1
|
Nguyen C, Eziama E, Dominguez AR, Cao JH. Janus kinase inhibitors in the treatment of refractory cicatrizing conjunctivitis in pemphigoid. Ocul Surf 2025; 38:1-7. [PMID: 40368030 DOI: 10.1016/j.jtos.2025.05.002] [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: 02/15/2025] [Revised: 04/19/2025] [Accepted: 05/05/2025] [Indexed: 05/16/2025]
Abstract
OBJECTIVE To evaluate the efficacy of Janus kinase inhibitor (JAKi) therapy in managing cicatrizing conjunctivitis associated with ocular cicatricial pemphigoid (OCP) and mucous membrane pemphigoid with ocular involvement (ocMMP). METHODS Retrospective chart review of patients with cicatrizing conjunctivitis secondary to OCP or ocMMP who underwent treatment with JAKi at a tertiary academic medical center from August 2015 to November 2024 for minimum follow-up of six months. Collected data included demographics, Foster stage of cicatrization, and treatment course. RESULTS Thirty-two patients met inclusion criteria: 23 (71.9 %) with OCP and 9 (28.1 %) with ocMMP. 96.9 % of patients demonstrated clinical improvement within twelve months of treatment initiation. Best response achieved were as follows: 1 (3.1 %) no response, 17 (53.1 %) partial response, 14 (43.8 %) complete remission, and 12 (37.5 %) steroid-free complete remission. The mean time to partial response, complete remission, steroid-free complete remission was 3.1 ± 1.8 (range, 0.9-8.3), 7.8 ± 3.3 months (range, 2.3-14.7 months), and 10.3 ± 7.4 months, (range, 2.3-31.4 months), respectively. Relapse in disease activity occurred in 8/32 (25.0 %) of patients. Side effects occurred in 8/32 (25.0 %) of patients. Four patients (12.5 %) discontinued therapy due to severe adverse events, including transient ischemic attack, pulmonary embolism, pyelonephritis, and cholecystitis. There was a significant association between lower Foster cicatrization stages and achieving remission (U = 630.0, p = 0.0036), with a rank-biserial correlation of 0.72. CONCLUSIONS JAK inhibitor therapy demonstrates efficacy in the management of recalcitrant cicatrizing conjunctivitis associated with pemphigoid. These findings highlight JAK inhibitors as a promising therapeutic option for refractory cases.
Collapse
Affiliation(s)
- Celine Nguyen
- University of Texas Southwestern Medical School, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ebuka Eziama
- University of Texas Southwestern Medical School, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Arturo R Dominguez
- University of Texas Southwestern Medical School, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Departments of Dermatology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jennifer H Cao
- University of Texas Southwestern Medical School, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA; Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
2
|
Schundler SF, Noot CH, Frost Z, Clarke J, Hopkins ZH. Treatment of recalcitrant hypertrophic lichen planus with upadacitinib. JAAD Case Rep 2025; 59:78-80. [PMID: 40276729 PMCID: PMC12018683 DOI: 10.1016/j.jdcr.2025.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025] Open
Affiliation(s)
| | - Christiaan H. Noot
- Department of Dermatology, University of Utah Spencer F. Eccles School of Medicine, Salt Lake City, Utah
| | | | - Jennie Clarke
- Department of Dermatology, University of Utah Spencer F. Eccles School of Medicine, Salt Lake City, Utah
| | - Zachary H. Hopkins
- Department of Dermatology, University of Utah Spencer F. Eccles School of Medicine, Salt Lake City, Utah
| |
Collapse
|
3
|
Ibba L, Falcidia C, Di Giulio S, Bianco M, Valenti M, Facheris P, Narcisi A, Costanzo A, Gargiulo L. Real-World Effectiveness and Safety of Upadacitinib and Abrocitinib in Moderate-to-Severe Atopic Dermatitis: A 52-Week Retrospective Study. J Clin Med 2025; 14:2953. [PMID: 40363984 PMCID: PMC12072195 DOI: 10.3390/jcm14092953] [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: 04/14/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Atopic dermatitis (AD) is a chronic pruritic inflammatory disease affecting children and adults. Upadacitinib and abrocitinib are selective Janus kinase 1 inhibitors approved for the treatment of moderate-to-severe AD. Although their efficacy and safety are described in phase 3 clinical trials, real-world data are limited. Objectives: We aimed to evaluate the effectiveness and safety of upadacitinib and abrocitinib treatment in a real-life adult population with moderate-to-severe AD throughout an extended observation period. Methods: This retrospective observational study was conducted by analyzing data from the electronic records of IRCCS Humanitas Research Hospital from January 2023 to December 2024. Patients were administered either upadacitinib (15 or 30 mg) or abrocitinib (100 or 200 mg). Effectiveness was evaluated by using clinician-reported scores (Investigator Global Assessment [IGA] and Eczema Area and Severity Index [EASI]) and patient-reported outcomes (peak pruritus numerical rating scale [PP-NRS]) at weeks 8, 16, 32 and 52. Statistical significance was set at a probability value (p-value) < 0.05. Adverse events were also collected. Results: In total, 129 patients were included in the study, and 84 of them reached 52 weeks. At week 52, the EASI 75, 90, and 100 responses were 88.9%, 70.8%, and 54.2% for upadacitinib, and 100%, 91.7%, and 75% for abrocitinib. An IGA score equal to 0 or 1 at 52 weeks was achieved by 84.7% of patients treated with upadacitinib and 100% of those receiving abrocitinib. A four-point reduction from baseline PP-NRS was reported by 86.1% for upadacitinib and by 83.3% of patients for abrocitinib after one year of follow-up. Conclusions: Our study showed comparable or even higher effectiveness outcomes in terms of EASI 75, EASI 90, and EASI 100 at week 52 compared to phase-3 clinical trials, with no new safety concerns, supporting the real-world effectiveness of abrocitinib and upadacitinib in moderate-to-severe AD.
Collapse
Affiliation(s)
- Luciano Ibba
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (C.F.); (S.D.G.); (M.B.); (M.V.); (P.F.); (A.N.); (A.C.); (L.G.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Costanza Falcidia
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (C.F.); (S.D.G.); (M.B.); (M.V.); (P.F.); (A.N.); (A.C.); (L.G.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Sara Di Giulio
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (C.F.); (S.D.G.); (M.B.); (M.V.); (P.F.); (A.N.); (A.C.); (L.G.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Matteo Bianco
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (C.F.); (S.D.G.); (M.B.); (M.V.); (P.F.); (A.N.); (A.C.); (L.G.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Mario Valenti
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (C.F.); (S.D.G.); (M.B.); (M.V.); (P.F.); (A.N.); (A.C.); (L.G.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Paola Facheris
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (C.F.); (S.D.G.); (M.B.); (M.V.); (P.F.); (A.N.); (A.C.); (L.G.)
| | - Alessandra Narcisi
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (C.F.); (S.D.G.); (M.B.); (M.V.); (P.F.); (A.N.); (A.C.); (L.G.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Antonio Costanzo
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (C.F.); (S.D.G.); (M.B.); (M.V.); (P.F.); (A.N.); (A.C.); (L.G.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Luigi Gargiulo
- Dermatology Unit, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (C.F.); (S.D.G.); (M.B.); (M.V.); (P.F.); (A.N.); (A.C.); (L.G.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| |
Collapse
|
4
|
Lin X, Li X, Zhai Z, Zhang M. JAK-STAT pathway, type I/II cytokines, and new potential therapeutic strategy for autoimmune bullous diseases: update on pemphigus vulgaris and bullous pemphigoid. Front Immunol 2025; 16:1563286. [PMID: 40264772 PMCID: PMC12011800 DOI: 10.3389/fimmu.2025.1563286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/20/2025] [Indexed: 04/24/2025] Open
Abstract
Autoimmune Bullous Diseases (AIBDs), characterized by the formation of blisters due to autoantibodies targeting structural proteins, pose significant therapeutic challenges. Current treatments, often involving glucocorticoids or traditional immunosuppressants, are limited by their non-specificity and side effects. Cytokines play a pivotal role in AIBDs pathogenesis by driving inflammation and immune responses. The JAK-STAT pathway is central to the biological effects of various type I and II cytokines, making it an attractive therapeutic target. Preliminary reports suggest that JAK inhibitors may be a promising approach in PV and BP, but further clinical validation is required. In AIBDs, particularly bullous pemphigoid (BP) and pemphigus vulgaris (PV), JAK inhibitors have shown promise in modulating pathogenic cytokine signaling. However, the safety and selectivity of JAK inhibitors remain critical considerations, with the potential for adverse effects and the need for tailored treatment strategies. This review explores the role of cytokines and the JAK-STAT pathway in BP and PV, evaluating the therapeutic potential and challenges associated with JAK inhibitors in managing these complex disorders.
Collapse
Affiliation(s)
| | | | - Zhifang Zhai
- Department of Dermatology, The First Affiliated Hospital, Army Medical University, Chongqing, China
| | - Mingwang Zhang
- Department of Dermatology, The First Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
5
|
Waeckel L, Talon C, Barrau M, Berger AE, Roblin X, Paul S. Development and evaluation of two whole-blood flow cytometry protocols for monitoring patients treated with JAK inhibitors. IMMUNOTHERAPY ADVANCES 2025; 5:ltaf006. [PMID: 40265078 PMCID: PMC12012447 DOI: 10.1093/immadv/ltaf006] [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: 09/19/2024] [Accepted: 02/27/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction The clinical efficacy of Janus kinase inhibitors (JAKinibs) is highly variable and their safety profiles are poorly understood. Methods We established two flow cytometry panels for the assessment of two promising leukocyte biomarkers: signal transducer and activator of transcription (STAT) phosphorylation and cytokine receptor expression. We evaluated the first panel, which assesses phosphorylation levels for STAT1, STAT3, and STAT5 after cytokine stimulation, with or without in vitro pretreatment with JAKinibs, in 10 healthy donors. We then evaluated the second panel, which assesses cytokine receptor expression on T cells and B cells, in five healthy donors. Results Stimulation with interleukin (IL)-2 or IL-7 increased STAT5 phosphorylation in T cells but not in B cells or monocytes. IL-6 stimulation induced STAT3 phosphorylation in monocytes and CD4 T cells and, to a lesser extent, in CD8 T cells, but not in B cells. IL-21 stimulation led to STAT3 phosphorylation in T cells and, to a lesser extent, in B cells, but not in monocytes. Interferon-α stimulation increased STAT1 phosphorylation in all cell types. STAT phosphorylation levels were lower after pretreatment with JAKinibs. A dose-response curve was plotted, confirming the correlation between JAKinib concentration and STAT phosphorylation inhibition. The second panel showed that each cell type displayed a distinct pattern of cytokine receptors expression, and that this pattern might be modified by in vitro treatment with JAKinibs. Conclusion This preliminary study confirms the utility of flow cytometry for monitoring the biological effects of JAKinibs. Further studies on treated patients are now required to evaluate the clinical value of these two flow cytometry panels.
Collapse
Affiliation(s)
- Louis Waeckel
- Immunology Laboratory, iBiothera Reference Center, CHU Saint-Etienne, F42055 Saint-Etienne, France
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, F42023 Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, CHU Saint-Etienne, F42055 Saint-Etienne, France
| | - Chloé Talon
- Immunology Laboratory, iBiothera Reference Center, CHU Saint-Etienne, F42055 Saint-Etienne, France
| | - Mathilde Barrau
- Department of Gastroenterology, CHU Saint-Etienne, Saint-Etienne, France
| | - Anne-Emmanuelle Berger
- Immunology Laboratory, iBiothera Reference Center, CHU Saint-Etienne, F42055 Saint-Etienne, France
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, F42023 Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, CHU Saint-Etienne, F42055 Saint-Etienne, France
| | - Xavier Roblin
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, F42023 Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, CHU Saint-Etienne, F42055 Saint-Etienne, France
- Department of Gastroenterology, CHU Saint-Etienne, Saint-Etienne, France
| | - Stéphane Paul
- Immunology Laboratory, iBiothera Reference Center, CHU Saint-Etienne, F42055 Saint-Etienne, France
- CIRI - Centre International de Recherche en Infectiologie, Team GIMAP, Université Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, F42023 Saint-Etienne, France
- CIC 1408 Inserm Vaccinology, CHU Saint-Etienne, F42055 Saint-Etienne, France
| |
Collapse
|
6
|
Ciccia F, McGonagle D, Thomas R, Marzo-Ortega H, Martin DA, Yndestad A, Volkov M. JAK inhibition and axial spondyloarthritis: new steps on the path to understanding pathophysiology. Front Immunol 2025; 16:1488357. [PMID: 40103808 PMCID: PMC11913702 DOI: 10.3389/fimmu.2025.1488357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 02/13/2025] [Indexed: 03/20/2025] Open
Abstract
Axial spondyloarthritis (axSpA) is a chronic inflammatory disease that predominantly affects the sacroiliac joints and spine. Tumor necrosis factor (TNF) and interleukin (IL)-17A are key cytokines in disease pathogenesis and are established axSpA treatment targets. Recently, axSpA treatment options have been complemented by Janus kinase inhibitors (JAKi), which inhibit various cytokines without directly impacting TNF or IL-17 signaling. The effect of JAKi on axSpA remains under investigation: besides a JAK2-mediated (and potentially tyrosine kinase 2 [TYK2]-mediated) effect on the IL-23/IL-17 axis, emerging evidence suggests γδ T cells, type 3 innate lymphoid cells, and mucosa-associated invariant T cells, which are dependent on IL-7 and/or IL-15 and thus on JAK1, are strongly inhibited by JAKi used to treat axSpA. This review summarizes potential effects of JAKi on axSpA and shows evidence from pre-clinical/clinical studies. Greater understanding of the mechanisms of action of available treatments may improve knowledge of axSpA and pave the road for future therapies.
Collapse
Affiliation(s)
- Francesco Ciccia
- Dipartimento di Medicina di Precisione, Università della Campania L. Vanvitelli, Naples, Italy
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, The Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | - Ranjeny Thomas
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Helena Marzo-Ortega
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds and NIHR Leeds Biomedical Research Centre, The Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom
| | | | | | | |
Collapse
|
7
|
Li J, Li X, Wang Y, Zhou C. Clinical response and safety of upadacitinib for alopecia areata: A retrospective study. J Am Acad Dermatol 2025:S0190-9622(25)00390-1. [PMID: 40043887 DOI: 10.1016/j.jaad.2025.02.083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 04/02/2025]
Affiliation(s)
- Jiecheng Li
- Department of Dermatology, Peking University People's Hospital, Beijing, China
| | - Xiangqian Li
- Department of Dermatology, Peking University People's Hospital, Beijing, China
| | - Yuxin Wang
- Department of Dermatology, Peking University People's Hospital, Beijing, China
| | - Cheng Zhou
- Department of Dermatology, Peking University People's Hospital, Beijing, China.
| |
Collapse
|
8
|
Huang W, de Vries C, Sharma RK, Wangriatisak K, Chatzidionysiou K, Malmström V, Grönwall C. JAK Inhibitors and B Cell Function: A Comparative Study of Their Impact on Plasma Cell Differentiation, Cytokine Production, and Naïve B Cell Activation. Eur J Immunol 2025; 55:e202451437. [PMID: 40098342 PMCID: PMC11914861 DOI: 10.1002/eji.202451437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/19/2025]
Abstract
B cells play a crucial role in autoimmune diseases, as evidenced by autoantibody responses and the effectiveness of B cell-targeted therapies. Janus kinase inhibitors (JAKi), which target downstream signaling of cytokine receptors, are potent rheumatic disease-modifying drugs. However, besides reducing inflammation, JAKi may impact the adaptive immune system. In this study, we examined the effects of JAKi on B-cell function using in vitro cultures and multiparameter flow cytometry. The results show a JAKi-mediated reduction in plasma cell differentiation, primarily by inhibition of memory B-cell stimulation and proliferation. JAKi exposure resulted in stalling R848, IL-2, and IL-21 stimulated B cells in an intermediate activated state with elevated naïve cells displaying increased expression of CXCR5, CD71, CD22, and CD20. In addition, the data demonstrate a moderate JAKi-mediated reduction of B cell TNF and IL-8 cytokine expression following stimulation. Importantly, the efficacy varied greatly between drugs; tofacitinib and upadacitinib (pan JAKi; JAK1i) exhibited the strongest impact, while baricitinib (JAK1/JAK2i) showed donor-dependent variation, and filgotinib (JAK1i) had no effect. All JAKi, except filgotinib, inhibited IL-2 or IL-21-induced STAT3 phosphorylation. Still, filgotinib demonstrated similar inhibition of phospho-STAT5 as other JAKi following IL-21. These findings underscore the therapeutic impact of JAKi through the modulation of B-cell functions.
Collapse
Affiliation(s)
- Wenqi Huang
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| | - Charlotte de Vries
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| | - Ravi Kumar Sharma
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular MedicineKarolinska University HospitalStockholmSweden
- Department of Clinical Immunology and RheumatologyAll India Institute of Medical SciencesBilaspurIndia
| | - Kittikorn Wangriatisak
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| | - Katerina Chatzidionysiou
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| | - Vivianne Malmström
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| | - Caroline Grönwall
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| |
Collapse
|
9
|
Picchianti-Diamanti A, Aiello A, De Lorenzo C, Migliori GB, Goletti D. Management of tuberculosis risk, screening and preventive therapy in patients with chronic autoimmune arthritis undergoing biotechnological and targeted immunosuppressive agents. Front Immunol 2025; 16:1494283. [PMID: 39963138 PMCID: PMC11830708 DOI: 10.3389/fimmu.2025.1494283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
Tuberculosis (TB) is the leading cause of death in the world from an infectious disease. Its etiologic agent, the Mycobacterium tuberculosis (Mtb), is a slow-growing bacterium that has coexisted in humans for thousands of years. According to the World Health Organization, 10.6 million new cases of TB and over 1 million deaths were reported in 2022. It is widely recognized that patients affected by chronic autoimmune arthritis such as rheumatoid arthritis (RA), psoriatic arthritis (PsA), and ankylosing spondylitis (AS) have an increased incidence rate of TB disease compared to the general population. As conceivable, the risk is associated with age ≥65 years and is higher in endemic regions, but immunosuppressive therapy plays a pivotal role. Several systematic reviews have analysed the impact of anti-TNF-α agents on the risk of TB in patients with chronic autoimmune arthritis, as well as for other biologic disease-modifying immunosuppressive anti-rheumatic drugs (bDMARDs) such as rituximab, abatacept, tocilizumab, ustekinumab, and secukinumab. However, the data are less robust compared to those available with TNF-α inhibitors. Conversely, data on anti-IL23 agents and JAK inhibitors (JAK-i), which have been more recently introduced for the treatment of RA and PsA/AS, are limited. TB screening and preventive therapy are recommended in Mtb-infected patients undergoing bDMARDs and targeted synthetic (ts)DMARDs. In this review, we evaluate the current evidence from randomized clinical trials, long-term extension studies, and real-life studies regarding the risk of TB in patients with RA, PsA, and AS treated with bDMARDs and tsDMARDs. According to the current evidence, TNF-α inhibitors carry the greatest risk of TB progression among bDMARDs and tsDMARDs, such as JAK inhibitors and anti-IL-6R agents. The management of TB screening and the updated preventive therapy are reported.
Collapse
Affiliation(s)
- Andrea Picchianti-Diamanti
- Department of Clinical and Molecular Medicine, “Sapienza” University, S. Andrea University Hospital, Rome, Italy
| | - Alessandra Aiello
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Chiara De Lorenzo
- Department of Clinical and Molecular Medicine, “Sapienza” University, S. Andrea University Hospital, Rome, Italy
| | - Giovanni Battista Migliori
- Istituti Clinici Scientifici Maugeri, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Tradate, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
10
|
Chiu TM, Chen CB, Lu CW, Hui RCY, Chi MH, Chang YC, Wu J, Chen KY, Lin YYW, Lo PC, Hsu TC, Wang CW, Chung WH. CCR8/CCL1 and CXCR3/CXCL10 axis-mediated memory T-cell activation in patients with recalcitrant drug-induced hypersensitivity. Br J Dermatol 2025; 192:293-305. [PMID: 39503255 DOI: 10.1093/bjd/ljae375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 08/19/2024] [Accepted: 11/04/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND As a drug-induced hypersensitivity syndrome, drug reaction with eosinophilia and systemic symptoms (DRESS) is potentially fatal. Most patients with DRESS recover within a few weeks; however, some patients may suffer from a prolonged disease course and develop autoimmune sequelae. OBJECTIVES To investigate the immune mechanism and therapeutic targets of patients with recalcitrant DRESS with a prolonged disease course. METHODS Thirty-two patients with recalcitrant DRESS with a prolonged treatment course (≥ 8 weeks; 'prolonged DRESS'), 28 patients with DRESS with a short treatment course (< 2 weeks; 'short-duration DRESS') and 26 healthy donors (HDs) were enrolled. RESULTS Bulk transcriptome results showed that the mRNA expression levels of CCR8 and CXCR3 were significantly increased in blood samples from patients in the acute stage of prolonged DRESS [Padj = 1.50 × 10-9 (CCR8) and Padj = 2.60 × 10-4 (CXCR3), patients with prolonged DRESS compared with the HD group]. Serum and skin lesion concentrations of CCL1 and CXCL10 (ligands of CCR8 and CXCR3, respectively) were significantly increased in patients with prolonged DRESS compared with patients with short-duration DRESS. The results from high-parameter flow cytometry and autoantibody screening also identified significant increases in CD8+ GNLY+ CXCR3+ effector memory T cells, CD8+ central memory T cells, CD4+ CCR8+ T helper 2 cells and IgG anti-HES-6 autoantibodies in patients with prolonged DRESS. Furthermore, in vitro blocking assays revealed that Janus kinase inhibitors (JAKi; mainly tofacitinib and upadacitinib) significantly decreased the release of CCL1 and CXCL10. Some patients with prolonged DRESS were successfully treated with JAKi. CONCLUSIONS JAKi (tofacitinib and upadacitinib) were associated with decreased concentrations of CCL1 and CXCL10, suggesting that they may attenuate CCR8/CCL1 and CXCR3/CXCL10 axis-mediated memory T-cell activation, which contributes to disease pathogenesis in patients with recalcitrant DRESS and a long-term treatment course. GRAPHICAL ABSTRACT
Collapse
Affiliation(s)
- Tsu-Man Chiu
- Department of Dermatology, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Bing Chen
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei, Tucheng and Keelung, Taiwan
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chun-Wei Lu
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei, Tucheng and Keelung, Taiwan
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Rosaline Chung-Yee Hui
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei, Tucheng and Keelung, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Min-Hui Chi
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei, Tucheng and Keelung, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ya-Ching Chang
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei, Tucheng and Keelung, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jennifer Wu
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei, Tucheng and Keelung, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Kuan-Yu Chen
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei, Tucheng and Keelung, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yang Yu-Wei Lin
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei, Tucheng and Keelung, Taiwan
| | - Pei-Chi Lo
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei, Tucheng and Keelung, Taiwan
| | - Tsai-Ching Hsu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chuang-Wei Wang
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei, Tucheng and Keelung, Taiwan
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China
| | - Wen-Hung Chung
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou, Taipei, Tucheng and Keelung, Taiwan
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou, Taiwan
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
- Department of Dermatology, Beijing Tsinghua Chang Gung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
- Department of Dermatology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
| |
Collapse
|
11
|
Makhija M, Manchanda D, Sharma M. Nano-based Therapeutics for Rheumatoid Arthritis: Recent Patents and Development. RECENT PATENTS ON NANOTECHNOLOGY 2025; 19:56-75. [PMID: 37691226 DOI: 10.2174/1872210518666230905155459] [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: 02/22/2023] [Revised: 07/21/2023] [Accepted: 08/03/2023] [Indexed: 09/12/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease marked by inflammation of synovium and generation of autoantibodies. Bone and cartilage are frequently damaged along with weakening of tendons and ligaments resulting in disability. An effective RA treatment needs a multi-disciplinary approach which relies upon pathophysiology that is still partially understood. In RA patients, inflammation was induced by pro-inflammatory cytokines including IL-1, IL-6 & IL-10. The conventional dosage regimens for treating RA have drawbacks such as ineffectiveness, greater doses, frequent dosing, relatively expensive and serious adverse effects. To formulate an effective treatment plan for RA, research teams have recently focused on producing several nanoformulations containing anti-inflammatory APIs with an aim to target the inflamed area. Nanomedicines have recently gained popularity in the treatment of RA. Interestingly, unbelievable improvements have been observed in current years in diagnosis and management of RA utilizing nanotechnology. Various patents and clinical trial data have been reported in relevance to RA treatment.
Collapse
Affiliation(s)
- Manish Makhija
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, 304022, India
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, 123401, India
| | - Deeksha Manchanda
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, 304022, India
- Department of Pharmaceutical Sciences, Indira Gandhi University, Meerpur, Rewari, 123401, India
| | - Manu Sharma
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, 304022, India
| |
Collapse
|
12
|
Yang Y, Chen J, Xiong S, Zhang J, Ye Q, Xue R, Tian X, Zhong J, Zhu H, Gao A, Liu Y. Comparative effectiveness of upadacitinib versus dupilumab for moderate-to-severe atopic dermatitis: A retrospective cohort study. Int Immunopharmacol 2024; 143:113383. [PMID: 39405928 DOI: 10.1016/j.intimp.2024.113383] [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: 07/05/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Although efficacy and safety of Upadacitinib and Dupilumab in moderate to severe atopic dermatitis (AD) have been shown in clinical trials, real world data are still limited. The aim of this retrospective study is to indirectly compare the efficacy and safety of Upadacitinib and Dupilumab in patients with moderate to severe AD in real world practice. METHODS A single-center retrospective cohort study was conducted. The study included patients with moderate to severe AD, who were enrolled from May 2022 to March 2024, to indirectly compare the efficacy and safety of Upadacitinib and Dupilumab over 12 weeks duration. RESULTS Eighty-seven patients were included (46 received Upadacitinib and 41 Dupilumab). Compared with week 0, there was a significant decrease in EASI score, ADCT score and NRS score in patients of both groups in weeks 4, 8, and 12. In week 4, the reduction in EASI score, ADCT score and NRS score was significantly greater in patients of Upadacitinib group compared to those in Dupilumab group. Compared to baseline, in week 12, the decrease in IL-4, IL-13, and IL-31 level in the serum of patients in Upadacitinib group was significantly greater than that of patients in Dupilumab group. The total IgE of patients in Dupilumab group decreased significantly, while there was no significant change in patients of Upadacitinib group. Although Upadacitinib group reported more adverse events than Dupilumab group, no serious adverse events were observed. CONCLUSIONS Both Upadacitinib and Dupilumab groups showed effective trend in patients with moderate to severe AD. Upadacitinib has better efficacy and rapid onset in the treatment of patients with moderate to severe AD.
Collapse
Affiliation(s)
- Yan Yang
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, Guangdong 510095, PR China
| | - Jiaoquan Chen
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, Guangdong 510095, PR China
| | - Siyin Xiong
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, Guangdong 510095, PR China
| | - Jing Zhang
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, Guangdong 510095, PR China
| | - Qianru Ye
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, Guangdong 510095, PR China
| | - Rujun Xue
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, Guangdong 510095, PR China
| | - Xin Tian
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, Guangdong 510095, PR China
| | - Jiemin Zhong
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, Guangdong 510095, PR China
| | - Huilan Zhu
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, Guangdong 510095, PR China
| | - Aili Gao
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, Guangdong 510095, PR China.
| | - Yumei Liu
- Department of Dermatology, Guangzhou Dermatology Hospital, Guangzhou, Guangdong 510095, PR China.
| |
Collapse
|
13
|
Zerrouk N, Augé F, Niarakis A. Building a modular and multi-cellular virtual twin of the synovial joint in Rheumatoid Arthritis. NPJ Digit Med 2024; 7:379. [PMID: 39719524 DOI: 10.1038/s41746-024-01396-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
Rheumatoid arthritis is a complex disease marked by joint pain, stiffness, swelling, and chronic synovitis, arising from the dysregulated interaction between synoviocytes and immune cells. Its unclear etiology makes finding a cure challenging. The concept of digital twins, used in engineering, can be applied to healthcare to improve diagnosis and treatment for complex diseases like rheumatoid arthritis. In this work, we pave the path towards a digital twin of the arthritic joint by building a large, modular biochemical reaction map of intra- and intercellular interactions. This network, featuring over 1000 biomolecules, is then converted to one of the largest executable Boolean models for biological systems to date. Validated through existing knowledge and gene expression data, our model is used to explore current treatments and identify new therapeutic targets for rheumatoid arthritis.
Collapse
Affiliation(s)
- Naouel Zerrouk
- GenHotel, Laboratoire Européen de Recherche Pour La Polyarthrite Rhumatoïde, University Paris-Saclay, University Evry, Evry, France
- Sanofi R&D Data and Data Science, Artificial Intelligence & Deep Analytics, Omics Data Science, Chilly-Mazarin, France
| | - Franck Augé
- Sanofi R&D Data and Data Science, Artificial Intelligence & Deep Analytics, Omics Data Science, Chilly-Mazarin, France
| | - Anna Niarakis
- GenHotel, Laboratoire Européen de Recherche Pour La Polyarthrite Rhumatoïde, University Paris-Saclay, University Evry, Evry, France.
- Lifeware Group, Inria Saclay, Palaiseau, France.
- University of Toulouse III-Paul Sabatier, Laboratory of Molecular, Cellular and Developmental Biology (MCD), Center of Integrative Biology (CBI), Toulouse, France.
| |
Collapse
|
14
|
Maeyama A, Kondo M, Harada H, Shono E, Nagamine R, Tsuru T, Inoue Y, Nakashima M, Yamasaki Y, Niiro H, Nakashima Y, Yamamoto T. Efficacy and safety of baricitinib in rheumatoid arthritis patients with moderate renal impairment: a multicenter propensity score matching study. BMC Rheumatol 2024; 8:69. [PMID: 39695823 DOI: 10.1186/s41927-024-00446-y] [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: 08/02/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND This study aimed to compare the efficacy and safety of baricitinib in patients with rheumatoid arthritis (RA) receiving different doses based on renal function. METHODS We conducted a retrospective study within the JAK Study Group, involving 23 facilities in Fukuoka Prefecture, examining patients treated with baricitinib for RA. Patients were categorized into two dose groups: 4 mg with normal/mild renal dysfunction and 2 mg with moderate renal dysfunction. Baricitinib's efficacy, retention rate, and safety were compared between the groups after propensity score matching. RESULTS After propensity score matching, disease duration, methotrexate dosage, and anti-cyclic citrullinated peptide antibody positivity rate were balanced across 33 patients in both groups. No significant differences were observed between the groups in tender/swollen joint counts, changes in evaluator/patient global assessments, achievement rate of low disease activity, remission rate on clinical/simplified disease activity indices, or retention rate. Additionally, the incidence of adverse events aligned with previous reports, indicating similar drug safety profiles. CONCLUSIONS Baricitinib 2 mg in RA patients with moderate renal dysfunction showed comparable efficacy and retention rate to 4 mg in patients with normal/mild renal dysfunction. The incidence and types of adverse events were consistent with previous studies, indicating the safety of the drug at these dosages.
Collapse
Affiliation(s)
- Akira Maeyama
- Department of Orthopaedic Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan.
| | - Masakazu Kondo
- Kondo Clinic of Rheumatology and Orthopaedic Surgery, 3-10-11 Tenjin, Chuo-ku, Fukuoka, 810-0001, Japan
| | - Hiroshi Harada
- Department of Orthopaedic Surgery Rheumatology Center, Yagi Hospital, 2-21-25, Maidashi, Higashi-ku, Fukuoka, 812-0054, Japan
| | - Eisuke Shono
- Shono Rheumatology Clinic, Nishijin Prime Building, 1-10-27, Nishijin, Sawara-ku, Fukuoka, 814-0002, Japan
| | - Ryuji Nagamine
- Nagamine Rheumatology and Orthopaedic Clinic, 1-11-1, Kashiiekimae, Higashi-ku, Fukuoka, 813-0013, Japan
| | - Tomomi Tsuru
- PS Clinic, Random Square, 6-18 Tenya-cho, Hakata-ku, Fukuoka, 812-0025, Japan
| | - Yasushi Inoue
- Department of Rheumatology, Fukuoka Red Cross Hospital, 3-1-1 Ogusu, Minami-ku, Fukuoka, 815-8555, Japan
| | - Munetoshi Nakashima
- Division of Rheumatology, Kurume University Medical Center, 155-1 Kokubu-machi, Kurume, 839-0863, Fukuoka, Japan
| | - Yutaro Yamasaki
- Department of Orthopaedic Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| | - Hiroaki Niiro
- Department of Medical Education, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yasuharu Nakashima
- Department of Orthopaedic Surgery, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takuaki Yamamoto
- Department of Orthopaedic Surgery, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka, 814-0180, Japan
| |
Collapse
|
15
|
Nguyen HN, Jeong Y, Kim Y, Kamiya M, Kim Y, Athar H, Castaldi PJ, Hersh CP, Menon JA, Wong J, Chan I, Oldham WM, Padera RF, Sharma NS, Sholl LM, Vivero M, Watts GFM, Knipe RS, Black KE, Hariri LP, Yun JH, Merriam LT, Yuan K, Kim EY, Brenner MB. Leukemia inhibitory factor (LIF) receptor amplifies pathogenic activation of fibroblasts in lung fibrosis. Proc Natl Acad Sci U S A 2024; 121:e2401899121. [PMID: 39636853 PMCID: PMC11648669 DOI: 10.1073/pnas.2401899121] [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: 02/01/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024] Open
Abstract
Fibrosis drives end-organ damage in many diseases. However, clinical trials targeting individual upstream activators of fibroblasts, such as TGFβ, have largely failed. Here, we target the leukemia inhibitory factor receptor (LIFR) as an "autocrine master amplifier" of multiple upstream activators of lung fibroblasts. In idiopathic pulmonary fibrosis (IPF), the most common fibrotic lung disease, we found that lung myofibroblasts had high LIF expression, and the fibroblasts in fibroblastic foci coexpressed LIF and LIFR. In IPF, fibroblastic foci are the "leading edge" of fibrosis and a key site of disease pathogenesis. TGFβ1, one of the principal drivers of fibrosis, up-regulated LIF expression in IPF fibroblasts. We found that TGFβ1, IL-4, and IL-13 stimulations of fibroblasts require the LIF-LIFR axis to evoke a strong fibrogenic effector response in fibroblasts. In vitro antibody blockade of LIFR on IPF lung fibroblasts reduced the induction of profibrotic genes after TGFβ1 stimulation. Silencing LIF and LIFR reduced profibrotic fibroblast activation following TGFβ1, IL-4, and IL-13 stimulations. We also demonstrated that LIFR amplified profibrotic stimuli in precision-cut lung slices from IPF patients. These LIFR signals were transduced via JAK2, and STAT1 in IPF lung fibroblasts. Together, we find that LIFR drives an autocrine circuit that amplifies and sustains pathogenic activation of IPF fibroblasts. Targeting a single, downstream master amplifier on fibroblasts, like LIFR, is an alternative therapeutic strategy that simultaneously attenuates the profibrotic effects of multiple upstream stimuli.
Collapse
Affiliation(s)
- Hung N. Nguyen
- Department of Medicine, Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Yunju Jeong
- Harvard Medical School, Boston, MA02115
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Department of Food and Nutrition, College of Human Ecology, Kyung Hee University, Seoul02447, Republic of Korea
| | - Yunhye Kim
- Harvard Medical School, Boston, MA02115
- Department of Pediatrics, Division of Pulmonary Medicine, Boston Children’s Hospital, Boston, MA02115
| | - Mari Kamiya
- Harvard Medical School, Boston, MA02115
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Yaunghyun Kim
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Humra Athar
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Peter J. Castaldi
- Harvard Medical School, Boston, MA02115
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Craig P. Hersh
- Harvard Medical School, Boston, MA02115
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Jaivardhan A. Menon
- Harvard Medical School, Boston, MA02115
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | | | - Ian Chan
- Abpro Corporation, Woburn, MA01801
| | - William M. Oldham
- Harvard Medical School, Boston, MA02115
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Robert F. Padera
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA02115
| | - Nirmal S. Sharma
- Harvard Medical School, Boston, MA02115
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Lynette M. Sholl
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA02115
| | - Marina Vivero
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA02115
| | - Gerald F. M. Watts
- Department of Medicine, Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Rachel S. Knipe
- Harvard Medical School, Boston, MA02115
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA02114
| | - Katharine E. Black
- Harvard Medical School, Boston, MA02115
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA02114
| | - Lida P. Hariri
- Harvard Medical School, Boston, MA02115
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA02114
- Department of Pathology, Massachusetts General Hospital, Boston, MA02114
| | - Jeong H. Yun
- Harvard Medical School, Boston, MA02115
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Louis T. Merriam
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Ke Yuan
- Harvard Medical School, Boston, MA02115
- Department of Pediatrics, Division of Pulmonary Medicine, Boston Children’s Hospital, Boston, MA02115
| | - Edy Y. Kim
- Harvard Medical School, Boston, MA02115
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Michael B. Brenner
- Department of Medicine, Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| |
Collapse
|
16
|
Mammoliti O, Menet C, Cottereaux C, Blanc J, De Blieck A, Coti G, Geney R, Oste L, Ostyn K, Palisse A, Quinton E, Schmitt B, Borgonovi M, Parent I, Jagerschmidt C, De Vos S, Vayssiere B, López-Ramos M, Shoji K, Brys R, Amantini D, Galien R, Joannesse C. Design of a potent and selective dual JAK1/TYK2 inhibitor. Bioorg Med Chem 2024; 114:117932. [PMID: 39447537 DOI: 10.1016/j.bmc.2024.117932] [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: 05/07/2024] [Revised: 09/18/2024] [Accepted: 09/28/2024] [Indexed: 10/26/2024]
Abstract
Janus kinase (JAK) inhibitors have gathered interest as treatments for several inflammatory and autoimmune diseases. The four first marketed inhibitors target JAK1, with varying selectivity towards other JAK family members, but none inhibit tyrosine kinase-2 (TYK2) at clinically relevant doses. TYK2 is required for the signaling of the interleukin (IL)-12 and IL-23 cytokines, which are key to the polarization of TH1 and TH17 cells, respectively; two cell subtypes that play major roles in inflammatory diseases. Herein, we report our effort towards the optimization of a potent and selective dual JAK1/TYK2 inhibitor series starting from a HTS hit. Structural information revealed vectors required to improve both JAK1 and TYK2 potency as well as selectivity towards JAK2. The potent inhibition of both JAK1 (3.5 nM) and TYK2 (5.7 nM) in biochemical assays by our optimized lead compound, as well as its notable selectivity against JAK2, were confirmed in cellular and whole blood assays. Inhibition of TYK2 by the lead compound was demonstrated by dose-dependent efficacy in an IL-23-induced psoriasis-like inflammation mouse model.
Collapse
Affiliation(s)
- Oscar Mammoliti
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Christel Menet
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Céline Cottereaux
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Javier Blanc
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Ann De Blieck
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Ghjuvanni Coti
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Raphaël Geney
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Line Oste
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Koen Ostyn
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Adeline Palisse
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Evelyne Quinton
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Benoit Schmitt
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - Monica Borgonovi
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Isabelle Parent
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | | - Steve De Vos
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | | | | | - Kenji Shoji
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Reginald Brys
- Galapagos NV, Generaal De Wittelaan L11, 2800 Mechelen, Belgium
| | - David Amantini
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - René Galien
- Galapagos SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | |
Collapse
|
17
|
Ahmed S, Yesudian R, Ubaide H, Coates LC. Rationale and concerns for using JAK inhibitors in axial spondyloarthritis. Rheumatol Adv Pract 2024; 8:rkae141. [PMID: 39660106 PMCID: PMC11630911 DOI: 10.1093/rap/rkae141] [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: 07/23/2024] [Accepted: 11/03/2024] [Indexed: 12/12/2024] Open
Abstract
Axial spondyloarthritis (axSpA) is a chronic illness with limited treatment options. The role of Janus kinase (JAK) inhibition as a therapeutic option has increasingly become a focus of research in recent years as they have brought a new mode of action to the clinical armamentarium. This review assesses the efficacy and safety profile of these drugs in axSpA. The current phase 2 and 3 clinical trials data are summarized across tofacitinib, upadacitinib and filgotinib. Moreover, the safety profiles of these drugs, in the context of emerging safety signals such as during the ORAL surveillance study, are reviewed. In summary, JAK inhibitors offer a novel therapeutic target for axSpA and appear to address some of the unmet needs for patients who have either failed to respond to current treatment options or in whom they are contraindicated. There is a relative lack of evidence in non-radiographic axSpA and longer-term trials are needed to establish true efficacy and safety profile in radiographic axSpA.
Collapse
Affiliation(s)
- Saad Ahmed
- General Medicine, Colchester Hospital, Colchester, UK
| | - Rohan Yesudian
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Hassan Ubaide
- General Medicine, Colchester Hospital, Colchester, UK
| | - Laura C Coates
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals NHS Trust, Oxford, UK
| |
Collapse
|
18
|
Perricone C, Dal Pozzolo R, Cafaro G, Calvacchi S, Bruno L, Tromby F, Colangelo A, Gerli R, Bartoloni E. Sudden improvement of alopecia universalis and psoriatic arthritis while receiving upadacitinib: a case-based review. Reumatismo 2024; 77. [PMID: 39470166 DOI: 10.4081/reumatismo.2024.1685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 06/06/2024] [Indexed: 10/30/2024] Open
Abstract
Alopecia universalis (AU), an advanced form of alopecia areata (AA), is a condition characterized by the complete loss of hair over the entire skin surface. Recent progress has significantly enhanced our understanding of the pathogenesis of AU. In particular, interferon-γ (IFN-γ) and interleukin (IL)-15 seem to play a pivotal role in the pathogenesis of the disease. Nonetheless, a variety of medications has been used to treat the disease with frequently inconsistent results. Given the broad modulation of the immune system and inhibition of key molecules, including IFN-γ and IL-15, oral janus kinase (JAK) inhibitors represent a treatment option for moderate to severe cases of AA, as demonstrated in case reports supporting their efficacy and tolerability. We present the case of a patient suffering from psoriatic arthritis and AU who experienced a sudden improvement in peripheral arthritis and AU while receiving JAK1 selective treatment with upadacitinib. So far, there are very limited case reports of successful upadacitinib treatment for patients with AA, mostly in patients also suffering from atopic dermatitis. Thus, we provide evidence for the efficacy of upadacitinib in managing AU in adults, as well as in the context of inflammatory arthritis such as psoriatic arthritis.
Collapse
Affiliation(s)
- Carlo Perricone
- Rheumatology, Department of Medicine and Surgery, University of Perugia
| | | | - Giacomo Cafaro
- Rheumatology, Department of Medicine and Surgery, University of Perugia
| | - Santina Calvacchi
- Rheumatology, Department of Medicine and Surgery, University of Perugia
| | - Lorenza Bruno
- Rheumatology, Department of Medicine and Surgery, University of Perugia
| | - Francesco Tromby
- Rheumatology, Department of Medicine and Surgery, University of Perugia
| | - Anna Colangelo
- Rheumatology, Department of Medicine and Surgery, University of Perugia
| | - Roberto Gerli
- Rheumatology, Department of Medicine and Surgery, University of Perugia
| | - Elena Bartoloni
- Rheumatology, Department of Medicine and Surgery, University of Perugia
| |
Collapse
|
19
|
Matsumoto S, Mashima H. Clinical Profiles of Leucine-Rich Alpha-2 Glycoprotein for Indicating Mucosal Healing in Ulcerative Colitis Patients under Administration of Molecular-Targeted Drug. Dig Dis 2024; 43:11-18. [PMID: 39462503 DOI: 10.1159/000542062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 10/12/2024] [Indexed: 10/29/2024]
Abstract
INTRODUCTION Leucine-rich alpha-2 glycoprotein (LRG) is a useful serum biomarker for monitoring disease activity during remission in ulcerative colitis (UC). Because LRG levels differ among patients, it is necessary to assess them after profiling patients, especially in patients with refractory UC undergoing treatment with molecular-targeted drugs. This study aimed to analyze LRG levels that indicate mucosal healing according to clinical characteristics and molecular-targeted drugs. METHODS Among 214 patients with UC treated with biologics or Janus kinase (JAK) inhibitors, this study evaluated 111 patients (174 measurements) who achieved mucosal healing based on colonoscopy performed within 2 months before and after LRG measurement and experienced no changes in disease status or treatment during the same period. We analyzed the relationship of LRG with clinical characteristics (including sex, age, body mass index, and disease type and duration) and molecular-targeted drugs. RESULTS Compared with men, women had significantly higher LRG levels (9.5 μg/mL vs. 11.3 μg/mL, p < 0.001). In addition, LRG levels were significantly higher in older patients (12.0 μg/mL vs. 9.8 μg/mL, p < 0.01). LRG levels were the highest in patients treated with vedolizumab and lower in patients treated with JAK inhibitors (vedolizumab: 12.7 μg/mL; tofacitinib: 8.9 μg/mL; upadacitinib: 8.5 μg/mL; and filgotinib: 9.1 μg/mL; p < 0.0001). CONCLUSION Among the patients who achieved mucosal healing, LRG levels were significantly higher in women and older patients. LRG levels differed according to the molecular-targeted drug used and were higher with vedolizumab and lower with JAK inhibitors.
Collapse
Affiliation(s)
- Satohiro Matsumoto
- Department of Gastroenterology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Hirosato Mashima
- Department of Gastroenterology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| |
Collapse
|
20
|
Li X, Zhang L, Gu H, He W, Zhai Z, Zhang M. Treatment and molecular analysis of bullous pemphigoid with tofacitinib: a case report and review of current literature. Front Immunol 2024; 15:1464474. [PMID: 39497828 PMCID: PMC11532171 DOI: 10.3389/fimmu.2024.1464474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 09/27/2024] [Indexed: 11/07/2024] Open
Abstract
Background Bullous pemphigoid (BP) is a rare, life-threatening autoimmune blistering disease with pruritus and tension blisters/bullous as the main clinical manifestations. Glucocorticosteroids are the main therapeutic agents for it, but their efficacy is poor in some patients. Tofacitinib, a small molecule agent that inhibits JAK1/3, has shown incredible efficacy in a wide range of autoimmune diseases and maybe a new valuable treatment option for refractory BP. Objective To report a case of refractory BP successfully treated with tofacitinib, then explore the underlying mechanism behind the treatment, and finally review similarities to other cases reported in the literature. Methods Case report and literature review of published cases of successful BP treatment with JAK inhibitors. The case report describes a 73-year-old male with refractory BP that was successfully managed with the combination therapy of tofacitinib and low-dose glucocorticoids for 28 weeks. Immunohistochemistry and RNA sequencing were performed to analyze the underlying mechanism of tofacitinib therapy. A systematic literature search was conducted to identify other cases of treatment with JAK inhibitors. Results Throughout the 28-week treatment period, the patient experienced clinical, autoantibody and histologic resolution. Immunohistochemical analysis showed tofacitinib significantly decreased the pSTAT3 and pSTAT6 levels in the skin lesions of this patient. RNA sequencing and immunohistochemical testing of lesion samples from other BP patients identified activation of the JAK-STAT signaling pathway. Literature review revealed 17 previously reported cases of BP treated with four kinds of JAK inhibitors successfully, including tofacitinib (10), baricitinib (1), upadacitinib (3) and abrocitinib (3). Conclusions Our findings support the potential of tofacitinib as a safe and effective treatment option for BP. Larger studies are underway to better understand this efficacy and safety.
Collapse
Affiliation(s)
| | | | | | | | - Zhifang Zhai
- Department of Dermatology, Southwest Hospital, Army Medical University, Chongqing, China
| | - Mingwang Zhang
- Department of Dermatology, Southwest Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
21
|
Chatterjee B, Thakur SS. Valuable Contributions and Lessons Learned from Proteomics and Metabolomics Studies of COVID-19. J Proteome Res 2024; 23:4171-4187. [PMID: 39157976 DOI: 10.1021/acs.jproteome.4c00340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
The COVID-19 pandemic caused by the SARS-CoV-2 virus infected more than 775,686,716 humans and was responsible for the death of more than 7,054,093 individuals. COVID-19 has taught us that the development of vaccines, repurposing of drugs, and understanding the mechanism of a disease can be done within a short time. The COVID-19 proteomics and metabolomics has contributed to its diagnosis, understanding of its progression, host-virus interaction, disease mechanism, and also in the search of suitable anti-COVID therapeutics. Mass spectrometry based proteomics was used to find the potential biomarkers of different stages of COVID-19 including severe and nonsevere cases in the blood serum. Notably, protein-protein interaction techniques to understand host-virus interactions were also significantly useful. The single-cell proteomics studies were carried out to ascertain the changes in immune cell composition and its activation in mild COVID-19 patients versus severe COVID-19 patients using whole-blood and peripheral-blood mononuclear cells. Modern technologies were helpful to deal with the pandemic; however, there is still scope for further development. Further, attempts were made to understand the protein-protein, metabolite-metabolite, and protein-metabolite interactomes, derived from proteins and metabolite fingerprints of COVID-19 patients by reanalysis of COVID-19 public mass spectrometry based proteomics and metabolomics studies. Further, some of these interactions were supported by the literature as validations in the COVID-19 studies.
Collapse
Affiliation(s)
| | - Suman S Thakur
- Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500007, India
| |
Collapse
|
22
|
Virtanen A, Spinelli FR, Telliez JB, O'Shea JJ, Silvennoinen O, Gadina M. JAK inhibitor selectivity: new opportunities, better drugs? Nat Rev Rheumatol 2024; 20:649-665. [PMID: 39251770 DOI: 10.1038/s41584-024-01153-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/11/2024]
Abstract
Cytokines function as communication tools of the immune system, serving critical functions in many biological responses and shaping the immune response. When cytokine production or their biological activity goes awry, the homeostatic balance of the immune response is altered, leading to the development of several pathologies such as autoimmune and inflammatory disorders. Cytokines bind to specific receptors on cells, triggering the activation of intracellular enzymes known as Janus kinases (JAKs). The JAK family comprises four members, JAK1, JAK2, JAK3 and tyrosine kinase 2, which are critical for intracellular cytokine signalling. Since the mid-2010s multiple JAK inhibitors have been approved for inflammatory and haematological indications. Currently, approved JAK inhibitors have demonstrated clinical efficacy; however, improved selectivity for specific JAKs is likely to enhance safety profiles, and different strategies have been used to accomplish enhanced JAK selectivity. In this update, we discuss the background of JAK inhibitors, current approved indications and adverse effects, along with new developments in this field. We address the issue of JAK selectivity and its relevance in terms of efficacy, and describe new modalities of JAK targeting, as well as new aspects of JAK inhibitor action.
Collapse
Affiliation(s)
- Anniina Virtanen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Francesca Romana Spinelli
- Dipartimento di Scienze Cliniche Internistiche, Anestesiologiche e Cardiovascolari-Reumatologia, Sapienza Universitá di Roma, Rome, Italy
| | | | - John J O'Shea
- Lymphocyte Cell Biology Section, Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Olli Silvennoinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Fimlab laboratories, Tampere, Finland
| | - Massimo Gadina
- Translational Immunology Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
23
|
Picchianti Diamanti A, Cattaruzza MS, Salemi S, Di Rosa R, Sesti G, De Lorenzo C, Felice GM, Frediani B, Baldi C, Chimenti MS, D'Antonio A, Crepaldi G, Luchetti MM, Paci V, Zabotti A, Giovannini I, Canzoni M, Sebastiani G, Scirocco C, Perricone C, Laganà B, Iagnocco A. Clinical and Ultrasonographic Remission in Bio-naïve and Bio-failure Patients with Rheumatoid Arthritis at 24 Weeks of Upadacitinib Treatment: The UPARAREMUS Real-Life Study. Rheumatol Ther 2024; 11:1347-1361. [PMID: 39177745 PMCID: PMC11422397 DOI: 10.1007/s40744-024-00712-y] [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: 07/03/2024] [Accepted: 08/01/2024] [Indexed: 08/24/2024] Open
Abstract
INTRODUCTION Clinical remission is the main target in the management of patients with rheumatoid arthritis (RA). However, several authors found synovitis in patients with RA in clinical remission at ultrasonography (US). Upadacitinib is a selective Janus kinase 1 inhibitor that achieved significantly higher remission rates than adalimumab and abatacept in patients with RA. Here we present the 24-week data of the UPAdacitinib Rheumatoid Arthritis REmission UltraSonography (UPARAREMUS) study. METHODS This is a longitudinal multicenter observational study, enrolling bio-naïve and bio-inadequate responder patients affected by RA. The primary endpoint was the proportion of patients achieving both clinical and US remission at week 24. The proportion of patients achieving clinical remission with different composite indexes at week 12 and 24 was also evaluated. US of four target joints (wrists and second metacarpophalangeal bilaterally) was performed at baseline and weeks 12/24, and US remission was defined as the absence of power Doppler (PD) signal ≥ 2 in one target joint, or PD ≥ 1 in two target joints. RESULTS After 12 weeks and 24 weeks, 40% and 63.6% of patients achieved US plus clinical remission. The following parameters were associated with US plus clinical remission: being bio-naïve and having a shorter disease duration, although at multivariate analysis significant odds ratio (OR) was found only for being bio-naïve. CONCLUSIONS UPARAREMUS is the first study evaluating the efficacy of upadacitinib in reaching both clinical and US remission in patients with RA. At 24 weeks, 63.6% of patients reached the primary endpoint, the only baseline associated parameter was being bio-naïve.
Collapse
Affiliation(s)
- Andrea Picchianti Diamanti
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, S. Andrea University Hospital, 00189, Rome, Italy.
| | - Maria Sofia Cattaruzza
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185, Rome, Italy
| | - Simonetta Salemi
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, S. Andrea University Hospital, 00189, Rome, Italy
| | - Roberta Di Rosa
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, S. Andrea University Hospital, 00189, Rome, Italy
| | - Giorgio Sesti
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, S. Andrea University Hospital, 00189, Rome, Italy
| | - Chiara De Lorenzo
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, S. Andrea University Hospital, 00189, Rome, Italy
| | - Gloria Maria Felice
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, S. Andrea University Hospital, 00189, Rome, Italy
| | - Bruno Frediani
- Department of Medicine, Surgery and Neurosciences, Rheumatology Unit, University of Siena, Viale Mario Bracci, 53100, Siena, Italy
| | - Caterina Baldi
- Department of Medicine, Surgery and Neurosciences, Rheumatology Unit, University of Siena, Viale Mario Bracci, 53100, Siena, Italy
| | - Maria Sole Chimenti
- Department of System Medicine, Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Arianna D'Antonio
- Department of System Medicine, Rheumatology, Allergology and Clinical Immunology, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Gloria Crepaldi
- Academic Rheumatology Centre, Department of Clinical and Biological Science, University of Turin, AO Mauriziano Torino, 10128, Turin, Italy
| | - Michele Maria Luchetti
- Clinica Medica, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | - Valentino Paci
- Clinica Medica, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | - Alen Zabotti
- Rheumatology Clinic, Department of Medicine, University of Udine, c/o Azienda Sanitaria Universitaria Friuli Centrale, 33100, Udine, Italy
| | - Ivan Giovannini
- Rheumatology Clinic, Department of Medicine, University of Udine, c/o Azienda Sanitaria Universitaria Friuli Centrale, 33100, Udine, Italy
| | - Marco Canzoni
- ASL Rome 1 UOSD Reumatologia, Ospedale Nuovo Regina Margherita, 00153, Rome, Italy
| | | | - Chiara Scirocco
- U.O.C. Reumatologia, A.O. San Camillo-Forlanini, Rome, Italy
| | - Carlo Perricone
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Bruno Laganà
- Department of Clinical and Molecular Medicine, "Sapienza" University of Rome, S. Andrea University Hospital, 00189, Rome, Italy
| | - Annamaria Iagnocco
- Academic Rheumatology Centre, Department of Clinical and Biological Science, University of Turin, AO Mauriziano Torino, 10128, Turin, Italy
| |
Collapse
|
24
|
Olatunji AO, Maqbool M, Abid MA, Makineni KS, Almadhoun MKIK, Meer HB, Ansari F, Alfakhori AM, Bedros AW, Banu N, Bokhari SFH. Safety and Efficacy of Upadacitinib in Crohn's Disease: An Updated Systematic Review. Cureus 2024; 16:e70125. [PMID: 39463627 PMCID: PMC11506236 DOI: 10.7759/cureus.70125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2024] [Indexed: 10/29/2024] Open
Abstract
Crohn's disease (CD) is a chronic inflammatory bowel disease that significantly impacts patient quality of life. This systematic review evaluates the safety and efficacy of upadacitinib, a selective Janus kinase (JAK) inhibitor, in the treatment of CD. A comprehensive literature search was conducted across multiple databases, yielding seven studies published between 2020 and 2024, encompassing 1,481 patients. The review includes randomized controlled trials, post hoc analyses of phase 3 trials, and observational studies. Findings consistently demonstrate upadacitinib's superiority over placebo in inducing and maintaining clinical remission, achieving endoscopic response, and normalizing inflammatory markers. Notably, upadacitinib showed rapid symptom relief, with clinical remission observed as early as five to six days after treatment initiation. Efficacy was observed across various patient populations, including those with prior biologic failure. Long-term studies indicated sustained clinical and endoscopic improvements, with remission rates maintained for up to 30 months. Upadacitinib also demonstrated effectiveness in real-world, treatment-refractory cohorts. Safety profiles were generally consistent with those of other JAK inhibitors. Common adverse events included infections, particularly herpes zoster, and laboratory abnormalities such as neutropenia and elevated creatine kinase. Serious adverse events were infrequent, although careful monitoring is warranted. This review suggests that upadacitinib is a promising treatment option for moderate to severe CD, offering rapid and sustained efficacy with an acceptable safety profile.
Collapse
Affiliation(s)
| | - Muhammad Maqbool
- Internal Medicine, Shaheed Mohtarma Benazir Bhutto Medical College Lyari, Karachi, PAK
| | | | - Karthik Sai Makineni
- Medicine, Sri Ramaswamy Memorial (SRM) Medical College Hospital and Research Centre, Chennai, IND
| | | | - Hamdah B Meer
- Medicine and Surgery, Dubai Medical College for Girls, Dubai, ARE
| | - Fazeela Ansari
- Medicine and Surgery, Dubai Medical College for Girls, Dubai, ARE
| | - Alma M Alfakhori
- Medicine and Surgery, Dubai Medical College for Girls, Dubai, ARE
| | - Adees W Bedros
- Internal Medicine, School of Medicine, The University of Jordan, Amman, JOR
| | - Nasreen Banu
- Medicine, Shadan Institute of Medical Sciences College, Hyderabad, IND
| | | |
Collapse
|
25
|
Yoon H, Ye BD, Kang SB, Lee KM, Choi CH, Jo JY, Woo J, Cheon JH. Safety and effectiveness of tofacitinib in Korean adult patients with ulcerative colitis: post-marketing surveillance study. BMC Gastroenterol 2024; 24:273. [PMID: 39160459 PMCID: PMC11331763 DOI: 10.1186/s12876-024-03336-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Tofacitinib is an oral Janus kinase inhibitor for the treatment of ulcerative colitis (UC). We aimed to identify the safety and effectiveness of tofacitinib in patients with UC in routine clinical settings in Korea. METHODS This open-label, observational, prospective, post-marketing surveillance study was conducted at 22 hospitals in the Republic of Korea. Patients with moderate to severe active UC who received tofacitinib were included and followed up for up to 52 weeks. Tofacitinib was administered at a dosage of 10 mg twice daily for at least 8 weeks, followed by 5 or 10 mg twice daily at the investigator's discretion based on clinical evaluation according to the approved Korean label. Safety including adverse events (AEs) and effectiveness including clinical remission, clinical response, and endoscopic mucosal healing were evaluated. Safety analysis set was defined as all patients registered for this study who received at least one dose of tofacitinib according to the approved Korean label and followed up for safety data. Effectiveness analysis set included patients in the safety analysis set who were evaluated for overall effectiveness assessment and excluded patients who had received tofacitinib less than 8 weeks. RESULTS A total of 110 patients were enrolled, of whom 106 patients were included in the safety population. The median duration of treatment was 370 days and the treatment duration ranged from 16 to 684 days for the safety population. AEs occurred in 42 patients (39.6%). Serious AEs (SAEs) occurred in 7 patients (6.6%) and of them, there were 2 cases of serious infections. These serious infections were reported as Adverse Event of Special Interest (AESI) in this study and no other AESI were reported. There were no cases of death during the study period. Clinical remission rates were 40.0%, 46.7%, 57.6%, and 55.1% at 8, 16, 24, and 52 weeks, and clinical response rates were 77.8%, 87.9%, 56.6%, and 81.4% at each visit, respectively. Endoscopic mucosal healing rates were 58.7% at 16 weeks and 46.2% at 52 weeks. CONCLUSION Tofacitinib was effective in Korean patients with moderate to severe active UC and the safety findings were consistent with the known safety profile of tofacitinib. This study confirmed the safety and effectiveness of tofacitinib in Korean patients with moderate to severe active UC in routine clinical settings. TRIAL REGISTRATION This study is registered in the ClinicalTrials.gov under the identifier NCT04071405, registered on 28 August 2019.
Collapse
Affiliation(s)
- Hyuk Yoon
- Seoul National University Bundang Hospital, Seongnam, Gyeonggi-do, Korea
| | - Byong Duk Ye
- University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Sang-Bum Kang
- The Catholic University of Korea, Daejeon ST. Mary's Hospital, Daejeon, Korea
| | - Kang-Moon Lee
- The Catholic University of Korea, ST. Vincent's Hospital, Suwon, Gyeonggi-do, Korea
| | | | | | - Juwon Woo
- Pfizer Pharmaceutical Korea Ltd, Seoul, Korea
| | - Jae Hee Cheon
- Department of Internal Medicine, College of Medicine, Yonsei University, 50-1, Yonsei-ro, Seodaemun-gu, Seoul, Republic of Korea.
| |
Collapse
|
26
|
Fujimoto K, Kameda Y, Nagano Y, Deguchi S, Yamamoto T, Krol RP, Gee P, Matsumura Y, Okamoto T, Nagao M, Takayama K, Yokokawa R. SARS-CoV-2-induced disruption of a vascular bed in a microphysiological system caused by type-I interferon from bronchial organoids. LAB ON A CHIP 2024; 24:3863-3879. [PMID: 38252025 DOI: 10.1039/d3lc00768e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Blood vessels show various COVID-19-related conditions including thrombosis and cytokine propagation. Existing in vitro blood vessel models cannot represent the consequent changes in the vascular structure or determine the initial infection site, making it difficult to evaluate how epithelial and endothelial tissues are damaged. Here, we developed a microphysiological system (MPS) that co-culture the bronchial organoids and the vascular bed to analyze infection site and interactions. In this system, virus-infected organoids caused damage in vascular structure. However, vasculature was not damaged or infected when the virus was directly introduced to vascular bed. The knockout of interferon-related genes and inhibition of the JAK/STAT pathway reduced the vascular damage, indicating the protective effect of interferon response suppression. The results demonstrate selective infection of bronchial epithelial cells and vascular damage by cytokines and also indicate the applicability of MPS to investigate how the infection influences vascular structure and functions.
Collapse
Affiliation(s)
- Kazuya Fujimoto
- Department of Micro Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| | - Yoshikazu Kameda
- Department of Micro Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| | - Yuta Nagano
- Department of Micro Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| | - Sayaka Deguchi
- Center for iPS cell Research and Application (CiRA), Kyoto University, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Takuya Yamamoto
- Center for iPS cell Research and Application (CiRA), Kyoto University, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan.
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
- Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Rafal P Krol
- Research and Development Center, CiRA Foundation, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8397, Japan
| | - Peter Gee
- MaxCyte Inc., Gaithersburg, MD 20878, USA
| | - Yasufumi Matsumura
- Department of Clinical Laboratory medicine, Kyoto University Graduate School of Medicine, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Toru Okamoto
- Department of Microbiology, School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Miki Nagao
- Department of Clinical Laboratory medicine, Kyoto University Graduate School of Medicine, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kazuo Takayama
- Center for iPS cell Research and Application (CiRA), Kyoto University, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| |
Collapse
|
27
|
Rael VE, Yano JA, Huizar JP, Slayden LC, Weiss MA, Turcotte EA, Terry JM, Zuo W, Thiffault I, Pastinen T, Farrow EG, Jenkins JL, Becker ML, Wong SC, Stevens AM, Otten C, Allenspach EJ, Bonner DE, Bernstein JA, Wheeler MT, Saxton RA, Liu B, Majer O, Barton GM. Large-scale mutational analysis identifies UNC93B1 variants that drive TLR-mediated autoimmunity in mice and humans. J Exp Med 2024; 221:e20232005. [PMID: 38780621 PMCID: PMC11116816 DOI: 10.1084/jem.20232005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/21/2024] [Accepted: 04/17/2024] [Indexed: 05/25/2024] Open
Abstract
Nucleic acid-sensing Toll-like receptors (TLR) 3, 7/8, and 9 are key innate immune sensors whose activities must be tightly regulated to prevent systemic autoimmune or autoinflammatory disease or virus-associated immunopathology. Here, we report a systematic scanning-alanine mutagenesis screen of all cytosolic and luminal residues of the TLR chaperone protein UNC93B1, which identified both negative and positive regulatory regions affecting TLR3, TLR7, and TLR9 responses. We subsequently identified two families harboring heterozygous coding mutations in UNC93B1, UNC93B1+/T93I and UNC93B1+/R336C, both in key negative regulatory regions identified in our screen. These patients presented with cutaneous tumid lupus and juvenile idiopathic arthritis plus neuroinflammatory disease, respectively. Disruption of UNC93B1-mediated regulation by these mutations led to enhanced TLR7/8 responses, and both variants resulted in systemic autoimmune or inflammatory disease when introduced into mice via genome editing. Altogether, our results implicate the UNC93B1-TLR7/8 axis in human monogenic autoimmune diseases and provide a functional resource to assess the impact of yet-to-be-reported UNC93B1 mutations.
Collapse
Affiliation(s)
- Victoria E. Rael
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Julian A. Yano
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - John P. Huizar
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Leianna C. Slayden
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Madeleine A. Weiss
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Elizabeth A. Turcotte
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Jacob M. Terry
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Wenqi Zuo
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Isabelle Thiffault
- Department of Pathology and Laboratory Medicine, Children’s Mercy Hospital, Kansas City, MO, USA
- Genomic Medicine Center, Children’s Mercy Hospital, Kansas City, MO, USA
- University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| | - Tomi Pastinen
- Genomic Medicine Center, Children’s Mercy Hospital, Kansas City, MO, USA
- University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| | - Emily G. Farrow
- Department of Pathology and Laboratory Medicine, Children’s Mercy Hospital, Kansas City, MO, USA
- Genomic Medicine Center, Children’s Mercy Hospital, Kansas City, MO, USA
- University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| | - Janda L. Jenkins
- Department of Genetics, Children’s Mercy Hospital, Kansas City, MO, USA
| | - Mara L. Becker
- Division of Rheumatology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Stephen C. Wong
- Division of Rheumatology, Department of Pediatrics, Seattle Children’s Hospital, Seattle, WA, USA
| | - Anne M. Stevens
- Division of Rheumatology, Department of Pediatrics, Seattle Children’s Hospital, Seattle, WA, USA
- Johnson & Johnson Innovative Medicine, Spring House, PA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Catherine Otten
- Division of Pediatric Neurology, Department of Neurology, Seattle Children’s Hospital, University of Washington, Seattle, WA, USA
| | - Eric J. Allenspach
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Devon E. Bonner
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, USA
- Division of Medical Genetics, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jonathan A. Bernstein
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, USA
- Division of Medical Genetics, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew T. Wheeler
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, USA
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert A. Saxton
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Chemistry, University of California, Berkeley, CA, USA
| | - Bo Liu
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Olivia Majer
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Gregory M. Barton
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
28
|
Dreo B, Muralikrishnan AS, Husic R, Lackner A, Brügmann T, Haudum P, Bosch P, Thiel J, Fessler J, Stradner M. JAK/STAT signaling in rheumatoid arthritis leukocytes is uncoupled from serum cytokines in a subset of patients. Clin Immunol 2024; 264:110238. [PMID: 38729230 DOI: 10.1016/j.clim.2024.110238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
OBJECTIVE Rheumatoid Arthritis (RA) is a systemic autoimmune disease involving pro-inflammatory cytokines that can be therapeutically targeted by antibodies or kinase inhibitors. Nevertheless, these drugs fail in a subset of patients independent of the abundance of the targeted cytokines. We aim to explore the cellular basis of this phenomenon by analyzing the relation of cytokine abundance and activation of downstream signaling pathways in RA. METHODS The study included 62 RA patients and 9 healthy controls (HC). Phosphorylation of STAT 1-6 in various immune cell subsets was determined ex vivo using a novel robust flow cytometry-based protocol. Serum concentrations of IL-6, IL-10, IL-12p70, IL-17 A, interferon gamma, and TNFα in the same samples were measured using highly sensitive single molecule array (SIMOA). RESULTS We found an increase in circulating cytokines in RA patients, while STAT activity was lower in RA patients compared to HC. Based on STAT activity we determined three endotypes in active RA patients (cDAI>10, n = 28): 1) those with active STAT5a/b signaling in T cells (n = 7/28), 2) those with a low STAT activity in all assessed cell types (n = 14/28), and 3) those with active STAT1 and STAT3 signaling mainly in myeloid cells (n = 7/28). Integrating intracellular STAT activation and cytokine analysis revealed diminished JAK/STAT signaling in a subset of patients (n = 8/20) despite elevated serum cytokine concentrations. CONCLUSION Diminished JAK/STAT signaling in active RA may partly explain unresponsiveness to therapy targeting cytokine signaling. Analysis of JAK/STAT phosphorylation may identify patients at risk for non-response to these therapies.
Collapse
Affiliation(s)
- Barbara Dreo
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| | | | - Rusmir Husic
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| | - Angelika Lackner
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| | - Theresa Brügmann
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| | - Patrizia Haudum
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| | - Philipp Bosch
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| | - Jens Thiel
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| | - Johannes Fessler
- Division of Immunology, Otto Loewi Research Center, Medical University of Graz, Austria.
| | - Martin Stradner
- Division of Rheumatology and Immunology, Medical University of Graz, Austria
| |
Collapse
|
29
|
Cao H, Wang S. G-CSF promotes the development of hepatocellular carcinoma by activating the PI3K/AKT/mTOR pathway in TAM. Aging (Albany NY) 2024; 16:10799-10812. [PMID: 38967628 PMCID: PMC11272108 DOI: 10.18632/aging.205922] [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: 07/27/2023] [Accepted: 04/23/2024] [Indexed: 07/06/2024]
Abstract
OBJECTIVE This investigation seeks to elucidate the role of the Granulocyte Colony-Stimulating Factor (G-CSF) in the progression of hepatocellular carcinoma (HCC), as well as the impact of the substance on related signaling pathways within the disease matrix. METHODS Nude mouse tumor-bearing assay was used to detect tumor progression. Levels of Mannose/CD68 and CD34/Mannose within these samples and the concentrations of Mannose and inducible Nitric Oxide Synthase (iNOS) in macrophages were quantified using immunofluorescence techniques. The angiogenic capability was assessed via tube formation assays, and protein expressions of G-CSF, Vascular Endothelial Growth Factor (VEGF), Transforming Growth Factor-beta (TGF-β), Matrix Metalloproteinases 2 and 9 (MMP2/9), SH2-containing protein tyrosine phosphatase-2 (SHP-2), phosphorylated PI3K/total PI3K (P-PI3K/t-PI3K), phosphorylated AKT/total AKT (P-AKT/t-AKT), and phosphorylated mTOR/total mTOR (P-mTOR/t-mTOR) were measured through Western Blot analysis in both tumor tissues and macrophages. RESULTS Administration of G-CSF resulted in a marked augmentation of tumor volume. Macrophage Mannose expression was significantly elevated upon G-CSF treatment, while iNOS levels were conspicuously diminished. G-CSF substantially enhanced the secretion of VEGF, TGF-β, and MMPs in tumor tissues. Macrophage parameters, following incubation in G-CSF pre-treated conditioned medium, indicated enhanced tube-forming capabilities relative to the control, an effect mitigated by the introduction of specific inhibitors. Furthermore, the G-CSF group exhibited a notable reduction in SHP-2 expression, alongside a substantial elevation in the phosphorylation levels of the PI3K/AKT/mTOR pathway proteins across all tumor-bearing paradigms. CONCLUSION G-CSF ostensibly facilitates the advancement of hepatocellular carcinoma by activating the PI3K/AKT/mTOR signaling cascade within Tumor-Associated Macrophages (TAM).
Collapse
Affiliation(s)
- Heng Cao
- Department of Hepatobiliary Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Shunxiang Wang
- Department of Hepatobiliary Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
30
|
Mammoliti O, Martina S, Claes P, Coti G, Blanque R, Jagerschmidt C, Shoji K, Borgonovi M, De Vos S, Marsais F, Oste L, Quinton E, López-Ramos M, Amantini D, Brys R, Jimenez JM, Galien R, van der Plas S. Discovery of GLPG3667, a Selective ATP Competitive Tyrosine Kinase 2 Inhibitor for the Treatment of Autoimmune Diseases. J Med Chem 2024; 67:8545-8568. [PMID: 38805213 PMCID: PMC11181332 DOI: 10.1021/acs.jmedchem.4c00769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024]
Abstract
Tyrosine kinase 2 (TYK2) mediates cytokine signaling through type 1 interferon, interleukin (IL)-12/IL-23, and the IL-10 family. There appears to be an association between TYK2 genetic variants and inflammatory conditions, and clinical evidence suggests that selective inhibition of TYK2 could produce a unique therapeutic profile. Here, we describe the discovery of compound 9 (GLPG3667), a reversible and selective TYK2 adenosine triphosphate competitive inhibitor in development for the treatment of inflammatory and autoimmune diseases. The preclinical pharmacokinetic profile was favorable, and TYK2 selectivity was confirmed in peripheral blood mononuclear cells and whole blood assays. Dermal ear inflammation was reduced in an IL-23-induced in vivo mouse model of psoriasis. GLPG3667 also completed a phase 1b study (NCT04594928) in patients with moderate-to-severe psoriasis where clinical effect was shown within the 4 weeks of treatment and it is now in phase 2 trials for the treatment of dermatomyositis (NCT05695950) and systemic lupus erythematosus (NCT05856448).
Collapse
Affiliation(s)
- Oscar Mammoliti
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | | | - Pieter Claes
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - Ghjuvanni Coti
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - Roland Blanque
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | | - Kenji Shoji
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Monica Borgonovi
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Steve De Vos
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - Florence Marsais
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Line Oste
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - Evelyne Quinton
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | | - David Amantini
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Reginald Brys
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | | | - René Galien
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | |
Collapse
|
31
|
Grisé A, Valere LC, Weinstein D, Sami N. Janus kinase inhibitors in the treatment of pyoderma gangrenosum: case report and review. Arch Dermatol Res 2024; 316:238. [PMID: 38795155 DOI: 10.1007/s00403-024-02958-6] [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: 11/19/2023] [Revised: 04/23/2024] [Accepted: 04/26/2024] [Indexed: 05/27/2024]
Abstract
Pyoderma gangrenosum (PG) is a rare inflammatory dermatologic condition with neutrophilic infiltration of the skin that causes pustules and ulcerations. Janus kinase (JAK) inhibitors are immunomodulating agents that have been recently described in the literature as an effective treatment for PG. We describe a patient with PG on the lower extremities successfully treated with baricitinib. We also conducted a narrative review of the literature of PG patients treated with JAK inhibitors who were refractory to other treatments.
Collapse
Affiliation(s)
- Alison Grisé
- University of Central Florida College of Medicine, Orlando, FL, USA
| | | | - David Weinstein
- University of Central Florida College of Medicine, Orlando, FL, USA
- Florida State University College of Medicine, Tallahassee, FL, USA
| | - Naveed Sami
- University of Central Florida College of Medicine, Orlando, FL, USA.
- Department of Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, USA.
- Department of Medicine, Health Sciences Campus at Lake Nona, 32827-7408, Orlando, FL, USA.
| |
Collapse
|
32
|
Nguyen HN, Jeong Y, Kim Y, Kim YH, Athar H, Castaldi PJ, Hersh CP, Padera RF, Sholl LM, Vivero M, Sharma NS, Yun J, Merriam LT, Yuan K, Kim EY, Brenner MB. Leukemia inhibitory factor (LIF) receptor amplifies pathogenic activation of fibroblasts in lung fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595153. [PMID: 38826450 PMCID: PMC11142130 DOI: 10.1101/2024.05.21.595153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Fibrosis drives end-organ damage in many diseases. However, clinical trials targeting individual upstream activators of fibroblasts, such as TGFβ, have largely failed. Here, we target the leukemia inhibitory factor receptor (LIFR) as a "master amplifier" of multiple upstream activators of lung fibroblasts. In idiopathic pulmonary fibrosis (IPF), the most common fibrotic lung disease, we found that lung myofibroblasts had high LIF expression. Further, TGFβ1, one of the key drivers of fibrosis, upregulated LIF expression in IPF fibroblasts. In vitro anti-LIFR antibody blocking on human IPF lung fibroblasts reduced induction of profibrotic genes downstream of TGFβ1, IL-4 and IL-13. Further, siRNA silencing of LIFR in IPF precision cut lung slices reduced expression of fibrotic proteins. Together, we find that LIFR drives an autocrine positive feedback loop that amplifies and sustains pathogenic activation of IPF fibroblasts downstream of multiple external stimuli, implicating LIFR as a therapeutic target in fibrosis. Significance Statement Fibroblasts have a central role in the pathogenesis of fibrotic diseases. However, due to in part to multiple profibrotic stimuli, targeting a single activator of fibroblasts, like TGFβ, has not yielded successful clinical treatments. We hypothesized that a more effective therapeutic strategy is identifying a downstream "master amplifier" of a range of upstream profibrotic stimuli. This study identifies the leukemia inhibitory factor receptor (LIFR) on fibrotic lung fibroblasts amplifies multiple profibrotic stimuli, such as IL-13 and TGFβ. Blocking LIFR reduced fibrosis in ex vivo lung tissue from patients with idiopathic pulmonary fibrosis (IPF). LIFR, acting as a master amplifier downstream of fibroblast activation, offers an alternative therapeutic strategy for fibrotic diseases.
Collapse
|
33
|
Miller M, Patel AS, Pasternak B. Rescue therapy with upadacitinib in medically refractory pediatric ulcerative colitis. JPGN REPORTS 2024; 5:197-199. [PMID: 38756133 PMCID: PMC11093917 DOI: 10.1002/jpr3.12067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/13/2024] [Accepted: 03/06/2024] [Indexed: 05/18/2024]
Abstract
Approved options for advanced therapy in pediatric inflammatory bowel disease (IBD) are limited. Although Janus kinase (JAK) inhibitors are approved in adult IBD, their benefit in pediatric populations is not yet delineated. We present a 13-year-old female patient with ulcerative colitis (UC) refractory to numerous therapies and courses of prednisone that ultimately responded to a JAK inhibitor. Initial treatment consisted of 5-aminosalicylate and azathioprine. This was changed to adalimumab due to persistent symptoms. Repeat colonoscopy revealed pancolitis, thus she was transitioned to vedolizumab. She was hospitalized twice for uncontrolled symptoms on vedolizumab and subsequent scope showed continued pancolitis. As a result, she transitioned to ustekinumab without symptomatic relief after adjusting to monthly dosing. The family declined colectomy, opting to exhaust all medical therapies. Upadacitinib was started and her symptoms resolved within 1 week, and she remains in steroid-free remission. This case illustrates the possible role of JAK inhibitors in extensively refractory pediatric UC patients before colectomy.
Collapse
Affiliation(s)
- Maria Miller
- Division of Pediatric GastroenterologyPhoenix Children's HospitalPhoenixArizonaUSA
- Creighton University School of MedicinePhoenixArizonaUSA
| | - Ashish S. Patel
- Division of Pediatric GastroenterologyPhoenix Children's HospitalPhoenixArizonaUSA
- Creighton University School of MedicinePhoenixArizonaUSA
| | - Brad Pasternak
- Division of Pediatric GastroenterologyPhoenix Children's HospitalPhoenixArizonaUSA
- Creighton University School of MedicinePhoenixArizonaUSA
| |
Collapse
|
34
|
Hagino T, Hamada R, Yoshida M, Fujimoto E, Saeki H, Kanda N. Total eosinophil count as a biomarker for therapeutic effects of upadacitinib in atopic dermatitis over 48 weeks. Front Immunol 2024; 15:1365544. [PMID: 38745653 PMCID: PMC11091278 DOI: 10.3389/fimmu.2024.1365544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/18/2024] [Indexed: 05/16/2024] Open
Abstract
Background Atopic dermatitis (AD) is a chronic skin disease characterized by type 2-skewed immune responses, and significantly influenced by cytokines dependent on Janus kinases (JAKs). Upadacitinib, a JAK1 inhibitor, is effective for moderate-to-severe AD. This study aims to identify biomarkers that reflect long-term therapeutic effects of upadacitinib 15 mg or 30 mg. Methods A retrospective study from August 2021 to July 2023 included 213 AD patients treated with upadacitinib 15 mg and 70 AD patients with 30 mg. We analyzed eczema area and severity index (EASI), peak pruritus-numerical rating scale (PP-NRS), serum immunoglobulin E (IgE), thymus and activation-regulated chemokine (TARC), lactate dehydrogenase (LDH), and total eosinophil count (TEC) at weeks 0, 4, 12, 24, 36, and 48 of treatment. Results Both treatments with upadacitinib 15 mg and 30 mg significantly reduced EASI and PP-NRS scores over week 4 to 48 compared to baseline. Upadacitinib 15 mg or 30 mg treatment significantly decreased TEC compared to baseline through week 4 to 36 or week 4 to 48, respectively. The percent reduction of TEC correlated with those of EASI and PP-NRS through week 4 to 48 of treatment with upadacitinib 15 mg, or through week 12 to 48 with 30 mg, respectively. After adjusting for % reductions of other laboratory markers, the significance of correlations was preserved at weeks 36 and 48 of 15 mg treatment, while at weeks 4 and 36 of 30 mg treatment. Conclusion The % reduction of TEC correlated with those of EASI and PP-NRS during upadacitinib treatment, indicating its potential as a biomarker reflecting treatment responses to upadacitinib in AD patients. However, the variability of significant correlation during treatment indicates that further inspection is needed for its usefulness in monitoring responses to upadacitinib treatment for AD.
Collapse
Affiliation(s)
- Teppei Hagino
- Department of Dermatology, Nippon Medical School Chiba Hokusoh Hospital, Inzai, Japan
| | - Risa Hamada
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | - Mai Yoshida
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | | | - Hidehisa Saeki
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | - Naoko Kanda
- Department of Dermatology, Nippon Medical School Chiba Hokusoh Hospital, Inzai, Japan
| |
Collapse
|
35
|
Kwon OC, Choi W, Ahn SM, Oh JS, Hong S, Lee CK, Yoo B, Park MC, Kim YG. Drug survival and change of disease activity using a second janus kinase inhibitor in patients with difficult-to-treat rheumatoid arthritis who failed to a janus kinase inhibitor and subsequent biologics. Adv Rheumatol 2024; 64:26. [PMID: 38622706 DOI: 10.1186/s42358-024-00368-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 03/30/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND To assess the drug survival and change of disease activity using a second Janus kinase inhibitor (JAKi) after failure to a JAKi and subsequent biologic disease-modifying anti-rheumatic drugs (bDMARDs) in patients with difficult-to-treat rheumatoid arthritis (RA). METHODS This retrospective cohort study included 32 patients with difficult-to-treat RA who failed to a JAKi and subsequently to one or more bDMARDs and then switched to a second JAKi. To assess drug survival, electronic medical records of each patient were reviewed. Data on whether the second JAKi was discontinued, and the reasons for discontinuation were collected. The change of disease activity was assessed by analyzing changes in tender joint count (TJC), swollen joint count (SJC), patient's global assessment of disease activity on a visual-analogue scale (VAS), erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), Disease Activity Score for 28 joints with ESR (DAS28-ESR), and DAS28-CRP from baseline to that at six months from initiation of the second JAKi. RESULTS Overall, discontinuation of the second JAKi occurred in 20 (62.5%) patients. Primary failure, secondary failure, adverse events, and insurance coverage issues were the reasons for discontinuation in 9 (45.0%), 5 (25.0%), 2 (10.0%), and 4 (20.0%) patients, respectively. The estimated 2-year drug survival rate was 39.3%. In terms of change of disease activity, the second JAKi significantly improved TJC (p < 0.001), SJC (p < 0.001), VAS (p < 0.001), CRP (p = 0.026), DAS28-ESR (p < 0.001), and DAS28-CRP (p < 0.001) at 6-month compared with that at the baseline. CONCLUSIONS Second JAKi could be a therapeutic option in patients with difficult-to-treat RA who have failed to a JAKi and subsequent bDMARDs.
Collapse
Affiliation(s)
- Oh Chan Kwon
- Division of Rheumatology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonjuro, Gangnam- gu, 06273, Seoul, Korea
| | - Wonho Choi
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, University of Ulsan, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Korea
| | - Soo Min Ahn
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, University of Ulsan, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Korea
| | - Ji Seon Oh
- Department of Biomedical Informatics, Asan Medical Center, Seoul, Korea
| | - Seokchan Hong
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, University of Ulsan, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Korea
| | - Chang-Keun Lee
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, University of Ulsan, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Korea
| | - Bin Yoo
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, University of Ulsan, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Korea
| | - Min-Chan Park
- Division of Rheumatology, Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonjuro, Gangnam- gu, 06273, Seoul, Korea.
| | - Yong-Gil Kim
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, University of Ulsan, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, 05505, Seoul, Korea.
| |
Collapse
|
36
|
Hagino T, Hamada R, Yoshida M, Saeki H, Fujimoto E, Kanda N. Effectiveness of Dose Increase in Upadacitinib from 15 mg to 30 mg for Patients with Moderate-to-Severe Atopic Dermatitis: A Real-World Clinical Practice in Japan. Clin Drug Investig 2024; 44:261-269. [PMID: 38446396 DOI: 10.1007/s40261-024-01352-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Atopic dermatitis is characterized by persistent eczema and pruritus. Janus kinase inhibitors, including upadacitinib, are effective treatments for moderate-to-severe atopic dermatitis. If patients do not respond well to a certain dose of a Janus kinase inhibitor, increasing the dose may improve their treatment responsiveness. OBJECTIVES We assessed the outcomes of a dose increase in upadacitinib from 15 mg to 30 mg for Japanese patients with moderate-to-severe atopic dermatitis. METHODS In 23 patients who showed insufficient responses to upadacitinib 15-mg treatment, the dose of upadacitinib was increased to 30 mg. We evaluated total Eczema Area and Severity Index (EASI), EASI on the head and neck, trunk, upper, or lower limbs, EASI of erythema, edema/papulation, excoriation, or lichenification, and Peak Pruritus Numerical-Rating Scale at baseline (onset of upadactinib 15 mg), week 0 (time of increase), and weeks 4 and 12 after the increase. RESULTS Total EASI, EASI on each anatomical site, EASI of each clinical sign, and Peak Pruritus Numerical-Rating Scale were markedly reduced at weeks 4 or 12 compared with week 0. After the dose increase, the achievement rates of EASI 75 and EASI 90 significantly improved; EASI 75 4.3%, 68.2%, and 66.7%; EASI 90 0%, 18.2%, and 38.1% at weeks 0, 4, and 12, respectively. CONCLUSIONS These results suggest that upadacitinib 30 mg can ameliorate rash and pruritus insufficiently improved by upadacitinib 15 mg, and that the dose increase to 30 mg may be considered as a treatment option for patients with atopic dermatitis with a limited response to upadacitinib 15 mg.
Collapse
Affiliation(s)
- Teppei Hagino
- Department of Dermatology, Nippon Medical School, Chiba Hokusoh Hospital, Kamagari 1715, Inzai, Chiba, 270-1694, Japan.
| | - Risa Hamada
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | - Mai Yoshida
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | - Hidehisa Saeki
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | | | - Naoko Kanda
- Department of Dermatology, Nippon Medical School, Chiba Hokusoh Hospital, Kamagari 1715, Inzai, Chiba, 270-1694, Japan
| |
Collapse
|
37
|
Rosenberg FM, Kamali Z, Voorberg AN, Oude Munnink TH, van der Most PJ, Snieder H, Vaez A, Schuttelaar MLA. Transcriptomics- and Genomics-Guided Drug Repurposing for the Treatment of Vesicular Hand Eczema. Pharmaceutics 2024; 16:476. [PMID: 38675137 PMCID: PMC11054470 DOI: 10.3390/pharmaceutics16040476] [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: 02/19/2024] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Vesicular hand eczema (VHE), a clinical subtype of hand eczema (HE), showed limited responsiveness to alitretinoin, the only approved systemic treatment for severe chronic HE. This emphasizes the need for alternative treatment approaches. Therefore, our study aimed to identify drug repurposing opportunities for VHE using transcriptomics and genomics data. We constructed a gene network by combining 52 differentially expressed genes (DEGs) from a VHE transcriptomics study with 3 quantitative trait locus (QTL) genes associated with HE. Through network analysis, clustering, and functional enrichment analyses, we investigated the underlying biological mechanisms of this network. Next, we leveraged drug-gene interactions and retrieved pharmaco-transcriptomics data from the DrugBank database to identify drug repurposing opportunities for (V)HE. We developed a drug ranking system, primarily based on efficacy, safety, and practical and pricing factors, to select the most promising drug repurposing candidates. Our results revealed that the (V)HE network comprised 78 genes that yielded several biological pathways underlying the disease. The drug-gene interaction search together with pharmaco-transcriptomics lookups revealed 123 unique drug repurposing opportunities. Based on our drug ranking system, our study identified the most promising drug repurposing opportunities (e.g., vitamin D analogues, retinoids, and immunomodulating drugs) that might be effective in treating (V)HE.
Collapse
Affiliation(s)
- Fieke M. Rosenberg
- Department of Dermatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (F.M.R.); (A.N.V.)
| | - Zoha Kamali
- Department of Epidemiology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands (H.S.)
- Department of Bioinformatics, School of Advanced Medical Technologies, Isfahan University of Medical Sciences, Isfahan P.O. Box 81746-7346, Iran
| | - Angelique N. Voorberg
- Department of Dermatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (F.M.R.); (A.N.V.)
| | - Thijs H. Oude Munnink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands;
| | - Peter J. van der Most
- Department of Epidemiology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands (H.S.)
| | - Harold Snieder
- Department of Epidemiology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands (H.S.)
| | - Ahmad Vaez
- Department of Epidemiology, University Medical Centre Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands (H.S.)
- Department of Bioinformatics, School of Advanced Medical Technologies, Isfahan University of Medical Sciences, Isfahan P.O. Box 81746-7346, Iran
| | - Marie L. A. Schuttelaar
- Department of Dermatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (F.M.R.); (A.N.V.)
| |
Collapse
|
38
|
Inoue S, Suzuki T, Sano S, Katayama I. JAK inhibitors for the treatment of vitiligo. J Dermatol Sci 2024; 113:86-92. [PMID: 38326166 DOI: 10.1016/j.jdermsci.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 12/28/2023] [Indexed: 02/09/2024]
Abstract
Vitiligo is an autoimmune disease involving melanocyte-targeting T cells initiated by environmental and genetic factors. Steroids and tacrolimus have been used as topical treatments. Recently, novel topical agents targeting Janus kinase (JAK), a family of tyrosine kinases that regulates cytokine signaling, have emerged. Ruxolitinib is the first approved in vitiligo therapy. Furthermore, ritlecitinib is currently under clinical trials for oral treatment of active vitiligo. In this review, we discuss the possibility of topical JAK inhibitors as promising options for the treatment of vitiligo with regard to their mechanism of action, efficacy and safety.
Collapse
Affiliation(s)
- Shintaro Inoue
- Department of Cosmetic Health Science, Gifu Pharmaceutical University, Gifu, Japan.
| | - Tamio Suzuki
- Department of Dermatology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Shigetoshi Sano
- Department of Dermatology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Ichiro Katayama
- Department of Pigmentation Research and Therapeutics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
39
|
Xiong N, Sun Q. Identifying COVID-19 subtypes by single-sample gene set enrichemnt analysis and providing guidance for sensitive drug selection. J Med Virol 2024; 96:e29497. [PMID: 38436142 DOI: 10.1002/jmv.29497] [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: 11/07/2023] [Revised: 02/02/2024] [Accepted: 02/20/2024] [Indexed: 03/05/2024]
Abstract
This study aimed at using single-sample gene set enrichment analysis scores to cluster naso/pharyngeal swab specimen samples from coronavirus disease 2019 (COVID-19) patients into two clusters. One cluster with higher fractions of immune cells and more active inflammatory-related pathways was called the Immunity-High (Immunity-H) group, and the other one was called the Immunity-Low group. We explored impacts of the method on COVID-19 treatment. First, given that the Immunity-H group was mainly enriched in inflammatory-related pathways and had higher fractions of inflammatory cells, the Immunity-H group may obtain more curative effects from anti-inflammatory treatment. Second, we searched some hot genes from the PubMed platform that had been studied by researchers and found these genes upregulated in the Immunity-H group, so we speculated the Immunity-H group and Immunity-Low group may have different curative effects from drugs targeting these genes. Finally, we screened out hub genes for the Immunity-H group and predicted potential drugs for these hub genes by a public data set (http://dgidb.genome.wustl.edu). These hub genes are significantly upregulated in the Immunity-H group and neutrophils so that the Immunity-H group may obtain different treatment results from potential drugs compared with the Immunity-Low group. Therefore, the cluster method may provide help in drug development and administration for COVID-19 patients.
Collapse
Affiliation(s)
- Nan Xiong
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
- Graduate School of Kunming Medical University, Kunming, China
| | - Qiangming Sun
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, China
- Yunnan Key Laboratory of Vaccine Research & Development on Severe Infectious Diseases, Kunming, China
| |
Collapse
|
40
|
Selmi C, Chimenti MS, Novelli L, Parikh BK, Morello F, de Vlam K, Ciccia F. Pain in axial spondyloarthritis: role of the JAK/STAT pathway. Front Immunol 2024; 15:1341981. [PMID: 38464510 PMCID: PMC10921361 DOI: 10.3389/fimmu.2024.1341981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/05/2024] [Indexed: 03/12/2024] Open
Abstract
Axial spondyloarthritis (axSpA) is a chronic inflammatory disease that is characterized by new bone formation in the axial musculoskeletal system, with X-ray discriminating between radiographic and non-radiographic forms. Current therapeutic options include non-steroidal anti-inflammatory drugs in addition to biological disease-modifying anti-rheumatic drugs that specifically target tumor necrosis factor-alpha (TNFα) or interleukin (IL)-17. Pain is the most critical symptom for axSpA patients, significantly contributing to the burden of disease and impacting daily life. While the inflammatory process exerts a major role in determining pain in the early phases of the disease, the symptom may also result from mechanical and neuromuscular causes that require complex, multi-faceted pharmacologic and non-pharmacologic treatment, especially in the later phases. In clinical practice, pain often persists and does not respond further despite the absence of inflammatory disease activity. Cytokines involved in axSpA pathogenesis interact directly/indirectly with the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling cascade, a fundamental component in the origin and development of spondyloarthropathies. The JAK/STAT pathway also plays an important role in nociception, and new-generation JAK inhibitors have demonstrated rapid pain relief. We provide a comprehensive review of the different pain types observed in axSpA and the potential role of JAK/STAT signaling in this context, with specific focus on data from preclinical studies and data from clinical trials with JAK inhibitors.
Collapse
Affiliation(s)
- Carlo Selmi
- Department of Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Maria Sole Chimenti
- Rheumatology, Allergology and Clinical Immunology, Department of “Medicina dei Sistemi”, University of Rome Tor Vergata, Rome, Italy
| | | | - Bhumik K. Parikh
- Global Medical Affairs, AbbVie, Inc., Mettawa, IL, United States
| | | | - Kurt de Vlam
- Department of Rheumatology, University Hospital Leuven, Leuven, Belgium
- Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KULeuven, Leuven, Belgium
| | - Francesco Ciccia
- Department of Precision Medicine Napoli, Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
| |
Collapse
|
41
|
Charles-Schoeman C, Giles JT, Lane NE, Choy E, Furst DE, Vencovský J, Wilson AG, Burmester GR, Coombs D, Penn SK, Khan N, Yee JB, Rahawi K, McInnes IB. Impact of Upadacitinib on Laboratory Parameters and Related Adverse Events in Patients with RA: Integrated Data Up to 6.5 Years. Rheumatol Ther 2024; 11:157-175. [PMID: 38180720 PMCID: PMC10796877 DOI: 10.1007/s40744-023-00624-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/26/2023] [Indexed: 01/06/2024] Open
Abstract
INTRODUCTION Upadacitinib (UPA) is a Janus kinase inhibitor that has demonstrated efficacy in moderate-to-severe rheumatoid arthritis (RA) with an acceptable safety profile. We investigated laboratory parameter changes in UPA RA clinical trials. METHODS Pooled data from six randomized trials in the SELECT phase 3 program were included. Key laboratory parameters and safety data were measured for UPA 15 and 30 mg once daily (QD), adalimumab (ADA) 40 mg every other week + methotrexate (MTX), and MTX monotherapy. Exposure-adjusted event rates (EAERs) of adverse events were calculated. RESULTS A total of 3209 patients receiving UPA 15 mg QD (10 782.7 patient-years [PY]), 1204 patients receiving UPA 30 mg QD (3162.5 PY), 579 patients receiving ADA + MTX (1573.2 PY), and 314 patients receiving MTX monotherapy (865.1 PY) were included, representing up to 6.5 years of total exposure. Decreases in mean levels of hemoglobin, neutrophils, and lymphocytes, and increases in mean levels of liver enzymes and creatinine phosphokinase were observed with UPA, with grade 3 or 4 changes observed in some patients. Mean low- and high-density lipoprotein cholesterol ratios remained stable for patients receiving UPA 15 mg QD. EAERs of anemia and neutropenia occurred at generally consistent rates between UPA and active comparators (3.1-4.3 and 1.7-5.0 events [E]/100 PY across treatment groups, respectively). Rates of hepatic disorder were higher with MTX monotherapy, UPA 15 mg and UPA 30 mg (10.8, 9.7, and 11.0 E/100 PY, respectively) versus ADA + MTX (6.4 E/100 PY). Rates of lymphopenia were highest with MTX monotherapy (3.2 E/100 PY). Treatment discontinuations due to laboratory-related events were rare, occurring in 1.1% and 2.2% of patients treated with UPA 15 and 30 mg QD, respectively. CONCLUSIONS The results of this integrated long-term analysis of laboratory parameters continue to support an acceptable safety profile of UPA 15 mg QD for moderate-to-severe RA.
Collapse
Affiliation(s)
| | | | - Nancy E Lane
- University of California Davis, Sacramento, CA, USA
| | - Ernest Choy
- CREATE Centre, Cardiff University, Cardiff, UK
| | - Daniel E Furst
- Division of Rheumatology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Jiří Vencovský
- Department of Rheumatology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Institute of Rheumatology, Na Slupi 4, 12850, Prague, Czech Republic
| | - Anthony G Wilson
- Center for Arthritis Research, Conway Institute, University College Dublin, Dublin, Ireland
| | | | | | | | | | | | | | - Iain B McInnes
- College of Medical, Veterinary, and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
42
|
Scheepers L, Yang Y, Chen YL, Jones G. Persistence of Janus-kinase (JAK) inhibitors in rheumatoid arthritis: Australia wide study. Semin Arthritis Rheum 2024; 64:152314. [PMID: 38029717 DOI: 10.1016/j.semarthrit.2023.152314] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/27/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND To compare persistence of disease-modifying antirheumatic (DMARDs), with a focus on Janus kinase (JAK) inhibitors in Australian rheumatoid arthritis (RA) patients. METHODS A retrospective observational study was conducted among 4,521 RA patients (females n=3,181 [70.4%]), using data from the Services Australia 10% Pharmaceuticals Benefits Scheme (PBS) dataset, aged ≥18 years and initiating a DMARD between 2011 to 2021. Kaplan-Meier analysis was used to estimate persistence rates, defined as occurrence of 6 months gap after the end of a drug dispensing. RESULTS Twelve-month persistence rates were 72% for upadacitinib, 61% for baricitinib, 58% for subcutaneous tumor necrosis factor-alpha inhibitors (TNFi), 55% for tocilizumab, 53% for tofacitinib, and 49% for abatacept. Median treatment persistence was not reached for upadacitinib (n=574) and baricitinib (n=553); and was 15.0 months for tofacitinib (95% CI 13.5-19.5), 20.5 months for TNFi (95% CI 19.0-22.4), 19.1 months for tocilizumab (95% CI 17.9-23.6), and 12.5 months for abatacept (95% CI 10.4-14.9). Persistence rates on first-line JAK inhibitors were 68% for upadacitinib and baricitinib and 55% for tofacitinib, and 49% for TNFi, 55% for abatacept, and 57% for tocilizumab; rates were sustained for upadacitinib, TNFi, and tocilizumab but dropped to 59% for baricitinib and 47% for abatacept in the second-line treatment. For each b/tsDMARD, persistence rates were higher when combined with methotrexate or other conventional synthetic DMARDs. The median oral glucocorticoid dose decreased from 4.3 mg/day (range:0-40) to 2.3 mg/day (range:0-22) over 2 years. Changes were significant for all RA DMARDs, tofacitinib and baricitinib combined (1-2 years post initiation only), TNFi, abatacept, and tocilizumab. CONCLUSIONS In a real-world setting, we showed highest persistence rates on upadacitinib, followed by baricitinib and then TNFi therapy and was improved by co-therapy. All agents appeared to be corticosteroid sparing.
Collapse
Affiliation(s)
- Lieke Scheepers
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.
| | | | | | - Graeme Jones
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
43
|
Taylor PC, Choy E, Baraliakos X, Szekanecz Z, Xavier RM, Isaacs JD, Strengholt S, Parmentier JM, Lippe R, Tanaka Y. Differential properties of Janus kinase inhibitors in the treatment of immune-mediated inflammatory diseases. Rheumatology (Oxford) 2024; 63:298-308. [PMID: 37624925 PMCID: PMC10836981 DOI: 10.1093/rheumatology/kead448] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/03/2023] [Accepted: 08/13/2023] [Indexed: 08/27/2023] Open
Abstract
Janus kinases (JAKs) are a family of cytosolic tyrosine kinases that regulate cytokine signal transduction, including cytokines involved in a range of inflammatory diseases, such as RA, psoriasis, atopic dermatitis and IBD. Several small-molecule JAK inhibitors (JAKis) are now approved for the treatment of various immune-mediated inflammatory diseases. There are, however, key differences between these agents that could potentially translate into unique clinical profiles. Each JAKi has a unique chemical structure, resulting in a distinctive mode of binding within the catalytic cleft of the target JAK, and giving rise to distinct pharmacological characteristics. In addition, the available agents have differing selectivity for JAK isoforms, as well as off-target effects against non-JAKs. Other differences include effects on haematological parameters, DNA damage repair, reproductive toxicity and metabolism/elimination. Here we review the pharmacological profiles of the JAKis abrocitinib, baricitinib, filgotinib, peficitinib, tofacitinib and upadacitinib.
Collapse
Affiliation(s)
- Peter C Taylor
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Ernest Choy
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | | | - Zoltan Szekanecz
- Faculty of Medicine, Department of Rheumatology, University of Debrecen, Debrecen, Hungary
| | - Ricardo M Xavier
- Serviço de Reumatologia, Hospital de Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - John D Isaacs
- Translational and Clinical Research Institute, Newcastle University and Musculoskeletal Unit, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Julie M Parmentier
- Immunology Precision Medicine, AbbVie Bioresearch Center, Worcester, MA, USA
| | - Ralph Lippe
- AbbVie Deutschland GmbH & Co. KG, Wiesbaden, Germany
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
44
|
Gadina M. JAK inhibitors: Is specificity at all relevant? Semin Arthritis Rheum 2024; 64S:152327. [PMID: 38007359 PMCID: PMC10939910 DOI: 10.1016/j.semarthrit.2023.152327] [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: 08/17/2023] [Accepted: 09/23/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND Cytokines are soluble factors that affect host defense and maintain immune homeostasis. Altered cytokine production leads to a dysfunctional immune responses and immune-related diseases. Cytokines bind to specific receptors and trigger various intracellular signaling cascades and targeting cytokines and/or their receptors has been effective in treating inflammatory diseases. OBJECTIVES Type I and II cytokine receptors activate four Janus kinases (JAKs), namely JAK1, JAK2, JAK3 and TYK2 and targeting of these enzymes resulted in the development of successful drugs now referred as JAK inhibitors or JAKinibs. RESULTS JAKinibs can be divided in three "generations." First-generation JAKinibs, molecules acting in an orthosteric manner, inhibit multiple JAKs and interfere with the biologic activity of many factors. With the idea of reducing side effects, second-generation JAKinibs, still orthosteric ATP competitors, have been developed with increased selectivity towards one or two JAKs. Third-generation JAKinibs have exploited our increased understanding of the structure and function of JAK domains and are allosteric inhibitors as they bind to specific residues in the pseudokinase domain. These third generation JAKInb indeed seems to possess a better safety profile. Notably, inhibition of cytokine activity in specific tissues could be more important than selective enzymatic blockade and for this reason, topical, inhaled, or as a non-absorbable JAKinibs are also being developed. CONCLUSIONS While JAKinibs entered the clinical arena about ten years ago, our understanding of these drugs and their selectivity relative to their activity and safety is still incomplete. More research is therefore needed to achieve better usage of these class of drugs.
Collapse
Affiliation(s)
- Massimo Gadina
- Translational Immunology Section, Office of Science and Technology, National Institute of Arthritis Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
45
|
Zerrouk N, Alcraft R, Hall BA, Augé F, Niarakis A. Large-scale computational modelling of the M1 and M2 synovial macrophages in rheumatoid arthritis. NPJ Syst Biol Appl 2024; 10:10. [PMID: 38272919 PMCID: PMC10811231 DOI: 10.1038/s41540-024-00337-5] [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: 09/18/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
Macrophages play an essential role in rheumatoid arthritis. Depending on their phenotype (M1 or M2), they can play a role in the initiation or resolution of inflammation. The M1/M2 ratio in rheumatoid arthritis is higher than in healthy controls. Despite this, no treatment targeting specifically macrophages is currently used in clinics. Thus, devising strategies to selectively deplete proinflammatory macrophages and promote anti-inflammatory macrophages could be a promising therapeutic approach. State-of-the-art molecular interaction maps of M1 and M2 macrophages in rheumatoid arthritis are available and represent a dense source of knowledge; however, these maps remain limited by their static nature. Discrete dynamic modelling can be employed to study the emergent behaviours of these systems. Nevertheless, handling such large-scale models is challenging. Due to their massive size, it is computationally demanding to identify biologically relevant states in a cell- and disease-specific context. In this work, we developed an efficient computational framework that converts molecular interaction maps into Boolean models using the CaSQ tool. Next, we used a newly developed version of the BMA tool deployed to a high-performance computing cluster to identify the models' steady states. The identified attractors are then validated using gene expression data sets and prior knowledge. We successfully applied our framework to generate and calibrate the M1 and M2 macrophage Boolean models for rheumatoid arthritis. Using KO simulations, we identified NFkB, JAK1/JAK2, and ERK1/Notch1 as potential targets that could selectively suppress proinflammatory macrophages and GSK3B as a promising target that could promote anti-inflammatory macrophages in rheumatoid arthritis.
Collapse
Affiliation(s)
- Naouel Zerrouk
- GenHotel, Laboratoire Européen de Recherche Pour La Polyarthrite Rhumatoïde, University Paris-Saclay, University Evry, Evry, France
- Sanofi R&D Data and Data Science, Artificial Intelligence & Deep Analytics, Omics Data Science, 1, Av Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Rachel Alcraft
- Advanced Research Computing Centre, University College London, London, UK
| | - Benjamin A Hall
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - Franck Augé
- Sanofi R&D Data and Data Science, Artificial Intelligence & Deep Analytics, Omics Data Science, 1, Av Pierre Brossolette, 91385, Chilly-Mazarin, France
| | - Anna Niarakis
- GenHotel, Laboratoire Européen de Recherche Pour La Polyarthrite Rhumatoïde, University Paris-Saclay, University Evry, Evry, France.
- Lifeware Group, Inria Saclay, Palaiseau, France.
| |
Collapse
|
46
|
Beckett M, Dutz J, Huang K. Upadacitinib therapy in refractory inflammatory myositis: a case series of 10 patients. RMD Open 2024; 10:e003837. [PMID: 38242552 PMCID: PMC10806474 DOI: 10.1136/rmdopen-2023-003837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024] Open
Abstract
OBJECTIVES To evaluate the effectiveness and safety of upadacitinib in treatment-refractory inflammatory myositis. METHODS Patients with refractory inflammatory myositis treated with upadacitinib from a single urban centre in Vancouver, British Columbia, Canada, were included from September 2020 to June 2023. The medical records of these patients were retrospectively reviewed. RESULTS 10 total patients were identified for review, including 5 classic dermatomyositis (DM), 3 amyopathic DM (ADM) and 2 antisynthetase syndrome. The patients failed an average of four immunosuppressants before initiation of upadacitinib. Three had prior Janus kinase inhibitor therapy with tofacitinib. In the classic DM and ADM aggregate group, upadacitinib offered clinically and statistically significant cutaneous improvement. Lack of active muscle disease at baseline precluded analysis of the effect of upadacitinib on muscle weakness. Nine patients remained on upadacitinib at the end of the study period. One patient discontinued upadacitinib due to severe facial acne. CONCLUSION Upadacitinib appears to be effective in targeting cutaneous manifestations of refractory inflammatory DM. Further research is still needed to validate its efficacy on a broader population scale.
Collapse
Affiliation(s)
- Madelaine Beckett
- Internal Medicine, The University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada
| | - Jan Dutz
- Dermatology, The University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada
- Vancouver General Hospital, Vancouver, British Columbia, Canada
- Rheumatology, The University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada
| | - Kun Huang
- Vancouver General Hospital, Vancouver, British Columbia, Canada
- Rheumatology, The University of British Columbia Faculty of Medicine, Vancouver, British Columbia, Canada
- Medicine, Surrey Memorial Hospital, Surrey, British Columbia, Canada
- Arthritis Research Canada, Vancouver, British Columbia, Canada
| |
Collapse
|
47
|
Bonelli M, Kerschbaumer A, Kastrati K, Ghoreschi K, Gadina M, Heinz LX, Smolen JS, Aletaha D, O'Shea J, Laurence A. Selectivity, efficacy and safety of JAKinibs: new evidence for a still evolving story. Ann Rheum Dis 2024; 83:139-160. [PMID: 37923366 PMCID: PMC10850682 DOI: 10.1136/ard-2023-223850] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/18/2023] [Indexed: 11/07/2023]
Abstract
Fundamental insight gained over the last decades led to the discovery of cytokines as pivotal drivers of inflammatory diseases such as rheumatoid arthritis, psoriasis/psoriasis arthritis, inflammatory bowel diseases, atopic dermatitis and spondylarthritis. A deeper understanding of the pro-inflammatory and anti-inflammatory effects of various cytokines has prompted new cytokine-targeting therapies, which revolutionised the treatment options in the last years for patients with inflammatory disorders. Disease-associated immune responses typically involve a complex interplay of multiple cytokines. Therefore, blockade of one single cytokine does not necessarily lead to a persistent remission in all patients with inflammatory disorders and fostered new therapeutic strategies targeting intracellular pathways shared by multiple cytokines. By inhibiting JAK-STAT signalling pathways common to families of cytokines, JAK-inhibitors (JAKinibs) have created a new paradigm for the treatment of inflammatory diseases. Multiple agents have been approved for various disorders and more are being investigated for several new indications. Second-generation selective JAKinibs have been devised with the aim to achieve an increased selectivity and a possible reduced risk of side effects. In the current review, we will summarise the current body of evidence of pan versus selective JAKinibs and the most recent insights on new side effects and indications, including COVID-19.
Collapse
Affiliation(s)
- Michael Bonelli
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Kastriot Kastrati
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Massimo Gadina
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Leonhard X Heinz
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Josef S Smolen
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Daniel Aletaha
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - John O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Arian Laurence
- Translational Gastroenterology Unit, Department of Haematology, University College Hospital, UCLH Hospitals NHS Trust, University of Oxford, Oxford, UK
| |
Collapse
|
48
|
Zheng DY, Wang YN, Huang YH, Jiang M, Dai C. Effectiveness and safety of upadacitinib for inflammatory bowel disease: A systematic review and meta-analysis of RCT and real-world observational studies. Int Immunopharmacol 2024; 126:111229. [PMID: 37977068 DOI: 10.1016/j.intimp.2023.111229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/23/2023] [Accepted: 11/11/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Upadacitinib, a novel and selective inhibitor of Janus kinase 1, has demonstrated promising efficacy in managing inflammatory bowel disease (IBD). In this systematic review and meta-analysis, our primary aim was to comprehensively assess the therapeutic effectiveness and safety profile of upadacitinib in the treatment of patients with IBD. METHODS We conducted an extensive literature search across prominent databases, including Medline, Embase, Web of Science, and Cochrane Central, to identify pertinent studies providing insights into the efficacy and safety of upadacitinib in IBD. The primary endpoint was the achievement of clinical remission, while secondary endpoints encompassed clinical response, endoscopic response, endoscopic remission, and the evaluation of adverse events (AEs). RESULTS In this meta-analysis of nine studies, we categorized results by study type. Clinical remission rates were: RCTs 36 % (95 % CI = 30-42 %), real-world studies 25 % (95 % CI = 1-49 %), retrospective studies 40 % (95 % CI = 24-56 %), cohort studies 55 % (95 % CI = 25-85 %). Clinical response rates were: RCTs 61 % (95 % CI = 55-67 %), real-world studies 42 % (95 % CI = 14-70 %), cohort studies 65 % (95 % CI = 57-73 %). Endoscopic remission rates were: RCTs 19 % (95 % CI = 15-24 %), cohort studies 29 % (95 % CI = 5-52 %). Endoscopic response rates were: RCTs 41 % (95 % CI = 36-47 %), cohort studies 57 % (95 % CI = 31-83 %). Incidence rate for any AEs: IBD 69 % (95 % CI = 63-76 %), UC 65 % (95 % CI = 57-74 %), CD 75 % (95 % CI = 67-82 %). CONCLUSION Cumulative data from real-world studies and trials confirm the efficacy of upadacitinib in IBD induction and maintenance, with consistent safety. However, further long-term studies are needed to understand its sustained effectiveness and safety.
Collapse
Affiliation(s)
- Dian-Yu Zheng
- Department of Gastroenterology, First Hospital of China Medical University, Shenyang City, Liaoning Province, China
| | - Yi-Nuo Wang
- Department of Gastroenterology, First Hospital of China Medical University, Shenyang City, Liaoning Province, China
| | - Yu-Hong Huang
- Department of Gastroenterology, First Hospital of China Medical University, Shenyang City, Liaoning Province, China
| | - Min Jiang
- Department of Gastroenterology, First Hospital of China Medical University, Shenyang City, Liaoning Province, China
| | - Cong Dai
- Department of Gastroenterology, First Hospital of China Medical University, Shenyang City, Liaoning Province, China.
| |
Collapse
|
49
|
Faquetti ML, Slappendel L, Bigonne H, Grisoni F, Schneider P, Aichinger G, Schneider G, Sturla SJ, Burden AM. Baricitinib and tofacitinib off-target profile, with a focus on Alzheimer's disease. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12445. [PMID: 38528988 PMCID: PMC10962475 DOI: 10.1002/trc2.12445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/10/2023] [Accepted: 12/27/2023] [Indexed: 03/27/2024]
Abstract
INTRODUCTION Janus kinase (JAK) inhibitors were recently identified as promising drug candidates for repurposing in Alzheimer's disease (AD) due to their capacity to suppress inflammation via modulation of JAK/STAT signaling pathways. Besides interaction with primary therapeutic targets, JAK inhibitor drugs frequently interact with unintended, often unknown, biological off-targets, leading to associated effects. Nevertheless, the relevance of JAK inhibitors' off-target interactions in the context of AD remains unclear. METHODS Putative off-targets of baricitinib and tofacitinib were predicted using a machine learning (ML) approach. After screening scientific literature, off-targets were filtered based on their relevance to AD. Targets that had not been previously identified as off-targets of baricitinib or tofacitinib were subsequently tested using biochemical or cell-based assays. From those, active concentrations were compared to bioavailable concentrations in the brain predicted by physiologically based pharmacokinetic (PBPK) modeling. RESULTS With the aid of ML and in vitro activity assays, we identified two enzymes previously unknown to be inhibited by baricitinib, namely casein kinase 2 subunit alpha 2 (CK2-α2) and dual leucine zipper kinase (MAP3K12), both with binding constant (K d) values of 5.8 μM. Predicted maximum concentrations of baricitinib in brain tissue using PBPK modeling range from 1.3 to 23 nM, which is two to three orders of magnitude below the corresponding binding constant. CONCLUSION In this study, we extended the list of baricitinib off-targets that are potentially relevant for AD progression and predicted drug distribution in the brain. The results suggest a low likelihood of successful repurposing in AD due to low brain permeability, even at the maximum recommended daily dose. While additional research is needed to evaluate the potential impact of the off-target interaction on AD, the combined approach of ML-based target prediction, in vitro confirmation, and PBPK modeling may help prioritize drugs with a high likelihood of being effectively repurposed for AD. Highlights This study explored JAK inhibitors' off-targets in AD using a multidisciplinary approach.We combined machine learning, in vitro tests, and PBPK modelling to predict and validate new off-target interactions of tofacitinib and baricitinib in AD.Previously unknown inhibition of two enzymes (CK2-a2 and MAP3K12) by baricitinib were confirmed using in vitro experiments.Our PBPK model indicates that baricitinib low brain permeability limits AD repurposing.The proposed multidisciplinary approach optimizes drug repurposing efforts in AD research.
Collapse
Affiliation(s)
- Maria L. Faquetti
- Department of Chemistry and Applied BiosciencesInstitute of Pharmaceutical SciencesETH ZurichZurichSwitzerland
| | - Laura Slappendel
- Department of Health Sciences and TechnologyInstitute of Food, Nutrition and Health, ETH ZurichZurichSwitzerland
| | - Hélène Bigonne
- Department of Health Sciences and TechnologyInstitute of Food, Nutrition and Health, ETH ZurichZurichSwitzerland
| | - Francesca Grisoni
- Department of Biomedical EngineeringInstitute for Complex Molecular SystemsEindhoven University of TechnologyEindhoventhe Netherlands
- Centre for Living TechnologiesAlliance TU/e, WUR, UU, UMC UtrechtUtrechtthe Netherlands
| | - Petra Schneider
- Department of Chemistry and Applied BiosciencesInstitute of Pharmaceutical SciencesETH ZurichZurichSwitzerland
- inSili.com LLCZurichSwitzerland
| | - Georg Aichinger
- Department of Health Sciences and TechnologyInstitute of Food, Nutrition and Health, ETH ZurichZurichSwitzerland
| | - Gisbert Schneider
- Department of Chemistry and Applied BiosciencesInstitute of Pharmaceutical SciencesETH ZurichZurichSwitzerland
- ETH Singapore SEC LtdSingaporeSingapore
| | - Shana J. Sturla
- Department of Health Sciences and TechnologyInstitute of Food, Nutrition and Health, ETH ZurichZurichSwitzerland
| | - Andrea M. Burden
- Department of Chemistry and Applied BiosciencesInstitute of Pharmaceutical SciencesETH ZurichZurichSwitzerland
| |
Collapse
|
50
|
Dhir B, Meena SK, Sardana K, Sharath S. Use of tofacitinib in baricitinib-refractory atopic dermatitis: An example of JAK inhibitor switching. Pediatr Dermatol 2024; 41:139-140. [PMID: 37495552 DOI: 10.1111/pde.15386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/20/2023] [Indexed: 07/28/2023]
Abstract
Atopic dermatitis (AD) is predominantly mediated by a T-helper 2 immune system response. While JAK inhibitors have gained treatment approval for AD, data regarding interclass efficacy and therapeutic rationale is lacking. We report a patient with recalcitrant AD who failed baricitinib but demonstrated an excellent response to the pan-JAK inhibitor tofacitinib.
Collapse
Affiliation(s)
- Bhawuk Dhir
- Department of Dermatology, Venereology and Leprosy, Atal Bihari Vajpayee Institute of Medical Sciences and Research Institute and Dr Ram Manohar Lohia Hospital, New Delhi, India
| | - Sumitra Kumari Meena
- Department of Dermatology, Venereology and Leprosy, Atal Bihari Vajpayee Institute of Medical Sciences and Research Institute and Dr Ram Manohar Lohia Hospital, New Delhi, India
| | - Kabir Sardana
- Department of Dermatology, Venereology and Leprosy, Atal Bihari Vajpayee Institute of Medical Sciences and Research Institute and Dr Ram Manohar Lohia Hospital, New Delhi, India
| | - Savitha Sharath
- Department of Dermatology, Venereology and Leprosy, Atal Bihari Vajpayee Institute of Medical Sciences and Research Institute and Dr Ram Manohar Lohia Hospital, New Delhi, India
| |
Collapse
|