1
|
Kharouf F, Gladman DD. Treatment controversies in spondyloarthritis and psoriatic arthritis: focus on biologics and targeted therapies. Expert Rev Clin Immunol 2024; 20:1381-1400. [PMID: 39072530 DOI: 10.1080/1744666x.2024.2384705] [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: 01/20/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION There are several treatment controversies that have emerged in spondyloarthritis and psoriatic arthritis. These are related to the nature of the conditions as well as to the use of medications. AREAS COVERED This review, which included a search of PubMed database as well as the references within the articles provides an overview of the nature of spondyloarthritis, controversy over the inclusion of psoriatic arthritis (PsA) as a peripheral spondyloarthritis, and a summary of current treatments for both PsA and axial spondyloarthritis (axSpA), with special emphasis on targeted therapy. The review highlights the differences in response to certain medications, particularly biologic therapy and summarizes the randomized controlled trials in psoriatic arthritis and axial spondyloarthritis providing data about the responses in table format. EXPERT OPINION There is a need for better outcome measures in axSpA. Currently, the measures are subjective. Imaging may be more appropriate but there is a need for research into the reliability and responsiveness of imaging techniques. In PsA, there may also be better response measures and research into the reliability and responsiveness of available measures is underway. There is also a need for novel therapies as well as biomarkers for response in both diseases.
Collapse
Affiliation(s)
- Fadi Kharouf
- Division of Rheumatology, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Gladman-Krembil Psoriatic Disease Program, Schroeder Arthritis Institute, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Dafna D Gladman
- Division of Rheumatology, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Gladman-Krembil Psoriatic Disease Program, Schroeder Arthritis Institute, Krembil Research Institute, Toronto Western Hospital, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Proft F, Duran TI, Ghoreschi K, Pleyer U, Siegmund B, Poddubnyy D. Treatment strategies for Spondyloarthritis: Implementation of precision medicine - Or "one size fits all" concept? Autoimmun Rev 2024; 23:103638. [PMID: 39276959 DOI: 10.1016/j.autrev.2024.103638] [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: 07/02/2024] [Revised: 09/05/2024] [Accepted: 09/07/2024] [Indexed: 09/17/2024]
Abstract
Spondyloarthritis (SpA) is a term to describe a group of chronic inflammatory rheumatic diseases, which have common pathophysiological, genetic, and clinical features. Under the umbrella term SpA, two main groups are subsumed: axial SpA (radiographic axSpA and non-radiographic axSpA) and peripheral SpA (with the leading representative being psoriatic arthritis (PsA) but also arthritis associated with inflammatory bowel disease (IBD), reactive arthritis, and undifferentiated pSpA). The key clinical symptom in axSpA is chronic back pain, typically with inflammatory characteristics, which starts in early adulthood, while the leading clinical manifestations of peripheral SpA (pSpA) are arthritis, enthesitis, and/or dactylitis. Furthermore, extra-musculoskeletal manifestations (EMMs) (acute anterior uveitis, psoriasis, and IBD) can accompany axial or peripheral symptoms. All these factors need to be taken into account when making treatment decisions in SpA patients. Despite the major advances in the treatment landscape over the past two decades with the introduction of biological disease-modifying anti-rheumatic drugs (bDMARDs) and most recently targeted synthetic DMARDs (tsDMARDs), a relevant proportion of patients still does not achieve the desired state of remission (=absence of disease activity). With this implementation of new treatment modalities, clinicians now have more choices to make in the treatment algorithms. However, despite generalized treatment recommendations, all factors need to be carefully considered when deciding on the optimal treatment strategy for an individual patient in clinical practice, aiming at an important first step towards personalized treatment strategies in SpA. In this narrative review, we focus on the efficacy of approved and emerging treatment options in axSpA and PsA as the main representative of pSpA and discuss their selective effect on the different manifestations associated with SpA to provide guidance on drivers of treatment decisions in specific situations.
Collapse
Affiliation(s)
- Fabian Proft
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| | - Tugba Izci Duran
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Clinic of Rheumatology, Denizli State Hospital, Denizli, Turkey
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Uwe Pleyer
- Department of Ophthalmology Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin; Berlin, Germany and (5)Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Denis Poddubnyy
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Epidemiology unit, German Rheumatism Research Centre, Berlin, Germany; Division of Rheumatology, Department of Medicine, University Health Network and University of Toronto, Toronto, Canada
| |
Collapse
|
3
|
Dalix E, Marcelli C, Bejan-Angoulvant T, Finckh A, Rancon F, Akrour M, De Araujo L, Presles E, Marotte H. Rotation or change of biotherapy after TNF blocker treatment failure for axial spondyloarthritis: the ROC-SpA study, a randomised controlled study protocol. BMJ Open 2024; 14:e087872. [PMID: 39260856 PMCID: PMC11409346 DOI: 10.1136/bmjopen-2024-087872] [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: 09/13/2024] Open
Abstract
INTRODUCTION Axial spondyloarthritis (axSpA) is a chronic inflammatory disease characterised by inflammatory low back pain. Non-steroidal anti-inflammatory drugs (NSAIDs) are recommended as a first treatment in axSpA. In case of inadequate response to NSAIDs, biological disease-modifying antirheumatic drugs (bDMARDs) should be introduced according to the recommendations of the European League Against Rheumatism (EULAR) and the American College of Rheumatology. Until 2015, only bDMARD was recommended for axSpA in case of failure to anti-tumour necrosis factor (TNF). The 2022 Assessment of SpondyloArthritis International Society (ASAS)-EULAR recommendation proposed to start an alternative bDMARD but without advocating a switch in mode of action as proposed in rheumatoid arthritis. Since 2015, the inhibition of interleukin (IL)-17 has demonstrated efficacy in axSpA. Then, we designed a randomised multicentre clinical trial to identify the more effective treatment after a first anti-TNF failure in axSpA, comparing an anti-IL-17 to a second anti-TNF. METHODS AND ANALYSIS The ROC-SpA (Rotation Or Change of biotherapy after first anti-TNF treatment failure in axSpA patients) study is a prospective, randomised, multicentre, superiority open-label phase IV trial comparing an anti-IL-17 strategy (secukinumab or ixekizumab) to a second TNF blocker in a 1:1 ratio. Patients with an active axSpA (Bath Ankylosing Spondylitis Disease Activity Index >4 or ankylosing spondylitis disease activity score (ASDAS) >3.5) with inadequate 3 months response to a first anti-TNF and with a stable dose of conventional synthetic DMARDs, oral corticosteroids and/or NSAIDs for at least 1 month are included in 31 hospital centres in France and Monaco. The primary outcome is the ASAS40 response at week 24. The secondary outcomes are ASAS40 at weeks 12 and 52, other clinical scores (ASAS20, partial remission rate, ASDAS major improvement rate) at weeks 12, 24 and 52 with the drugs and anti-drugs concentrations at baseline, weeks 12, 24 and 52. The primary analysis is performed at the end of the study according to the intent-to-treat principle. ETHICS AND DISSEMINATION Ethics approval was obtained from the committee for the protection of persons (Comité de protection des personnes Ouest IV #12/18_1, 6 February 2018) and registered in ClinicalTrials.gov and in EudraCT. Results of this study, whether positive or negative, will be presented at national and international congresses, to national axSpA patient associations and published in a peer-reviewed journal. It could also impact the international recommendation to manage patients with axSpA. TRIAL REGISTRATION NUMBER NCT03445845 and EudraCT2017-004700-22.
Collapse
Affiliation(s)
- Elisa Dalix
- University Jean Monnet Saint-Etienne, Mines Saint-Etienne, INSERM, SAINBIOSE U1059, Saint-Etienne, France
| | - Christian Marcelli
- Department of Rheumatology, Caen University Hospital University and INSERM, UniCaen, U1075, COMETE, PFRS, Normandie University, Caen, France
| | - Theodora Bejan-Angoulvant
- Centre Hospitalier Universitaire Et Faculté de Médecine, Pharmacologie Médicale, EA4245, Université de Tours, Tours, France
| | - Axel Finckh
- Division of Rheumatology, Department of Medicine, Faculty of Medicine and Geneva University Hospitals, Genev, Switzerland
| | - Florence Rancon
- Unité de Recherche Clinique, Centre d'Investigation Clinique 1408, CHU de Saint-Etienne, Saint-Etienne, France
| | - Madjid Akrour
- Unité de Recherche Clinique, Centre d'Investigation Clinique 1408, CHU de Saint-Etienne, Saint-Etienne, France
| | - Liliane De Araujo
- Unité de Recherche Clinique, Centre d'Investigation Clinique 1408, CHU de Saint-Etienne, Saint-Etienne, France
| | - Emilie Presles
- Unité de Recherche Clinique, Centre d'Investigation Clinique 1408, CHU de Saint-Etienne, Saint-Etienne, France
| | - Hubert Marotte
- CHU Saint-Etienne, Mines Saint-Etienne, Centre d'Investigation Clinique 1408, INSERM, SAINBIOSE U1059, University Jean Monnet Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
4
|
Lopalco G, Cito A, Venerito V, Iannone F, Proft F. The management of axial spondyloarthritis with cutting-edge therapies: advancements and innovations. Expert Opin Biol Ther 2024; 24:835-853. [PMID: 39109494 DOI: 10.1080/14712598.2024.2389987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/05/2024] [Indexed: 08/28/2024]
Abstract
INTRODUCTION Axial involvement in spondyloarthritis has significantly evolved from the original 1984 New York criteria for ankylosing spondylitis, leading to an improved understanding of axial spondyloarthritis (axSpA) as a disease continuum encompassing non- radiographic axSpA (nr-axSpA) and radiographic axSpA (r-axSpA). A clear definition for early axSpA has been established, underscoring the need for early intervention with biological and targeted synthetic drugs to mitigate pain, reduce functional impairment, and prevent radiographic progression. AREAS COVERED This review explores therapeutic strategies in axSpA management, focusing on biological and targeted synthetic therapies and recent advancements. Biologics targeting TNFα or IL-17 and targeted synthetic disease-modifying antirheumatic drugs (DMARDs) are primary treatment options. These therapies significantly impact clinical outcomes, radiographic progression, and patient-reported functional improvement. EXPERT OPINION AxSpA treatment has evolved significantly, offering various therapeutic options. Biological DMARDs, particularly TNFα inhibitors, have transformed treatment, significantly enhancing patient outcomes. However, challenges persist for patients unresponsive or intolerant to existing therapies. Emerging therapeutic targets promise to address these challenges. Comprehensive management strategies and personalized approaches, considering extra-articular manifestations and individual patient factors, are crucial for achieving optimal outcomes in axSpA management.
Collapse
Affiliation(s)
- Giuseppe Lopalco
- Department of Precision Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Bari, Italy
| | - Andrea Cito
- Department of Precision Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Venerito
- Department of Precision Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Bari, Italy
| | - Florenzo Iannone
- Department of Precision Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Bari, Italy
| | - Fabian Proft
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
5
|
Calderón P, Núñez P, Nos P, Quera R. Personalised therapy in inflammatory bowel disease. GASTROENTEROLOGIA Y HEPATOLOGIA 2024; 47:763-770. [PMID: 38101615 DOI: 10.1016/j.gastrohep.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/27/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023]
Abstract
Inflammatory bowel diseases (IBD), with ulcerative colitis and Crohn's disease being their most common presentations, comprise a spectrum of diverse disease phenotypes, exhibiting variable behaviors ranging from an indolent course to aggressive phenotypes that impact quality of life of these patients. The last two decades have been marked by the development of new medications (biological therapy and novel small molecules) with diverse mechanisms of action, which have revolutionized the management of IBD, thereby enhancing the quality of life for these patients. This landscape of multiple therapeutic options underscores the need to define which medication will benefit each patient the most and at what speed it should be started. The objective of this review is to present personalized approaches for patients with IBD, thus contributing to therapeutic management.
Collapse
Affiliation(s)
- Paula Calderón
- Programa de Enfermedad Inflamatoria Intestinal, Centro de Enfermedades Digestivas, Clínica Universidad de Los Andes, Santiago, Chile
| | - Paulina Núñez
- Programa de Enfermedad Inflamatoria Intestinal, Centro de Enfermedades Digestivas, Clínica Universidad de Los Andes, Santiago, Chile; Sección de Gastroenterología, Departamento de Medicina Interna, Universidad de los Andes, Santiago, Chile; Hospital San Juan de Dios, Facultad de Medicina Occidente, Universidad de Chile, Santiago, Chile
| | - Pilar Nos
- Servicio de Aparato Digestivo en Hospital Universitari y Politécnic la Fe de Valencia, Valencia, España
| | - Rodrigo Quera
- Programa de Enfermedad Inflamatoria Intestinal, Centro de Enfermedades Digestivas, Clínica Universidad de Los Andes, Santiago, Chile; Sección de Gastroenterología, Departamento de Medicina Interna, Universidad de los Andes, Santiago, Chile.
| |
Collapse
|
6
|
Alascio L, Azuaga-Piñango AB, Frade-Sosa B, Sarmiento-Monroy JC, Ponce A, Farietta S, Gómez-Puerta JA, Sanmartí R, Cañete JD, Ramírez J. Axial Disease in Psoriatic Arthritis: A Challenging Domain in Clinical Practice. Diagnostics (Basel) 2024; 14:1637. [PMID: 39125513 PMCID: PMC11311426 DOI: 10.3390/diagnostics14151637] [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: 06/27/2024] [Revised: 07/24/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory condition affecting about one-third of individuals with psoriasis. Defining axial involvement in PsA (axPsA) remains debated. While rheumatologists guide clinical practice, consensus on axPsA is still lacking. This paper explores historical and upcoming definitions from the Axial Involvement in Psoriatic Arthritis (AXIS) study, which aims to establish a validated axPsA definition. Epidemiological data reveal diverse axPsA prevalence rates, emphasizing its complex relationship with peripheral arthritis and enthesitis. Unique genetic, clinical, and radiological features differentiate axPsA from ankylosing spondylitis (AS), necessitating refined classification criteria. The recommendations from the Assessment of Spondylarthritis international Society (ASAS) provide valuable guidance due to the limited direct evidence. Emerging therapies, including interleukin-23 (IL-23) inhibitors or Janus kinase (JAK) inhibitors, are under investigation for axPsA. Currently, secukinumab, an interleukin-17 (IL-17) inhibitor, is an evidence-based option for axPsA management. However, given the variability in individual patient responses and disease manifestations, personalized, evidence-based treatment approaches remain essential for optimizing patient outcomes. In the final section, two real-life cases illustrate the challenges in managing axPsA, emphasizing the importance of tailored therapies. Achieving precision in defining axPsA remains a formidable task, making detailed criteria essential for effective strategies and improving patient outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Julio Ramírez
- Arthritis Unit, Rheumatology Department, Hospital Clínic Barcelona, Villarroel Street, 170, 08036 Barcelona, Spain; (L.A.); (A.B.A.-P.); (B.F.-S.); (J.C.S.-M.); (A.P.); (S.F.); (J.A.G.-P.); (R.S.); (J.D.C.)
| |
Collapse
|
7
|
Hen O, Harrison SR, De Marco G, Marzo-Ortega H. Early psoriatic arthritis: when is the right time to start advanced therapy? Ther Adv Musculoskelet Dis 2024; 16:1759720X241266727. [PMID: 39071239 PMCID: PMC11283661 DOI: 10.1177/1759720x241266727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 06/20/2024] [Indexed: 07/30/2024] Open
Abstract
Despite significant advances in the treatment of psoriatic arthritis (PsA) in the last two decades, remission remains elusive and there is no cure. Evidence from rheumatoid arthritis (RA) confirming enhanced response and outcome from earlier treatment intervention suggests the plausibility of the window of opportunity in the pathogenesis of RA. Yet, data are lacking in PsA. Although treatment response may be enhanced in shorter disease duration, it is unknown how this early intervention may impact long-term outcomes. Furthermore, it remains to be demonstrated whether there is a best treatment strategy and time of intervention. Crucially, the main hurdle when aiming for early treatment intervention is the ability to achieve a timely diagnosis that highlights the need to focus research efforts on characterizing the very early disease stages including the transition to PsA in the at-risk psoriasis population.
Collapse
Affiliation(s)
- Or Hen
- NIHR Leeds Biomedical Research Centre, The Leeds Teaching Hospitals NHS Trust, Leeds
- Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- Department of Medicine ‘C’, Sheba Medical Center, Tel HaShomer, Ramat Gan, Israel
| | - Stephanie R. Harrison
- NIHR Leeds Biomedical Research Centre, The Leeds Teaching Hospitals NHS Trust, Leeds
- Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- Stephanie R. Harrison is also affiliated to Leeds Institute of Data Analytics, University of Leeds, Leeds
| | - Gabriele De Marco
- NIHR Leeds Biomedical Research Centre, The Leeds Teaching Hospitals NHS Trust, Leeds
- Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- Stephanie R. Harrison is also affiliated to Leeds Institute of Data Analytics, University of Leeds, Leeds
| | - Helena Marzo-Ortega
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Second Floor, Chapel Allerton Hospital, Chapeltown Road, Leeds LS7 4SA, UK
| |
Collapse
|
8
|
Baraliakos X, Gladman DD, Chakravarty SD, Gong C, Shawi M, Rampakakis E, Kishimoto M, Soriano ER, Mease PJ. BASDAI versus ASDAS in evaluating axial involvement in patients with psoriatic arthritis: a pooled analysis of two phase 3 studies. Rheumatol Adv Pract 2024; 8:rkae058. [PMID: 38765190 PMCID: PMC11099656 DOI: 10.1093/rap/rkae058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 03/18/2024] [Indexed: 05/21/2024] Open
Abstract
Objective In the absence of axial psoriatic arthritis (axPsA)-specific tools, the BASDAI and Ankylosing Spondylitis Disease Activity Score (ASDAS) are used to assess axial symptoms in patients with PsA. Here, we assessed the performance of BASDAI and ASDAS in patients with PsA. Methods Patients with active PsA in DISCOVER-1 and DISCOVER-2 (ClinicalTrials.gov: NCT03162796 and NCT03158285, respectively) with or without axPsA but with available baseline BASDAI information were analysed; those with investigator-identified axial symptoms and imaging-confirmed sacroiliitis comprised the axPsA cohort. Correlations between BASDAI/ASDAS and clinical variables were assessed with Pearson's coefficient (r). Longitudinal effects of enthesitis (Leeds Enthesitis Index [LEI]), swollen joint count and presence versus absence of axPsA on BASDAI/ASDAS (normalized 0-10 scale) were analysed with mixed models for repeated measures. Results At baseline in the axPsA (n = 312) and non-axPsA (n = 124) cohorts, BASDAI scores showed no or weak correlation with swollen joint count (0.18-0.20), tender joint count (0.12-0.29), LEI (-0.04 to 0.24) and physician global assessment (0.35-0.43); moderate correlation with fatigue (both -0.56); and strong correlation with patient global assessment of disease activity (0.62-0.69) and patient-reported pain (0.66-0.70). Similar correlations were observed for ASDAS. Axial involvement versus non-involvement was associated with higher BASDAI scores and ASDAS (all β ≥ 0.5), without differences between instruments; longitudinal associations between swollen joint count (β ≤ 0.06)/LEI (β ≤ 0.19) and BASDAI/ASDAS were clinically unimportant. Conclusion BASDAI and ASDAS performed similarly in patients with active PsA and axial involvement, independent of peripheral disease involvement, supporting their performance in assessing axial disease activity. Trial registration ClinicalTrials.gov, http://clinicaltrials.gov, NCT03162796 and NCT03158285.
Collapse
Affiliation(s)
| | - Dafna D Gladman
- Department of Medicine, University of Toronto, Schroeder Arthritis Institute; Krembil, Research Institute; Toronto Western Hospital, Toronto, ON, Canada
| | - Soumya D Chakravarty
- Immunology, Janssen Scientific Affairs, LLC, a Johnson & Johnson company, Horsham, PA, USA
- Division of Rheumatology, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Cinty Gong
- Immunology, Janssen Research & Development, LLC, a Johnson & Johnson company, Spring House, PA, USA
| | - May Shawi
- Immunology, Janssen Research & Development, LLC, a Johnson & Johnson company, Titusville, NJ, USA
| | - Emmanouil Rampakakis
- Department of Pediatrics, McGill University, Montreal, Canada
- Scientific Affairs, JSS Medical Research, Inc, Montreal, Canada
| | - Mitsumasa Kishimoto
- Department of Nephrology and Rheumatology, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Enrique R Soriano
- Rheumatology Section, Internal Medicine Service, Department of Medicine, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
- Department of Medicine, University Institute Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Philip J Mease
- Rheumatology Research, Providence Swedish Medical Center, Seattle, WA, USA
- University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
9
|
Reddy SM, Xue K, Husni ME, Scher JU, Stephens-Shields AJ, Goel N, Koplin J, Craig ET, Walsh JA, Ogdie A. Use of the Bath Ankylosing Spondylitis Disease Activity Index in Patients With Psoriatic Arthritis With and Without Axial Disease. J Rheumatol 2024; 51:139-143. [PMID: 38101918 DOI: 10.3899/jrheum.2023-0504] [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] [Accepted: 11/27/2023] [Indexed: 12/17/2023]
Abstract
OBJECTIVE To evaluate whether the Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) is a responsive instrument in psoriatic arthritis (PsA) and whether it differentiates between axial and peripheral disease activity in PsA. METHODS Individuals with PsA initiating therapy in a longitudinal cohort study based in the United States were included. Axial PsA (axPsA), most often also associated with peripheral disease, was defined as fulfillment of the Assessment of Spondyloarthritis international Society axial spondyloarthritis classification criteria or presence of axial disease imaging features. Baseline BASDAI, individual BASDAI items, patient global assessment, patient pain, and Routine Assessment of Patient Index Data 3, and score changes following therapy initiation were descriptively reported. Standardized response means (SRMs) were calculated as the mean change divided by the SD of the change. RESULTS The mean (SD) baseline BASDAI score at the time of therapy initiation was 5.0 (2.2) among those with axPsA (n = 40) and 4.8 (2.0) among those with peripheral-only disease (n = 79). There was no significant difference in patient-reported outcome scores between the groups. The mean change for BASDAI was similar among axial vs peripheral disease (-0.75 vs -0.83). SRMs were similar across axial vs peripheral disease for BASDAI (-0.37 vs -0.44) and the individual BASDAI items. CONCLUSION BASDAI has reasonable responsiveness in PsA but does not differentiate between axPsA and peripheral PsA. (ClinicalTrials.gov: NCT03378336).
Collapse
Affiliation(s)
- Soumya M Reddy
- S.M. Reddy, MD, J.U. Scher, MD, Department of Medicine and Rheumatology, New York University, New York, New York
| | - Katie Xue
- K. Xue, BS, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - M Elaine Husni
- M.E. Husni, MD, MPH, Department Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, Ohio
| | - Jose U Scher
- S.M. Reddy, MD, J.U. Scher, MD, Department of Medicine and Rheumatology, New York University, New York, New York
| | - Alisa J Stephens-Shields
- A.J. Stephens-Shields, PhD, Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Niti Goel
- N. Goel, MD, Patient Research Partner, and Department of Medicine, Division of Rheumatology, Duke University School of Medicine, Durham, North Carolina
| | - Joelle Koplin
- J. Koplin, CRNP, Department of Medicine/Rheumatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ethan T Craig
- E.T. Craig, MD, Department of Medicine/Rheumatology, Perelman School of Medicine, University of Pennsylvania, and Department of Medicine/Rheumatology, Michael J. Crescenz Veterans Medical Center, Philadelphia, Pennsylvania
| | - Jessica A Walsh
- J.A. Walsh, MD, MBA, Department of Medicine and Rheumatology, University of Utah, Salt Lake City, Utah
| | - Alexis Ogdie
- A. Ogdie, MD, MSCE, Department of Medicine/Rheumatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, and Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
10
|
Yousif P, Nahra V, Khan MA, Magrey M. Disease characteristics, pathogenesis, and treatment controversies of axial psoriatic arthritis. Joint Bone Spine 2024; 91:105625. [PMID: 37495073 DOI: 10.1016/j.jbspin.2023.105625] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/16/2023] [Accepted: 07/12/2023] [Indexed: 07/28/2023]
Abstract
Axial psoriatic arthritis (axPsA) has considerable overlap with axial spondyloarthritis (axSpA) but has some unique features that sometimes preclude classification into axSpA. It has some clinical and radiographic differences compared to axSpA. Imaging typically shows asymmetric syndesmophytes, mainly in the cervical spine, with less frequent sacroiliitis. It more commonly presents later in life and is associated with less severe inflammatory back pain than axSpA. The interleukin (IL) IL-23/IL-17 axis is central to the pathogenesis of both diseases. However, the response to therapies targeting these cytokines has been different. IL-23 inhibitors are ineffective in axSpA but may be effective in psoriatic arthritis (PsA). Recent post hoc analyses of clinical trial data with IL-23 inhibitors in PsA have raised the possibility of their efficacy in axPsA and need evaluation in future clinical trials. Moreover, there is a need for classification criteria for axPsA and better tools to assess therapeutic response.
Collapse
Affiliation(s)
- Patrick Yousif
- Division of Rheumatology, University Hospitals Cleveland Medical Center/Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Vicky Nahra
- Division of Rheumatology, University Hospitals Cleveland Medical Center/Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Muhammad A Khan
- Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Marina Magrey
- Division of Rheumatology, University Hospitals Cleveland Medical Center/Case Western Reserve University School of Medicine, Cleveland, OH, United States.
| |
Collapse
|
11
|
Mease PJ, Gladman DD, Poddubnyy D, Chakravarty SD, Shawi M, Kollmeier AP, Xu XL, Xu S, Deodhar A, Baraliakos X. Efficacy of Guselkumab on Axial-Related Symptoms Through up to 2 Years in Adults with Active Psoriatic Arthritis in the Phase 3, Randomized, Placebo-Controlled DISCOVER-2 Study. Rheumatol Ther 2023; 10:1637-1653. [PMID: 37819505 PMCID: PMC10654317 DOI: 10.1007/s40744-023-00592-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/03/2023] [Indexed: 10/13/2023] Open
Abstract
INTRODUCTION Guselkumab previously showed greater improvements versus placebo in axial symptoms in patients with psoriatic arthritis (PsA) (assessed by Bath Ankylosing Spondylitis Disease Activity Index [BASDAI] and Ankylosing Spondylitis Disease Activity Score [ASDAS]), in post hoc analyses of the phase 3, placebo-controlled, randomized DISCOVER-1 and DISCOVER-2 studies. We now evaluate durability of response in axial-related outcomes through 2 years of DISCOVER-2. METHODS DISCOVER-2 biologic-naive adults with active PsA (≥ 5 tender/ ≥ 5 swollen joints, C-reactive protein ≥ 0.6 mg/dl) were randomized to guselkumab 100 mg every 4 weeks (Q4W) or at week 0, week 4, then Q8W, or placebo → guselkumab Q4W at week 24. Among patients with imaging-confirmed sacroiliitis (investigator-identified), axial symptoms were assessed through 2 years utilizing BASDAI, BASDAI Question #2 (spinal pain), modified BASDAI (mBASDAI; excludes Question #3 [peripheral joint pain]), and ASDAS. Mean changes in scores and proportions of patients achieving ≥ 50% improvement in BASDAI (BASDAI 50) and ASDAS responses, including major improvement (decrease ≥ 2.0), were determined through week 100. Treatment failure rules (through week 24) and nonresponder imputation of missing data (post-week 24) were utilized. Mean BASDAI component scores were assessed through week 100 (observed data). Exploratory analyses evaluated efficacy by sex and HLA-B*27 status. RESULTS Among 246 patients with PsA and imaging-confirmed sacroiliitis, guselkumab-treated patients had greater mean improvements in BASDAI, mBASDAI, spinal pain, and ASDAS scores, lower mean BASDAI component scores, and greater response rates in achieving BASDAI 50 and ASDAS major improvement vs. placebo at week 24. Differences from placebo were observed for guselkumab-treated patients in selected endpoints regardless of sex or HLA-B*27 status. At week 100, mean improvements were ~ 3 points for all BASDAI scores and 1.6-1.7 for ASDAS; 49-54% achieved BASDAI 50 and 39% achieved ASDAS major improvement at week 100. CONCLUSIONS Guselkumab treatment provided durable and meaningful improvements in axial symptoms and disease activity in substantial proportions of patients with active PsA and imaging-confirmed sacroiliitis. TRIAL REGISTRATION Clinicaltrials.gov NCT03158285.
Collapse
Affiliation(s)
- Philip J Mease
- Department of Rheumatology, Swedish Medical Center/Providence St. Joseph Health and University of Washington, Rheumatology Research, 601 Broadway, Ste 600, Seattle, WA, 98122, USA.
| | - Dafna D Gladman
- Centre for Prognosis Studies in the Rheumatic Diseases, Toronto Western Hospital, Toronto, ON, Canada
| | | | - Soumya D Chakravarty
- Janssen Scientific Affairs, LLC, Horsham, PA, USA
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - May Shawi
- Janssen Research & Development, LLC, Titusville, NJ, USA
| | | | - Xie L Xu
- Janssen Research & Development, LLC, San Diego, CA, USA
| | - Stephen Xu
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Atul Deodhar
- Oregon Health & Science University, Portland, OR, USA
| | | |
Collapse
|
12
|
Kim SH, Lee SH. Updates on ankylosing spondylitis: pathogenesis and therapeutic agents. JOURNAL OF RHEUMATIC DISEASES 2023; 30:220-233. [PMID: 37736590 PMCID: PMC10509639 DOI: 10.4078/jrd.2023.0041] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 09/23/2023]
Abstract
Ankylosing spondylitis (AS) is an autoinflammatory disease that manifests with the unique feature of enthesitis. Gut microbiota, HLA-B*27, and biomechanical stress mutually influence and interact resulting in setting off a flame of inflammation. In the HLA-B*27 positive group, dysbiosis in the gut environment disrupts the barrier to exogenous bacteria or viruses. Additionally, biomechanical stress induces inflammation through enthesial resident or gut-origin immune cells. On this basis, innate and adaptive immunity can propagate inflammation and lead to chronic disease. Finally, bone homeostasis is regulated by cytokines, by which the inflamed region is substituted into new bone. Agents that block cytokines are constantly being developed to provide diverse therapeutic options for preventing the progression of inflammation. In addition, some antibodies have been shown to distinguish disease selectively, which support the involvement of autoimmune immunity in AS. In this review, we critically analyze the complexity and uniqueness of the pathogenesis with updates on the findings of immunity and provide new information about biologics and biomarkers.
Collapse
Affiliation(s)
- Se Hee Kim
- Division of Rheumatology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Sang-Hoon Lee
- Division of Rheumatology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
13
|
Nakamura A, Chandran V. Risankizumab for the treatment of active psoriatic arthritis in adults. Expert Rev Clin Immunol 2023; 19:1435-1448. [PMID: 37772959 DOI: 10.1080/1744666x.2023.2265567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/27/2023] [Indexed: 09/30/2023]
Abstract
INTRODUCTION Psoriatic arthritis (PsA) is a chronic rheumatic disease that displays a variety of clinical manifestations. Although new treatments have emerged over the last 2 decades, challenges remain in controlling inflammation in multiple PsA clinical domains. AREAS COVERED Risankizumab, one of the biologic disease modification anti-rheumatic drugs (bDMARDs) that target the interleukin (IL)-23 p19 subunit, was recently approved for PsA worldwide. This review primarily highlights the recent clinical trials of risankizumab covering its physiological evaluation, patient-reported outcomes, and safety profiles in patients with PsA. We also provide evidence for anti-IL-23 therapies against extra-articular manifestations and axial symptoms of PsA. Furthermore, potential distinct efficacies and mechanisms of action in anti-IL-23 therapies are discussed. Overall, risankizumab is effective in a variety of clinical signs and symptoms of PsA regardless of prior bDMARDs experience. EXPERT OPINION Accumulating evidence shows that anti-IL-23 drugs, including risankizumab, are promising treatments that can be used as first- or second-line therapies for PsA. However, multiple challenges remain, including confirming efficacy for axial symptoms and identifying the phenotype of specific patients who respond better to risankizumab than other drugs. Lastly, future data focusing on the long-term efficacy and safety of risankizumab beyond the 1-year observation period are also needed.
Collapse
Affiliation(s)
- Akihiro Nakamura
- Department of Medicine, Division of Rheumatology, Queen's University, Kingston, Ontario, Canada
- Translational Institute of Medicine, School of Medicine, Queen's University, Kingston, Ontario, Canada
- Rheumatology Clinic, Kingston Health Science Centre, Kingston, Ontario, Canada
| | - Vinod Chandran
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- Division of Rheumatology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Queiro R, Loredo M, Braña I, Pardo E, Alonso S, Alperi M. Managing psoriatic arthritis in different clinical scenarios. Expert Rev Clin Immunol 2023; 19:1469-1484. [PMID: 37589128 DOI: 10.1080/1744666x.2023.2249235] [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: 04/27/2023] [Accepted: 08/14/2023] [Indexed: 08/18/2023]
Abstract
INTRODUCTION Psoriatic arthritis (PsA) is a chronic, immune-mediated disease characterized by synovio-entheseal inflammation. It is estimated to affect around 30% of patients with psoriasis and significantly reduces patients' physical function and quality of life. There is a growing number of treatment options for PsA, but due to the heterogeneous clinical features of the disease and prevalence of comorbidities, managing PsA can be challenging. AREAS COVERED In this article, we review current understanding of the disease and available pharmacological options. Based on published treatment guidelines, emerging evidence and clinical experience, we provide our expert opinion on treatment strategies, taking into consideration the predominant disease domain and the presence of comorbidities, which can impact treatment decisions and clinical outcomes. EXPERT OPINION Biological and targeted synthetic disease-modifying agents are dramatically improving the lives of patients with PsA. Biosimilar TNF inhibitors offer a particularly versatile and cost-effective option, whilst newer biologics and targeted synthetic molecules that can be used to treat most domains of psoriatic disease are an attractive alternative to TNF inhibitors. Despite a lack of consensus on treatment sequencing and tapering, it is important that PsA patients, especially those with comorbidities, are looked after by a multidisciplinary team to optimize their care.
Collapse
Affiliation(s)
- Rubén Queiro
- Rheumatology & ISPA Translational Immunology Division, Central University Hospital of Asturias, Oviedo, Spain
- Department of Medicine, Oviedo University School of Medicine, Oviedo, Spain
| | - Marta Loredo
- Rheumatology Division, Central University Hospital of Asturias, Oviedo, Spain
| | - Ignacio Braña
- Rheumatology Division, Central University Hospital of Asturias, Oviedo, Spain
| | - Estefanía Pardo
- Rheumatology Division, Central University Hospital of Asturias, Oviedo, Spain
| | - Sara Alonso
- Rheumatology Division, Central University Hospital of Asturias, Oviedo, Spain
| | - Mercedes Alperi
- Rheumatology Division, Central University Hospital of Asturias, Oviedo, Spain
| |
Collapse
|
15
|
Kavanaugh A, Baraliakos X, Gao S, Chen W, Sweet K, Chakravarty SD, Song Q, Shawi M, Rahman P. Genetic and Molecular Distinctions Between Axial Psoriatic Arthritis and Radiographic Axial Spondyloarthritis: Post Hoc Analyses from Four Phase 3 Clinical Trials. Adv Ther 2023; 40:2439-2456. [PMID: 36995469 PMCID: PMC10129963 DOI: 10.1007/s12325-023-02475-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/20/2023] [Indexed: 03/31/2023]
Abstract
INTRODUCTION Emerging evidence suggests psoriatic arthritis (PsA) with axial involvement (axPsA) and radiographic axial spondyloarthritis (r-axSpA) may possibly represent distinct disorders, with some differing clinical manifestations, genetic associations, and radiographic findings. Moreover, axPsA and r-axSpA may respond differently to therapies: guselkumab (interleukin [IL]-23p19 subunit inhibitor [i]) and ustekinumab (IL-12/23p40i) demonstrated improvements in axial symptoms in patients with PsA; however, neither risankizumab (IL-23p19i) nor ustekinumab demonstrated efficacy versus placebo in patients with r-axSpA. Current analyses aim to further understand potential molecular distinctions between axPsA and r-axSpA and examine the pharmacodynamic effects of guselkumab in patients with axPsA and those with PsA without axial involvement (non-axPsA). METHODS Post hoc analyses utilized biomarker data from blood and serum samples collected from a subset of participants in phase 3 studies of ustekinumab in r-axSpA and guselkumab in PsA (DISCOVER-1 and DISCOVER-2). Participants with axPsA were identified by investigator-verified sacroiliitis (imaging-confirmed) and axial symptoms. HLA mapping, serum cytokine analysis, and whole-blood RNA sequencing were conducted. RESULTS Relative to r-axSpA, patients with axPsA had a lower prevalence of HLA-B27, HLA-C01, and HLA-C02 alleles and a higher prevalence of HLA-B13, HLA-B38, HLA-B57, HLA-C06, and HLA-C12 alleles. Compared with r-axSpA, patients with axPsA had elevated baseline levels of serum IL-17A and IL-17F cytokines, enrichment of IL-17 and IL-10 pathway-associated genes, and neutrophil gene markers. Across axPsA and non-axPsA cohorts, reductions in cytokine levels and normalization of pathway-associated gene expression with guselkumab treatment were comparable. CONCLUSION The differences in HLA genetic associations, serum cytokines, and enrichment scores support the concept that axPsA and r-axSpA may be distinct disorders. The comparable pharmacodynamic effects of guselkumab on cytokine levels and pathway-associated genes observed in patients with axPsA and non-axPsA are consistent with demonstrated clinical improvements across PsA cohorts. These findings contribute to the understanding of potential genetic and molecular distinctions between axPsA and r-axSpA. TRIAL REGISTRATION ClinicalTrials.gov identifiers, NCT03162796, NCT0315828, NCT02437162, and NCT02438787.
Collapse
Affiliation(s)
- Arthur Kavanaugh
- University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | | | - Sheng Gao
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Warner Chen
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Kristen Sweet
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - Soumya D Chakravarty
- Janssen Scientific Affairs, LLC, Horsham, PA, USA
- Drexel University College of Medicine, Philadelphia, PA, USA
| | - Qingxuan Song
- Janssen Research & Development, LLC, Spring House, PA, USA
| | - May Shawi
- Immunology Global Medical Affairs, Janssen Pharmaceutical Companies of Johnson & Johnson, Horsham, PA, USA
| | - Proton Rahman
- Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
16
|
Proft F, Käding H. Same, same or different? Commonalities and differences between spondyloarthritis and its subsets of axial and peripheral spondyloarthritis with psoriatic arthritis and its diverse phenotypes. RMD Open 2023; 9:e002872. [PMID: 37028815 PMCID: PMC10083843 DOI: 10.1136/rmdopen-2022-002872] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/23/2023] [Indexed: 04/09/2023] Open
Affiliation(s)
- Fabian Proft
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charite Universitatsmedizin Berlin, Berlin, Germany
| | - Henriette Käding
- Department of Gastroenterology, Infectiology and Rheumatology (including Nutrition Medicine), Charite Universitatsmedizin Berlin, Berlin, Germany
| |
Collapse
|
17
|
Del Vescovo S, Venerito V, Iannone C, Lopalco G. Uncovering the Underworld of Axial Spondyloarthritis. Int J Mol Sci 2023; 24:6463. [PMID: 37047435 PMCID: PMC10095023 DOI: 10.3390/ijms24076463] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Axial spondyloarthritis (axial-SpA) is a multifactorial disease characterized by inflammation in sacroiliac joints and spine, bone reabsorption, and aberrant bone deposition, which may lead to ankylosis. Disease pathogenesis depends on genetic, immunological, mechanical, and bioenvironmental factors. HLA-B27 represents the most important genetic factor, although the disease may also develop in its absence. This MHC class I molecule has been deeply studied from a molecular point of view. Different theories, including the arthritogenic peptide, the unfolded protein response, and HLA-B27 homodimers formation, have been proposed to explain its role. From an immunological point of view, a complex interplay between the innate and adaptive immune system is involved in disease onset. Unlike other systemic autoimmune diseases, the innate immune system in axial-SpA has a crucial role marked by abnormal activity of innate immune cells, including γδ T cells, type 3 innate lymphoid cells, neutrophils, and mucosal-associated invariant T cells, at tissue-specific sites prone to the disease. On the other hand, a T cell adaptive response would seem involved in axial-SpA pathogenesis as emphasized by several studies focusing on TCR low clonal heterogeneity and clonal expansions as well as an interindividual sharing of CD4/8 T cell receptors. As a result of this immune dysregulation, several proinflammatory molecules are produced following the activation of tangled intracellular pathways involved in pathomechanisms of axial-SpA. This review aims to expand the current understanding of axial-SpA pathogenesis, pointing out novel molecular mechanisms leading to disease development and to further investigate potential therapeutic targets.
Collapse
Affiliation(s)
- Sergio Del Vescovo
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Polyclinic Hospital, University of Bari, 70124 Bari, Italy
| | - Vincenzo Venerito
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Polyclinic Hospital, University of Bari, 70124 Bari, Italy
| | - Claudia Iannone
- Division of Clinical Rheumatology, ASST Gaetano Pini-CTO Institute, 20122 Milan, Italy
| | - Giuseppe Lopalco
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), Polyclinic Hospital, University of Bari, 70124 Bari, Italy
| |
Collapse
|
18
|
[Chronic back pain in axial spondylarthritis : Current diagnostic challenges and treatment possibilities]. Z Rheumatol 2023; 82:10-17. [PMID: 36044071 PMCID: PMC9894960 DOI: 10.1007/s00393-022-01256-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND Among chronic back diseases, axial spondylarthritis (axSpA) is the entity with the largest spectrum of specific anti-inflammatory treatment modalities; however, from a general medical perspective axSpA is only ranked as another etiology of back pain to be considered after spinal cord or cauda equina compression, bone metastases, epidural abscess or osteomyelitis of the vertebrae, radiculopathy or spinal stenosis. Due to its relatively low prevalence and mostly later occurring sequelae, there is a danger that axSpA will be a relatively neglected entity for specialists. RESULTS This article recapitulates the recommendations of the Assessment of Ankylosing Spondyloarthritis International Society (ASAS). This review addresses the practical aspects of the detailed evaluation of treatment attempts carried out so far with nonsteroidal anti-inflammatory drugs (NSAID) for back pain. Undesired effects on the symptoms of the lower intestinal tract could be of particular interest here. The sex-specific differences in the response to treatment with tumor necrosis factor (TNF) inhibitors in axSpA are mentioned. Further aspects of treatment options with biologics in axSpA are discussed based on a case of maintained remission after a course of interleukin (IL) 17 inhibitors in undifferentiated, HLA-B27 and magnetic resonance imaging (MRI) positive axSpA and long-standing good treatment response to IL-12/23 inhibitor treatment in axial psoriatic arthritis. Furthermore, the literature is discussed with respect to uveitis, carditis and amyloidosis in the context of axSpA. CONCLUSION The early diagnosis in the general medical context and the specific consideration of numerous predictive factors play an increasingly more important role in the personalized treatment of axSpA.
Collapse
|
19
|
Salaffi F, Siragusano C, Alciati A, Cassone G, D’Angelo S, Guiducci S, Favalli EG, Conti F, Gremese E, Iannone F, Caporali R, Sebastiani M, Ferraccioli GF, Lapadula G, Atzeni F. Axial Spondyloarthritis: Reshape the Future-From the "2022 GISEA International Symposium". J Clin Med 2022; 11:jcm11247537. [PMID: 36556152 PMCID: PMC9780899 DOI: 10.3390/jcm11247537] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/02/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
The term "axial spondyloarthritis" (axSpA) refers to a group of chronic rheumatic diseases that predominantly involve the axial skeleton and consist of ankylosing spondylitis, reactive arthritis, arthritis/spondylitis associated with psoriasis (PsA) and arthritis/spondylitis associated with inflammatory bowel diseases (IBD). Moreover, pain is an important and common symptom of axSpA. It may progress to chronic pain, a more complicated bio-psychosocial phenomena, leading to a significant worsening of quality of life. The development of the axSpA inflammatory process is grounded in the complex interaction between genetic (such as HLA B27), epigenetic, and environmental factors associated with a dysregulated immune response. Considering the pivotal contribution of IL-23 and IL-17 in axSpA inflammation, the inhibition of these cytokines has been evaluated as a potential therapeutic strategy. With this context, here we discuss the main pathogenetic mechanisms, therapeutic approaches and the role of pain in axSpA from the 2022 International GISEA/OEG Symposium.
Collapse
Affiliation(s)
- Fausto Salaffi
- Rheumatology Clinic, Ospedale Carlo Urbani, Università Politecnica delle Marche, 60035 Jesi, Italy
| | - Cesare Siragusano
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, 98125 Messina, Italy
| | - Alessandra Alciati
- Department of Clinical Neurosciences, Hermanas Hospitalarias, Villa San Benedetto Menni Hospital, Como, and Humanitas Clinical and Research Centre, Rozzano, 20089 Milan, Italy
| | - Giulia Cassone
- Rheumatology Unit, Azienda Ospedaliera Policlinico di Modena, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Salvatore D’Angelo
- Rheumatology Institute of Lucania and Rheumatology Department of Lucania, San Carlo Hospital of Potenza, 85100 Potenza, Italy
| | - Serena Guiducci
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Ennio Giulio Favalli
- Division of Clinical Rheumatology, ASST Gaetano Pini-CTO Institute, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, Research Center for Adult and Pediatric Rheumatic Diseases, University of Milan, 20122 Milan, Italy
| | - Fabrizio Conti
- Lupus Clinic, Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari, Sapienza University of Rome, 00161 Rome, Italy
| | - Elisa Gremese
- Rheumatology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Florenzo Iannone
- Rheumatology Unit, Department of Emergency Surgery and Organ Transplantations, University of Bari, 70121 Bari, Italy
| | - Roberto Caporali
- Division of Clinical Rheumatology, ASST Gaetano Pini-CTO Institute, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, Research Center for Adult and Pediatric Rheumatic Diseases, University of Milan, 20122 Milan, Italy
| | - Marco Sebastiani
- Rheumatology Unit, Azienda Ospedaliera Policlinico di Modena, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Correspondence:
| | | | - Giovanni Lapadula
- Rheumatology Unit, Department of Emergency Surgery and Organ Transplantations, University of Bari, 70121 Bari, Italy
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, 98125 Messina, Italy
| |
Collapse
|
20
|
Gladman DD, Mease PJ, Bird P, Soriano ER, Chakravarty SD, Shawi M, Xu S, Quinn ST, Gong C, Leibowitz E, Poddubnyy D, Tam LS, Helliwell PS, Kavanaugh A, Deodhar A, Østergaard M, Baraliakos X. Efficacy and safety of guselkumab in biologic-naïve patients with active axial psoriatic arthritis: study protocol for STAR, a phase 4, randomized, double-blinded, placebo-controlled trial. Trials 2022; 23:743. [PMID: 36064592 PMCID: PMC9444112 DOI: 10.1186/s13063-022-06589-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Axial involvement constitutes a specific domain of psoriatic arthritis (PsA). Interleukin (IL)-23 inhibitors have demonstrated improvement in axial PsA (axPsA) symptoms, but have not shown efficacy in treating ankylosing spondylitis (AS), suggesting differences in axPsA processes and treatments. In a post hoc, pooled analysis of patients with investigator- and imaging-confirmed sacroiliitis in two phase 3, randomized, placebo-controlled studies (DISCOVER-1 and DISCOVER-2), patients treated with guselkumab, an IL-23p19 inhibitor, had greater axial symptom improvements compared with placebo. Confirmatory imaging at baseline was restricted to the sacroiliac (SI) joints, occurred prior to/at screening, and was locally read. METHODS The STAR study will prospectively assess efficacy outcomes in PsA patients with magnetic resonance imaging (MRI)-confirmed axial inflammation. Eligible, biologic-naïve patients with PsA (N = 405) for ≥ 6 months and active disease (≥ 3 swollen and ≥ 3 tender joints, C-reactive protein [CRP] ≥ 0.3 mg/dL) despite prior non-biologic disease-modifying antirheumatic drugs, apremilast, and/or nonsteroidal anti-inflammatory drugs will be randomized (1:1:1) to guselkumab every 4 weeks (Q4W); guselkumab at week (W) 0, W4, then every 8 weeks (Q8W); or placebo with crossover to guselkumab at W24, W28, then Q8W. Patients will have Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) score ≥ 4, spinal pain component score (0-10 visual analog scale) ≥ 4, and screening MRI-confirmed axial involvement (positive spine and/or SI joints according to centrally read Spondyloarthritis Research Consortium of Canada [SPARCC] score ≥ 3 in ≥ 1 region). The primary endpoint is mean change from baseline in BASDAI at W24; multiplicity controlled secondary endpoints at W24 include AS Disease Activity Score employing CRP (ASDAS), Disease Activity Index for PsA (DAPSA), Health Assessment Questionnaire - Disability Index (HAQ-DI), Investigator's Global Assessment of skin disease (IGA), and mean changes from baseline in MRI SI joint SPARCC scores. Centrally read MRIs of spine and SI joints (scored using SPARCC) will be obtained at W0, W24, and W52, with readers blinded to treatment group and timepoint. Treatment group comparisons will be performed using a Cochran-Mantel-Haenszel or chi-square test for binary endpoints and analysis of covariance, mixed model for repeated measures, or constrained longitudinal data analysis for continuous endpoints. DISCUSSION This study will evaluate the ability of guselkumab to reduce both axial symptoms and inflammation in patients with active PsA. TRIAL REGISTRATION This trial was registered at ClinicalTrials.gov, NCT04929210 , on 18 June 2021. PROTOCOL VERSION Version 1.0 dated 14 April 2021.
Collapse
Affiliation(s)
- Dafna D. Gladman
- Centre for Prognosis Studies in The Rheumatic Diseases, Toronto Western Hospital, Toronto, ON Canada
| | - Philip J. Mease
- Swedish Medical Center/Providence St. Joseph Health and University of Washington, Rheumatology Research, Seattle, WA USA
| | - Paul Bird
- University of New South Wales, Randwick, NSW Australia
| | | | - Soumya D. Chakravarty
- Janssen Scientific Affairs, LLC, Horsham, PA USA
- Drexel University College of Medicine, Philadelphia, PA USA
| | - May Shawi
- Immunology Global Medical Affairs, Janssen Pharmaceutical Companies of Johnson & Johnson, Horsham, PA USA
| | - Stephen Xu
- Janssen Research & Development, LLC, Spring House, PA USA
| | | | - Cinty Gong
- Janssen Scientific Affairs, LLC, Horsham, PA USA
| | | | | | - Lai-Shan Tam
- The Chinese University of Hong Kong, Hong Kong, China
| | | | | | - Atul Deodhar
- Oregon Health & Science University, Portland, OR USA
| | | | | |
Collapse
|
21
|
Atzeni F, Siragusano C, Masala IF, Antonio C, Valentina P, D'Angelo S. IL-23 in axial spondyloarthritis and psoriatic arthritis: a good fit for biological treatment? Expert Opin Biol Ther 2022; 22:843-853. [PMID: 35722768 DOI: 10.1080/14712598.2022.2090834] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Interleukin 23 (IL-23) is a pro-inflammatory cytokine that plays a protective role against bacterial and fungal infections. However, the dysregulation of the IL-23/IL-17 axis provides a solid substrate for the development of various inflammatory diseases, such as psoriatic arthritis (PsA) and ankylosing spondylitis (AS). AREAS COVERED In different clinical trials, several drugs against IL-23 have shown efficacy and safety towards PsA, with excellent results on skin and joint scores. However, the same drugs did not show the same efficacy in AS, suggesting that IL-23 may not be a relevant driver of the pathobiology and clinical symptoms of active axial spondyloarthritis (axSpA). EXPERT OPINION These drugs have shown an excellent efficacy and a good safety profile towards PsA, while in AS the efficacy of the IL-23 blockade is lacking for reasons not yet known. Several hypotheses have been reported, but further studies will be needed for a greater understanding. This suggests the involvement of pathways or mechanisms for the development of SpA that remain unknown. In order to allow a wide use of IL-23 inhibitors, further clinical trials and long-term prospective studies are necessary.
Collapse
Affiliation(s)
- Fabiola Atzeni
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Cesare Siragusano
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | | | - Carriero Antonio
- Rheumatology Institute of Lucania (IReL): Rheumatology Department of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy.,PhD Scholarship in Translational and Clinical Medicine, Department of Medicine and Health Sciences, University of Molise, Campobasso, Italy
| | - Picerno Valentina
- Rheumatology Institute of Lucania (IReL): Rheumatology Department of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy
| | - Salvatore D'Angelo
- Rheumatology Institute of Lucania (IReL): Rheumatology Department of Lucania, San Carlo Hospital of Potenza and Madonna delle Grazie Hospital of Matera, Potenza, Italy
| |
Collapse
|
22
|
Mease PJ, Chakravarty SD, McLean RR, Blachley T, Kawashima T, Lin I, Kavanaugh A, Ogdie A. Treatment Responses in Patients With Psoriatic Arthritis Axial Disease According to Human Leukocyte Antigen-B27 Status: An Analysis From the CorEvitas Psoriatic Arthritis/Spondyloarthritis Registry. ACR Open Rheumatol 2022; 4:447-456. [PMID: 35218320 PMCID: PMC9096522 DOI: 10.1002/acr2.11416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/02/2021] [Accepted: 01/09/2022] [Indexed: 11/07/2022] Open
Abstract
OBJECTIVE Axial disease is common and burdensome in patients with psoriatic arthritis (PsA). Human leukocyte antigen-B27 (HLA-B27) is a risk factor for axial PsA; treatment response by HLA-B27 status is inadequately characterized. This study evaluated responses to biologic disease-modifying antirheumatic drugs (bDMARDs) or targeted synthetic DMARDs (tsDMARDs) overall and by HLA-B27 status in patients with PsA axial disease. METHODS This observational study included participants in the CorEvitas (formerly Corrona) PsA/Spondyloarthritis Registry who initiated bDMARD or tsDMARD treatment at baseline, had a 6-month follow-up visit, fulfilled Classification Criteria for Psoriatic Arthritis, had a baseline Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) score of ≥4, and had known HLA-B27 status. Disease characteristics at baseline and 6 months were evaluated overall and by HLA-B27 status. Association between HLA-B27 status and treatment response was evaluated using an analysis of covariance model. RESULTS The analysis included 173 bDMARD or tsDMARD treatment initiations (54 [31.2%] among patients with HLA-B27+ status and 119 [68.8%] among patients with HLA-B27- status). BASDAI total and component scores decreased by ≤0.84 across groups after 6 months of bDMARD or tsDMARD therapy; these changes are not considered clinically meaningful. HLA-B27 status was not statistically significantly associated with changes in axial-related outcomes. CONCLUSION In patients with PsA axial disease, 6 months of bDMARD or tsDMARD therapy provided only mild improvements in axial-related outcomes, irrespective of HLA-B27 status. This continued high disease activity reflects a critical unmet need for focus on the axial domain of PsA and for additional safe and effective therapies for psoriatic axial disease.
Collapse
Affiliation(s)
- Philip J. Mease
- Swedish Medical Center/Providence St. Joseph Health and University of Washington School of MedicineSeattle
| | - Soumya D. Chakravarty
- Janssen Scientific Affairs, LLC, Horsham, Pennsylvania, and Drexel University College of MedicinePhiladelphiaPennsylvania
| | | | | | | | - Iris Lin
- Janssen Scientific Affairs, LLCHorshamPennsylvania
| | | | - Alexis Ogdie
- University of Pennsylvania School of MedicinePhiladelphia
| |
Collapse
|
23
|
Gladman DD, Mease PJ, Bird P, Soriano ER, Chakravarty SD, Shawi M, Lavie F, Gong C, Leibowitz E, Poddubnyy D, Tam LS, Helliwell PS, Kavanaugh A, Deodhar AA, Østergaard M, Baraliakos X. Correspondence on 'No efficacy of anti-IL-23 therapy for axial spondyloarthritis in randomised controlled trials but in post-hoc analyses of psoriatic arthritis-related 'physician-reported spondylitis'?' by Braun and Landewé. Ann Rheum Dis 2022:annrheumdis-2022-222161. [PMID: 35487679 DOI: 10.1136/annrheumdis-2022-222161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 11/04/2022]
Affiliation(s)
- Dafna D Gladman
- Division of Rheumatology, University of Toronto and Toronto Western Hospital, Toronto, Ontario, Canada
| | - Philip J Mease
- Rheumatology Research, Swedish Medical Center/Providence St. Joseph Health and University of Washington, Seattle, Washington, USA
| | - Paul Bird
- Rheumatology, University of New Sounth Wales, Sydney, New South Wales, Australia
| | - Enrique R Soriano
- Rheumatology, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Soumya D Chakravarty
- Immunology, Janssen Scientific Affairs LLC, Horsham, Pennsylvania, USA.,Rheumatology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - May Shawi
- Immunology Global Medical Affairs, Janssen Pharmaceutical Companies of Johnson & Johnson, Horsham, PA, USA
| | - Frederic Lavie
- Immunology Global Medical Affairs, Janssen Pharmaceutical Companies of Johnson & Johnson, Paris, France
| | - Cinty Gong
- Immunology, Janssen Scientific Affairs LLC, Horsham, Pennsylvania, USA
| | - Evan Leibowitz
- Immunology, Janssen Scientific Affairs LLC, Horsham, Pennsylvania, USA
| | - Denis Poddubnyy
- Division of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Lai-Shan Tam
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Philip S Helliwell
- Academic Unit of Musculoskeletal Medicine, University of Leeds, School of Medicine, Leeds, UK
| | - Arthur Kavanaugh
- Department of Medicine, School of Medicine, University of California at San Diego, La Jolla, California, USA
| | - Atul A Deodhar
- Division of Arthritis/Rheumatic Diseases (OPO9), Oregon Health & Science University, Portland, Oregon, USA
| | - Mikkel Østergaard
- Copenhagen Center for Arthritis Research, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
24
|
Lenti MV, Dolby V, Clark T, Hall V, Tattersall S, Fairhurst F, Kenneth C, Walker R, Kemp K, Borg‐Bartolo S, Limdi JK, Taylor J, Townsend T, Subramanian S, Storey D, Assadsangabi A, Stansfield C, Smith P, Byrne D, De Silvestri A, Selinger C. A propensity score-matched, real-world comparison of ustekinumab vs vedolizumab as a second-line treatment for Crohn's disease. The Cross Pennine study II. Aliment Pharmacol Ther 2022; 55:856-866. [PMID: 34935160 PMCID: PMC9305775 DOI: 10.1111/apt.16742] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/06/2021] [Accepted: 12/09/2021] [Indexed: 12/20/2022]
Abstract
BACKGROUND The optimal choice of biological agents after failure of anti-tumour-necrosis-factor-(TNF)α agent in Crohn's disease (CD) is yet to be defined. AIMS To assess the effectiveness and safety of ustekinumab compared to vedolizumab as second-line treatment in CD patients who failed anti-TNFα therapy. METHODS Retrospective analysis of clinical response and remission at 14 and 52 weeks to ustekinumab by physician global assessment (PGA). A propensity score-matched analysis with a cohort treated with vedolizumab was performed. RESULTS Of 282 patients (mean age 40 ± 15, F:M ratio 1.7:1) treated with ustekinumab, clinical response or remission was reached by 200/282 patients (70.9%) at 14 weeks, and 162/259 patients (62.5%) at 52 weeks. Overall, 74 adverse events occurred, of which 26 were labelled as serious (8.3 per 100 person-year). After exclusion of patients without prior anti-TNFα exposure and patients previously exposed to vedolizumab or ustekinumab, we analysed 275/282 patients (97.5%) on ustekinumab and 118/135 patients (87.4%) on vedolizumab. Propensity score analysis revealed that at 14 weeks, patients treated with ustekinumab were 38% (95% CI 25%-50%; P < 0.001) more likely to achieve clinical remission, while at 52 weeks, the difference of 9% (95% CI -15% to 33%; P = 0.462) was not significant. CONCLUSIONS Ustekinumab was effective and well tolerated in this real-world cohort. While ustekinumab proved more effective at 14-weeks, we found no statistically significant differences at 52 weeks compared to vedolizumab.
Collapse
|
25
|
Lei C, Jiang J, Zhang Y, Xiong G. Role and Function of Regulatory T Cell in Chronic Rhinosinusitis with Nasal Polyposis. J Immunol Res 2022; 2022:1144563. [PMID: 35378904 PMCID: PMC8976649 DOI: 10.1155/2022/1144563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/28/2022] [Accepted: 03/14/2022] [Indexed: 11/21/2022] Open
Abstract
Chronic rhinosinusitis with nasal polyps (CRSwNP) is a subtype of chronic rhinosinusitis characterized by high edema in the stroma, albumin deposition, and formation of pseudocysts. The pathogenesis of CRSwNP is not yet fully understood. Regulatory T (Treg) cells are a subset of CD4+ T cells that play a suppressive immunoregulatory role in the process of CRSwNP. Recent studies have found that there was a significant reduction in Treg cells in polyp tissues, which leads to the onset of CRSwNP. An imbalance between Th17 and Treg cells can also aggravate inflammation toward the Th2 type. This review focuses on our understanding of the function and role of Treg cells and their regulatory factors and clinical significance in CRSwNP. We also summarize the current drug treatments for CRSwNP with Tregs as the potential therapeutic target, which will provide new ideas for the treatment of CRSwNP in the future.
Collapse
Affiliation(s)
- Chenyang Lei
- Department of Otorhinolaryngology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Juan Jiang
- Department of Otorhinolaryngology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Yanyan Zhang
- Department of Otorhinolaryngology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Gaoyun Xiong
- Department of Otorhinolaryngology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| |
Collapse
|
26
|
Miyagawa I, Tanaka Y. Dawn of Precision Medicine in Psoriatic Arthritis. Front Med (Lausanne) 2022; 9:851892. [PMID: 35372404 PMCID: PMC8973395 DOI: 10.3389/fmed.2022.851892] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/22/2022] [Indexed: 01/01/2023] Open
Abstract
The establishment of precision medicine is considered particularly important in heterogeneous autoimmune diseases (e.g., psoriatic arthritis, systemic lupus erythematosus), which reveal clinical and molecular heterogeneity. The selection of optimal treatment strategies for individual patients may be more important and complex in autoimmune diseases than in other diseases. Two factors are important in precision medicine: patient stratification and use of targeted. When both factors work, patients are likely to have good outcomes. However, research into precision medicine and its practice in systemic autoimmune diseases is lacking. In contrast, the usefulness of peripheral immune cell phenotyping in the evaluation of immunological characteristics and stratification into subgroups of individual patients with systemic autoimmune diseases such as immunoglobulin 4-related disease, systemic lupus erythematosus, and anti-neutrophil cytoplasmic antibody-related vasculitis was reported. Furthermore, the potential of precision medicine using biological disease-modifying antirheumatic drugs based on peripheral immune cell phenotyping was recently demonstrated for psoriatic arthritis in the clinical setting. Precision medicine has not yet been sufficiently investigated in real world clinical settings. However, a dawn of precision medicine has emerged. We should shed further light on precision medicine in PsA and other autoimmune diseases. Here, we first review the usefulness of peripheral immune cell phenotyping in systemic autoimmune diseases and the potential of precision medicine in PsA based on this method.
Collapse
|
27
|
Rakowsky S, Papamichael K, Cheifetz AS. Choosing the right biologic for complications of inflammatory bowel disease. Expert Rev Gastroenterol Hepatol 2022; 16:235-249. [PMID: 35094628 DOI: 10.1080/17474124.2022.2036122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Inflammatory bowel disease (IBD) is a chronic, inflammatory condition that involves the intestinal tract, and can also present with extra-intestinal manifestations (EIM). Choosing the right treatment for IBD is often nuanced and decisions can become even more complicated when a patient presents with or develops a complication of the disease. AREAS COVERED We aimed to provide an overview of the most common complications of IBD, specifically intestinal and EIM, and summarize the data regarding biologic therapy for treatment of these conditions. A comprehensive literature review was performed using PubMed and Medline databases to identify studies published in the English language relevant to the broad scope of this review. EXPERT OPINION There are still significant gaps in our understanding of the pathophysiology of IBD and its treatment, especially in regards to complications of the disease. As novel therapies continue to emerge for treatment of IBD, we feel concurrent examination of their impact on intestinal complications and EIM of IBD is important and should be a priority of future research.
Collapse
Affiliation(s)
- Shana Rakowsky
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA USA
| | - Konstantinos Papamichael
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA USA
| | - Adam S Cheifetz
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA USA
| |
Collapse
|
28
|
Gialouri CG, Evangelatos G, Fragoulis GE. Choosing the Appropriate Target for the Treatment of Psoriatic Arthritis: TNFα, IL-17, IL-23 or JAK Inhibitors? Mediterr J Rheumatol 2022; 33:150-161. [PMID: 36127928 PMCID: PMC9450184 DOI: 10.31138/mjr.33.1.150] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/25/2021] [Accepted: 11/15/2021] [Indexed: 02/05/2023] Open
Abstract
Psoriatic arthritis (PsA) is a highly heterogenous disease. Apart from arthritis and psoriasis, other manifestations can also occur, including enthesitis, dactylitis, axial-, nail-, eye- and bowel- involvement. Comorbidities are also frequent in the setting of PsA, with cardiovascular disease and mental-health disorders being the most frequent. The Rheumatologist's arsenal has many different treatment options for treating PsA. Despite their effectiveness, there are some differences in terms of efficacy and safety that might affect clinician's decision for one or the other drug. Comparing biologic DMARDs and JAK-inhibitors, one could say that they have similar effectiveness in terms of musculoskeletal manifestations. However, anti-IL-17 and anti-IL-23 drugs seem to be more effective for skin manifestations. In contrast, JAK-inhibitors and etanercept might be less effective for these manifestations. Inflammatory bowel disease and uveitis are non-responsive to etanercept and anti-IL-17 drugs. As regards to comorbidities, data are scarce, but future studies will shed light on possible differential effect of bDMARDs or JAK-inhibitors. Safety is always an important drive for choosing the appropriate treatment. Infections are the most common adverse event of these drugs. Etanercept and anti-IL-17 drugs are safer for patients having latent tuberculosis, while herpes zoster is more common in individuals receiving JAK-inhibitors. Finally, venous thromboembolism risk, should be taken into account when JAK-inhibitors are used. In this review, we comparatively present, as outlined above, the various aspects that could affect the choice of the appropriate bDMARD or JAK-inhibitor for the treatment of a PsA patient.
Collapse
Affiliation(s)
- Chrysoula G. Gialouri
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens,“Laiko” General Hospital, Athens, Greece
| | - Gerasimos Evangelatos
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens,“Laiko” General Hospital, Athens, Greece
| | - George E. Fragoulis
- First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens,“Laiko” General Hospital, Athens, Greece
- Corresponding Author: George E. Fragoulis, MD, PhD Rheumatology Unit, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens Laiko General Hospital, Mikras Asias 75 Str, 11527 Athens, Greece, Tel.: +30 210 746 2636, E-mail:
| |
Collapse
|
29
|
Zeidler HK. Psoriatic Spondylitis: A Disease Manifestation in Debate: Evidences to Know for the Clinical Rheumatologist. J Clin Rheumatol 2022; 28:38-43. [PMID: 34941618 DOI: 10.1097/rhu.0000000000001815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT With the advent of classification criteria for psoriatic arthritis (PsA) and axial spondyloarthritis (axSpA), patients with axial manifestations associated with psoriasis, initially described in the l950s as a specific entity termed psoriatic spondylitis (PS), are now categorized within PsA, ankylosing spondylitis (AS), and axSpA. Thus, different terms are used to describe axial disease in patients with PsA including PS, axial psoriatic arthritis (axPsA), and psoriatic spondyloarthritis. Patients with PS may present with inflammatory and/or mechanical back pain, but also may display axial disease on imaging despite not complaining of back pain. Cervical spondylitis has been reported in 35% to 75% of patients with PsA. Axial disease is silent in 20% and 25% of patients with axial PsA and PsA, respectively. The majority of axPsA patients have peripheral arthritis alongside the axial involvement, whereas only 2% to 5% of PsA patients have solely axial arthritis with no peripheral arthritis.A debate is currently underway as to whether inflammatory axial disease and psoriasis represent axSpA with psoriasis or a subset of PsA named axPsA. Studies have recognized that axial disease in PsA patients seems to be different demographically, genetically, clinically, and radiographically when compared with AS with or without psoriasis. This narrative review summarizes current knowledge regarding axial involvement of PsA in terms of history, terminology, classification, epidemiology, clinical presentation, imaging, diagnosis, and treatment, with the aim of providing advice for management of PS in clinical evidence-based practice. Data-driven studies are needed to develop clear, nonoverlapping classification criteria for spinal involvement in PsA.
Collapse
|
30
|
Findeisen KE, Östör AJK. The Role of Interleukin-23 Inhibition in the Treatment of Psoriatic Arthritis. Rheumatology (Oxford) 2022. [DOI: 10.17925/rmd.2022.1.1.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Psoriatic arthritis is a chronic inflammatory autoimmune disease with varied manifestations, including functional limitation and reduced quality of life. Improved understanding of the immunopathogenesis of psoriatic arthritis, particularly the role of the interleukin-23 and interleukin-17 axis, has led to the development of therapeutic targets to alter the natural history of the condition. In this article, we review the role of monoclonal antibodies targeting the p19 subunit of interleukin-23 in the treatment of psoriatic arthritis.
Collapse
|
31
|
Hoffmann T, Oelzner P, Busch M, Franz M, Teichgräber U, Kroegel C, Schulze PC, Wolf G, Pfeil A. Organ Manifestation and Systematic Organ Screening at the Onset of Inflammatory Rheumatic Diseases. Diagnostics (Basel) 2021; 12:67. [PMID: 35054234 PMCID: PMC8774450 DOI: 10.3390/diagnostics12010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/20/2021] [Accepted: 12/25/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Inflammatory rheumatic diseases (IRD) are often associated with the involvement of various organs. However, data regarding organ manifestation and organ spread are rare. To close this knowledge gap, this cross-sectional study was initiated to evaluate the extent of solid organ manifestations in newly diagnosed IRD patients, and to present a structured systematic organ screening algorithm. MATERIALS AND METHODS The study included 84 patients (63 women, 21 men) with newly diagnosed IRD. None of the patients received any rheumatic therapy. All patients underwent a standardised organ screening programme encompassing a basic screening (including lungs, heart, kidneys, and gastrointestinal tract) and an additional systematic screening (nose and throat, central and peripheral nervous system) on the basis of clinical, laboratory, and immunological findings. RESULTS Represented were patients with connective tissue diseases (CTD) (72.6%), small-vessel vasculitis (16.7%), and myositis (10.7%). In total, 39 participants (46.5%) had one or more organ manifestation(s) (one organ, 29.7%; two organs, 10.7%; ≥three organs, 6.0%). The most frequently involved organs were the lungs (34.5%), heart (11.9%), and kidneys (8.3%). Lastly, a diagnostic algorithm for organ manifestation was applied. CONCLUSION One-half of the patients presented with a solid organ involvement at initial diagnosis of IRD. Thus, in contrast to what has been described in the literature, organ manifestations were already present in a high proportion of patients at the time of diagnosis of IRD rather than after several years of disease. Therefore, in IRD patients, systematic organ screening is essential for treatment decisions.
Collapse
Affiliation(s)
- Tobias Hoffmann
- Department of Internal Medicine III, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (P.O.); (M.B.); (G.W.); (A.P.)
| | - Peter Oelzner
- Department of Internal Medicine III, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (P.O.); (M.B.); (G.W.); (A.P.)
| | - Martin Busch
- Department of Internal Medicine III, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (P.O.); (M.B.); (G.W.); (A.P.)
| | - Marcus Franz
- Department of Internal Medicine I, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (M.F.); (P.C.S.)
| | - Ulf Teichgräber
- Institute of Diagnostic and Interventional Radiology, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany;
| | - Claus Kroegel
- Department of Internal Medicine I, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (M.F.); (P.C.S.)
| | - Paul Christian Schulze
- Department of Internal Medicine I, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (M.F.); (P.C.S.)
| | - Gunter Wolf
- Department of Internal Medicine III, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (P.O.); (M.B.); (G.W.); (A.P.)
| | - Alexander Pfeil
- Department of Internal Medicine III, Jena University Hospital, Friedrich Schiller University Jena, Am Klinikum 1, 07747 Jena, Germany; (P.O.); (M.B.); (G.W.); (A.P.)
| |
Collapse
|
32
|
Ishchenko A, Joly J, Neerinckx B, Lories R, de Vlam K. Evolution of patient characteristics in the era of biologic treatment of psoriatic arthritis: 18-year Belgian experience from the Leuven Spondyloarthritis Biologics Cohort (BioSPAR). Rheumatol Adv Pract 2021; 5:rkab085. [PMID: 34888434 PMCID: PMC8651220 DOI: 10.1093/rap/rkab085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/22/2021] [Indexed: 11/13/2022] Open
Abstract
Objectives Biologic treatments have revolutionized the management of PsA by significantly improving clinical manifestations and preventing structural damage. Both result in better quality of life and improved physical functioning. Since the introduction of the first TNF inhibitor (TNFi) in the early 2000s, therapeutic options for PsA are increasing steadily, and a new generation of biologics, including anti-IL-17 and anti-IL-23 strategies, allows distinct targeted approaches. The purpose of this study was to investigate whether the demographic, clinical and disease characteristics of PsA patients who are selected for first-line biologic treatment has changed over time since the introduction of biologics. Methods Patients with a clinical diagnosis of PsA were included in the KU Leuven BioSPAR registry, a prospective cohort of SpA and PsA patients treated with biologics and targeted synthetic DMARDs (tsDMARDs), such as apremilast and Janus kinase inhibitors. Demographics, prior DMARD use, disease characteristics and disease activity parameters were recorded at the initiation of biologic treatment and subsequently every 3 months for the first 2 years and later every 6 months. The patient data were compared in three treatment periods, corresponding to availability of the first and second generation of TNFi and the third generation of biologics. Results Analysis of 185 Caucasian patients with PsA from our prospective cohort showed longer disease duration and higher disease activity, with higher tender joint count, swollen joint count and CRP in the first period compared with the later time periods. The demographic characteristics and prior DMARD use did not change over time. Skin and nail psoriasis were more frequent in earlier compared with the later treatment periods. The bio-DMARD survival rate was similar in the early and later treatment periods. Conclusion The population of patients selected for treatment escalation has changed over time since the introduction of biologics. Our results suggest that with years of experience, PsA patients might be considered earlier and for therapy intensification in patients with less active disease in comparison to profiles in the early days of biologic treatment.
Collapse
Affiliation(s)
- Alla Ishchenko
- Department of Rheumatology, University Hospitals Leuven.,Skeletal Biology and Engineering Research Centre, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Johan Joly
- Skeletal Biology and Engineering Research Centre, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Barbara Neerinckx
- Department of Rheumatology, University Hospitals Leuven.,Skeletal Biology and Engineering Research Centre, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Rik Lories
- Department of Rheumatology, University Hospitals Leuven.,Skeletal Biology and Engineering Research Centre, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Kurt de Vlam
- Department of Rheumatology, University Hospitals Leuven.,Skeletal Biology and Engineering Research Centre, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
33
|
Braun J, Landewé RB. No efficacy of anti-IL-23 therapy for axial spondyloarthritis in randomised controlled trials but in post-hoc analyses of psoriatic arthritis-related 'physician-reported spondylitis'? Ann Rheum Dis 2021; 81:466-468. [PMID: 34656991 DOI: 10.1136/annrheumdis-2021-221422] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/07/2021] [Indexed: 12/24/2022]
Abstract
The three monoclonal antibodies ustekinumab, guselkumab and risankizumab targeting the p 40 or the 19 subunit of interleukin -23 have now been approved for the indication psoriasis and the former two also for psoriatic arthritis (PsA). Ustekinumab and risankizumab have appeared ineffective in randomised controlled trials with patients with axial spondyloarthritis (axSpA), but post-hoc analyses of PsA trials have now suggested that they may improve back pain symptoms potentially induced by axial inflammation based on PsA. Here we argue that, based on the absence of efficacy in axSpA, this is unlikely and more probably due to generic, non-specific effects, which are not adequately covered by the tools developed for the assessment of inflammation in axSpA.
Collapse
Affiliation(s)
- Juergen Braun
- Rheumazentrum Ruhrgebiet, Ruhr University Bochum, Herne, Germany
| | - Robert Bm Landewé
- Amsterdam Rheumatology Center, AMC, Amsterdam, The Netherlands.,Rheumatology, Zuyderland Medical Center, Heerlen, The Netherlands
| |
Collapse
|
34
|
Bianchi E, Vecellio M, Rogge L. Editorial: Role of the IL-23/IL-17 Pathway in Chronic Immune-Mediated Inflammatory Diseases: Mechanisms and Targeted Therapies. Front Immunol 2021; 12:770275. [PMID: 34630440 PMCID: PMC8496837 DOI: 10.3389/fimmu.2021.770275] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 01/01/2023] Open
Affiliation(s)
- Elisabetta Bianchi
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France
- Unité Mixte de Recherche, Institut Pasteur/AP-HP Hôpital Cochin, Paris, France
| | - Matteo Vecellio
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| | - Lars Rogge
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Paris, France
- Unité Mixte de Recherche, Institut Pasteur/AP-HP Hôpital Cochin, Paris, France
| |
Collapse
|
35
|
Abstract
Psoriatic arthritis (PsA) is a complex inflammatory disease with heterogeneous clinical features, which complicates psoriasis in 30% of patients. There are no diagnostic criteria or tests available. Diagnosis is most commonly made by identifying inflammatory musculoskeletal features in joints, entheses or the spine in the presence of skin and/or nail psoriasis and in the usual absence of rheumatoid factor and anti-cyclic citrullinated peptide. The evolution of psoriasis to PsA may occur in stages, although the mechanisms are unclear. In many patients, there may be little or no relationship between severity of musculoskeletal inflammation and severity of skin or nail psoriasis. The reason for this disease heterogeneity may be explained by differences in genotype, especially in the HLA region. New targeted therapies for PsA have been approved with additional therapies in development. These developments have substantially improved both short-term and long-term outcomes including a reduction in musculoskeletal and skin manifestations and in radiographic damage. With efforts underway aimed at improving our understanding of the molecular basis for the heterogeneity of PsA, a personalized approach to treating PsA may become possible.
Collapse
|
36
|
Kaeley GS, Kaler JK. Peripheral Enthesitis in Spondyloarthritis: Lessons from Targeted Treatments. Drugs 2021; 80:1419-1441. [PMID: 32720292 DOI: 10.1007/s40265-020-01352-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A significant proportion of patients with spondyloarthritis (SpA) have peripheral enthesitis. Data suggest that psoriatic arthritis (PsA) patients with enthesitis have a higher disease burden than those without enthesitis. Over the past decade, there has been a proliferation of treatment options for spondyloarthropathy. These medications target multiple signaling pathways, including tumor necrosis factor (TNF), interleukin (IL)-17A, IL-12/23, IL-23, thymus (T)-cell co-stimulation, intracellular Janus kinases, and phosphodiesterase enzymes. As a key domain in SpA, enthesitis outcomes are included in pivotal trials of these agents and are reported as secondary outcome measures. One significant limitation is that the clinical evaluation of enthesitis relies on eliciting tenderness on palpation and is insensitive when compared with imaging. Furthermore, direct comparisons between studies are not available due to the use of different outcome measures, lack of consistent and comprehensive reporting outcomes, and subgroup analyses with a lower number of patients with enthesitis. This systematic review describes the epidemiology, pathophysiology, and available targeted therapies against enthesitis, as well as a detailed report of their efficacy. One major trend identified during this review is incomplete reporting of outcome measures, as many studies reported proportions of enthesitis prevalence. Factors that affected responsiveness in clinical trials included the entheseal instrument used, the number of subjects available for comparison, as well as the therapeutic agent. In general, anti-TNF and anti-IL-17 agents, as well as Janus kinase inhibitors, show moderate responsiveness for enthesitis. The data for IL-23 targeting is contradictory.
Collapse
Affiliation(s)
- Gurjit S Kaeley
- Division of Rheumatology, University of Florida College of Medicine Jacksonville, 653-1 West Eight Street, LRC 2nd Floor L-14, Jacksonville, FL, 32209-6561, USA.
| | - Jaspreet K Kaler
- Division of Rheumatology, University of Florida College of Medicine Jacksonville, 653-1 West Eight Street, LRC 2nd Floor L-14, Jacksonville, FL, 32209-6561, USA
| |
Collapse
|
37
|
Floris A, Congia M, Chessa E, Angioni MM, Piga M, Cauli A. Targeted Therapies in Axial Psoriatic Arthritis. Front Genet 2021; 12:689984. [PMID: 34262600 PMCID: PMC8273289 DOI: 10.3389/fgene.2021.689984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/25/2021] [Indexed: 11/13/2022] Open
Abstract
Specific and high-quality evidence on the efficacy of the current targeted therapies for axial disease in psoriatic arthritis (axPsA) is still scarce. Indeed, almost all the cohorts investigated in clinical trials on PsA consisted of patients with peripheral arthritis, where a small number of them also had axial involvement. Only one randomized controlled trial was so far specifically designed to assess the efficacy of a biological disease-modifying antirheumatic drug (DMARD) in axPsA. For other biological and synthetic targeted DMARDs, the most specific evidence for treatment in axPsA is extrapolated from post-hoc analyses based on PsA patients with concomitant peripheral and axial manifestations. Furthermore, the current trials and post-hoc analysis on axPsA are affected by major limitations, including the lack of a widely accepted definition of axPsA and the lack of specific and validated outcome measures. Finally, poor data are available on the genetics of axPsA, although alleles differentially expressed in different patterns of axPsA might offer advantages in the prospective of personalized medicine in axPsA patients. Overall, this review suggests that there is an urgent need for more reliable evidence derived from studies specifically designed for axPsA and based on a validated definition of axPsA and on specific outcome measures.
Collapse
Affiliation(s)
- Alberto Floris
- Unità Operativa Complessa di Reumatologia, Dipartimento di Scienze Mediche e Sanità Pubblica, Azienda Ospedaliero-Universitaria e Università di Cagliari, Monserrato, Italy
| | | | | | | | | | | |
Collapse
|
38
|
Fisher C, Ciurtin C, Leandro M, Sen D, Wedderburn LR. Similarities and Differences Between Juvenile and Adult Spondyloarthropathies. Front Med (Lausanne) 2021; 8:681621. [PMID: 34136509 PMCID: PMC8200411 DOI: 10.3389/fmed.2021.681621] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
Spondyloarthritis (SpA) encompasses a broad spectrum of conditions occurring from childhood to middle age. Key features of SpA include axial and peripheral arthritis, enthesitis, extra-articular manifestations, and a strong association with HLA-B27. These features are common across the ages but there are important differences between juvenile and adult onset disease. Juvenile SpA predominantly affects the peripheral joints and the incidence of axial arthritis increases with age. Enthesitis is important in early disease. This review article highlights the similarities and differences between juvenile and adult SpA including classification, pathogenesis, clinical features, imaging, therapeutic strategies, and disease outcomes. In addition, the impact of the biological transition from childhood to adulthood is explored including the importance of musculoskeletal and immunological maturation. We discuss how the changes associated with adolescence may be important in explaining age-related differences in the clinical phenotype between juvenile and adult SpA and their implications for the treatment of juvenile SpA.
Collapse
Affiliation(s)
- Corinne Fisher
- Centre for Adolescent Rheumatology Versus Arthritis at University College London, University College London Hospital and Great Ormond Street Hospital, London, United Kingdom.,Department of Adolescent Rheumatology, University College London Hospitals NHS Foundation Trust, London, United Kingdom.,National Institute for Health Research University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Coziana Ciurtin
- Centre for Adolescent Rheumatology Versus Arthritis at University College London, University College London Hospital and Great Ormond Street Hospital, London, United Kingdom.,Department of Adolescent Rheumatology, University College London Hospitals NHS Foundation Trust, London, United Kingdom.,Division of Medicine, Department of Rheumatology (Bloomsbury), University College London, London, United Kingdom
| | - Maria Leandro
- Centre for Adolescent Rheumatology Versus Arthritis at University College London, University College London Hospital and Great Ormond Street Hospital, London, United Kingdom.,Department of Adolescent Rheumatology, University College London Hospitals NHS Foundation Trust, London, United Kingdom.,Division of Medicine, Department of Rheumatology (Bloomsbury), University College London, London, United Kingdom
| | - Debajit Sen
- Centre for Adolescent Rheumatology Versus Arthritis at University College London, University College London Hospital and Great Ormond Street Hospital, London, United Kingdom.,Department of Adolescent Rheumatology, University College London Hospitals NHS Foundation Trust, London, United Kingdom.,National Institute for Health Research University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Lucy R Wedderburn
- Centre for Adolescent Rheumatology Versus Arthritis at University College London, University College London Hospital and Great Ormond Street Hospital, London, United Kingdom.,National Institute for Health Research Biomedical Research Centre at Great Ormond Street Hospital for Children, London, United Kingdom.,Infection, Immunity & Inflammation Teaching and Research Department University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| |
Collapse
|
39
|
Management of particular clinical situations in psoriatic arthritis: an expert's recommendation document based on systematic literature review and extended Delphi process. Rheumatol Int 2021; 41:1549-1565. [PMID: 33934175 PMCID: PMC8316175 DOI: 10.1007/s00296-021-04877-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023]
Abstract
To establish practical recommendations for the management of patients with psoriatic arthritis (PsA) with particular clinical situations that might lead to doubts in the pharmacological decision-making. A group of six expert rheumatologists on PsA identified particular clinical situations in PsA. Then, a systematic literature review (SLR) was performed to analyse the efficacy and safety of csDMARDs, b/tsDMARDs in PsA. In a nominal group meeting, the results of the SLR were discussed and a set of recommendations were proposed for a Delphi process. A total of 65 rheumatologists were invited to participate in the Delphi. Agreement was defined if ≥ 70% of the participants voted ≥ 7 (from 1, totally disagree to 10, totally agree). For each recommendation, the level of evidence and grade of recommendation was established based on the Oxford Evidence-Based Medicine categorisation. Particular clinical situations included monoarthritis, axial disease, or non-musculoskeletal manifestations. The SLR finally comprised 131 articles. A total of 16 recommendations were generated, all but 1 reached consensus. According to them, it is crucial to carefully analyse the impact of individual manifestations on patients (disability, quality of life, etc.), but also to recognise the impact of each drug singularities on selected clinical phenotypes to adopt the most appropriate treatment strategy. Early diagnosis and treatment to target approach, along with a close risk management, is also necessary. These recommendations are intended to complement gaps in national and international guidelines by helping health professionals address and manage particular clinical situations in PsA.
Collapse
|
40
|
Helliwell PS, Gladman DD, Chakravarty SD, Kafka S, Karyekar CS, You Y, Campbell K, Sweet K, Kavanaugh A, Gensler LS. Effects of ustekinumab on spondylitis-associated endpoints in TNFi-naïve active psoriatic arthritis patients with physician-reported spondylitis: pooled results from two phase 3, randomised, controlled trials. RMD Open 2021; 6:rmdopen-2019-001149. [PMID: 32209721 PMCID: PMC7046941 DOI: 10.1136/rmdopen-2019-001149] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/07/2020] [Accepted: 01/10/2020] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND The interleukin-12/23p40-subunit-inhibitor ustekinumab significantly improved spondylitis-related symptoms through Week 24 in psoriatic arthritis (PsA) patients with peripheral arthritis and physician-reported spondylitis (PA-PRS) in PSUMMIT-1&2. We further evaluated ustekinumab's effect on spondylitis-related endpoints in PSUMMIT-1&2 tumour necrosis factor-inhibitor (TNFi)-naïve patients with PA-PRS. METHODS Patients with active PsA (≥5 swollen and ≥5 tender joints, C-reactive-protein ≥ 3.0 mg/L) despite conventional (PSUMMIT-1&2) and/or prior TNFi (PSUMMIT-2) therapy received subcutaneous ustekinumab 45 mg, 90 mg or placebo (Week 0, Week 4, Week 16). Changes in Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) neck/back/hip pain question (#2) and modified BASDAI (mBASDAI, excluding PA) scores and Ankylosing Spondylitis Disease Activity Score (ASDAS) responses were assessed at Weeks 12 and 24. RESULTS The pooled PSUMMIT-1&2, TNFi-naïve (n=747), PA-PRS (n=223) subset (158 with human-leucocyte-antigen (HLA)-B27 results) presented with moderate-to-severe spondylitis-related symptoms (mean BASDAI-neck/back/hip pain-6.51, mBASDAI-6.54, BASDAI-6.51, ASDAS-3.81). Mean Week 24 changes were larger among ustekinumab than placebo-treated patients for both neck/back/hip pain (-1.99 vs -0.18) and mBASDAI (-2.09 vs -0.59). Improvements in neck/back/hip pain and fatigue appeared numerically greater in HLA-B27+ than HLA-B27 - patients; those for other domains were generally consistent. Greater proportions of ustekinumab versus placebo-treated patients achieved ASDAS clinically important improvement at Week 24 (decrease ≥ 1.1; 49.6% vs 12.7%; nominal p<0.05). CONCLUSIONS Improvements in BASDAI neck/back/hip pain and mBASDAI among ustekinumab-treated, TNFi-naïve, PsA patients with PA-PRS were clinically meaningful and consistent across assessment tools. Numerically greater improvements in neck/back/hip pain in HLA-B27+ than HLA-B27 - patients, noted in the context of similar overall mBASDAI improvements between the subgroups, suggest ustekinumab may improve disease activity in TNFi-naïve PsA patients likely to exhibit axial disease. CLINICAL TRIAL REGISTRATION NUMBERS PSUMMIT 1, NCT01009086; PSUMMIT 2, NCT01077362.
Collapse
Affiliation(s)
- Philip S Helliwell
- Section of Musculoskeletal Disease, NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds, UK
| | - Dafna D Gladman
- Krembil Institute, University of Toronto, Toronto, Ontario, Canada
| | - Soumya D Chakravarty
- Janssen Scientific Affairs LLC, Horsham, Pennsylvania, USA.,Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Shelly Kafka
- Janssen Scientific Affairs LLC, Horsham, Pennsylvania, USA
| | | | - Yin You
- Janssen Research & Development LLC, Spring House, Pennsylvania, USA
| | - Kim Campbell
- Janssen Research & Development LLC, Spring House, Pennsylvania, USA
| | - Kristen Sweet
- Janssen Research & Development LLC, Spring House, Pennsylvania, USA
| | | | - Lianne S Gensler
- University of California San Francisco School of Medicine, San Francisco, California, USA
| |
Collapse
|
41
|
Greuter T, Rieder F, Kucharzik T, Peyrin-Biroulet L, Schoepfer AM, Rubin DT, Vavricka SR. Emerging treatment options for extraintestinal manifestations in IBD. Gut 2021; 70:796-802. [PMID: 32847845 PMCID: PMC9014274 DOI: 10.1136/gutjnl-2020-322129] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/20/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022]
Abstract
Extraintestinal manifestations (EIMs) are frequently observed in IBDs and contribute considerably to morbidity and mortality. They have long been considered a difficult to treat entity due to limited therapy options, but the increasing use of anti-tumour necrosis factors has dramatically changed the therapeutic approach to EIM in recent years. Newly emerging therapies such as JAK inhibitors and anti-interleukin 12/23 will further shape the available armamentarium. Clinicians dealing with EIMs in everyday IBD practice may be puzzled by the numerous available biological agents and small molecules, their efficacy for EIMs and their potential off-label indications. Current guidelines on EIMs in IBD do not include treatment algorithms to help practitioners in the treatment decision-making process. Herein, we summarise knowledge on emerging biological treatment options and small molecules for EIMs, highlight current research gaps, provide therapeutic algorithms for EIM management and shed light on future strategies in the context of IBD-related EIMs.
Collapse
Affiliation(s)
- Thomas Greuter
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland .,Department of Internal Medicine, GZO - Zurich Regional Health Center, Wetzikon, Switzerland
| | - Florian Rieder
- Division of Gastroenterology, Hepatology & Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Torsten Kucharzik
- Division of Gastroenterology and Hepatology, Klinikum Lueneburg, Lueneburg, Germany
| | - Laurent Peyrin-Biroulet
- Inserm U954, Department of Hepato-Gastroenterology, University Hospital of Nancy, Université Henri Poincaré 1, Vandoeuvre-lès-Nancy, France
| | - Alain M Schoepfer
- Division of Gastroenterology and Hepatology, University Hospital Lausanne – CHUV, Lausanne, Switzerland,University of Lausanne, Lausanne, Switzerland
| | - David T Rubin
- Inflammatory Bowel Disease Center, University of Chicago, Chicago, IL, United States
| | - Stephan R Vavricka
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland .,Gastroenterology and Hepatology Center, Zurich, Switzerland
| |
Collapse
|
42
|
McGonagle D, Watad A, Sharif K, Bridgewood C. Why Inhibition of IL-23 Lacked Efficacy in Ankylosing Spondylitis. Front Immunol 2021; 12:614255. [PMID: 33815371 PMCID: PMC8017223 DOI: 10.3389/fimmu.2021.614255] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/18/2021] [Indexed: 12/24/2022] Open
Abstract
The term spondyloarthritis pertains to both axial and peripheral arthritis including ankylosing spondylitis (AS) and psoriatic arthritis (PsA), which is strongly linked to psoriasis and also the arthritis associated with inflammatory bowel disease. The argument supporting the role for IL-23 across the spectrum of SpA comes from 4 sources. First, genome wide associated studies (GWAS) have shown that all the aforementioned disorders exhibit IL-23R pathway SNPs, whereas HLA-B27 is not linked to all of these diseases-hence the IL-23 pathway represents the common genetic denominator. Secondly, experimental animal models have demonstrated a pivotal role for the IL-23/IL-17 axis in SpA related arthropathy that initially manifests as enthesitis, but also synovitis and axial inflammation and also associated aortic root and cutaneous inflammation. Thirdly, the emergent immunology of the human enthesis also supports the presence of IL-23 producing myeloid cells, not just at the enthesis but in other SpA associated sites including skin and gut. Finally, drugs that target the IL-23 pathway show excellent efficacy for skin disease, efficacy for IBD and also in peripheral arthropathy associated with SpA. The apparent failure of IL-23 blockade in the AS which is effectively a spinal polyenthesitis but evidence for efficacy of IL-23 inhibition for peripheral enthesitis in PsA and preliminary suggestions for benefit in axial PsA, raises many questions. Key amongst these is whether spinal inflammation may exhibit entheseal IL-17A production independent of IL-23 but peripheral enthesitis is largely dependent on IL-23 driven IL-17 production. Furthermore, IL-23 blocking strategies in animal models may prevent experimental SpA evolution but not prevent established disease, perhaps pointing towards a role for IL-23 in innate immune disease initiation whereas persistent disease is dependent on memory T-cell responses that drive IL-17A production independently of IL-23, but this needs further study. Furthermore, IL-12/23 posology in inflammatory bowel disease is substantially higher than that used in AS trials which merits consideration. Therefore, the IL-23 pathway is centrally involved in the SpA concept but the nuances and intricacies in axial inflammation that suggest non-response to IL-23 antagonism await formal definition. The absence of comparative immunology between the different skeletal sites renders explanations purely hypothetical at this juncture.
Collapse
Affiliation(s)
- Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
- National Institute for Health Research (NIHR), Leeds Biomedical Research Centre (BRC), Leeds Teaching Hospitals, Leeds, United Kingdom
| | - Abdulla Watad
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
- Department of Medicine ‘B’, Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Kassem Sharif
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
- Department of Medicine ‘B’, Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Charlie Bridgewood
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
43
|
Ustekinumab-induced chronic lymphocytic leukemia in a patient with psoriatic arthritis. Reumatologia 2021; 59:58-61. [PMID: 33707797 PMCID: PMC7944959 DOI: 10.5114/reum.2021.102618] [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: 05/03/2020] [Accepted: 12/29/2020] [Indexed: 11/17/2022] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease characterized by skin and joint involvement. The disease may present with various joint pattern involvement, which sometimes may lead to joint destruction and deformity. Early diagnosis and treatment with disease-modifying anti-rheumatic drugs may prevent joint deformity. Recently there are many new treatment options including biologic drugs. Ustekinumab, an interleukin 12/23 inhibitor, has proven efficacy in the treatment of psoriatic arthritis. Like other biologic drugs (anti-TNF-α), there are contradictory data about the safety of ustekinumab and possible relationship with cancer development. Herein we report the development of chronic lymphocytic leukemia in a patient with PsA treated with ustekinumab.
Collapse
|
44
|
Fiechter RH, de Jong HM, van Mens LJJ, Fluri IA, Tas SW, Baeten DLP, Yeremenko NG, van de Sande MGH. IL-12p40/IL-23p40 Blockade With Ustekinumab Decreases the Synovial Inflammatory Infiltrate Through Modulation of Multiple Signaling Pathways Including MAPK-ERK and Wnt. Front Immunol 2021; 12:611656. [PMID: 33746955 PMCID: PMC7971179 DOI: 10.3389/fimmu.2021.611656] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Psoriatic arthritis (PsA) is a chronic inflammatory joint disease within the spondyloarthritis spectrum. IL-12p40/IL-23p40 blockade reduces PsA disease activity, but its impact on synovial inflammation remains unclear. Objectives: To investigate the cellular and molecular pathways affected by IL-12p40/IL-23p40 blockade with ustekinumab in the synovium of PsA patients. Methods: Eleven PsA patients with at least one inflamed knee or ankle joint were included in a 24-week single-center open-label study and received ustekinumab 45 mg/sc according to standard care at week 0, 4, and 16. Besides clinical outcomes, synovial tissue (ST) samples were obtained by needle arthroscopy from an inflamed knee or ankle joint at baseline, week 12 and 24 and analyzed by immunohistochemistry, RNA-sequencing and real-time quantitative polymerase chain reaction (qPCR). Results: We obtained paired baseline and week 12, and paired baseline, week 12 and 24 ST samples from nine and six patients, respectively. Eight patients completed 24 weeks of clinical follow-up. At 12 weeks 6/11 patients met ACR20, 2/11 met ACR50 and 1/11 met ACR70 improvement criteria, at 24 weeks this was 3/8, 2/8 and 1/8 patients, respectively. Clinical and serological markers improved significantly. No serious adverse events occurred. We observed numerical decreases of all infiltrating cell subtypes at week 12, reaching statistical significance for CD68+ sublining macrophages. For some cell types this was even more pronounced at week 24, but clearly synovial inflammation was incompletely resolved. IL-17A and F, TNF, IL-6, IL-8, and IL-12p40 were not significantly downregulated in qPCR analysis of W12 total biopsies, only MMP3 and IL-23p19 were significantly decreased. RNA-seq analysis revealed 178 significantly differentially expressed genes between baseline and 12 weeks (FDR 0.1). Gene Ontology and KEGG terms enrichment analyses identified overrepresentation of biological processes as response to reactive oxygen species, chemotaxis, migration and angiogenesis as well as MAPK-ERK and PI3K-Akt signaling pathways among the downregulated genes and of Wnt signaling pathway among the upregulated genes. Furthermore, ACR20 responders and non-responders differed strikingly in gene expression profiles in a post-hoc exploratory analysis. Conclusions: Ustekinumab suppresses PsA synovial inflammation through modulation of multiple signal transduction pathways, including MAPK-ERK, Wnt and potentially PI3K-Akt signaling rather than by directly impacting the IL-17 pathway.
Collapse
Affiliation(s)
- Renée H Fiechter
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Henriëtte M de Jong
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Leonieke J J van Mens
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Inka A Fluri
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Sander W Tas
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Dominique L P Baeten
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Nataliya G Yeremenko
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| | - Marleen G H van de Sande
- Amsterdam UMC, Department of Rheumatology and Clinical Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam Rheumatology and Immunology Center, Amsterdam, Netherlands
| |
Collapse
|
45
|
Torres T, Barcelos A, Filipe P, Fonseca JE. A Systematic Review With Network Meta-Analysis of the Available Biologic Therapies for Psoriatic Disease Domains. Front Med (Lausanne) 2021; 7:618163. [PMID: 33521024 PMCID: PMC7843938 DOI: 10.3389/fmed.2020.618163] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 11/26/2020] [Indexed: 11/13/2022] Open
Abstract
Introduction: Several new treatments have been developed for psoriatic disease, an inflammatory condition that involves skin and joints. Notwithstanding, few studies have made direct comparisons between treatments and therefore it is difficult to select the ideal treatment for an individual patient. The aim of this systematic review with network meta-analysis (NMA) was to analyze available and approved biologic therapies for each domain of psoriatic disease: skin, peripheral arthritis, axial arthritis, enthesitis, dactylitis, and nail involvement. Methods: Data from randomized clinical trials (RCTs) were included. A systematic review was performed using the MEDLINE database (July 2020) using PICO criteria. Bayesian NMA was conducted to compare the clinical efficacy of biological therapy in terms of the American College of Rheumatology criteria (ACR, 24 weeks) and Psoriasis Area and Severity Index (PASI, 10-16 weeks). Results: Fifty-four RCTs were included in the systematic review. Due to the design of the RCTs, namely, outcomes and time points, network meta-analysis was performed for skin and peripheral arthritis domains. For the skin domain, 30 studies reporting PASI100 were included. The peripheral arthritis domain was analyzed through ACR70 in 12 studies. From the therapies approved for both domains, secukinumab and ixekizumab were the ones with the highest probability of reaching the proposed outcomes. There is a lack of outcome uniformization in the dactylitis, enthesitis, and nail domains, and therefore, an objective comparison of the studies was not feasible. Nevertheless, secukinumab was the treatment with the best compromise between the number of studies in each domain and the results obtained in the different outcomes. Conclusion: Secukinumab and ixekizumab were the treatments with the highest probability of reaching both PASI100 and ACR70 outcomes. Due to the lack of a standard evaluation of outcomes of the other psoriatic disease domains, a network meta-analysis for all the domains was not possible to perform.
Collapse
Affiliation(s)
- Tiago Torres
- Department of Dermatology, Centro Hospitalar Universitário do Porto, Porto, Portugal
- Multidisciplinar Medical Research Unit, Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Anabela Barcelos
- Rheumatology Department, Centro Hospitalar do Baixo Vouga, Aveiro, Portugal
- NOVA National School of Public Health, Public Health Research Centre, Universidade NOVA de Lisboa – Portugal, Lisbon, Portugal
- Comprehensive Health Research Center (CHRC), Universidade NOVA de Lisboa – Portugal, Lisbon, Portugal
| | - Paulo Filipe
- Serviço de Dermatologia e Venereologia, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
- Unidade de Investigação em Dermatologia, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
- Clínica Universitária de Dermatologia, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - João Eurico Fonseca
- Serviço de Reumatologia e Doenças Ósseas Metabólicas, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
- Unidade de Investigação em Reumatologia, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
46
|
Vecellio M, Hake VX, Davidson C, Carena MC, Wordsworth BP, Selmi C. The IL-17/IL-23 Axis and Its Genetic Contribution to Psoriatic Arthritis. Front Immunol 2021; 11:596086. [PMID: 33574815 PMCID: PMC7871349 DOI: 10.3389/fimmu.2020.596086] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease belonging to the family of spondyloarthropathies (SpA). PsA commonly aggravates psoriasis of the skin and frequently manifests as an oligoarthritis with axial skeletal involvement and extraarticular manifestations including dactylitis, enthesitis, and uveitis. The weight of genetic predisposition to psoriasis and PsA is illustrated by the concordance rates in monozygotic twins which clearly demonstrate that genomics is insufficient to induce the clinical phenotype. The association of PsA with several single nucleotide polymorphisms (SNPs) at the IL23R locus and the involvement of Th17 cells in the immunopathogenesis of PsA clearly put the IL-23/IL-17 axis in the spotlight. The IL-23 and IL-17 cytokines have a pivotal role in the chronic inflammation of the synovium in PsA and are also prominent in the skin lesions of those with PsA. In this review, we focus on the genetic association of the IL-23/IL-17 axis with PsA and the contribution of these master cytokines in the pathophysiology of the disease, highlighting the main cell types incriminated in PsA and their specific role in the peripheral blood, lesional skin and joints of patients. We then provide an overview of the approved biologic drugs targeting the IL-23/IL-17 axis and discuss the advantages of genetic stratification to enhance personalized therapies in PsA.
Collapse
Affiliation(s)
- Matteo Vecellio
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.,Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| | - Vivien Xanath Hake
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| | - Connor Davidson
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | | | - B Paul Wordsworth
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center, IRCCS, Milan, Italy
| |
Collapse
|
47
|
Helliwell PS, Deodhar A, Gottlieb AB, Boehncke WH, Xu XL, Xu S, Wang Y, Hsia EC, Gladman DD, Ritchlin CT. Composite Measures of Disease Activity in Psoriatic Arthritis: Comparative Instrument Performance Based on the Efficacy of Guselkumab in an Interventional Phase II Trial. Arthritis Care Res (Hoboken) 2020; 72:1579-1588. [PMID: 31421033 PMCID: PMC7702129 DOI: 10.1002/acr.24046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 08/13/2019] [Indexed: 11/10/2022]
Abstract
Objective To assess performance of psoriatic arthritis (PsA) composite indices and evaluate guselkumab’s effect on achieving low disease activity or remission. Methods In this phase II trial, patients with active PsA (≥3 tender and ≥3 swollen joints, C‐reactive protein level ≥0.3 mg/dl, ≥3% body surface‐area with psoriasis involvement) were randomized 2:1 to subcutaneous guselkumab 100 mg (n = 100) or placebo (n = 49) at week 0, week 4, and every 8 weeks through week 44. At week 16, patients with <5% improvement in swollen and tender joints could early escape to open‐label ustekinumab. Patients continuing placebo crossed over to receive guselkumab 100 mg at weeks 24, 28, 36, and 44 (placebo to guselkumab). PsA composite indices (Psoriatic Arthritis Disease Activity Score [PASDAS], Group for Research and Assessment of Psoriasis and Psoriatic Arthritis composite score [GRACE], modified Composite Psoriatic Disease Activity Index [mCPDAI], and Disease Activity in Psoriatic Arthritis [DAPSA]) were analyzed as secondary outcomes (last observation carried forward for missing/post–early escape data through week 24; observed data post–week 24). Instrument performance was assessed. Results Baseline PASDAS, GRACE, mCPDAI, and DAPSA scores indicated moderate‐to‐high disease activity. At week 24, mean changes in each of these composite indices showed significant improvement with guselkumab (–2.50, –2.73, –3.8, and –23.08, respectively) versus placebo (–0.49, 0.35, –0.8, and –4.98, respectively; P < 0.001 for all). Significantly more guselkumab‐treated patients achieved low/very low/remitted disease activity states according to PASDAS (very low + low 35% versus 4%; P < 0.001), GRACE (30% versus 2%; P < 0.001), mCPDAI (46% versus 10%; P < 0.001), and DAPSA (remission + low 40% versus 12%; P < 0.001). A total of 12% of guselkumab‐treated versus no placebo‐treated patients achieved DAPSA remission (P < 0.01). The PASDAS and GRACE instruments were more sensitive than the mCPDAI and DAPSA tools in detecting treatment effect. Residual skin disease and enthesitis were marginally more prominent in patients achieving DAPSA low disease activity versus other indices. Conclusion Guselkumab demonstrated efficacy in achieving low disease activity/remission based on all PsA composite indices assessed. Composite index use in PsA trials and the clinic requires careful consideration to optimize feasibility and instrument performance.
Collapse
Affiliation(s)
| | | | | | | | - Xie L Xu
- Janssen Research & Development, LLC, San Diego, California
| | - Stephen Xu
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Yuhua Wang
- Janssen Research & Development, LLC, Spring House, Pennsylvania
| | - Elizabeth C Hsia
- Janssen Research & Development, LLC, Spring House, and University of Pennsylvania Medical Center, Philadelphia, Pennsylvania
| | - Dafna D Gladman
- University of Toronto, Krembil Research Institute, and Toronto Western Hospital, Toronto, Ontario, Canada
| | | |
Collapse
|
48
|
Garcia‐Melendo C, Vilarrasa E, Cubiró X, Bittencourt F, Puig L. Sequential paradoxical psoriasiform reaction and sacroiliitis following adalimumab treatment of hidradenitis suppurativa, successfully treated with guselkumab. Dermatol Ther 2020; 33:e14180. [DOI: 10.1111/dth.14180] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/22/2020] [Accepted: 08/02/2020] [Indexed: 11/27/2022]
Affiliation(s)
- Cristina Garcia‐Melendo
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
| | - Eva Vilarrasa
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
| | - Xavier Cubiró
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
| | - Flavia Bittencourt
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
| | - Lluís Puig
- Department of Dermatology, Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
| |
Collapse
|
49
|
Miyagawa I, Kubo S, Tanaka Y. A wide perspective of targeted therapies for precision medicine in autoimmune diseases. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020. [DOI: 10.1080/23808993.2020.1804867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Ippei Miyagawa
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Satoshi Kubo
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
50
|
Rahman P, Arendse R, Khraishi M, Sholter D, Sheriff M, Rampakakis E, Lehman AJ, Nantel F. Long-term effectiveness and safety of infliximab, golimumab and ustekinumab in patients with psoriatic arthritis from a Canadian prospective observational registry. BMJ Open 2020; 10:e036245. [PMID: 32792436 PMCID: PMC7430557 DOI: 10.1136/bmjopen-2019-036245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES The objectives of this study were to describe the demographic profile and baseline disease characteristics of patients with psoriatic arthritis (PsA) treated with either infliximab (IFX), subcutaneous golimumab (GLM) or ustekinumab (UST) treatment in Canadian routine care setting along with assessing long-term effectiveness and safety. METHODS Patients with PsA were enrolled into the Biologic Treatment Registry Across Canada registry (ClinicalTrials.gov Identifier: NCT00741793) from 2005 to 2017. The study visits occurred at study enrolment (baseline) and every 6 months thereafter. Effectiveness was assessed by changes in disease parameters (joint counts, Psoriasis Area Severity Index (PASI), Health Assessment Questionnaire, patient/physician global, minimal disease activity, enthesitis, dactylitis, erythrocyte sedimentation rate, C reactive protein). Improvements from baseline were explored with the paired t-test and the McNemar's test. Safety was evaluated by assessing the incidence of adverse events (AEs) and drug survival rates. RESULTS A total of 111 IFX-treated, 281 GLM-treated and 70 UST-treated patients were enrolled. Most baseline disease parameters remained similar over time in all three cohorts. UST-treated patients had lower mean baseline Disease Activity Score in 28 joints CRP, swollen joint based on 28 joints and higher PASI compared with patients treated with GLM. Treatment with IFX, GLM and UST was associated with significant improvements in all disease parameters over time (p<0.001) from baseline up to 84, 84 and 40 months, respectively.AEs were reported for 74.8%, 69.8% and 52.9% (138, 114 and 115 events/100 patient-years (PYs)) covering 325, 567 and 87 years of exposure for IFX-treated, GLM-treated and UST-treated patients, respectively. Severe AEs were reported in 19.8%, 8.5% and 5.7% (8.8, 7.2 and 8.0 events/100 PYs) in IFX-treated, GLM-treated and UST-treated patients, respectively. The proportion of patients who discontinued treatment were 63.1%, 50.9% and 50.0%, respectively. CONCLUSIONS IFX, GLM and UST treatment significantly reduced disease activity and improved functionality in patients with PsA followed by routine clinical practice and had a safety profile similar to that previously reported in the literature. TRIAL REGISTRATION NUMBER NCT00741793.
Collapse
Affiliation(s)
- Proton Rahman
- Department of Medicine, Memorial University of Newfoundland, St. Johns, Newfoundland and Labrador, Canada
| | - Regan Arendse
- College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Majed Khraishi
- Department of Medicine, Memorial University of Newfoundland, St. Johns, Newfoundland and Labrador, Canada
| | - Dalton Sholter
- Faculty of Medicine & Dentistry - Medicine Dept, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|