1
|
Megna M, Potestio L, Ruggiero A, Cacciapuoti S, Maione F, Tasso M, Caso F, Costa L. JAK Inhibitors in Psoriatic Disease. Clin Cosmet Investig Dermatol 2023; 16:3129-3145. [PMID: 37927384 PMCID: PMC10625379 DOI: 10.2147/ccid.s433367] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/13/2023] [Indexed: 11/07/2023]
Abstract
Psoriasis is now considered to be the cutaneous phenotype of a systemic inflammatory condition, recognized under the term Psoriatic Disease (PsD). PsD has several extracutaneous manifestations, such as inflammatory articular and entheseal involvement, leading to psoriatic arthritis (PsA), and the less frequent intestinal and ocular manifestations with colitis/inflammatory bowel disease and uveitis, respectively. There have also been several reports of an increased frequency of comorbidities such as hypertension, diabetes, dyslipidemia, obesity, metabolic syndrome and cardiovascular manifestations during the course of PsD. The link between psoriasis and related comorbidities is considered a long-term disease sequela, often characterized by an unhealthy lifestyle and a consequence of systemic inflammation; hence, psoriasis requires adequate and prompt treatment, with the aim of controlling not only cutaneous manifestations but also extracutaneous manifestations and systemic inflammation. Pharmacological strategies for PsD have significantly increased over recent years. Recently, the targeted synthetic DMARDs, Janus kinase (JAK) inhibitors, tofacitinib and upadacitinib, were added to the therapeutic armamentarium for treating PsA, and deucravacitinib for psoriasis. These oral agents act directly on inflammatory mechanisms underlining the disease, as antagonists of the intracellular JAK signal pathway and, by STAT phosphorylation, inhibit gene proinflammatory cytokine transcription. JAK inhibitors represent a recent additional treatment strategy for PsD management and, among these, tofacitinib and upadacitinib have recently been approved for PsA, and deucravacitinib for psoriasis. In this review we describe ongoing and recent phase II and III randomized controlled trials (RCTs) evaluating the efficacy and safety of investigational JAK inhibitors in psoriasis and PsA.
Collapse
Affiliation(s)
- Matteo Megna
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Luca Potestio
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Angelo Ruggiero
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Sara Cacciapuoti
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Francesco Maione
- Immunopharmalab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Marco Tasso
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Francesco Caso
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Luisa Costa
- Rheumatology Research Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
2
|
Caso F, Costa L, Triggianese P, Maione F, Bertolini N, Vastarella M, Chimenti MS, Tasso M. Recent developments for new investigational JAK inhibitors in psoriatic arthritis. Expert Opin Investig Drugs 2023:1-11. [PMID: 37096862 DOI: 10.1080/13543784.2023.2207737] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
INTRODUCTION Psoriatic arthritis (PsA) is a chronic inflammatory disease affecting about one-third of subjects with psoriasis. Several treatment modalities targeting Janus Kinase pathways and intracellular inflammatory cascade are now available and under clinical investigation to treat this disease. AREAS COVERED This review describes ongoing and recently completed phase 2 and 3 Randomized Clinical Trials (RCTs) evaluating the efficacy and safety of approved JAK (Tofacitinib and Upadacitinib) and investigational JAK inhibitors (JAK1 inhibitors: Filgotinib and Ivarmacitinib (SHR0302); TYK2 inhibitors: Brepocitinib (PF-06700841) Deucravacitinib (BMS-986165), and NDI-034858) in PsA through February 2023. EXPERT OPINION Current standard of care has significantly improved the quality of life in PsA. Recently approved JAK inhibitors for PsA have addressed many of the unmet needs of PsA, particularly of those with severe phenotypes. Preliminary results from several RCTs have reported good and fast efficacy and an acceptable safety profile of investigational JAK inhibitors in PsA. Additional clinical trials and long-term outcome data on these agents are necessary for increasing available therapeutic options for PsA.
Collapse
Affiliation(s)
- Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II
| | - Paola Triggianese
- Rheumatology, allergology and clinical immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Francesco Maione
- ImmunoPharmaLab, Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Nicoletta Bertolini
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II
| | - Maria Vastarella
- Dermatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II
| | - Maria Sole Chimenti
- Rheumatology, allergology and clinical immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Marco Tasso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II
| |
Collapse
|
3
|
Letarouilly JG, Flachaire B, Labadie C, Kyheng M, Cohen N, Sellam J, Richette P, Dieude P, Claudepierre P, Fautrel B, Houvenagel E, Nguyen CD, Guyot MH, Segaud N, Marguerie L, Deprez X, Salmon JH, Baudens G, Miceli-Richard C, Gervais E, Chary-Valckenaere I, Lafforgue P, Philippe P, Loeuille D, Richez C, Tubach F, Pham T, Flipo RM. Secukinumab and ustekinumab treatment in psoriatic arthritis: results of a direct comparison. Rheumatology (Oxford) 2021; 60:2773-2782. [PMID: 33232465 DOI: 10.1093/rheumatology/keaa710] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/19/2020] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVES To evaluate the characteristics of patients (pts) with PsA treated by ustekinumab (UST) or secukinumab (SEK) and to compare real-world persistence of UST and SEK in PsA. METHODS In this retrospective, national, multicentre cohort study, pts with PsA (CASPAR criteria or diagnosis confirmed by the rheumatologist) initiating UST or SEK with a follow-up ≥6 months were included from January 2011 to April 2019. The persistence between SEK and UST was assessed after considering the potential confounding factors by using pre-specified propensity-score methods. Causes of discontinuation and tolerance were also collected. RESULTS A total of 406 pts were included: 245 with UST and 161 with SEK. The persistence rate was lower in the UST group compared with the SEK group [median persistence 9.4 vs 14.7 months; 26.4% vs 38.0% at 2 years; weighted hazard ratio (HR) = 1.42; 95% CI: 1.07, 1.92; P =0.015]. In subgroup analysis, the persistence rate of SEK associated with MTX was significantly higher than that of UST associated with MTX: HR = 2.20; 95% CI: 1.30, 3.51; P =0.001, in contrast to SEK vs UST monotherapy: HR = 1.06; 95% CI: 0.74, 1.53; P =0.75. Discontinuation due to inefficacy was reported in 91.7% (SEK) and 82.4% (UST) of pts. Discontinuation due to an adverse event was reported in 12.2% (SEK) and 7.7% (UST) of pts. CONCLUSION In this first study comparing UST and SEK, the persistence of SEK was higher than that of UST in PsA. In subgroup analysis, this difference was only found in association with MTX.
Collapse
Affiliation(s)
| | - Benoît Flachaire
- Service de Rhumatologie, Univ. Aix-Marseille, APHM, Marseille, France
| | - Céline Labadie
- Service de Rhumatologie, Université Bordeaux, CHU Bordeaux, Bordeaux, France
| | - Maéva Kyheng
- ULR 2694 - METRICS: Évaluation des Technologies de santé et Des Pratiques Médicales, Université Lille, France.,Département des Biostatistiques, CHU de Lille, Lille, France
| | - Nicolas Cohen
- Service de Rhumatologie, Univ. Aix-Marseille, APHM, Marseille, France
| | - Jérémie Sellam
- Service de Rhumatologie, APHP, Hôpital Saint Antoine, Sorbonne Université, CRSA Inserm UMRS_938, Paris, France
| | - Pascal Richette
- Service de Rhumatologie, APHP, Hôpital Lariboisière, Paris, France.,Inserm, UMR-S 1132, Bioscar, Université de Paris, Paris, France
| | - Philippe Dieude
- Service de Rhumatologie, Université de Paris, INSERM UMR1152, Groupe Hospitalier Universitaire Bichat-Claude Bernard, APHP, Paris, France
| | - Pascal Claudepierre
- Ep, iDermE, EA7379, Université Paris Est Creteil, Creteil, France.,Department of Rheumatology, Hopital Henri-Mondor, AP-HP, Creteil, France
| | - Bruno Fautrel
- Service de Rhumatologie, Hôpital Pitié, Sorbonne Université, AP-HP, Paris, France
| | - Eric Houvenagel
- Service de Rhumatologie, Institut Catholique de Lille, Hôpital Saint Philibert, Ghicl, Lomme, France
| | | | | | - Nicolas Segaud
- Service de Médecine Interne, CH Armentières, Armentières, France
| | | | - Xavier Deprez
- Service de Rhumatologie, CH Valenciennes, Valenciennes, France
| | - Jean-Hugues Salmon
- Service de Rhumatologie, Université Reims, Champagne-Ardenne, CHU Reims, Reims, France
| | - Guy Baudens
- Cabinet, Libéral de Rhumatologie, Valenciennes, France
| | | | - Elisabeth Gervais
- Service de Rhumatologie, Université de Poitiers, CHU Poitiers, Poitiers, France
| | | | - Pierre Lafforgue
- Service de Rhumatologie, Univ. Aix-Marseille, APHM, Marseille, France
| | - Peggy Philippe
- Service de Rhumatologie, Université Lille, CHU Lille, Lille, France
| | - Damien Loeuille
- Service de Rhumatologie, Université de Poitiers, CHU Poitiers, Poitiers, France
| | - Christophe Richez
- Service de Rhumatologie, Université Bordeaux, CHU Bordeaux, Bordeaux, France
| | - Florence Tubach
- Département de Santé Publique, Centre de Pharmacoépidémiologie (Cephepi), Institut Pierre Louis d'Epidémiologie et de Santé Publique, Hôpital Pitié Salpêtrière, Sorbonne Université, AP-HP, Paris, France
| | - Thao Pham
- Service de Rhumatologie, Univ. Aix-Marseille, APHM, Marseille, France
| | - René-Marc Flipo
- Service de Rhumatologie, Université Lille, CHU Lille, Lille, France
| |
Collapse
|
4
|
Navarini L, Currado D, Costa L, Tasso M, Chimenti MS, Caso F. Experimental and Investigational Pharmacotherapy for Psoriatic Arthritis: Drugs of the Future. J Exp Pharmacol 2020; 12:487-502. [PMID: 33235521 PMCID: PMC7679354 DOI: 10.2147/jep.s265633] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/24/2020] [Indexed: 12/28/2022] Open
Abstract
In recent years, different studies have shown in psoriatic arthritis (PsA), the pathogenetic role of multiple cytokines other than tumor necrosis factor-α, such as interleukin-17 (IL-17), and IL-23 and dysfunction of Janus kinase (JAK)-signal family pathway. These molecules also represent the target of recently developed biologic (bDMARDs) and targeted synthetic disease modifying antirheumatic drugs (DMARDs) (tsDMARDs) currently investigated in several Phase II and III randomized controlled trials (RCTs). This review examines the therapeutic efficacy and safety of most recent developed IL-17, IL-23 and JAK inhibitors and highlights how these new PsA therapies are going to revolutionize the management of PsA in the next few years. Ongoing RCTs of these molecules in PsA are also described. Available literature on new anti-IL-17 and anti-IL-23 agents and JAK inhibitors demonstrates the potential role of these molecules as effective therapeutic strategies across multiple PsA clinical domains, along with an acceptable tolerability and safety profile, thus expanding the treatment options available for PsA patients. Of note, other molecules are under investigation, and among those, potential therapeutic strategies seem to be represented by single antibodies blocking simultaneously two cytokines, the agents inhibiting mammalian target of rapamycin (mTOR), receptor retinoic acid receptor-related orphan receptor gamma (RORγt), A3 adenosine receptor (A3 AR), and K+ channel voltage channel inhibitors. Remarkable progress has been made in PsA pharmacotherapy, and novel bDMARDs targeting IL17A and tsDMARDs (JAK-inhibitors) represent promising therapies. More clinical trials are needed to better characterize the efficacy and safety profile of these therapeutic agents in PsA treatment.
Collapse
Affiliation(s)
- Luca Navarini
- Unit of Rheumatology, Immunology and Clinical Medicine, Università Campus Bio-Medico Di Roma, Rome, Italy
| | - Damiano Currado
- Unit of Rheumatology, Immunology and Clinical Medicine, Università Campus Bio-Medico Di Roma, Rome, Italy
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Marco Tasso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| |
Collapse
|
5
|
Chimenti MS, D’Antonio A, Conigliaro P, Ferrigno S, Vendola A, Ferraioli M, Triggianese P, Costa L, Caso F, Perricone R. An Update for the Clinician on Biologics for the Treatment of Psoriatic Arthritis. Biologics 2020; 14:53-75. [PMID: 32903867 PMCID: PMC7445514 DOI: 10.2147/btt.s260754] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/05/2020] [Indexed: 12/16/2022]
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory arthropathy typically associated with psoriasis (PsO). The pathogenesis is strictly related to the association among the presence of genetic risk alleles and innate and acquired immune response with dramatic consequences on bone remodeling. Clinically, PsA patients may present heterogenicity of articular and periarticular manifestations that may be associated with the presence of comorbidities making treatment decision challenging in patients management. The identification of patient-targeted therapies is still a critical issue. Actually, several biological and synthetic drugs are promising in terms of efficacy and safety profile. National and international treatment recommendations support clinicians in the decision of the best treatment, although they may have limits basically related to updates and different outcomes included in the clinical studies evaluated. The aim of this narrative review is therefore to give guidance for clinicians for PsA patients treatment. For this purpose, we evaluated evidence on biological therapies efficacy used for PsA treatment. Specifically, we reviewed data on biological therapies, Janus kinases (JAK) inhibitors, and drugs with a new mechanism of action that are part of the treatment pipeline. The concept of "switching" and "swapping" is also described, as well as data concerning special populations such as pregnant women and elderly patients.
Collapse
Affiliation(s)
- Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Arianna D’Antonio
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Conigliaro
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Sara Ferrigno
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Andrea Vendola
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Mario Ferraioli
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Triggianese
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University Federico II, Naples, Italy
| | - Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine and Surgery, University Federico II, Naples, Italy
| | - Roberto Perricone
- Rheumatology, Allergology and Clinical Immunology, Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
6
|
Chimenti MS, Perricone C, Conigliaro P, Triggianese P, D'Antonio A, de Martino E, Fonti GL, Caso F, Costa L, Perricone R. Tackling the autoimmune side in Spondyloarthritis: A systematic review. Autoimmun Rev 2020; 19:102648. [PMID: 32801035 DOI: 10.1016/j.autrev.2020.102648] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 02/24/2020] [Indexed: 02/07/2023]
Abstract
Spondyloarthritis (SpA) are a heterogeneous group of inflammatory chronic diseases characterized by sharing common pathogenic, clinical and radiologic features. The aim of this review is to support clinicians in understanding and managing this complex disease, from pathogenesis to therapeutic targets, through a systematic review of the current literature in accordance with PRISMA guidelines and checklist. HLA-B27 has been found to be associated with axial involvement either in SA and in PsA patients: it might be involved through presentation of an "arthritogenic peptide" to autoreactive CD8+ T cells or might accumulate in misfolded form and induce production pro-inflammatory cytokines by binding to several innate immune receptors. This genetic background in combination with mechanical stress leads to the activation of both innate and acquired immune responses as well as a possible role of autoimmunity in SpA pathogenesis. The release of IL-23 and IL-17 is relevant for their systemic and local effect on bone, inducing the activation of osteoclasts. Thus, the regulatory role of IL-17 on fibroblasts, osteoblasts and chondrocytes has an impact in both synovial inflammation and joint destruction. Innovative therapies targeting IL-12/23 and IL-17 and the use of small targeted synthetic molecules, as JAK-inhibitors, proved to be effective in SpA patients representing an alternative strategy to TNF-inhibitors.
Collapse
Affiliation(s)
- Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Carlo Perricone
- Rheumatology, Department of Medicine, University of Perugia, Piazzale Giorgio Menghini, 1, 06129 Perugia, Italy.
| | - Paola Conigliaro
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Triggianese
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Arianna D'Antonio
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Erica de Martino
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Giulia Lavinia Fonti
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131 Naples, Italy
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131 Naples, Italy
| | - Roberto Perricone
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
7
|
Capri S, Migliore A, Loconsole F, Barbieri M. A cost-effectiveness and budget impact analysis of apremilast in patients with psoriatic arthritis in Italy. J Med Econ 2020; 23:353-361. [PMID: 31856609 DOI: 10.1080/13696998.2019.1707208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Aims: The aim of this study was to conduct a cost-effectiveness analysis, as well as a budget impact analysis, on the use of apremilast for the treatment of adult patients with psoriatic arthritis (PsA), within the Italian National Health Service (NHS).Methods: A Markov state transition cohort model, which was adapted to the Italian context, was used to compare the costs of the currently available treatments and of the patients' quality of life with two alternative treatment sequences, with or without apremilast as pre-biologic therapy. Moreover, a budget impact model was developed based on the population of patients treated for PsA in Italy, who can be eligible for treatment with apremilast. The eligible population was represented by adult patients with PsA who had an inadequate response to or were intolerant to previous disease-modifying antirheumatic drugs (DMARDs), for the approved indication, and for the treatment studied in the economic analytic model.Results: This cost-effectiveness analysis estimated that the strategy of using apremilast before biologic therapy is cost-effective, with an incremental cost-effectiveness ratio of €32,263.00 per QALY gained which is slightly over the normal threshold found in other Italian economic studies, which usually considers a 40-year-period. Conversely, the budget impact analysis was conducted over 3 years, and it led to an estimated annual saving of €1.6 million, €4.6 million and €5.5 million in the first, second and third year of apremilast commercialization, respectively, for a total saving of €11.75 million in 3 years.Limitations: Limitations of this analysis include the absence of head-to-head trials comparing therapies included in the economic model, the lack of comparative long-term data on treatment efficacy, and the assumption of complete independence between the considered response rates to therapy.Conclusion: The use of apremilast as a first option before the use of biologic agents may represent a cost-effective treatment strategy for patients with PsA who fail to respond to, or are intolerant to, previous DMARD therapy. In addition, based on a budget impact perspective, the use of apremilast may lead to cost savings to the Italian healthcare system.
Collapse
Affiliation(s)
- Stefano Capri
- School of Economics and Management, Università Cattaneo-LIUC, Castellanza, Italy
| | - A Migliore
- Rheumatology Unit, S. Pietro Fatebenefratelli Hospital, Rome, Italy
| | - F Loconsole
- Department of Dermatology, University of Bari, Bari, Italy
| | - Marco Barbieri
- Centre for Health Economics, University of York, York, UK
| |
Collapse
|
8
|
Caso F, Chimenti MS, Navarini L, Ruscitti P, Peluso R, Girolimetto N, Del Puente A, Giacomelli R, Scarpa R, Costa L. Metabolic Syndrome and psoriatic arthritis: considerations for the clinician. Expert Rev Clin Immunol 2020; 16:409-420. [PMID: 32149545 DOI: 10.1080/1744666x.2020.1740593] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Metabolic Syndrome (MetS) is strictly interconnected with systemic inflammation, and increased evidence has described a close link between this condition and Psoriatic Arthritis (PsA).Areas covered: This review summarizes main studies exploring clinical aspects and prevalence of MetS in PsA cohorts. Further, there is accumulating evidence showing shared inflammatory pathways between MetS, its components, and PsA.Expert opinion: The high prevalence of MetS in PsA highlights the need for screening, evaluation, and close monitoring of MetS and its components (namely, diabetes mellitus, obesity, hypertension, and dyslipidemia) in psoriatic patients.Further studies should focus on the pathogenetic link between MetS and PsA. More studies are required to identify appropriate algorithms for the assessment and management of MetS in PsA patients.
Collapse
Affiliation(s)
- Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Luca Navarini
- Unit of Allergology, Clinical Immunology and Rheumatology, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Rosario Peluso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Nicolò Girolimetto
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy.,Department of Rheumatology, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Antonio Del Puente
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Roberto Giacomelli
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Raffaele Scarpa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| |
Collapse
|
9
|
Balogh EA, Bashyam AM, Ghamrawi RI, Feldman SR. Emerging systemic drugs in the treatment of plaque psoriasis. Expert Opin Emerg Drugs 2020; 25:89-100. [PMID: 32192366 DOI: 10.1080/14728214.2020.1745773] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Psoriasis is a common, chronic inflammatory skin condition that affects 2-3% of the US population and represents a large psychosocial burden for patients. Over the last decade, highly effective targeted therapies for psoriasis have been developed - namely, those targeting interleukin (IL)-17 and IL-23. The success of biologic agents targeting IL-17 and IL-23 underscores the importance of the IL-23/T helper (Th)17 cell axis in psoriasis pathogenesis. Oral small molecule drugs - such as Janus kinase (JAK) inhibitors, tyrosine kinase 2 (TYK2) inhibitors, and fumaric acid esters (FAEs) - are also being investigated for the treatment of psoriasis. AREAS COVERED This article reviews systemic biologic and oral small molecule drugs currently undergoing clinical trials for the treatment of plaque psoriasis. EXPERT OPINION Many patients with psoriasis have mild disease, and many with mild disease do not seek medical care for their condition. Many patients with mild disease could be adequately treated with topical treatments and phototherapy; however, adherence and feasibility have often been an issue with these treatment types. There seems to be limited room for development of novel biologics, as the existing ones are extraordinarily safe, effective, and convenient with few injections. Patients would prefer a safe, effective oral treatment; however, JAK inhibitors seem unlikely to fill this role completely.
Collapse
Affiliation(s)
- Esther A Balogh
- Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine , Winston-Salem, NC, USA
| | - Arjun M Bashyam
- Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine , Winston-Salem, NC, USA
| | - Rima I Ghamrawi
- Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine , Winston-Salem, NC, USA
| | - Steven R Feldman
- Center for Dermatology Research, Department of Dermatology, Wake Forest School of Medicine , Winston-Salem, NC, USA.,Department of Pathology, Wake Forest School of Medicine , Winston-Salem, NC, USA.,Department of Social Sciences & Health Policy, Wake Forest School of Medicine , Winston-Salem, NC, USA.,Department of Dermatology, University of Southern Denmark , Odense, Denmark
| |
Collapse
|
10
|
Navarini L, Costa L, Tasso M, Chimenti MS, Currado D, Fonti GL, Ciccozzi M, Margiotta DPE, Benigno C, De Martino E, Perricone R, Afeltra A, Scarpa R, Caso F. Retention rates and identification of factors associated with anti-TNFα, anti-IL17, and anti-IL12/23R agents discontinuation in psoriatic arthritis patients: results from a real-world clinical setting. Clin Rheumatol 2020; 39:2663-2670. [PMID: 32189149 DOI: 10.1007/s10067-020-05027-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 02/12/2020] [Accepted: 03/05/2020] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Biologic disease-modifying antirheumatic drugs (bDMARDs) play a pivotal role in the treatment of psoriatic arthritis (PsA). Despite this, their discontinuation due to inefficacy or adverse events is often observed. The aims of this study are to describe retention rates and treatment trends of anti-TNFα, anti-IL17, and anti-IL12/23R agents in patients with PsA and to identify factors associated with bDMARDs discontinuation in a real-world clinical setting. METHODS A retrospective cohort study based on the analysis of the three Italian prescription cohorts of patients with PsA has been performed. Survival analysis was performed using Kaplan-Meier curves and Cox proportional-hazards model. RESULTS During the follow up, which lasted 25.5 (12-60) months, 68 patients discontinued a bDMARD: 13 for primary failure, 12 for secondary failure, 15 for adverse events, 5 for remission, 12 because of lost at follow-up, and 11 for other causes. Cox proportional-hazards demonstrated that a shorter disease duration (HR 0.994991, 95% CI 0.9910336-0.9989647, p = 0.014) and first-line bDMARD (HR 0.5090986, 95% CI 0.3073519-0.8432722, p = 0.009) have a protective role on bDMARD retention rate, while the multivariable analysis failed in demonstrating an independent protective role of male sex on drug retention rate (p = 0.083). No significant differences in retention rate have been found regarding biologic drugs, combination therapy or monotherapy, and class of bDMARD (anti-TNFα or anti-pIL12/23R and anti-IL-17). CONCLUSIONS This study shows that a shorter disease duration and treatment with a first-line bDMARD are predictors of bDMARDs retention rate, further highlighting the importance of early diagnosis of PsA. Key Points • No significant difference in retention among patients treated with anti-IL17A, anti-IL12/23R, and anti-TNFα agents has been demonstrated. • A shorter disease duration and first-line bDMARD treatment are associated with persistence in biologic treatment.
Collapse
Affiliation(s)
- Luca Navarini
- Unit of Allergology, Clinical Immunology and Rheumatology, Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy.
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Marco Tasso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Damiano Currado
- Unit of Allergology, Clinical Immunology and Rheumatology, Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy
| | - Giulia Lavinia Fonti
- Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Massimo Ciccozzi
- Unit of Clinical Laboratory Science, Department of Medicine, Campus Bio-Medico of Rome University, Rome, Italy
| | - Domenico Paolo Emanuele Margiotta
- Unit of Allergology, Clinical Immunology and Rheumatology, Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy
| | - Carolina Benigno
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Erica De Martino
- Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Roberto Perricone
- Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, Rome, Italy
| | - Antonella Afeltra
- Unit of Allergology, Clinical Immunology and Rheumatology, Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy
| | - Raffaele Scarpa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, School of Medicine, University of Naples Federico II, Naples, Italy
| |
Collapse
|
11
|
Caso F, Navarini L, Ruscitti P, Chimenti MS, Girolimetto N, Del Puente A, Giacomelli R, Scarpa R, Costa L. Targeted synthetic pharmacotherapy for psoriatic arthritis: state of the art. Expert Opin Pharmacother 2020; 21:785-796. [PMID: 32057269 DOI: 10.1080/14656566.2020.1726317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION In recent years, different studies regarding psoriatic arthritis (PsA) have shown the pathogenetic role of dysfunction of signaling pathways involving the phosphodiesterase-4 enzyme and transcription factors or enzymes belonging to the kinase (JAK)-signal family pathway. These also represent the target of several drugs known as targeted synthetic disease-modifying antirheumatic drugs (tsDMARDs). AREAS COVERED The authors performed a systematic literature search using the PubMed database, as well as through retrieving data from randomized controlled trials, their post-hoc analysis, and pooled data analysis on the efficacy and safety profile of the PDE4 inhibitor (PDE4i), apremilast, and the inhibitors of JAK (JAKis), tofacitinib, filgotinib, baricitinib, and upadacitinib, in PsA. EXPERT OPINION In PsA, the PDE4i, apremilast, and the JAKi, tofacitinib, are effective across multiple clinical domains and have an acceptable tolerability profile, thus expanding the treatment options available for PsA patients. Apremilast and tofacitinib show several advantages mainly represented by their oral administration, a fast onset of action, and a short half-life. Data on tsDMARDs in PsA are still limited, and randomized trials and real-life studies are advocated.
Collapse
Affiliation(s)
- Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II , Naples, Italy
| | - Luca Navarini
- Unit of Allergology, Clinical Immunology and Rheumatology, Università Campus Bio-Medico Di Roma , Rome, Italy
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila , L'Aquila, Italy
| | - Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of System Medicine, University of Rome Tor Vergata , Rome, Italy
| | - Nicolò Girolimetto
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II , Naples, Italy.,Department of Rheumatology, Azienda USL-IRCCS Di Reggio Emilia , Reggio Emilia, Italy
| | - Antonio Del Puente
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II , Naples, Italy
| | - Roberto Giacomelli
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila , L'Aquila, Italy
| | - Raffaele Scarpa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II , Naples, Italy
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II , Naples, Italy
| |
Collapse
|
12
|
Peluso R, Caso F, Tasso M, Sabbatino V, Lupoli R, Dario Di Minno MN, Ursini F, Costa L, Scarpa R. Biomarkers of subclinical atherosclerosis in patients with psoriatic arthritis. Open Access Rheumatol 2019; 11:143-156. [PMID: 31388317 PMCID: PMC6607207 DOI: 10.2147/oarrr.s206931] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/15/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Psoriatic arthritis (PsA) is a chronic immune-mediated disease. It is associated with an increase in cardiovascular risk factors (obesity, hypertension, diabetes, and dyslipidemia), giving a higher risk of major adverse cardiovascular events. Patients with PsA have an increased incidence of subclinical atherosclerosis and endothelial dysfunction. The aim of this study is to perform a review of the biomarkers of subclinical atherosclerosis in patients with PsA. Methods: A search was performed in the electronic databases (PubMed, Web of Science, Scopus, and Embase) up until July 2017. Studies were considered if they included data on biomarkers of subclinical atherosclerosis in PsA, and each article was then reviewed for quality and clinical relevance. After completing the literature search, all screened literature was summarized and discussed in our study group (CaRRDs study group). Results: The initial search produced 532 abstracts, which were limited to 258 potentially relevant articles by preliminary review of the titles and by excluding review articles and case reports (n=274). A further 102 articles were deemed ineligible after examining the abstracts. Full texts of the remaining 156 articles were retrieved. Most articles were excluded because they were not relevant to the biomarkers of subclinical atherosclerosis in psoriasis and/or PsA. In the end, 54 articles were deemed eligible for this review. Conclusion: Patients with PsA showed more severe atherosclerotic disease compared with patients with only psoriasis. This may have been due to the higher systemic inflammatory burden from the combination of both diseases. In patients with PsA some molecules may be considered as markers of atherosclerotic disease, and their detection may be a prognostic marker, in addition to imaging procedures, for the development of atherosclerotic disease, and could be suitable for the management of patients with PsA.
Collapse
Affiliation(s)
- Rosario Peluso
- Department of Clinical Medicine and Surgery, Rheumatology Research Unit, Federico II University, Naples, Italy
| | - Francesco Caso
- Department of Clinical Medicine and Surgery, Rheumatology Research Unit, Federico II University, Naples, Italy
| | - Marco Tasso
- Department of Clinical Medicine and Surgery, Rheumatology Research Unit, Federico II University, Naples, Italy
| | - Vincenzo Sabbatino
- Department of Clinical Medicine and Surgery, Rheumatology Research Unit, Federico II University, Naples, Italy
| | - Roberta Lupoli
- Department of Clinical Medicine and Surgery, Division of Internal Medicine, Federico II University, Naples, Italy
| | | | - Francesco Ursini
- Internal Medicine Unit, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Luisa Costa
- Department of Clinical Medicine and Surgery, Rheumatology Research Unit, Federico II University, Naples, Italy
| | - Raffaele Scarpa
- Department of Clinical Medicine and Surgery, Rheumatology Research Unit, Federico II University, Naples, Italy
| |
Collapse
|
13
|
|
14
|
Chimenti MS, Caso F, Alivernini S, De Martino E, Costa L, Tolusso B, Triggianese P, Conigliaro P, Gremese E, Scarpa R, Perricone R. Amplifying the concept of psoriatic arthritis: The role of autoimmunity in systemic psoriatic disease. Autoimmun Rev 2019; 18:565-575. [DOI: 10.1016/j.autrev.2018.11.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 11/30/2018] [Indexed: 02/08/2023]
|
15
|
Vandendorpe AS, de Vlam K, Lories R. Evolution of psoriatic arthritis study patient population characteristics in the era of biological treatments. RMD Open 2019; 5:e000779. [PMID: 30740243 PMCID: PMC6347028 DOI: 10.1136/rmdopen-2018-000779] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/29/2018] [Accepted: 12/17/2018] [Indexed: 12/12/2022] Open
Abstract
Objectives Psoriatic arthritis is a chronic inflammatory disease that affects the musculoskeletal system. It can include arthritis, spondylitis, dactylitis and enthesitis, and is strongly associated with the presence of psoriasis. The introduction of biological therapies as a treatment option has brought a significant improvement in disease control for patients with psoriatic arthritis. Here, we aimed to detect emerging differences in demographic and clinical characteristics of the psoriatic arthritis patient study population since the introduction of biologicals. We hypothesised that evolving views on control of disease activity and increased experience in the management of psoriatic arthritis have affected the patient population considered for clinical trials and that this may serve as a proxy for changes in clinical practice. Methods We systematically searched for and selected 12 phase II and phase III trials and divided them into three treatment periods based on different time periods and working mechanisms of the particular biologicals. We made a selection of patient and disease parameters for which data were available in all three periods, calculated those data per period and looked for statistically significant differences between the treatment periods. Results Statistical analysis showed significant differences in patient characteristics, disease characteristics, disease activity, disease effects and use of prior treatments between the patient populations of the three periods. Conclusion This study shows a clear evolution of the patient population considered for clinical trials since the introduction of biologicals. Further research is needed to see if those changes can be detected in the daily clinical practice.
Collapse
Affiliation(s)
- Ann-Sophie Vandendorpe
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Kurt de Vlam
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Rik Lories
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium.,Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Caso F, Costa L, Nucera V, Barilaro G, Masala IF, Talotta R, Caso P, Scarpa R, Sarzi-Puttini P, Atzeni F. From autoinflammation to autoimmunity: old and recent findings. Clin Rheumatol 2018; 37:2305-2321. [PMID: 30014358 DOI: 10.1007/s10067-018-4209-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 12/13/2022]
Abstract
Autoimmune diseases and autoinflammatory diseases have a number of similar etiopathogenetic and clinical characteristics, including genetic predisposition and recurrent systemic inflammatory flares. The first phase of ADs involves innate immunity: by means of TLRs, autoantigen presentation, B and T cell recruitment and autoantibody synthesis. The second phase involves adaptive immunity, a self-sustaining process in which immune complexes containing nucleic acids and autoantibodies activate self-directed inflammation. The link between autoimmunity and autoinflammation is IL-1ß, which is crucial in connecting the innate immune response due to NLR activation and the adaptive immune responses of T and B cells. In conclusion, although ADs are still considered adaptive immunity-mediated disorders, there is increasing evidence that innate immunity and inflammasomes are also involved. The aim of this review is to highlight the link between the innate and adaptive immune mechanisms involved in autoimmune diseases.
Collapse
Affiliation(s)
- Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, Via Sergio Pansini, 5, Naples, Italy
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, Via Sergio Pansini, 5, Naples, Italy
| | - Valeria Nucera
- Rheumatology Unit, University of Messina, Messina, Italy
| | - Giuseppe Barilaro
- Department of Internal Medicine, IRCCS San Raffaele Pisana, Rome, Italy
| | | | - Rossella Talotta
- Rheumatology Unit, ASST Fatebenefratelli Sacco Buzzi, Milan, Italy
| | - Paolo Caso
- Geriatric Unit, Faculty of Medicine and Psychology, S. Andrea Hospital, "Sapienza" University of Rome, Rome, Italy
| | - Raffaele Scarpa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, Via Sergio Pansini, 5, Naples, Italy.
| | | | - Fabiola Atzeni
- Rheumatology Unit, University of Messina, Messina, Italy
| |
Collapse
|
17
|
Thibodaux RJ, Triche MW, Espinoza LR. Ustekinumab for the treatment of psoriasis and psoriatic arthritis: a drug evaluation and literature review. Expert Opin Biol Ther 2018; 18:821-827. [PMID: 29949399 DOI: 10.1080/14712598.2018.1492545] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Psoriasis (PsO) is an inflammatory disorder characterized by proliferation of keratinocytes, and it may be associated with a systemic inflammatory articular disorder, psoriatic arthritis (PsA). The presentations of PsO and PsA are heterogeneous, and our understanding of pathogenesis has led to a better understanding of the role of the interleukin (IL)-23/T-helper 17 (Th17) axis. Areas covered: Ustekinumab is a monoclonal antibody against IL-12 and IL-23. The pathogenesis of PsO and PsA is a multifactorial process involving genetic, environmental, and lifestyle factors. IL-23 signaling and activation of Th17 cells leads to a self-perpetuating inflammatory loop resulting in continuous keratinocyte proliferation and synovitis. Treatment options are varied, ranging from topical therapy to injection of targeted biologic disease-modifying antirheumatic drugs (bDMARDs). Evidence on the use of ustekinumab in the management of PsO is strong, but it is not as impressive in management of PsA. Expert opinion: IL-12/23 inhibition appears to be a good first-line option for plaque PsO, but efficacy in PsA does not compare favorably to IL-17 inhibition. In general, poorer responses to therapy with any bDMARD in PsA cohorts highlight psoriatic disease heterogeneity. Until new knowledge can remedy the failure of monotherapy, synergistic methods may have to be explored, including combination biologic therapy.
Collapse
Affiliation(s)
- Ross J Thibodaux
- a Section of Rheumatology, Department of Internal Medicine , LSU Health Sciences Center at New Orleans , New Orleans , LA , USA
| | - Mallory W Triche
- b Clinical Pharmacy Department , Thibodaux Regional Medical Center , Thibodaux , LA , USA
| | - Luis R Espinoza
- a Section of Rheumatology, Department of Internal Medicine , LSU Health Sciences Center at New Orleans , New Orleans , LA , USA
| |
Collapse
|
18
|
Costa L, Perricone C, Chimenti MS, Del Puente A, Caso P, Peluso R, Bottiglieri P, Scarpa R, Caso F. Switching Between Biological Treatments in Psoriatic Arthritis: A Review of the Evidence. Drugs R D 2018; 17:509-522. [PMID: 29058302 PMCID: PMC5694428 DOI: 10.1007/s40268-017-0215-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory arthropathy. Therapy with anti-tumor necrosis factor (TNF)-α agents represents the first therapeutic choice for moderate and severe forms; however, PsA patients can experience anti-TNFα failure, lack of efficacy, or adverse events. Several evidences exist on the effectiveness of switching among different TNFα inhibitors, and we reviewed the published data on the effectiveness of anti-TNFα first-, second- and third-line. Most of the studies report that the main reason for switching to a second anti-TNFα agent is represented by lack of efficacy (primary or secondary) and, more rarely, adverse events. Switchers receiving their second anti-TNFα agent have considerably poorer responses compared with non-switchers. Survival of anti-TNFα treatment appears to be superior in PsA patients when compared with rheumatoid arthritis patients. Switching from anti-TNF agents to ustekinumab or secukinumab or apremilast can represent a valid alternative therapeutic strategy.
Collapse
Affiliation(s)
- Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131, Naples, Italy
| | - Carlo Perricone
- Rheumatology, Department of Internal Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of "Medicina dei Sistemi", University of Rome "Tor Vergata", Rome, Italy
| | - Antonio Del Puente
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131, Naples, Italy
| | - Paolo Caso
- Geriatric Unit, Faculty of Medicine and Psychology, S. Andrea Hospital, "Sapienza" University of Rome, Rome, Italy
| | - Rosario Peluso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131, Naples, Italy
| | - Paolo Bottiglieri
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131, Naples, Italy
| | - Raffaele Scarpa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131, Naples, Italy.
| | - Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131, Naples, Italy
| |
Collapse
|
19
|
Psoriatic disease treatment nowadays: unmet needs among the “jungle of biologic drugs and small molecules”. Clin Rheumatol 2018; 37:1739-1741. [DOI: 10.1007/s10067-018-4090-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 03/24/2018] [Accepted: 03/28/2018] [Indexed: 12/17/2022]
|
20
|
Livshits G, Kalinkovich A. Hierarchical, imbalanced pro-inflammatory cytokine networks govern the pathogenesis of chronic arthropathies. Osteoarthritis Cartilage 2018; 26:7-17. [PMID: 29074297 DOI: 10.1016/j.joca.2017.10.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/15/2017] [Accepted: 10/10/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Chronic inflammatory arthropathies, such as rheumatoid arthritis (RA), spondyloarthritis, including psoriatic arthritis (PsA), ankylosing spondyloarthritis (AS), osteoarthritis (OA), and intervertebral disc degenerative disease (DDD) constitute major public health problems that are anticipated to grow significantly as the human population ages. However, many aspects concerning the molecular mechanisms underlying their onset and progression remain unclear. DESIGN This narrative review critically analyzes the molecular mechanisms underlying the inflammation-associated pathogenesis of the aforementioned joint diseases. This includes, in particular, the major role played by several key soluble factors (such as cytokines and the associated signaling pathways, designated as "fragile nodes") produced by local cells and recruited to the joints' immune cells, whose elimination by specific drugs has dramatically improved the diseases' symptomatology and outcome in human clinical trials or in rodent arthritis models. HYPOTHESIS AND THE AIM OF THIS REVIEW We hypothesize that the pathogenesis of chronic inflammatory arthropathies is governed by hierarchical, imbalanced pro-inflammatory cytokine networks (HIPICNs) (comprising a combination of fragile nodes) that are created during the development of both autoimmune (RA, PsA, and AS) and non-autoimmune (OA and DDD) disorders. The main aim of this review is to provide evidence that despite substantial pathobiological differences between these arthropathies, the HIPICNs created are quite common, thus justifying the merging of these disorders mechanistically and suggesting that these common mechanisms exist in the onset and progression of different joint diseases.
Collapse
Affiliation(s)
- G Livshits
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, Tel Aviv 69978, Israel.
| | - A Kalinkovich
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| |
Collapse
|
21
|
Wang Y, Mao Y, Zhang J, Shi G, Cheng L, Lin Y, Li Y, Zhang X, Zhang Y, Chen X, Deng J, Su X, Dai L, Yang Y, Zhang S, Yu D, Wei Y, Deng H. IL-35 recombinant protein reverses inflammatory bowel disease and psoriasis through regulation of inflammatory cytokines and immune cells. J Cell Mol Med 2017; 22:1014-1025. [PMID: 29193791 PMCID: PMC5783847 DOI: 10.1111/jcmm.13428] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 09/22/2017] [Indexed: 02/05/2023] Open
Abstract
Interleukin‐35 (IL‐35), a member of the IL‐12 family, functions as a new anti‐inflammatory factor involved in arthritis, psoriasis, inflammatory bowel disease (IBD) and other immune diseases. Although IL‐35 can significantly prevent the development of inflammation in many diseases, there have been no early studies accounting for the role of IL‐35 recombinant protein in IBD and psoriasis. In this study, we assessed the therapeutic potential of IL‐35 recombinant protein in three well‐known mouse models: the dextransulfate sodium (DSS)‐induced colitis mouse model, the keratin14 (K14)‐vascular endothelial growth factor A (VEGF‐A)‐transgenic (Tg) psoriasis mouse model and the imiquimod (IMQ)‐induced psoriasis mouse model. Our results indicated that IL‐35 recombinant protein can slow down the pathologic process in DSS‐induced acute colitis mouse model by decreasing the infiltrations of macrophages, CD4+T and CD8+T cells and by promoting the infiltration of Treg cells. Further analysis demonstrated that IL‐35 recombinant protein may regulate inflammation through promoting the secretion of IL‐10 and inhibiting the expression of pro‐inflammatory cytokines such as IL‐6, TNF‐α and IL‐17 in acute colitis model. In addition, lower dose of IL‐35 recombinant protein could achieve long‐term treatment effects as TNF‐α monoclonal antibody did in the psoriasis mouse. In summary, the remarkable therapeutic effects of IL‐35 recombinant protein in acute colitis and psoriasis mouse models indicated that IL‐35 recombinant protein had a variety of anti‐inflammatory effects and was expected to become an effective candidate drug for the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Yuan Wang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Mao
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Junfeng Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Gang Shi
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Cheng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Lin
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yiming Li
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaomei Zhang
- Laboratory Animal Center, Sichuan University, Chengdu, China
| | - Yujing Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaolei Chen
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Deng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaolan Su
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shuang Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Dechao Yu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Costa L, Del Puente A, Peluso R, Tasso M, Caso P, Chimenti MS, Sabbatino V, Girolimetto N, Benigno C, Bertolini N, Del Puente A, Perricone R, Scarpa R, Caso F. Small molecule therapy for managing moderate to severe psoriatic arthritis. Expert Opin Pharmacother 2017; 18:1557-1567. [PMID: 28891341 DOI: 10.1080/14656566.2017.1378343] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION The majority of psoriatic arthritis (PsA) patients experience a good clinical response to conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) and biologic therapies (bDMARDs). However, treatment failure with these drugs can represent a relevant clinical problem. Moreover, in daily clinical practice, the appropriate identification of patients eligible for these agents can be conditioned by numerous aspects, mainly represented by comorbidities, such as history of malignancies, chronic and recurrent infectious diseases. Areas covered: We searched in the PUBMED database and review published data on the efficacy and safety profile of the small molecules, inhibitor of phosphodiesterase 4, apremilast, and of JAK/STAT pathways, tofacitinib, in PsA. Moreover, we report data on the other JAK inhibitor, baricitinib, and the A(3) adenosine receptors agonist, CF101, emerging by studies conducted in psoriasis patients. Expert opinion: In Psoriatic Arthritis, apremilast appears promising for PsA and recent studies have shown a good efficacy and an acceptable safety profile. Data on tofacitinib in PsA are limited. Studies on the small molecules, baricitinib and CF101 are still incomplete and limited to trials conducted in Rheumatoid Arthritis and in psoriasis. Further studies on small molecules and on their underlining mechanisms are advocated in PsA.
Collapse
Affiliation(s)
- Luisa Costa
- a Rheumatology Unit, Department of Clinical Medicine and Surgery , University Federico II , Naples , Italy
| | - Antonio Del Puente
- a Rheumatology Unit, Department of Clinical Medicine and Surgery , University Federico II , Naples , Italy
| | - Rosario Peluso
- a Rheumatology Unit, Department of Clinical Medicine and Surgery , University Federico II , Naples , Italy
| | - Marco Tasso
- a Rheumatology Unit, Department of Clinical Medicine and Surgery , University Federico II , Naples , Italy
| | - Paolo Caso
- b Geriatric Unit, Faculty of Medicine and Psychology , "Sapienza" University of Rome, S. Andrea, Hospital , Rome , Italy
| | - Maria Sole Chimenti
- c Rheumatology, allergology and clinical immunology, Department of System Medicine , University of Rome Tor Vergata , Rome , Italy
| | - Vincenzo Sabbatino
- a Rheumatology Unit, Department of Clinical Medicine and Surgery , University Federico II , Naples , Italy
| | - Nicolò Girolimetto
- a Rheumatology Unit, Department of Clinical Medicine and Surgery , University Federico II , Naples , Italy
| | - Carolina Benigno
- a Rheumatology Unit, Department of Clinical Medicine and Surgery , University Federico II , Naples , Italy
| | - Nicoletta Bertolini
- a Rheumatology Unit, Department of Clinical Medicine and Surgery , University Federico II , Naples , Italy
| | - Aurora Del Puente
- d Department of Medicine and Surgery , University of Milan "Bicocca" , Naples , Italy
| | - Roberto Perricone
- c Rheumatology, allergology and clinical immunology, Department of System Medicine , University of Rome Tor Vergata , Rome , Italy
| | - Raffaele Scarpa
- a Rheumatology Unit, Department of Clinical Medicine and Surgery , University Federico II , Naples , Italy
| | - Francesco Caso
- a Rheumatology Unit, Department of Clinical Medicine and Surgery , University Federico II , Naples , Italy
| |
Collapse
|
23
|
SCARPA RAFFAELE, CASO FRANCESCO, COSTA LUISA, PELUSO ROSARIO, DEL PUENTE ANTONIO, OLIVIERI IGNAZIO. Psoriatic Disease 10 Years Later. J Rheumatol 2017; 44:1298-1301. [DOI: 10.3899/jrheum.161402] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
24
|
Abstract
Alopecia areata (AA), a prevalent inflammatory cause of hair loss, lacks FDA-approved therapeutics for extensive cases, which are associated with very poor rates of spontaneous hair regrowth and major psychological distress. Current treatments for severe cases include broad immune-suppressants, which are associated with significant adverse effects, precluding long-term use, with rapid hair loss following treatment termination. As a result of the extent of the disease in severe cases, topical contact sensitizers and intralesional treatments are of limited use. The pathogenesis of AA is not yet fully understood, but recent investigations of the immune activation in AA skin reveal Th1/IFN-γ, as well as Th2, PDE4, IL-23, and IL-9 upregulations. Tissue analyses of both animal models and human lesions following broad-acting and cytokine-specific therapeutics (such as JAK inhibitors and ustekinumab, respectively) provide another opportunity for important insights into the pathogenesis of AA. As reviewed in this paper, numerous novel therapeutics are undergoing clinical trials for AA, emphasizing the potential transformation of the clinical practice of AA, which is currently lacking. Dermatologists are already familiar with the revolution in disease management of psoriasis, stemming from better understanding of immune dysregulations, and atopic dermatitis will soon follow a similar path. In light of these recent developments, the therapeutic arena of AA treatments is finally getting more exciting. AA will join the lengthening list of dermatologic diseases with mechanism-targeted drugs, thus changing the face of AA.
Collapse
Affiliation(s)
- Yael Renert-Yuval
- Department of Dermatology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Emma Guttman-Yassky
- Department of Dermatology and the Laboratory for Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
25
|
Minimal disease activity in patients with psoriatic arthritis treated with ustekinumab: results from a 24-week real-world study. Clin Rheumatol 2017; 36:1589-1593. [DOI: 10.1007/s10067-017-3700-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 05/17/2017] [Accepted: 05/22/2017] [Indexed: 12/29/2022]
|
26
|
Ishii M. Immunology proves a great success for treating systemic autoimmune diseases - a perspective on immunopharmacology: IUPHAR Review 23. Br J Pharmacol 2017; 174:1875-1880. [PMID: 28299772 DOI: 10.1111/bph.13784] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 02/27/2017] [Accepted: 03/03/2017] [Indexed: 11/29/2022] Open
Abstract
Recent advances in the bioengineering of monoclonal antibodies (mAbs) have revolutionized the treatment of several immunological and rheumatic diseases. mAbs exhibit high specificity and affinity, and are very effective targeting agents, associated with minimal off-target adverse effects. Of several relevant immunological diseases, rheumatoid arthritis was the condition initially treated with mAbs, with great success. Currently, many immunological disorders are targeted and successfully treated using such novel approaches; these include inflammatory bowel diseases, multiple sclerosis, lupus and psoriasis. Today, the efforts of researchers in basic immunology (with a long history) have borne fruit; bioengineered mAbs are employed in clinical practice. In this brief review, I will describe the current and emerging therapeutic mAbs and molecular targeted agents, and discuss the future of the field, especially from the viewpoint of pharmacology.
Collapse
Affiliation(s)
- Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine and Frontier Biosciences, Osaka University, Osaka, Japan
| |
Collapse
|
27
|
Banerjee S, Biehl A, Gadina M, Hasni S, Schwartz DM. JAK-STAT Signaling as a Target for Inflammatory and Autoimmune Diseases: Current and Future Prospects. Drugs 2017; 77:521-546. [PMID: 28255960 PMCID: PMC7102286 DOI: 10.1007/s40265-017-0701-9] [Citation(s) in RCA: 777] [Impact Index Per Article: 97.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The Janus kinase/signal transduction and activator of transcription (JAK-STAT) signaling pathway is implicated in the pathogenesis of inflammatory and autoimmune diseases including rheumatoid arthritis, psoriasis, and inflammatory bowel disease. Many cytokines involved in the pathogenesis of autoimmune and inflammatory diseases use JAKs and STATs to transduce intracellular signals. Mutations in JAK and STAT genes cause a number of immunodeficiency syndromes, and polymorphisms in these genes are associated with autoimmune diseases. The success of small-molecule JAK inhibitors (Jakinibs) in the treatment of rheumatologic disease demonstrates that intracellular signaling pathways can be targeted therapeutically to treat autoimmunity. Tofacitinib, the first rheumatologic Jakinib, is US Food and Drug Administration (FDA) approved for rheumatoid arthritis and is currently under investigation for other autoimmune diseases. Many other Jakinibs are in preclinical development or in various phases of clinical trials. This review describes the JAK-STAT pathway, outlines its role in autoimmunity, and explains the rationale/pre-clinical evidence for targeting JAK-STAT signaling. The safety and clinical efficacy of the Jakinibs are reviewed, starting with the FDA-approved Jakinib tofacitinib, and continuing on to next-generation Jakinibs. Recent and ongoing studies are emphasized, with a focus on emerging indications for JAK inhibition and novel mechanisms of JAK-STAT signaling blockade.
Collapse
Affiliation(s)
- Shubhasree Banerjee
- Rheumatology Fellowship and Training Branch, National Institute of Arthritis Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA.
| | - Ann Biehl
- Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Massimo Gadina
- Translational Immunology Section, National Institute of Arthritis Musculoskeletal and Skin diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sarfaraz Hasni
- Lupus Clinical Research Program, National Institute of Arthritis Musculoskeletal and Skin diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniella M Schwartz
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis Musculoskeletal and Skin diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
28
|
Reed M, Crosbie D. Apremilast in the treatment of psoriatic arthritis: a perspective review. Ther Adv Musculoskelet Dis 2017; 9:45-53. [PMID: 28255338 DOI: 10.1177/1759720x16673786] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Apremilast is an orally-active small molecule which inhibits phosphodiesterase-4 (PDE4). Clinical trials have demonstrated its efficacy and safety in psoriatic arthritis (PsA) and psoriasis. Established therapeutic options have variable effectiveness across the different domains of psoriatic disease. Whilst biologic therapies have proven to be of significant benefit to many patients, not all patients respond, and others are not eligible or do not tolerate biologic therapy. We review the mechanism of action, pharmacokinetics and clinical trial data with regards to both efficacy and safety for apremilast and consider where this new treatment may be positioned in the treatment of PsA.
Collapse
Affiliation(s)
- Michael Reed
- Department of Rheumatology, Queen Elizabeth University Hospital, UK
| | - David Crosbie
- Department of Rheumatology, Queen Elizabeth University Hospital, 1345 Govan Road, Glasgow G51 4TF, UK
| |
Collapse
|
29
|
Quantitative imaging by pixel-based contrast-enhanced ultrasound reveals a linear relationship between synovial vascular perfusion and the recruitment of pathogenic IL-17A-F +IL-23 + CD161 + CD4 + T helper cells in psoriatic arthritis joints. Clin Rheumatol 2016; 36:391-399. [PMID: 27995384 DOI: 10.1007/s10067-016-3500-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 11/27/2016] [Indexed: 12/14/2022]
Abstract
To develop quantitative imaging biomarkers of synovial tissue perfusion by pixel-based contrast-enhanced ultrasound (CEUS), we studied the relationship between CEUS synovial vascular perfusion and the frequencies of pathogenic T helper (Th)-17 cells in psoriatic arthritis (PsA) joints. Eight consecutive patients with PsA were enrolled in this study. Gray scale CEUS evaluation was performed on the same joint immediately after joint aspiration, by automatic assessment perfusion data, using a new quantification approach of pixel-based analysis and the gamma-variate model. The set of perfusional parameters considered by the time intensity curve includes the maximum value (peak) of the signal intensity curve, the blood volume index or area under the curve, (BVI, AUC) and the contrast mean transit time (MTT). The direct ex vivo analysis of the frequencies of SF IL17A-F+CD161+IL23+ CD4+ T cells subsets were quantified by fluorescence-activated cell sorter (FACS). In cross-sectional analyses, when tested for multiple comparison setting, a false discovery rate at 10%, a common pattern of correlations between CEUS Peak, AUC (BVI) and MTT parameters with the IL17A-F+IL23+ - IL17A-F+CD161+ - and IL17A-F+CD161+IL23+ CD4+ T cells subsets, as well as lack of correlation between both peak and AUC values and both CD4+T and CD4+IL23+ T cells, was observed. The pixel-based CEUS assessment is a truly measure synovial inflammation, as a useful tool to develop quantitative imaging biomarker for monitoring target therapeutics in PsA.
Collapse
|
30
|
Renert-Yuval Y, Guttman-Yassky E. A novel therapeutic paradigm for patients with extensive alopecia areata. Expert Opin Biol Ther 2016; 16:1005-14. [DOI: 10.1080/14712598.2016.1188076] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
31
|
Napolitano M, Caso F, Scarpa R, Megna M, Patrì A, Balato N, Costa L. Psoriatic arthritis and psoriasis: differential diagnosis. Clin Rheumatol 2016; 35:1893-1901. [PMID: 27156076 DOI: 10.1007/s10067-016-3295-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 04/27/2016] [Accepted: 04/28/2016] [Indexed: 12/19/2022]
Abstract
Psoriasis frequency ranges from 1 to 3 % in white population, and arthritis occurs in 10-40 % of psoriasis patients, representing a relevant health issue. Psoriatic arthritis (PsA) is an inflammatory arthropathy, associated with psoriasis, in which ocular-, intestinal-, metabolic-, and cardiovascular-related manifestations can variably coexist. In order to favor early PsA and psoriasis diagnosis, it is crucial to rule out other conditions that can resemble the disease and delay appropriate therapeutic approach. Therefore, the aim of this review is to focus on PsA and psoriasis differential diagnosis.
Collapse
Affiliation(s)
- Maddalena Napolitano
- Dermatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131, Naples, Italy
| | - Francesco Caso
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131, Naples, Italy
| | - Raffaele Scarpa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131, Naples, Italy
| | - Matteo Megna
- Dermatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131, Naples, Italy
| | - Angela Patrì
- Dermatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131, Naples, Italy
| | - Nicola Balato
- Dermatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131, Naples, Italy
| | - Luisa Costa
- Rheumatology Unit, Department of Clinical Medicine and Surgery, University Federico II, via S. Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
32
|
Qu X, Zhang S, Tao L, Song Y. A meta-analysis of apremilast on psoriatic arthritis long-term assessment of clinical efficacy (PALACE). Expert Rev Clin Pharmacol 2016; 9:799-805. [PMID: 26918950 DOI: 10.1586/17512433.2016.1159130] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Xiaoyu Qu
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, PR China
| | - Sixi Zhang
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, PR China
| | - Lina Tao
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, PR China
| | - Yanqing Song
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, PR China
| |
Collapse
|