic disease and psoriasis. Trial Registration ClinicalTrials.gov Identifier: NCT03082729.
Collapse
Affiliation(s)
- Joel M. Gelfand
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Daniel B. Shin
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - April W. Armstrong
- Department of Dermatology, Keck School of Medicine, University of Southern California, Los Angeles
| | - Stephen K. Tyring
- Department of Dermatology, University of Texas Health Science Center at Houston, Houston
| | | | - Scott Gottlieb
- Dermatology and Skin Surgery Center, Exton, Pennsylvania
| | | | - Robert E. Kalb
- SUNY at Buffalo School of Medicine and Biomedical Sciences, Department of Dermatology, Buffalo Medical Group, Buffalo, New York
| | - Robert Fitzsimmons
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Justin Rodante
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Philip Parel
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Grigory A. Manyak
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Laurel Mendelsohn
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Megan H. Noe
- Department of Dermatology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maryte Papadopoulos
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Maha N. Syed
- Department of Dermatology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Thomas J. Werner
- Department of Radiology (Nuclear Medicine), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Joy Wan
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Martin P. Playford
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Abass Alavi
- Department of Radiology (Nuclear Medicine), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Nehal N. Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| |
Collapse
|
56
|
Vieujean S, D’Amico F, Netter P, Danese S, Peyrin‐Biroulet L. Landscape of new drugs and targets in inflammatory bowel disease. United European Gastroenterol J 2022; 10:1129-1166. [PMID: 36112543 PMCID: PMC9752289 DOI: 10.1002/ueg2.12305] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/22/2022] [Indexed: 01/13/2023] Open
Abstract
Although the therapeutic armamentarium of Inflammatory bowel diseases (IBD) physicians has expanded rapidly in recent years, a proportion of patients remain with a suboptimal response to medical treatment due to primary no response, loss of response or intolerance to currently available drugs. Our growing knowledges of IBD pathophysiology has led to the development of a multitude of new therapies over time, which may, 1 day, be able to address this unmet medical need. This review aims to provide physicians an update of emerging therapies in IBD by focusing on drugs currently in phase 3 clinical trials. Among the most promising molecules are anti-IL-23, JAK-inhibitors, anti-integrins and S1P modulators. While the results in terms of efficacy and safety are fairly clear for some classes, the question of safety remains more uncertain for other classes. Molecules at a more preliminary stage of development (phase 1 and 2), one of which may 1 day offer an optimal benefit-risk ratio, will also be presented as well as their respective mechanisms of action.
Collapse
Affiliation(s)
- Sophie Vieujean
- Hepato‐Gastroenterology and Digestive OncologyUniversity Hospital CHU of LiègeLiègeBelgium
| | - Ferdinando D’Amico
- Department of Gastroenterology and EndoscopyIRCCS San Raffaele Hospital and Vita‐Salute San Raffaele UniversityMilanItaly
- Department of Biomedical SciencesHumanitas UniversityPieve EmanueleMilanItaly
| | | | - Silvio Danese
- Department of Gastroenterology and EndoscopyIRCCS San Raffaele Hospital and Vita‐Salute San Raffaele UniversityMilanItaly
| | - Laurent Peyrin‐Biroulet
- Department of GastroenterologyUniversity of LorraineCHRU‐NancyNancyFrance
- University of LorraineInserm, NGERENancyFrance
| |
Collapse
|
57
|
|
58
|
Yu J, Zhao Q, Wang X, Zhou H, Hu J, Gu L, Hu Y, Zeng F, Zhao F, Yue C, Zhou P, Li G, Li Y, Wu W, Zhou Y, Li J. Pathogenesis, multi-omics research, and clinical treatment of psoriasis. J Autoimmun 2022; 133:102916. [PMID: 36209691 DOI: 10.1016/j.jaut.2022.102916] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 11/07/2022]
Abstract
Psoriasis is a common inflammatory skin disease involving interactions between keratinocytes and immune cells that significantly affects the quality of life. It is characterized by hyperproliferation and abnormal differentiation of keratinocytes and excessive infiltration of immune cells in the dermis and epidermis. The immune mechanism underlying this disease has been elucidated in the past few years. Research shows that psoriasis is regulated by the complex interactions among immune cells, such as keratinocytes, dendritic cells, T lymphocytes, neutrophils, macrophages, natural killer cells, mast cells, and other immune cells. An increasing number of signaling pathways have been found to be involved in the pathogenesis of psoriasis, which has prompted the search for new treatment targets. In the past decades, studies on the pathogenesis of psoriasis have focused on the development of targeted and highly effective therapies. In this review, we have discussed the relationship between various types of immune cells and psoriasis and summarized the major signaling pathways involved in the pathogenesis of psoriasis, including the PI3K/AKT/mTOR, JAK-STAT, JNK, and WNT pathways. In addition, we have discussed the results of the latest omics research on psoriasis and the epigenetics of the disease, which provide insights regarding its pathogenesis and therapeutic prospects; we have also summarized its treatment strategies and observations of clinical trials. In this paper, the various aspects of psoriasis are described in detail, and the limitations of the current treatment methods are emphasized. It is necessary to improve and innovate treatment methods from the molecular level of pathogenesis, and further provide new ideas for the treatment and research of psoriasis.
Collapse
Affiliation(s)
- Jiadong Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Qixiang Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Xiaoyan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Hong Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Jing Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Linna Gu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Yawen Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Fanlian Zeng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Fulei Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Chengcheng Yue
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Pei Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Guolin Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Ya Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Wenling Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Yifan Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China
| | - Jiong Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, and Collaborative Innovation Center for Biotherapy, 1 Keyuan 4th Road, Gaopeng Street, High Technological Development Zone, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
59
|
Safety of Apremilast in Patients with Psoriasis and Psoriatic Arthritis: Findings from the UK Clinical Practice Research Datalink. Drug Saf 2022; 45:1403-1411. [PMID: 36151359 PMCID: PMC9510500 DOI: 10.1007/s40264-022-01235-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2022] [Indexed: 11/03/2022]
Abstract
INTRODUCTION This real-world safety analysis was requested by the European Medicines Agency following approval of apremilast, an oral treatment for psoriasis or psoriatic arthritis. OBJECTIVE We aimed to compare incidence rates of adverse events of special interest identified a priori, in patients receiving apremilast with those receiving other systemic treatments for psoriasis or psoriatic arthritis. METHODS This 5-year cohort study was conducted in Clinical Practice Research Datalink GOLD between January 2015 and June 2020. Incidence rates of adverse events of special interest were estimated for four matched cohorts: apremilast-exposed and three matched non-apremilast cohorts (oral only, injectable only, and oral and injectable psoriasis or psoriatic arthritis treatments). RESULTS The apremilast-exposed cohort included 341 patients and the three non-apremilast cohorts included 4981 patients. There were no incident cases of vasculitis, hematologic malignancy, non-melanoma skin malignancy, treated depression, treated anxiety, or suicidal behaviors in the apremilast-exposed cohort during the follow-up. Similar incidence rates of all-cause mortality, major adverse cardiac events, tachyarrhythmias, and solid malignancies were recorded in the apremilast and non-apremilast cohorts. The incidence rate (95% confidence interval) per 1000 person-years of opportunistic and serious infections in the apremilast-exposed cohort (64 [40-102])) was similar to incidence rates in the oral (50 [42-60]) and oral and injectable non-apremilast cohorts (57 [47-69]), while the incidence rates were lower in the injectable treatment-only cohort (20 [10-41]). Limitations include small numbers of apremilast-exposed patients and potential exposure misclassification partly owing to missing information on biologic and other specialty treatment use. CONCLUSIONS No new apremilast safety signals were identified in this study. These results provide evidence that the long-term safety of apremilast in psoriasis and psoriatic arthritis in a real-world setting is comparable to that reported in clinical trials.
Collapse
|
60
|
Altered platelet functions during treatment with apremilast for psoriatic arthritis: A case report. Curr Res Transl Med 2022; 70:103358. [PMID: 35724504 DOI: 10.1016/j.retram.2022.103358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 06/05/2022] [Indexed: 01/31/2023]
|
61
|
Quartuccio L, Sebastiani M, Spinelli FR, Di Marco F, Peluso R, D'Angelo S, Cauli A, Rossini M, Atzeni F. More than a random association between chronic obstructive pulmonary disease and psoriatic arthritis: shared pathogenic features and implications for treatment. Expert Rev Clin Immunol 2022; 18:983-990. [PMID: 35881045 DOI: 10.1080/1744666x.2022.2106969] [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: 11/04/2022]
Abstract
INTRODUCTION Psoriatic arthritis (PsA) is a chronic inflammatory condition characterized by skin and joints involvement, and with a great burden of comorbidity that could affect the choice of treatment. Chronic obstructive pulmonary disease (COPD) is one of the primary causes of morbidity and mortality. Medical therapy can improve symptoms and the frequency and severity of exacerbations. A variety of evidence showed an increasing association between COPD and PsA. AREAS COVERED Psoriatic disease and COPD appear to have a possible pathophysiologic link. The inhibition of intracellular molecules responsible for pro-inflammatory responses could be a therapeutic approach for both psoriatic diseases and COPD. Inhibitors of phosphodiesterase 4 (PDE-4) were developed to treat chronic inflammatory conditions such as psoriasis, PsA and COPD. Roflumilast has been used to treat COPD and asthma, while Apremilast to treat psoriasis and PsA. Given the efficacy and safety of these treatments, we can speculate that blocking PDE-4 might also provide clinical benefits in patients with co-existing COPD and PsA. EXPERT OPINION This hypothesis could offer the opportunity to screen patients for both diseases. Furthermore, this approach would increase the involvement of other specialists in the management of PsA, and it would improve the use of a tailored treatment for each patient.
Collapse
Affiliation(s)
- Luca Quartuccio
- Department of Medicine, Rheumatology Unit, University of Udine, Udine, Italy
| | - Marco Sebastiani
- Rheumatology Unit, Azienda Policlinico di Modena, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Romana Spinelli
- Reumatologia, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, Rome, Italy
| | - Fabiano Di Marco
- Respiratory Unit, Department of Health Sciences, University of Milan, Milan, Italy
| | - Rosario Peluso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II of Naples, Naples, Italy
| | - Salvatore D'Angelo
- Rheumatology Institute of Lucania (IReL) and Rheumatology Department of Lucania, San Carlo Hospital of Potenza, Potenza, Italy
| | - Alberto Cauli
- Rheumatology Unit, Department of Medical Sciences and Public Health, AOU and University of Cagliari, Cagliari, Italy
| | - Maurizio Rossini
- Department of Medicine, University of Verona, Rheumatology Unit, Azienda Ospedaliera Universitaria Integrata di Verona, Policlinico Borgo Roma, Verona, Italy
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, Messina, Italy
| |
Collapse
|
62
|
Merzel Šabović EK, Starbek Zorko M, Janić M. Killing Two Birds with One Stone: Potential Therapies Targeting Psoriasis and Atherosclerosis at the Same Time. Int J Mol Sci 2022; 23:ijms23126648. [PMID: 35743091 PMCID: PMC9224172 DOI: 10.3390/ijms23126648] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/10/2022] [Accepted: 06/12/2022] [Indexed: 01/27/2023] Open
Abstract
Psoriasis is a chronic systemic inflammatory disease. Due to systemic inflammation, it is associated with many comorbidities. Among them, cardiovascular diseases represent the most common causes of morbidity and mortality in this population. Therefore, physicians treating patients with psoriasis should keep in mind that, as important as the treatment of psoriasis, awareness of cardiovascular risk deserves additional attention. Thus, in parallel with psoriasis treatment, a cardiovascular risk assessment must also be performed and addressed accordingly. In addition to encouraging non-pharmacologic strategies for a healthy lifestyle, physicians should be familiar with different pharmacologic options that can target psoriasis and reduce cardiovascular risk. In the present article, we present the pathophysiological mechanisms of the psoriasis and cardiometabolic interplay, our view on the interaction of psoriasis and cardiovascular disease, review the atherosclerotic effect of therapeutic options used in psoriasis, and vice versa, i.e., what the effect of medications used in the prevention of atherosclerosis could be on psoriasis.
Collapse
Affiliation(s)
- Eva Klara Merzel Šabović
- Department of Dermatovenerology, University Medical Centre Ljubljana, Gradiškova Ulica 10, SI-1000 Ljubljana, Slovenia;
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia;
- Correspondence:
| | - Mateja Starbek Zorko
- Department of Dermatovenerology, University Medical Centre Ljubljana, Gradiškova Ulica 10, SI-1000 Ljubljana, Slovenia;
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia;
| | - Miodrag Janić
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, SI-1000 Ljubljana, Slovenia;
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, Zaloška Cesta 7, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
63
|
Wakiya R, Ushio Y, Ueeda K, Kameda T, Shimada H, Nakashima S, Kato M, Miyagi T, Sugihara K, Mizusaki M, Mino R, Kadowaki N, Dobashi H. Efficacy and safety of apremilast and its impact on serum cytokine levels in patients with Behçet's disease. Dermatol Ther 2022; 35:e15616. [DOI: 10.1111/dth.15616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Risa Wakiya
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine Kagawa University
| | - Yusuke Ushio
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine Kagawa University
| | - Kiyo Ueeda
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine Kagawa University
| | - Tomohiro Kameda
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine Kagawa University
| | - Hiromi Shimada
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine Kagawa University
| | - Shusaku Nakashima
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine Kagawa University
| | - Mikiya Kato
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine Kagawa University
| | - Taichi Miyagi
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine Kagawa University
| | - Koichi Sugihara
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine Kagawa University
| | - Mao Mizusaki
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine Kagawa University
| | - Rina Mino
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine Kagawa University
| | - Norimitsu Kadowaki
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine Kagawa University
| | - Hiroaki Dobashi
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine Kagawa University
| |
Collapse
|
64
|
Chiapparoli I, Galluzzo C, Salvarani C, Pipitone N. A glance into the future of myositis therapy. Ther Adv Musculoskelet Dis 2022; 14:1759720X221100299. [PMID: 35634354 PMCID: PMC9136432 DOI: 10.1177/1759720x221100299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/26/2022] [Indexed: 12/24/2022] Open
Abstract
The idiopathic inflammatory myopathies are chronic diseases of the skeletal muscle that comprise various conditions, including dermatomyositis, polymyositis, immune-mediated necrotizing myopathy, and the antisynthetase syndrome. Although there are a number of distinguishing features, all these disorders are characterized by an immune and inflammatory response mainly directed against the muscle. Hence, therapy is geared toward curbing the autoimmune and inflammatory response. A quite wide range of medications are currently available to treat these disorders, but despite all therapeutic progress still a number of patients are unable to maintain a sustained remission. In this review article, we have marshaled a variety of potential therapeutic agents that may hold promise for the future treatment of the idiopathic inflammatory myopathies. It is to be expected that by increasing the therapeutic armamentarium with agents that have different mechanisms of action even challenging cases could be successfully managed, thus reducing disease burden and disability.
Collapse
Affiliation(s)
| | | | - Carlo Salvarani
- Azienda USL-IRCCS di Reggio Emilia,
Reggio Emilia, Italy
- Università di Modena e Reggio Emilia,
Reggio Emilia, Italy
| | - Nicolò Pipitone
- Arcispedale Santa Maria Nuova, Viale
Risorgimento 80, 42123 Reggio Emilia, Italy
- Azienda USL-IRCCS di Reggio Emilia,
Reggio Emilia, Italy
| |
Collapse
|
65
|
Abushal AS, Aleanizy FS, Alqahtani FY, Shakeel F, Iqbal M, Haq N, Alsarra IA. Self-Nanoemulsifying Drug Delivery System (SNEDDS) of Apremilast: In Vitro Evaluation and Pharmacokinetics Studies. Molecules 2022; 27:3085. [PMID: 35630561 PMCID: PMC9145325 DOI: 10.3390/molecules27103085] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/08/2022] [Accepted: 05/11/2022] [Indexed: 12/21/2022] Open
Abstract
Psoriatic arthritis is an autoimmune disease of the joints that can lead to persistent inflammation, irreversible joint damage and disability. The current treatments are of limited efficacy and inconvenient. Apremilast (APR) immediate release tablets Otezla® have 20-33% bioavailability compared to the APR absolute bioavailability of 73%. As a result, self-nanoemulsifying drug delivery systems (SNEDDS) of APR were formulated to enhance APR's solubility, dissolution, and oral bioavailability. The drug assay was carried out using a developed and validated HPLC method. Various thermodynamic tests were carried out on APR-SNEDDS. Stable SNEDDS were characterized then subjected to in vitro drug release studies via dialysis membrane. The optimum formulation was F9, which showed the maximum in vitro drug release (94.9%) over 24 h, and this was further investigated in in vivo studies. F9 was composed of 15% oil, 60% Smix, and 25% water and had the lowest droplet size (17.505 ± 0.247 nm), low PDI (0.147 ± 0.014), low ZP (-13.35 mV), highest %T (99.15 ± 0.131) and optimum increases in the relative bioavailability (703.66%) compared to APR suspension (100%) over 24 h. These findings showed that APR-SNEDDS is a possible alternative delivery system for APR. Further studies are warranted to evaluate the major factors that influence the encapsulation efficiency and stability of APR-containing SNEDDS.
Collapse
Affiliation(s)
- Ahad S. Abushal
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (F.S.A.); (F.Y.A.); (F.S.); (N.H.)
| | - Fadilah S. Aleanizy
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (F.S.A.); (F.Y.A.); (F.S.); (N.H.)
| | - Fulwah Y. Alqahtani
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (F.S.A.); (F.Y.A.); (F.S.); (N.H.)
| | - Faiyaz Shakeel
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (F.S.A.); (F.Y.A.); (F.S.); (N.H.)
| | - Muzaffar Iqbal
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
- Central Laboratory, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Nazrul Haq
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (F.S.A.); (F.Y.A.); (F.S.); (N.H.)
| | - Ibrahim A. Alsarra
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.S.A.); (F.S.A.); (F.Y.A.); (F.S.); (N.H.)
| |
Collapse
|
66
|
Scott KA, Cox PB, Njardarson JT. Phenols in Pharmaceuticals: Analysis of a Recurring Motif. J Med Chem 2022; 65:7044-7072. [PMID: 35533692 DOI: 10.1021/acs.jmedchem.2c00223] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Phenols and phenolic ethers are significant scaffolds recurring both in nature and among approved small-molecule pharmaceuticals. This compendium presents the first comprehensive compilation and analysis of the structures of U.S. FDA-approved molecules containing phenol or phenolic ether fragments. This dataset comprises 371 structures, which are strongly represented by natural products. A total of 55 of the compounds described here are on the World Health Organization's list of essential medicines. Structural analysis reveals significant differences in the physicochemical properties imparted by phenols versus phenol ethers, each having benefits and drawbacks for drug developability. Despite trends over the past decade to increase the fraction of sp3 centers in drug leads, thereby "escaping flatland", phenols and phenolic ethers are represented in 62% of small-molecule drugs approved in 2020, suggesting that this aromatic moiety holds a special place in drugs and natural products.
Collapse
Affiliation(s)
- Kevin A Scott
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States.,Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85721, United States
| | - Philip B Cox
- Drug Discovery Science and Technology, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Jon T Njardarson
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
67
|
Infante M, Padilla N, Alejandro R, Caprio M, Della-Morte D, Fabbri A, Ricordi C. Diabetes-Modifying Antirheumatic Drugs: The Roles of DMARDs as Glucose-Lowering Agents. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:571. [PMID: 35629988 PMCID: PMC9143119 DOI: 10.3390/medicina58050571] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023]
Abstract
Systemic inflammation represents a shared pathophysiological mechanism which underlies the frequent clinical associations among chronic inflammatory rheumatic diseases (CIRDs), insulin resistance, type 2 diabetes (T2D), and chronic diabetes complications, including cardiovascular disease. Therefore, targeted anti-inflammatory therapies are attractive and highly desirable interventions to concomitantly reduce rheumatic disease activity and to improve glucose control in patients with CIRDs and comorbid T2D. Therapeutic approaches targeting inflammation may also play a role in the prevention of prediabetes and diabetes in patients with CIRDs, particularly in those with traditional risk factors and/or on high-dose corticosteroid therapy. Recently, several studies have shown that different disease-modifying antirheumatic drugs (DMARDs) used for the treatment of CIRDs exert antihyperglycemic properties by virtue of their anti-inflammatory, insulin-sensitizing, and/or insulinotropic effects. In this view, DMARDs are promising drug candidates that may potentially reduce rheumatic disease activity, ameliorate glucose control, and at the same time, prevent the development of diabetes-associated cardiovascular complications and metabolic dysfunctions. In light of their substantial antidiabetic actions, some DMARDs (such as hydroxychloroquine and anakinra) could be alternatively termed "diabetes-modifying antirheumatic drugs", since they may be repurposed for co-treatment of rheumatic diseases and comorbid T2D. However, there is a need for future randomized controlled trials to confirm the beneficial metabolic and cardiovascular effects as well as the safety profile of distinct DMARDs in the long term. This narrative review aims to discuss the current knowledge about the mechanisms behind the antihyperglycemic properties exerted by a variety of DMARDs (including synthetic and biologic DMARDs) and the potential use of these agents as antidiabetic medications in clinical settings.
Collapse
Affiliation(s)
- Marco Infante
- Clinical Cell Transplant Program (CCTP), Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL 33136, USA; (R.A.); (C.R.)
- Department of Systems Medicine, Diabetes Research Institute Federation (DRIF), University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
- Section of Endocrinology, UniCamillus, Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Via Cola di Rienzo 28, 00192 Rome, Italy
| | - Nathalia Padilla
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Colonia Centroamérica L-823, Managua 14048, Nicaragua;
| | - Rodolfo Alejandro
- Clinical Cell Transplant Program (CCTP), Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL 33136, USA; (R.A.); (C.R.)
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy;
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta 247, 00166 Rome, Italy;
| | - David Della-Morte
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta 247, 00166 Rome, Italy;
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, 1120 NW 14th St., Miami, FL 33136, USA
| | - Andrea Fabbri
- Department of Systems Medicine, Diabetes Research Institute Federation (DRIF), University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| | - Camillo Ricordi
- Clinical Cell Transplant Program (CCTP), Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL 33136, USA; (R.A.); (C.R.)
| |
Collapse
|
68
|
Viswanath V, Joshi P, Lawate P, Tare D, Dhoot D, Mahadkar N, Barkate H. An Open-Label, Randomized, Prospective, Comparative, Three-Arm Clinical Trial to Evaluate the Safety and Effectiveness of Apremilast with Three Different Titration Methods in Patients with Chronic Plaque Psoriasis in India. Psoriasis (Auckl) 2022; 12:53-61. [PMID: 35496380 PMCID: PMC9041601 DOI: 10.2147/ptt.s357184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/17/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose To minimize adverse effects (AEs), apremilast is recommended to titrate at the initiation of therapy. But still, many patients experience AEs, resulting in discontinuation of therapy. As a result, many dermatologists have adapted to further titrate apremilast in different ways. The present study was planned to evaluate the safety and effectiveness of apremilast in different dose titration methods as initiation therapy in the treatment of plaque psoriasis. Patients and Methods In this open-label, randomized, prospective, comparative, three-arm, single center study, 128 plaque psoriasis patients were included. Patients were randomized into three groups. Group I received standard titration for the first 6 days; Group II received all tablets in a starter pack as once a day (OD) total for 13 days; and Group III received two starter packs as 8 tablets each of apremilast 10 mg and 20 mg as OD and 10 tablets of 30 mg as OD, in total for 26 days. All groups received apremilast 30 mg as twice a day after initial titration. The total duration of apremilast therapy in all groups was 16 weeks. Results In safety assessment, AEs were reported in 50%, 41.3% and 25% in Groups I, II and III, respectively (p <0.05) with nausea being the most common AE. In Group I, 10.53% of patients discontinued apremilast whereas 6.52% and 2.27% discontinued in Groups II and III respectively. Maximum number of AEs were seen in Group I in first week only (74.19%) compared with other groups. At week 16, on the Psoriasis Area and Severity Index, PASI 75 was achieved in 31.43%, 42.4% and 33.3% of patients in Groups I, II and III, respectively with no statistical difference between any groups. Conclusion It can be concluded that slower titration is a useful strategy for minimizing AEs while at the same time maintaining effectiveness of apremilast.
Collapse
Affiliation(s)
| | - Pradnya Joshi
- Department of Dermatology, Rajiv Gandhi Medical College, Thane, Mumbai, India
| | - Prakash Lawate
- Department of Dermatology, Rajiv Gandhi Medical College, Thane, Mumbai, India
| | - Dakshata Tare
- Department of Dermatology, Rajiv Gandhi Medical College, Thane, Mumbai, India
| | - Dhiraj Dhoot
- Department of Global Medical Affairs, Glenmark Pharmaceuticals Ltd, Mumbai, Maharashtra, India
- Correspondence: Dhiraj Dhoot, Department of Global Medical Affairs, Glenmark Pharmaceuticals Ltd, B D Sawant Marg, Andheri (E), Mumbai, Maharashtra, 400099, India, Tel +91 9619811219, Email
| | - Namrata Mahadkar
- Department of Global Medical Affairs, Glenmark Pharmaceuticals Ltd, Mumbai, Maharashtra, India
| | - Hanmant Barkate
- Department of Global Medical Affairs, Glenmark Pharmaceuticals Ltd, Mumbai, Maharashtra, India
| |
Collapse
|
69
|
Gao JC, Wu AG, Contento MN, Maher JM, Cline A. Apremilast in the Treatment of Plaque Psoriasis: Differential Use in Psoriasis. Clin Cosmet Investig Dermatol 2022; 15:395-402. [PMID: 35300435 PMCID: PMC8921676 DOI: 10.2147/ccid.s266036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/03/2022] [Indexed: 12/15/2022]
Abstract
Small molecule medications like apremilast are emerging as promising options for patients with psoriasis and other inflammatory conditions. Apremilast was approved by the Food and Drug Administration in 2014 for the management of both psoriasis and psoriatic arthritis. Apremilast inhibits phosphodiesterase-4, which increases the intracellular levels of cyclic AMP, thereby reducing inflammatory cytokine production. This review aims to discuss the published evidence and evaluate the differential use of apremilast in plaque psoriasis of the body and scalp, nail psoriasis, and palmoplantar psoriasis. In clinical trials, apremilast effectively reduced the severity of different dermatological manifestations of psoriasis and improved patients' quality of life. It has an acceptable safety profile and is generally well-tolerated. Oral medications like apremilast offer an alternative route of administration which can be more convenient and appropriate for some patients. Additionally, pharmacoeconomic analyses of available anti-psoriatic systemic agents favor apremilast as a cost-effective therapeutic option.
Collapse
Affiliation(s)
- Jia C Gao
- School of Medicine, New York Medical College, Valhalla, NY, USA
| | - Albert G Wu
- School of Medicine, New York Medical College, Valhalla, NY, USA
| | | | | | - Abigail Cline
- Department of Dermatology, New York Medical College, New York, NY, USA
| |
Collapse
|
70
|
Morgulchik N, Athanasopoulou F, Chu E, Lam Y, Kamaly N. Potential therapeutic approaches for targeted inhibition of inflammatory cytokines following COVID-19 infection-induced cytokine storm. Interface Focus 2022; 12:20210006. [PMID: 34956607 PMCID: PMC8662389 DOI: 10.1098/rsfs.2021.0006] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 10/14/2021] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a deadly respiratory disease caused by severe acute respiratory syndrome coronavirus 2, which has caused a global pandemic since early 2020 and severely threatened people's livelihoods and health. Patients with pre-diagnosed conditions admitted to hospital often develop complications leading to mortality due to acute respiratory distress syndrome (ARDS) and associated multiorgan failure and blood clots. ARDS is associated with a cytokine storm. Cytokine storms arise due to elevated levels of circulating cytokines and are associated with infections. Targeting various pro-inflammatory cytokines in a specific manner can result in a potent therapeutic approach with minimal host collateral damage. Immunoregulatory therapies are now of interest in order to regulate the cytokine storm, and this review will summarize and discuss advances in targeted therapies against cytokine storms induced by COVID-19.
Collapse
Affiliation(s)
- Nelli Morgulchik
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Foteini Athanasopoulou
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Edmund Chu
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Yoriko Lam
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Nazila Kamaly
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| |
Collapse
|
71
|
Ghislain PD, Lambert J, Lam Hoai XL, Hillary T, Roquet-Gravy PP, de la Brassinne M, Segaert S. Real-Life Effectiveness of Apremilast for the Treatment of Psoriasis in Belgium: Results From the Observational OTELO Study. Adv Ther 2022; 39:1068-1080. [PMID: 34977985 PMCID: PMC8866388 DOI: 10.1007/s12325-021-01981-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/27/2021] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Apremilast is approved for the treatment of psoriasis and psoriatic arthritis. However, data on the efficacy and safety of apremilast in clinical practice are limited. We assessed the real-world use and effectiveness of apremilast in patients with moderate to severe plaque psoriasis visiting dermatologist practices in Belgium, from the perspectives of the patient and the physician. METHODS This prospective observational study enrolled adults aged 18 years or more initiating apremilast between 6 April 2017 and 30 June 2018, per Belgian reimbursement criteria. Primary outcome was the Patient Benefit Index for Skin Diseases (PBI-S). Secondary outcomes included the Patient Global Assessment (PtGA), Dermatology Life Quality Index (DLQI), Psoriasis Area and Severity Index (PASI), and body surface area (BSA). Patients were followed up for up to 18 months. RESULTS Overall, 122 enrolled patients received at least one dose of apremilast, of which 89 received treatment for more than 150 days and were included in the reference population. Treatment goals most frequently identified (at least 70% of patients) as "very important" in the PBI-S were related to physical impairments. After 6 months of apremilast treatment, 61-78% of patients reported they had achieved these goals; only 12.5% assessed their disease as severe (PtGA, 53.6% at apremilast initiation) and over half reported a DLQI score of 5 or less, indicating improved quality of life. As assessed by the physician, 68.4% and 35.1% of patients achieved at least a 50% and 75% reduction in PASI, respectively, at month 6. Apremilast was well tolerated with no new safety signals identified. CONCLUSIONS Our real-world data indicate that apremilast fulfils the expectations of Belgian patients with moderate to severe psoriasis, and from the perspectives of both the patient and physician, apremilast has a positive impact on their disease. TRIAL REGISTRATION ClinicalTrials.gov Identifier NCT03097003.
Collapse
Affiliation(s)
- Pierre-Dominique Ghislain
- Department of Dermatology, University Hospitals Saint-Luc, Av. Hippocrate 10, 1200, Brussels, Belgium.
| | - Jo Lambert
- Department of Dermatology, Ghent University Hospital, Ghent, Belgium
| | - X L Lam Hoai
- Department of Dermatology, University Hospital Saint-Pierre, Brussels, Belgium
| | - Tom Hillary
- Department of Dermatology, University Hospital Leuven, Leuven, Belgium
| | | | | | | |
Collapse
|
72
|
Schick MA, Schlegel N. Clinical Implication of Phosphodiesterase-4-Inhibition. Int J Mol Sci 2022; 23:1209. [PMID: 35163131 PMCID: PMC8835523 DOI: 10.3390/ijms23031209] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 01/08/2023] Open
Abstract
The pleiotropic function of 3',5'-cyclic adenosine monophosphate (cAMP)-dependent pathways in health and disease led to the development of pharmacological phosphodiesterase inhibitors (PDE-I) to attenuate cAMP degradation. While there are many isotypes of PDE, a predominant role of PDE4 is to regulate fundamental functions, including endothelial and epithelial barrier stability, modulation of inflammatory responses and cognitive and/or mood functions. This makes the use of PDE4-I an interesting tool for various therapeutic approaches. However, due to the presence of PDE4 in many tissues, there is a significant danger for serious side effects. Based on this, the aim of this review is to provide a comprehensive overview of the approaches and effects of PDE4-I for different therapeutic applications. In summary, despite many obstacles to use of PDE4-I for different therapeutic approaches, the current data warrant future research to utilize the therapeutic potential of phosphodiesterase 4 inhibition.
Collapse
Affiliation(s)
- Martin Alexander Schick
- Department of Anesthesiology and Critical Care, Medical Center—University of Freiburg, 79106 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79110 Freiburg, Germany
| | - Nicolas Schlegel
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Wuerzburg, 97080 Würzburg, Germany;
| |
Collapse
|
73
|
Zhang Q, Durig T, Blass B, Fassihi R. Development of an amorphous based sustained release system for apremilast a selective phosphodiesterase 4 (PDE4) inhibitor. Int J Pharm 2022; 615:121516. [DOI: 10.1016/j.ijpharm.2022.121516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 10/19/2022]
|
74
|
Yamamoto-Furusho JK, Parra-Holguín NN. Emerging therapeutic options in inflammatory bowel disease. World J Gastroenterol 2021; 27:8242-8261. [PMID: 35068868 PMCID: PMC8717021 DOI: 10.3748/wjg.v27.i48.8242] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/04/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic disease that requires chronic treatment throughout the evolution of the disease, with a complex physiopathology that entails great challenges for the development of new and specific treatments for ulcerative colitis and Crohn´s disease. The anti-tumor necrosis factor alpha therapy has impacted the clinical course of IBD in those patients who do not respond to conventional treatment, so there is a need to develop new therapies and markers of treatment response. Various pathways involved in the development of the disease are known and the new therapies have focused on blocking the inflammatory process at the gastrointestinal level by oral, intravenous, subcutaneous, and topical route. All these new therapies can lead to more personalized treatments with higher success rates and fewer relapses. These treatments have not only focused on clinical remission, but also on achieving macroscopic changes at the endoscopic level and microscopic changes by achieving mucosal healing. These treatments are mainly based on modifying signaling pathways, by blocking receptors or ligands, reducing cell migration and maintaining the integrity of the epithelial barrier. Therefore, this review presents the efficacy and safety of the new treatments that are currently under study and the advances that have been made in this area in recent years.
Collapse
Affiliation(s)
- Jesus K Yamamoto-Furusho
- Gastroenterology Unit, Inflammatory Bowel Disease Clinic, Instituto Nacional de Ciencias Medicas y Nutricion, Mexico City 14080, Mexico
| | - Norma N Parra-Holguín
- Gastroenterology Unit, Inflammatory Bowel Disease Clinic, Instituto Nacional de Ciencias Medicas y Nutricion, Mexico City 14080, Mexico
| |
Collapse
|
75
|
Hidradenitis Suppurativa: Host-Microbe and Immune Pathogenesis Underlie Important Future Directions. JID INNOVATIONS 2021; 1:100001. [PMID: 34909706 PMCID: PMC8659377 DOI: 10.1016/j.xjidi.2021.100001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/24/2020] [Accepted: 12/10/2020] [Indexed: 12/17/2022] Open
Abstract
Hidradenitis suppurativa (HS) is an inflammatory disease of the skin with a chronic, relapsing-remitting course. The pathogenesis of the disease is poorly understood and involves multiple factors, including genetics, environment, host-microbe interactions, and immune dysregulation. In particular, the composition of the cutaneous microbiome shifts as the disease progresses, although it is unclear whether this is a primary or secondary process. Trials with immunomodulatory therapy elucidate the role of specific immune pathways and cytokine signaling in disease mechanism, such as TNF-α, IL-1β, IL-12, IL-17, IL-23, and complement. Future studies should continue examining the causes of and contributing factors to microbial changes and immune dysregulation in HS pathogenesis.
Collapse
Key Words
- AMP, antimicrobial peptide
- BD, β-defensin
- BMI, body mass index
- DC, dendritic cell
- DCD, dermcidin
- GSC, γ-secretase complex
- HS, hidradenitis suppurativa
- HiSCR, hidradenitis suppurativa clinical response
- IBD, inflammatory bowel disease
- IHS4, International Hidradenitis Suppurativa Severity Score System
- KC, keratinocyte
- MMP, matrix metalloproteinase
- NET, neutrophil extracellular traps
- NMSC, nonmelanoma skin cancer
- PG, pyoderma gangrenosum
- RCT, randomized controlled trial
- SAPHO, synovitis, acne, pustulosis, hyperostosis, and osteitis
- TLR, toll-like receptor
- Th, T helper type
- iNOS, inducible nitric oxide synthase
- pDC, plasmacytoid dendritic cell
Collapse
|
76
|
Reich K, Korge B, Magnolo N, Manasterski M, Schwichtenberg U, Staubach-Renz P, Kaiser S, Roemmler-Zehrer J, Gómez NN, Lorenz-Baath K. Quality-of-Life Outcomes, Effectiveness and Tolerability of Apremilast in Patients with Plaque Psoriasis and Routine German Dermatology Care: Results from LAPIS-PSO. Dermatol Ther (Heidelb) 2021; 12:203-221. [PMID: 34913153 PMCID: PMC8776950 DOI: 10.1007/s13555-021-00658-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/27/2021] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Psoriasis is a systemic inflammatory disease characterised by pruritic skin lesions that impair quality of life (QOL). Long-Term Documentation of the Utilization of Apremilast in Patients with Plaque Psoriasis under Routine Conditions (LAPIS-PSO; ClinicalTrials.gov: NCT02626793) was a 52-week, prospective, multicentre, observational cohort study conducted in real-world dermatology clinical settings in Germany. We evaluated physician- and patient-reported outcomes for QOL, effectiveness and tolerability in patients with moderate to severe psoriasis vulgaris in LAPIS-PSO. METHODS The primary endpoint was the percentage of patients achieving Dermatology Life Quality Index (DLQI) score ≤ 5 or ≥ 5-point improvement from baseline in DLQI score at visit 2 (~ 4 months after baseline). Secondary endpoints included assessments of symptoms and disease severity. Tolerability was evaluated based on adverse events (AEs). A pre-defined subgroup analysis based on baseline Physician's Global Assessment (PGA) score (2 or 3 versus 4) was performed. Data were examined descriptively through visit 5 (~ 13 months) using the last-observation-carried-forward (LOCF) approach and data as observed. RESULTS In total, 257 patients were included for efficacy assessment. On LOCF analysis, most patients achieved the primary endpoint at visit 2 (66.5%); DLQI response was maintained at visit 5 (72.4%). Earlier treatment response was observed in patients with a PGA score of 2 or 3 versus 4 (visit 1 PASI ≤ 3: 20.5% versus 10.8%). Adverse events were consistent with the known safety profile of apremilast. CONCLUSIONS In routine clinical care in Germany, patients with moderate to severe plaque psoriasis benefited from apremilast treatment up to ~ 13 months, consistent with findings from clinical trials, with a good safety profile.
Collapse
Affiliation(s)
- Kristian Reich
- Translational Research in Inflammatory Skin Diseases, Institute for Health Services Research in Dermatology and Nursing, University Medical Center Hamburg-Eppendorf, Martinistraße 52, Building West 38/Room 514, 20246, Hamburg, Germany.
| | - Bernhard Korge
- Hautarztpraxis Priv. Doz. Dr. med. Bernhard Korge, Düren, Germany
| | - Nina Magnolo
- Department of Dermatology, University Hospital Muenster, Münster, Germany
| | | | | | | | | | | | | | | |
Collapse
|
77
|
Meier-Schiesser B, Mellett M, Ramirez-Fort MK, Maul JT, Klug A, Winkelbeiner N, Fenini G, Schafer P, Contassot E, French LE. Phosphodiesterase-4 Inhibition Reduces Cutaneous Inflammation and IL-1β Expression in a Psoriasiform Mouse Model but Does Not Inhibit Inflammasome Activation. Int J Mol Sci 2021; 22:ijms222312878. [PMID: 34884681 PMCID: PMC8657753 DOI: 10.3390/ijms222312878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 11/14/2021] [Accepted: 11/24/2021] [Indexed: 11/16/2022] Open
Abstract
Apremilast (Otezla®) is an oral small molecule phosphodiesterase 4 (PDE4) inhibitor approved for the treatment of psoriasis, psoriatic arthritis, and oral ulcers associated with Behçet’s disease. While PDE4 inhibition overall is mechanistically understood, the effect of apremilast on the innate immune response, particularly inflammasome activation, remains unknown. Here, we assessed the effect of apremilast in a psoriasis mouse model and primary human cells. Psoriatic lesion development in vivo was studied in K5.Stat3C transgenic mice treated with apremilast for 2 weeks, resulting in a moderate (2 mg/kg/day) to significant (6 mg/kg/day) resolution of inflamed plaques after 2-week treatment. Concomitantly, epidermal thickness dramatically decreased, the cutaneous immune cell infiltrate was reduced, and proinflammatory cytokines were significantly downregulated. Additionally, apremilast significantly inhibited lipopolysaccharide- or anti-CD3-induced expression of proinflammatory cytokines in peripheral mononuclear cells (PBMCs). Notably, inflammasome activation and secretion of IL-1β were not inhibited by apremilast in PBMCs and in human primary keratinocytes. Collectively, apremilast effectively alleviated the psoriatic phenotype of K5.Stat3 transgenic mice, further substantiating PDE4 inhibitor-efficiency in targeting key clinical, histopathological and inflammatory features of psoriasis. Despite lacking direct effect on inflammasome activation, reduced priming of inflammasome components upon apremilast treatment reflected the indirect benefit of PDE4 inhibition in reducing inflammation.
Collapse
Affiliation(s)
- Barbara Meier-Schiesser
- Department of Dermatology, University Hospital Zurich (USZ), Raemistrasse 100, 8091 Zurich, Switzerland; (M.M.); (J.-T.M.); (A.K.); (N.W.); (G.F.)
- Correspondence: ; Tel.: +41-43-255-11-11
| | - Mark Mellett
- Department of Dermatology, University Hospital Zurich (USZ), Raemistrasse 100, 8091 Zurich, Switzerland; (M.M.); (J.-T.M.); (A.K.); (N.W.); (G.F.)
| | | | - Julia-Tatjana Maul
- Department of Dermatology, University Hospital Zurich (USZ), Raemistrasse 100, 8091 Zurich, Switzerland; (M.M.); (J.-T.M.); (A.K.); (N.W.); (G.F.)
| | - Annika Klug
- Department of Dermatology, University Hospital Zurich (USZ), Raemistrasse 100, 8091 Zurich, Switzerland; (M.M.); (J.-T.M.); (A.K.); (N.W.); (G.F.)
| | - Nicola Winkelbeiner
- Department of Dermatology, University Hospital Zurich (USZ), Raemistrasse 100, 8091 Zurich, Switzerland; (M.M.); (J.-T.M.); (A.K.); (N.W.); (G.F.)
| | - Gabriele Fenini
- Department of Dermatology, University Hospital Zurich (USZ), Raemistrasse 100, 8091 Zurich, Switzerland; (M.M.); (J.-T.M.); (A.K.); (N.W.); (G.F.)
| | - Peter Schafer
- Bristol Myers Squibb, 100 Nassau Park Blvd #300, Princeton, NJ 08540, USA;
| | - Emmanuel Contassot
- Department of Biomedicine, Dermatology Department, Basel University Hospital, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland;
| | - Lars E. French
- Department of Dermatology, Ludwigs-Maximilians-University, Frauenlobstraße 9-11, 80337 Munich, Germany;
- Dr. Philip Frost Department of Dermatology and Cutaneous Surgery, Miller School of Medicine, University of Miami, 1600 NW 10th Avenue, Miami, FL 33136, USA
| |
Collapse
|
78
|
Sharma K, Choudhary S, Silakari O. Portraying molecular modulation and therapeutic aspects of psoriasis: Retrospection and current status. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
79
|
Frank B, Guo H, Lebrec H, Wang X. Application of a newly-developed cynomolgus macaque BiTE-mediated cytotoxic T-lymphocyte activity assay to various immunomodulatory agents in vitro. J Immunotoxicol 2021; 18:154-162. [PMID: 34714999 DOI: 10.1080/1547691x.2021.1992687] [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: 10/20/2022] Open
Abstract
The immunotoxic potential of drug candidates is assessed through the examination of results from a variety of in vitro and in vivo immunophenotyping and functional study endpoints in pre-clinical studies. CD8+ cytotoxic T-lymphocyte (CTL) activity impairment by immunosuppressive agents is recognized to be a potentiating factor for decreased antiviral defense and increased cancer risk. A bi-specific T-cell engager (BiTE®)-mediated CTL activity assay that applies to ex vivo experimentation in non-human primates in the context of toxicology studies was successfully developed and applied in cynomolgus monkey regulatory studies. While an ex vivo analysis conducted in the context of repeat-dose toxicology studies focuses on the long-term impact on CTL function, an in vitro assay with the same experimental design captures acute effects in the presence of the test article. Here, the in vitro assay was applied to a list of drugs with known clinical immunomodulatory impact to understand the applicability of the assay. The results showed this assay was sensitive to a wide range of immunosuppressants directly targeting cell-intrinsic signaling pathways in activated CTL. However, agents executing immuno-modulation through inhibiting cytokines/cytokine receptors, co-stimulatory molecules, and cell adhesion and migration pathways did not impair the CTL activity in this short-term in vitro culture. In addition, anti-PD-1/PD-L1 immune checkpoint blockers enhanced the CTL activity. Taken together, the results here demonstrate that in concordance with their mechanism of action, the in vitro BiTE®-mediated CTL assay is applicable and sensitive to immunomodulatory agents acting via a variety of mechanisms.
Collapse
Affiliation(s)
- Brendon Frank
- Translational Safety and Bioanalytical Sciences, Amgen Research, Amgen Inc., South San Francisco, CA, USA
| | - Hao Guo
- Translational Safety and Bioanalytical Sciences, Amgen Research, Amgen Inc., South San Francisco, CA, USA
| | - Hervé Lebrec
- Translational Safety and Bioanalytical Sciences, Amgen Research, Amgen Inc., South San Francisco, CA, USA
| | - Xiaoting Wang
- Translational Safety and Bioanalytical Sciences, Amgen Research, Amgen Inc., South San Francisco, CA, USA
| |
Collapse
|
80
|
McMichael J, Stoff BK. Treatment of a perforating dermatosis with apremilast. JAAD Case Rep 2021; 16:155-157. [PMID: 34632026 PMCID: PMC8488176 DOI: 10.1016/j.jdcr.2021.08.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Josette McMichael
- Correspondence to: Josette McMichael, MD, Department of Dermatology, Emory University School of Medicine, 1525 Clifton Road NE 1st Floor, Atlanta, GA 30322. @globaldermie
| | | |
Collapse
|
81
|
Zhang Q, Li A, Yan Y, Wu Y, Zhang X. Systematic thermodynamic analysis of apremilast polymorphs via solubility measurement with modeling: Mechanism evaluation through molecular simulation. Eur J Pharm Sci 2021; 165:105958. [PMID: 34314840 DOI: 10.1016/j.ejps.2021.105958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/28/2021] [Accepted: 07/23/2021] [Indexed: 11/25/2022]
Abstract
The polymorphism of apremilast has been investigated. Two polymorphs have been identified and characterized by differential scanning calorimeter, fourier transform infrared spectroscopy, and powder X-ray diffractometer. Solubilities of apremilast forms B and E in three binary solvents of methanol-water, acetonitrile-water, and acetonitrile-methanol have been measured using the static method at a temperature ranging from 288.15 K to 328.15 K under standard atmospheric pressure. Subsequently, the solubility data have been analyzed using the Wilson, NRTL, and UNIQUAC thermodynamic models, respectively. Furthermore, the Gibbs energy of solution and the radial distribution function have been calculated using the molecular simulation method to evaluate the dissolution mechanism. The Gibbs energy of solution reveals that the rank of solute-solvent interaction correlated well with solubility order in binary solvent mixtures, and the radial distribution function indicates that weakening of solvent-solvent interaction led to an increase in solubility.
Collapse
Affiliation(s)
- Qi Zhang
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Ang Li
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yizhen Yan
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yanyang Wu
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Xiangyang Zhang
- State Key Laboratory of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
82
|
Larsson J, Hoppe E, Gautrois M, Cvijovic M, Jirstrand M. Second-generation TNFα turnover model for improved analysis of test compound interventions in LPS challenge studies. Eur J Pharm Sci 2021; 165:105937. [PMID: 34260892 DOI: 10.1016/j.ejps.2021.105937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/31/2021] [Accepted: 07/04/2021] [Indexed: 11/30/2022]
Abstract
This study presents a non-linear mixed effects model describing tumour necrosis factor alpha (TNFα) release after lipopolysaccharide (LPS) provocations in absence or presence of anti-inflammatory test compounds. Inter-occasion variability and the pharmacokinetics of two test compounds have been added to this second-generation model, and the goal is to produce a framework of how to model TNFα response in LPS challenge studies in vivo and demonstrate its general applicability regardless of occasion or type of test compound. Model improvements based on experimental data were successfully implemented and provided a robust model for TNFα response after LPS provocation, as well as reliable estimates of the median pharmacodynamic parameters. The two test compounds, Test Compound A and roflumilast, showed 81.1% and 74.9% partial reduction of TNFα response, respectively, and the potency of Test Compound A was estimated to 0.166 µmol/L. Comparing this study with previously published work reveals that our model leads to biologically reasonable output, handles complex data pooled from different studies, and highlights the importance of accurately distinguishing the stimulatory effect of LPS from the inhibitory effect of the test compound.
Collapse
Affiliation(s)
- Julia Larsson
- Fraunhofer-Chalmers Centre, Chalmers Science Park, 412 88 Gothenburg, Sweden.; Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, 412 96 Gothenburg, Sweden..
| | | | | | - Marija Cvijovic
- Department of Mathematical Sciences, Chalmers University of Technology and University of Gothenburg, 412 96 Gothenburg, Sweden
| | - Mats Jirstrand
- Fraunhofer-Chalmers Centre, Chalmers Science Park, 412 88 Gothenburg, Sweden
| |
Collapse
|
83
|
Kolla AM, Liu L, Shaw K, Shapiro J, Femia A, Lo Sicco K. A narrative review of therapies for scalp dermatomyositis. Dermatol Ther 2021; 34:e15138. [PMID: 34549494 DOI: 10.1111/dth.15138] [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: 06/08/2021] [Revised: 09/08/2021] [Accepted: 09/19/2021] [Indexed: 11/30/2022]
Abstract
Cutaneous involvement of the scalp is a common manifestation of dermatomyositis (DM), occurring in up to 82% of adults with DM. Scalp DM predominantly affects women and is characterized by dermatitis, alopecia, pruritus, and/or burning. While cutaneous DM negatively impacts quality-of-life, scalp symptoms in particular are often severe, debilitating, and recalcitrant to standard DM therapies. Currently, there is a paucity of guidelines to inform management of scalp symptoms in patients with cutaneous DM. In this narrative review, we summarize the treatments utilized to manage scalp DM and highlight potential areas for future research. We identified eight studies that reported on 27 treatments focused on cutaneous DM and described outcomes on scalp symptoms. A majority of the treatments were standard therapies for cutaneous DM and resulted in no or minimal improvement in scalp symptoms. Five therapies did result in complete resolution of scalp symptoms and were recommended as potential areas of future research. These included low-dose naltrexone and platelet-rich plasma, as well as two frequent and one less common therapy for cutaneous DM respectively: intravenous immunoglobulin, rituximab, and apremilast. Though the literature was not systematically assessed in this review, these findings illustrate not only that strategies for refractory scalp DM are lacking, but also that those demonstrating potential efficacy are limited by low levels of evidence. Additional studies, especially randomized controlled trials, are needed to better inform management of scalp DM.
Collapse
Affiliation(s)
- Avani M Kolla
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York, USA
| | - Lynn Liu
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York, USA
| | - Katharina Shaw
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York, USA
| | - Jerry Shapiro
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York, USA
| | - Alisa Femia
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York, USA
| | - Kristen Lo Sicco
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
84
|
Practical and Asymmetric Synthesis of Apremilast Using Ellman’s Sulfinamide as a Chiral Auxiliary. MOLBANK 2021. [DOI: 10.3390/m1275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Herein, we described a new protocol for the asymmetric synthesis of apremilast using tert-butanesulfinamide as a chiral auxiliary. This synthetic route consisted of four steps starting from the commercially available 3-hydroxy-4-methoxybenzaldehyde, and apremilast was accordingly obtained in an overall 56% yield and with 95.5% ee.
Collapse
|
85
|
Sule RR, Choudhary P, Salunke TS. Apremilast induced tinnitus: An unreported side effect. Indian J Pharmacol 2021; 53:328-329. [PMID: 34414912 PMCID: PMC8411969 DOI: 10.4103/ijp.ijp_533_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Rajiv Ramesh Sule
- Department of Dermatology, Deenanath Mangeshkar Hospital and Research Centre, Pune, Maharashtra, India
| | - Paras Choudhary
- Department of Dermatology, Deenanath Mangeshkar Hospital and Research Centre, Pune, Maharashtra, India
| | | |
Collapse
|
86
|
Okubo Y, Ohtsuki M, Komine M, Imafuku S, Kassir N, Petric R, Nemoto O. Population pharmacokinetic and exposure-response analysis of apremilast in Japanese subjects with moderate to severe psoriasis. J Dermatol 2021; 48:1652-1664. [PMID: 34396569 PMCID: PMC9290614 DOI: 10.1111/1346-8138.16068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/02/2021] [Indexed: 01/08/2023]
Abstract
Apremilast is an orally available phosphodiesterase 4 inhibitor used for the treatment of moderate to severe psoriasis. The aims of this analysis were to develop a population pharmacokinetic (PPK) model of apremilast based on observed data from phase 1 studies combined with clinical trial data from subjects with moderate to severe psoriasis, and to develop exposure–response (E‐R) models to determine whether Japanese subjects with moderate to severe psoriasis achieve response to apremilast treatment similar to that observed in non‐Japanese, predominantly Caucasian subjects with moderate to severe psoriasis. The PPK model demonstrated that apremilast plasma concentrations and overall apparent clearance rate were comparable between the Japanese and Caucasian subgroups. The E‐R analyses of ≥75% or ≥50% improvement from baseline in Psoriasis Area and Severity Index score and achievement of static Physician Global Assessment score of 0 (clear) or 1 (almost clear) at week 16 indicated that apremilast treatment in Japanese subjects approached the maximal effect with response rates comparable to those in predominantly Caucasian subjects. Overall, the analyses confirm that the approved apremilast 30 mg b.i.d. dose is appropriate for Japanese subjects with moderate to severe psoriasis, with an efficacy profile similar to that previously observed in Caucasian subjects.
Collapse
Affiliation(s)
| | | | | | | | - Nastya Kassir
- Certara Strategic Consulting, Princeton, New Jersey, USA
| | | | | |
Collapse
|
87
|
Takeno M, Dobashi H, Tanaka Y, Kono H, Sugii S, Kishimoto M, Cheng S, McCue S, Paris M, Chen M, Ishigatsubo Y. Apremilast in a Japanese subgroup with Behçet's syndrome: Results from a Phase 3, randomised, double-blind, placebo-controlled study. Mod Rheumatol 2021; 32:413-421. [PMID: 34894266 DOI: 10.1093/mr/roab008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/01/2021] [Accepted: 05/18/2021] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Apremilast efficacy and safety was assessed in a prespecified subgroup of Japanese patients with oral ulcers associated with Behçet's syndrome from a Phase 3 randomized, placebo-controlled, double-blind study of apremilast (RELIEF). METHODS The primary end point was area under the curve for number of oral ulcers during the 12-week placebo-controlled phase (AUCWk0-12). Key secondary end points were change from baseline in oral ulcer pain, complete oral ulcer resolution, and measures of disease activity and quality of life (QoL). RESULTS Thirty-nine Japanese patients were randomised (apremilast 30 mg BID: n = 19; placebo: n = 20). Improvements at Week 12 were observed for apremilast vs. placebo in AUCWk0-12 for the number of oral ulcers (115.9 vs. 253.3; nominal P = 0.0168); 57.9% vs. 25.0% achieved complete oral ulcer resolution, 47.4% vs. 0.0% achieved oral ulcer resolution by Week 6 and maintained oral ulcer-free status for ≥6 additional weeks; mean change from baseline in BSAS was -10.5 vs. 0.5. Favourable effects were observed for apremilast vs. placebo in other secondary end points, including QoL. Clinical benefits were sustained over 28 weeks of continued apremilast treatment. Adverse events were consistent with apremilast's known safety profile. CONCLUSIONS Apremilast reduced the number of oral ulcers and overall disease activity in this Japanese subgroup with Behçet's syndrome.
Collapse
Affiliation(s)
- Mitsuhiro Takeno
- Department of Allergy and Rheumatology, Nippon Medical School, Musashi Kosugi Hospital, Kanagawa, Japan
| | - Hiroaki Dobashi
- Division of Hematology, Rheumatology and Respiratory Medicine, Department of Internal Medicine, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | | | - Hajime Kono
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Shouji Sugii
- Department of Rheumatic Diseases, Tokyo Metropolitan Matsuzawa Hospital, Tokyo, Japan
| | - Mitsumasa Kishimoto
- Department of Nephrology and Rheumatology, Kyorin University School of Medicine, Tokyo, Japan
| | | | - Shannon McCue
- Celgene Corporation, Summit, NJ, USA.,The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | | | | | - Yoshiaki Ishigatsubo
- Department of Internal Medicine and Clinical Immunology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
88
|
Jia Y, Chen X, Sun J. Apremilast ameliorates IL-1α-induced dysfunction in epidermal stem cells. Aging (Albany NY) 2021; 13:19293-19305. [PMID: 34375302 PMCID: PMC8386542 DOI: 10.18632/aging.203265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/09/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND PURPOSE Skin tissue is the natural barrier that protects our body, the damage of which can be repaired by the epidermal stem cells (ESCs). However, external factors abolish the self-repair ability of ESCs by inducing oxidative stress and severe inflammation. Apremilast is a small molecular inhibitor of phosphodiesterase 4 that was approved for the treatment of psoriasis. In the present study, the protective property of Apremilast against IL-1α-induced dysfunction on epidermal stem cells, as well as the preliminary mechanism, will be investigated. METHODS ESCs were isolated from neonatal mice. The expression levels of TNF-α, IL-8, IL-12, MMP-2, and MMP-9 were detected using real-time PCR and ELISA. MitoSOX Red assay was used to determine the level of mitochondrial reactive oxygen species (ROS). Western blot and real-time PCR were utilized to determine the expression levels of IL-1R1, Myd88, and TRAF6. Activation of NF-κB was assessed by measuring the p-NF-κB p65 and luciferase activity. Capacities of ESCs were evaluated by measuring the gene expressions of integrin β1 and Krt19 using real-time PCR. RESULTS Firstly, the expression levels of TNF-α, IL-8, IL-12, MMP-2, MMP-9 and IL-1R1, as well as the ROS level, were significantly elevated by IL-1α but greatly suppressed by treatment with Apremilast. Subsequently, we found that the activated Myd88/TRAF6/NF-κB signaling pathway induced by stimulation with IL-1α was significantly inhibited by the introduction of Apremilast. As a result, Apremilast protected ESCs against IL-1α-induced impairment in capacities of ESCs, this was verified by the elevated expression levels of integrin β1 and Krt19. CONCLUSIONS Apremilast might ameliorate IL-1α-induced dysfunction in ESCs by mitigating oxidative stress and inflammation through inhibiting the activation of the Myd88/TRAF6/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yuxi Jia
- Department of Dermatology, The China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Xiangru Chen
- Department of Dermatology, The China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| | - Jing Sun
- Department of Dermatology, The China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, China
| |
Collapse
|
89
|
Rajagopalan M, Dogra S, Saraswat A, Varma S, Banodkar P. The Use of Apremilast in Psoriasis: An Indian Perspective on Real-World Scenarios. Psoriasis (Auckl) 2021; 11:109-122. [PMID: 34430450 PMCID: PMC8375310 DOI: 10.2147/ptt.s320810] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/20/2021] [Indexed: 12/27/2022] Open
Abstract
Apremilast, an oral phosphodiesterase-4 inhibitor, is approved for use in the management of psoriasis and psoriatic arthritis. Although its efficacy and safety have been well established in clinical studies, in real-world settings, different practice scenarios have been reported. This review paper serves to evaluate clinical real-world scenarios and aspects of treatment for which the information in the literature was considered to be lacking or controversial. Following a literature review, a panel of five dermatologists with expertise in psoriasis considered five scenarios; namely, the positioning of apremilast in psoriasis, its use in difficult-to-treat areas, special conditions and populations, safety, dose titration and dose in maintenance therapy. These were then assessed with psoriasis experts in India using a web-based questionnaire. A total of 28 questions were discussed regarding these scenarios. According to the responses, apremilast is effective in stable mild to moderate psoriasis as monotherapy and in severe psoriasis in combination. Also, a positive response was received with regard to its effectiveness in difficult locations such as the scalp, palms and soles. To reduce adverse effects, prolonged titration therapy over 4 weeks is required and lower doses can be prescribed to maintain remission. Apremilast therapy should be continued for a minimum of 8 weeks once initiated to achieve the desired results, and the total duration of therapy should be about 24 weeks for better efficacy. It is also effective in many other cases, such as obese patients, patients with hepatitis B or C and HIV, or patients on polypharmacy. It was also reported that apremilast requires less prescreening and monitoring than other conventional and biologic systemic therapies. Overall, apremilast is an attractive option for the individualized treatment of psoriasis owing to its favorable safety profile, its ease of oral administration without the need for screening or ongoing laboratory monitoring, and its positive impact on symptoms and lesions in difficult-to-treat areas.
Collapse
Affiliation(s)
- Murlidhar Rajagopalan
- Department of Dermatology, Apollo Hospitals, Chennai, Tamilnadu, India
- Correspondence: Murlidhar Rajagopalan Dermatology, Apollo Hospital, No 21, Greams Lane, Off Greams Road, Chennai, 600006, Tamil Nadu, IndiaTel +91 9840045500 Email
| | - Sunil Dogra
- Department of Dermatology, Venereology & Leprology, PGIMER, Chandigarh, India
| | - Abir Saraswat
- Indushree Skin Clinic, Lucknow, Uttar Pradesh, India
| | - Sachin Varma
- Skinvita Clinic Kolkata, Department of Dermatology, Apollo Hospital, Kolkata, West Bengal, India
| | - Pravin Banodkar
- Skin Crest Clinic, Mumbai, Department of Dermatology, Saifee Hospital, Breach Candy Hospital, and St. Elizabeth’s Hospital, Mumbai, Maharashtra, India
| |
Collapse
|
90
|
Cohen-Sors R, Fougerousse AC, Reguiai Z, Maccari F, Mahé E, Delaunay J, Roussel A, de la Breteque MA, Cottencin C, Bertolotti A, Kemp H, Chaby G. Biological Therapies or Apremilast in the Treatment of Psoriasis in Patients with a History of Hematologic Malignancy: Results from a Retrospective Study in 21 Patients. Clin Cosmet Investig Dermatol 2021; 14:845-854. [PMID: 34267533 PMCID: PMC8275167 DOI: 10.2147/ccid.s320098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/25/2021] [Indexed: 01/30/2023]
Abstract
Background Few studies addressing the safety and efficacy of biological therapy (BT) or apremilast (APR) in patients with psoriasis with a history of hematologic malignancy (HM) exist. Aim To describe the tolerance and efficacy of BT and APR in moderate-to-severe psoriasis in patients with a history of in-remission or evolving HM. Methodology A retrospective, multicenter chart review of the tolerance and efficacy of BT or APR in patients with moderate-to-severe psoriasis and a clinical history of in-remission or evolving HM. Results Twenty-one patients with severe psoriasis and a history of HM were included in France by the GEM Resopso study group. Of the 16 patients treated with one or more BT lines, none showed recurrence of their HM which was considered as stable or in remission, and only 2 patients showed an evolution of their HM which had been considered as stable at the beginning of treatment. In the 10 patients treated with APR, the HM of one patient who also received BT worsened. The 3 evolutions did not impact the treatment with BT or APR. Tolerance was very satisfactory, with a low occurrence of infections. Regarding efficacy, only one patient treated with APR did not achieve any notable clinical improvement. Conclusion Despite supportive data regarding tolerance, the heterogeneity of the analyzed population and limited available data, BT and APR should be used with caution in this patient population and investigations on larger cohorts should be conducted to further assess their tolerance in this patient population.
Collapse
Affiliation(s)
- Raphaella Cohen-Sors
- Dermatology Department, Amiens-Picardie University Hospital Center, Amiens, France
| | | | - Ziad Reguiai
- Dermatology Department, Polyclinic Courlancy, Reims, France
| | - Francois Maccari
- Dermatology Department, Military Teaching Hospital Bégin, Saint-Mandé, France
| | - Emmanuel Mahé
- Dermatology Department, Argenteuil Hospital, Argenteuil, France
| | | | - Aude Roussel
- Dermatology Department, Military Teaching Hospital Bégin, Saint-Mandé, France
| | | | | | - Antoine Bertolotti
- Dermatology Department, University Hospital, Saint-Pierre, Ile de la Réunion, France
| | - Hélène Kemp
- Hematology Department, Amiens-Picardie University Hospital Center, Amiens, France
| | - Guillaume Chaby
- Dermatology Department, Amiens-Picardie University Hospital Center, Amiens, France
| |
Collapse
|
91
|
Narode H, Gayke M, Eppa G, Yadav JS. A Review on Synthetic Advances toward the Synthesis of Apremilast, an Anti-inflammatory Drug. Org Process Res Dev 2021. [DOI: 10.1021/acs.oprd.1c00107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Hanuman Narode
- Department of Chemistry, School of Science, Indrashil University, Kadi, Mehsana 382715, India
| | - Manoj Gayke
- Department of Chemistry, School of Science, Indrashil University, Kadi, Mehsana 382715, India
| | - Gyanchander Eppa
- Department of Chemistry, School of Science, Indrashil University, Kadi, Mehsana 382715, India
| | - Jhillu Singh Yadav
- Department of Chemistry, School of Science, Indrashil University, Kadi, Mehsana 382715, India
| |
Collapse
|
92
|
Wegesser T, Coppi A, Harper T, Paris M, Minocherhomji S. Nonclinical genotoxicity and carcinogenicity profile of apremilast, an oral selective inhibitor of PDE4. Regul Toxicol Pharmacol 2021; 125:104985. [PMID: 34237378 DOI: 10.1016/j.yrtph.2021.104985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/16/2021] [Accepted: 06/28/2021] [Indexed: 11/26/2022]
Abstract
Apremilast is an oral, selective small molecule inhibitor of phosphodiesterase-4 (PDE4) that has been approved for the treatment of active psoriatic arthritis, moderate to severe plaque psoriasis, and for patients with oral ulcers associated with Behçet's disease. Apremilast modulates the inflammatory cascade in cells by inhibiting PDE4, thus preventing the degradation of cyclic adenosine monophosphate, resulting in the upregulation of interleukin (IL)-10 and the downregulation of proinflammatory cytokines, including IL-23, interferon gamma (IFNγ), and tumor necrosis factor alpha (TNFα). Here, we evaluated the genotoxic and carcinogenic potential of apremilast using Good Laboratory Practice (GLP)-compliant in vitro and in vivo studies. Apremilast was not genotoxic in the genetic toxicology battery, as evaluated for mutagenicity in the Ames test up to concentrations of 5000 μg/plate, clastogenicity in cultured human peripheral blood lymphocytes up to concentrations of 700 ug/mL was in excess of the solubility limit in culture medium and not able to assess; and negative for the induction of micronuclei in the bone marrow micronucleus test in mice up to doses of 2000 mg/kg/day. Furthermore, apremilast did not increase the incidence of tumors in lifetime rat or mouse carcinogenicity studies up to the maximum tolerated dose. In summary, in non-clinical studies, apremilast is not genotoxic and is not carcinogenic.
Collapse
Affiliation(s)
| | - Aldo Coppi
- Amgen Inc., Thousand Oaks, CA, 91320, USA
| | - Tod Harper
- Amgen Inc., Thousand Oaks, CA, 91320, USA
| | | | | |
Collapse
|
93
|
Özdede A, Hatemi G. An evaluation of apremilast for the treatment of adult patients with oral ulcers associated with Behçet's syndrome. Expert Opin Pharmacother 2021; 22:1533-1537. [PMID: 34218739 DOI: 10.1080/14656566.2021.1939307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Behçet's syndrome is a chronic, multi-system, variable vasculitis of unknown etiology that can result in significant morbidity and mortality. Mucocutaneous lesions such as oral ulcers and genital ulcers are common manifestations that can affect the quality of life of patients significantly. Treatment for mucocutaneous lesions in Behçet's syndrome continues to be critical, and an unmet need remains a significant issue. AREAS COVERED This review evaluates the mechanism of action of apremilast, its effect on the number and pain of oral ulcers, other manifestations, such as genital ulcers, disease activity, quality of life and safety profile in Behçet's syndrome patients. Data from clinical trials as well as observational studies were included. EXPERT OPINION Two randomized placebo-controlled trials and real-world observational data suggest that apremilast is an effective and well-tolerated treatment modality for oral and genital ulcers in Behçet's syndrome. Observational studies additionally showed beneficial results for skin lesions, arthritis, and intestinal involvement.
Collapse
Affiliation(s)
- Ayşe Özdede
- Division of Rheumatology, Department of Internal Medicine, Istanbul University -Cerrahpasa, Istanbul, Turkey.,Professor of Medicine, Division of Rheumatology, Department of Internal Medicine and Behçet's Disease Research Center, Istanbul University - Cerrahpasa, Istanbul, Turkey
| | - Gülen Hatemi
- Division of Rheumatology, Department of Internal Medicine, Istanbul University -Cerrahpasa, Istanbul, Turkey.,Professor of Medicine, Division of Rheumatology, Department of Internal Medicine and Behçet's Disease Research Center, Istanbul University - Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
94
|
Ben Abdallah H, Johansen C, Iversen L. Key Signaling Pathways in Psoriasis: Recent Insights from Antipsoriatic Therapeutics. PSORIASIS-TARGETS AND THERAPY 2021; 11:83-97. [PMID: 34235053 PMCID: PMC8254604 DOI: 10.2147/ptt.s294173] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/05/2021] [Indexed: 12/27/2022]
Abstract
Psoriasis is a common chronic inflammatory skin disease associated with several comorbidities and reduced quality of life. In the past decades, highly effective targeted therapies have led to breakthroughs in the management of psoriasis, providing important insights into the pathogenesis. This article reviews the current concepts of the pathophysiological pathways and the recent progress in antipsoriatic therapeutics, highlighting key targets, signaling pathways and clinical effects in psoriasis.
Collapse
Affiliation(s)
| | - Claus Johansen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Lars Iversen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
95
|
Anti-inflammatory effects of a novel phosphodiesterase-4 inhibitor, AA6216, in mouse dermatitis models. Eur J Pharmacol 2021; 906:174258. [PMID: 34139195 DOI: 10.1016/j.ejphar.2021.174258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/02/2021] [Accepted: 06/11/2021] [Indexed: 01/25/2023]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease that is commonly treated with corticosteroids. However, these drugs have long-term adverse effects, representing an unmet need for new treatments. AD is associated with dysregulation of phosphodiesterase 4 (PDE4) activity in inflammatory cells and the topical PDE4 inhibitor, crisaborole, is approved by the US FDA for mild-to-moderate AD. In this study, we compared the effects of a novel PDE4 inhibitor, AA6216, with those of crisaborole on skin inflammation. We found that AA6216 is a more potent inhibitor of PDE4 and of cytokine production (TNF-α, IL-12/23p40, IL-4, IL-13, and IFN-γ) by human peripheral blood mononuclear cells (PBMCs) stimulated by phytohemagglutinin (PHA) or anti-CD3 antibodies, with IC50 values ranging from 5.9 to 47 nM. AA6216 also significantly suppressed skin inflammation in three mouse models of dermatitis. In acute and chronic oxazolone-induced dermatitis models, topical AA6216 exhibited stronger inhibitory effects on ear inflammation and cytokine production (TNFα, IL-1β, and IL-4) in skin lesions compared with crisaborole. In a Dermatophagoides farinae-induced dermatitis model, AA6216 significantly reduced the dermatitis score, based on the development of erythema/hemorrhage, scarring/dryness, edema, and excoriation/erosion, compared with a clinically used topical AD drug, tacrolimus. These results suggest the possibility that AA6216 is a novel and effective topical therapeutic agent for the treatment of dermatitis including AD.
Collapse
|
96
|
Ferguson LD, Cathcart S, Rimmer D, Semple G, Brooksbank K, Paterson C, Brown R, Harvie J, Gao X, Radjenovic A, Welsh P, McInnes IB, Sattar N, Siebert S. Effect of the phosphodiesterase 4 inhibitor apremilast on cardiometabolic outcomes in psoriatic disease-results of the IMAPA study. Rheumatology (Oxford) 2021; 61:1026-1034. [PMID: 34097014 PMCID: PMC8889283 DOI: 10.1093/rheumatology/keab474] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/19/2021] [Indexed: 11/22/2022] Open
Abstract
Objectives Studies have suggested phosphodiesterase 4 (PDE4) inhibition may be associated with weight loss and other cardiometabolic benefits. We evaluated the effect of the PDE4 inhibitor apremilast on body weight and composition, glucose homeostasis, lipid profiles and vascular function in psoriatic disease and whether weight change correlated with therapeutic response. Methods We conducted a prospective, open-label study (Immune Metabolic Associations in Psoriatic Arthritis) of adults receiving apremilast 30 mg as part of routine care for PsA and/or psoriasis. Cardiometabolic, anthropometric and disease activity assessments were performed at baseline (pre-apremilast) and at months 1, 3 and 6 of apremilast treatment in 60 patients. A subgroup underwent further assessment of endothelial function, body composition and adipocyte morphology. Results In patients (median age 54.5 years, 63% women, median BMI 33.2 kg/m2), apremilast was associated with a mean weight loss of 2.2 kg (95% CI 1.4, 3.0; P < 0.001) and a mean BMI decrease of 0.8 kg/m2 (95% CI 0.5, 1.2; P < 0.001) after 6 months of treatment. Body composition analysis demonstrated a reduction in total abdominal fat [mean decrease 0.52 L (95% CI 0.08, 0.96), P = 0.022], principally subcutaneous adipose tissue [mean decrease 0.37 L (95% CI 0.05, 0.68), P = 0.022]. There was no change in adipocyte diameter, haemoglobin A1c, lipid, glucagon-like peptide-1 or vascular function. Psoriatic disease activity improved with apremilast, although this was not correlated with weight change. Conclusion Following apremilast treatment, we observed weight loss, principally abdominal subcutaneous fat, and improvement in psoriatic disease activity. The latter was independent of weight change, suggesting apremilast likely acts through direct immunological mechanisms.
Collapse
Affiliation(s)
- Lyn D Ferguson
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Susanne Cathcart
- Glasgow Clinical Research Facility, Glasgow Royal Infirmary, Glasgow, UK
| | - Dominic Rimmer
- Glasgow Clinical Research Facility, Glasgow Royal Infirmary, Glasgow, UK
| | - Gary Semple
- Glasgow Clinical Research Facility, Glasgow Royal Infirmary, Glasgow, UK
| | - Katriona Brooksbank
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Caron Paterson
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK
| | - Rosemary Brown
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | - Xuan Gao
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Aleksandra Radjenovic
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Paul Welsh
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Iain B McInnes
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Stefan Siebert
- Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
97
|
Keeling S, Maksymowych WP. JAK inhibitors, psoriatic arthritis, and axial spondyloarthritis: a critical review of clinical trials. Expert Rev Clin Immunol 2021; 17:701-715. [PMID: 33944642 DOI: 10.1080/1744666x.2021.1925541] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Psoriatic arthritis (PsA) and spondyloarthritis (SpA) are inflammatory arthritides associated with progressive damage, deformity and morbidity. Janus kinase (JAK) inhibitors block JAKs, cytoplasmic protein tyrosine kinases important in signal transduction and immune processes that are currently being studied as synthetic disease modifying anti-rheumatic drugs (tsDMARDs) in psoriatic arthritis and spondyloarthritis. AREAS COVERED This review evaluates published phase 2 and 3 clinical trial data for JAK kinase inhibitors for psoriatic arthritis and spondyloarthritis. A literature search using PubMed was conducted using the following keywords: 'psoriatic arthritis', 'ankylosing spondylitis', 'axial spondyloarthritis', 'non-radiographic axial spondyloarthritis', 'tofacitinib', 'baricitinib', 'filgotinib' and 'upadacitinib'. Mechanism of action, phase 2 and 3 clinical trial data, including efficacy and safety, are discussed. EXPERT OPINION JAK inhibitors are important orally administered agents conferring different degrees of selectivity toward JAK1, JAK2, and JAK3 which may have implications on efficacy and safety in PsA and SpA. Phase 2 and 3 clinical trials in PsA for tofacitinib and upadacitinib and phase 2 for filgotinib confirmed efficacy comparable to biologic DMARDs. In SpA, phase 2 and 2/3 studies confirmed significant efficacy of tofacitinib, filgotinib and upadacitinib compared to placebo. Safety was comparable to clinical trial, long-term extension, and registry data for rheumatoid arthritis.
Collapse
Affiliation(s)
- Stephanie Keeling
- Division of Rheumatology, University of Alberta, Edmonton, Alberta, Canada
| | | |
Collapse
|
98
|
Yang L, Wu P, Xu J, Xie D, Wang Z, Wang Q, Chen Y, Li CH, Zhang J, Chen H, Quan G. Development of Apremilast Solid Dispersion Using TPGS and PVPVA with Enhanced Solubility and Bioavailability. AAPS PharmSciTech 2021; 22:142. [PMID: 33893566 DOI: 10.1208/s12249-021-02005-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/27/2021] [Indexed: 12/16/2022] Open
Abstract
Apremilast (APST) is an effective inhibitor of phosphodieasterase 4 (PDE4) which is the first oral drug for the treatment of adult patients with active psoriatic arthritis. However, Apremilast's low solubility restricts its dissolution and bioavailability. In this study, APST solid dispersion with D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) and Poly(1-vinylpyrrolidone-co-vinyl acetate) (PVPVA) was developed to improve the dissolution and bioavailability of APST by spray drying. A series of TPGS were synthesized to elucidate the effect of the ratio of monoester to diester on solubilizing capacity. X-ray powder diffraction (XRPD), differential scanning calorimetry (DSC), and Fourier transform infrared spectrophotometry (FT-IR) were used to characterize the solid dispersion, and the results showed that APST was amorphous in solid dispersion. In vitro dissolution study showed that the dissolution rate of solid dispersion in phosphate buffered saline (pH 6.8) was remarkably increased, reaching a release of 90% within 10 min. Moreover, in vivo pharmacokinetics study revealed that the bioavailability of solid dispersion in rats had significant improvement. In particular, its Cmax and AUClast were nearly 22- and 12.9-fold greater as compared to APST form B, respectively. In conclusion, APST solid dispersion with TPGS and PVPVA is an alternative drug delivery system to improve the solubility and oral bioavailability of APST.
Collapse
Affiliation(s)
- Liuhong Yang
- HEC Research and Development Center, HEC Pharm Group, Dongguan, 523871, China
| | - Penghui Wu
- HEC Research and Development Center, HEC Pharm Group, Dongguan, 523871, China
| | - Jinchao Xu
- HEC Research and Development Center, HEC Pharm Group, Dongguan, 523871, China
| | - Dihuan Xie
- HEC Research and Development Center, HEC Pharm Group, Dongguan, 523871, China
| | - Zhongqing Wang
- HEC Research and Development Center, HEC Pharm Group, Dongguan, 523871, China
| | - Qian Wang
- HEC Research and Development Center, HEC Pharm Group, Dongguan, 523871, China
| | - Yong Chen
- HEC Research and Development Center, HEC Pharm Group, Dongguan, 523871, China.
- Hunan Provincial Key Laboratory of Xiangnan Rare-Precious Metals Compounds and Applications, College of Chemical Biology and Environmental Engineering, Xiangnan University, Chenzhou, 423043, China.
| | - Chuan Hua Li
- Hunan Provincial Key Laboratory of Xiangnan Rare-Precious Metals Compounds and Applications, College of Chemical Biology and Environmental Engineering, Xiangnan University, Chenzhou, 423043, China
| | - Jiaxin Zhang
- College of Pharmacy, Yanbian University, Yanji, 133002, China
| | - Hangping Chen
- College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China
| | - Guilan Quan
- College of Pharmacy, Jinan University, 601 Huangpu Avenue West, Guangzhou, 510632, China.
| |
Collapse
|
99
|
Epstein PM, Basole C, Brocke S. The Role of PDE8 in T Cell Recruitment and Function in Inflammation. Front Cell Dev Biol 2021; 9:636778. [PMID: 33937235 PMCID: PMC8085600 DOI: 10.3389/fcell.2021.636778] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/29/2021] [Indexed: 01/07/2023] Open
Abstract
Inhibitors targeting cyclic nucleotide phosphodiesterases (PDEs) expressed in leukocytes have entered clinical practice to treat inflammatory disorders, with three PDE4 inhibitors currently in clinical use as therapeutics for psoriasis, psoriatic arthritis, atopic dermatitis and chronic obstructive pulmonary disease. In contrast, the PDE8 family that is upregulated in pro-inflammatory T cells is a largely unexplored therapeutic target. It was shown that PDE8A plays a major role in controlling T cell and breast cancer cell motility, including adhesion to endothelial cells under physiological shear stress and chemotaxis. This is a unique function of PDE8 not shared by PDE4, another cAMP specific PDE, employed, as noted, as an anti-inflammatory therapeutic. Additionally, a regulatory role was shown for the PDE8A-rapidly accelerated fibrosarcoma (Raf)-1 kinase signaling complex in myelin antigen reactive CD4+ effector T cell adhesion and locomotion by a mechanism differing from that of PDE4. The PDE8A-Raf-1 kinase signaling complex affects T cell motility, at least in part, via regulating the LFA-1 integrin mediated adhesion to ICAM-1. The findings that PDE8A and its isoforms are expressed at higher levels in naive and myelin oligodendrocyte glycoprotein (MOG)35–55 activated effector T (Teff) cells compared to regulatory T (Treg) cells and that PDE8 inhibition specifically affects MOG35–55 activated Teff cell adhesion, indicates that PDE8A could represent a new beneficial target expressed in pathogenic Teff cells in CNS inflammation. The implications of this work for targeting PDE8 in inflammation will be discussed in this review.
Collapse
Affiliation(s)
- Paul M Epstein
- Department of Cell Biology, UConn Health, Farmington, CT, United States
| | - Chaitali Basole
- Department of Immunology, UConn Health, Farmington, CT, United States
| | - Stefan Brocke
- Department of Immunology, UConn Health, Farmington, CT, United States
| |
Collapse
|
100
|
Milakovic M, Gooderham MJ. Phosphodiesterase-4 Inhibition in Psoriasis. PSORIASIS-TARGETS AND THERAPY 2021; 11:21-29. [PMID: 33763335 PMCID: PMC7982714 DOI: 10.2147/ptt.s303634] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/04/2021] [Indexed: 01/13/2023]
Abstract
Psoriasis is a chronic immune-mediated inflammatory disorder. Phosphodiesterase-4 (PDE-4) is an enzyme that mediates inflammatory responses and plays a role in psoriasis pathogenesis. PDE-4 degrades its substrate cyclic adenosine monophosphate (cAMP) to adenosine monophosphate (AMP), which subsequently leads to the production of pro-inflammatory mediators. Inhibitors of PDE-4 work by blocking the degradation of cAMP, which leads to a reduction in inflammation. Apremilast is the only approved oral PDE-4 inhibitor for the treatment of psoriasis. While it is effective for some patients, it may be limited by adverse effects in others. A topical PDE-4 inhibitor, roflumilast, is being investigated in psoriasis and showing promising results. Crisaborole, a topical PDE-4 inhibitor approved for use in atopic dermatitis, has also been investigated in psoriasis. This is an updated comprehensive review to summarize the currently available evidence for the PDE-4 inhibitors apremilast, roflumilast and crisaborole in the treatment of psoriasis, with a focus on data from randomized clinical trials.
Collapse
Affiliation(s)
| | - Melinda J Gooderham
- Skin Centre for Dermatology, Peterborough, ON, K9J 5K2, Canada.,Probity Medical Research, Waterloo, ON, N2J 1C4, Canada.,Department of Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| |
Collapse
|