1
|
Karjalainen A, Witalisz-Siepracka A, Prchal-Murphy M, Martin D, Sternberg F, Krunic M, Dolezal M, Fortelny N, Farlik M, Macho-Maschler S, Lassnig C, Meissl K, Amenitsch L, Lederer T, Pohl E, Gotthardt D, Bock C, Decker T, Strobl B, Müller M. Cell-type-specific requirement for TYK2 in murine immune cells under steady state and challenged conditions. Cell Mol Life Sci 2025; 82:98. [PMID: 40025196 PMCID: PMC11872851 DOI: 10.1007/s00018-025-05625-9] [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: 11/12/2024] [Revised: 01/31/2025] [Accepted: 02/17/2025] [Indexed: 03/04/2025]
Abstract
Tyrosine kinase 2 (TYK2) deficiency and loss or inhibition of kinase activity in men and mice leads to similar immune compromised phenotypes, predominantly through impairment of interferon (IFN) and interleukin 12 family responses. Here we relate the transcriptome changes to phenotypical changes observed in TYK2-deficient (Tyk2-/-) and TYK2 kinase-inactive (Tyk2K923E) mice in naïve splenic immune cells and upon ex vivo IFN treatment or in vivo tumor transplant infiltration. The TYK2 activities under homeostatic and both challenged conditions are highly cell-type-specific with respect to quantity and quality of transcriptionally dependent genes. The major impact of loss of TYK2 protein or kinase activity in splenic homeostatic macrophages, NK and CD8+ T cells and tumor-derived cytolytic cells is on IFN responses. While reportedly TYK2 deficiency leads to partial impairment of IFN-I responses, we identified cell-type-specific IFN-I-repressed gene sets completely dependent on TYK2 kinase activity. Reported kinase-inactive functions of TYK2 relate to signaling crosstalk, metabolic functions and cell differentiation or maturation. None of these phenotypes relates to respective enriched gene sets in the TYK2 kinase-inactive cell types. Nonetheless, the scaffolding functions of TYK2 are capable to change transcriptional activities at single gene levels and chromatin accessibility at promoter-distal regions upon cytokine treatment most prominently in CD8+ T cells. The cell-type-specific transcriptomic and epigenetic effects of TYK2 shed new light on the biology of this JAK family member and are relevant for current and future treatment of autoimmune and inflammatory diseases with TYK2 inhibitors.
Collapse
Affiliation(s)
- Anzhelika Karjalainen
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Agnieszka Witalisz-Siepracka
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems an Der Donau, Austria
| | - Michaela Prchal-Murphy
- Pharmacology and Toxicology, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - David Martin
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Felix Sternberg
- Physiology and Biophysics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Department of Nutritional Sciences, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Milica Krunic
- Campus Tulln, University of Applied Sciences Wiener Neustadt, Wiener Neustadt, Austria
| | - Marlies Dolezal
- Platform Biostatistics and Bioinformatics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Nikolaus Fortelny
- Department of Biosciences and Medical Biology, Center for Tumor Biology and Immunology, Paris-Lodron University Salzburg, Salzburg, Austria
| | - Matthias Farlik
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sabine Macho-Maschler
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Caroline Lassnig
- Core Facility VetBiomodels, University of Veterinary Medicine, Vienna, Austria
| | - Katrin Meissl
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Lena Amenitsch
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Therese Lederer
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Elena Pohl
- Physiology and Biophysics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Dagmar Gotthardt
- Division Pharmacology, Karl Landsteiner University of Health Sciences, Krems an Der Donau, Austria
| | - Christoph Bock
- Cemm Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Institute of Artificial Intelligence, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Thomas Decker
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- Center for Molecular Biology, Department of Microbiology, Immunobiology and Genetics, University of Vienna, Vienna, Austria
| | - Birgit Strobl
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mathias Müller
- Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria.
| |
Collapse
|
2
|
Navid F, Chen L, Bowness P, Colbert RA. HLA-B27 and spondyloarthritis: at the crossroads of innate and adaptive immunity. Nat Rev Rheumatol 2025; 21:77-87. [PMID: 39623156 DOI: 10.1038/s41584-024-01189-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2024] [Indexed: 01/29/2025]
Abstract
HLA-B*27 confers a strong risk of developing spondyloarthritis (SpA), which includes axial SpA with or without peripheral arthritis, enthesitis, acute anterior uveitis and gastrointestinal inflammation. Although no definitive mechanism has been established to explain the role of this HLA class I protein in the pathogenesis of SpA, three main hypotheses have emerged. First is the idea that self-peptides displayed by HLA-B27 resemble microorganism-derived peptides, leading to the expansion of autoreactive CD8+ T cells that trigger disease. The second and third hypotheses focus on aberrant properties of HLA-B27, including its tendency to form cell-surface dimers that can activate innate killer immunoglobulin-like receptors on CD4+ T helper 17 cells, triggering the production of pathogenic cytokines. HLA-B27 also misfolds in the endoplasmic reticulum, which can activate the unfolded protein response, increasing IL-23 expression and thereby promoting the production of type 17 cytokines. HLA-B27 misfolding in mesenchymal stem cells has also been linked to enhanced bone formation by mesenchymal stem cell-derived osteoblasts, which could contribute to structural damage in axial SpA. In this Review we summarize prevailing ideas about the role of HLA-B27 in SpA, discuss the latest developments as well as the gaps in current knowledge, and provide recommendations for future research to address these unmet needs.
Collapse
Affiliation(s)
- Fatemeh Navid
- Pediatric Translational Research Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Liye Chen
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK
| | - Paul Bowness
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Oxford University, Oxford, UK
| | - Robert A Colbert
- Pediatric Translational Research Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
3
|
Raychaudhuri SP, Shah RJ, Banerjee S, Raychaudhuri SK. JAK-STAT Signaling and Beyond in the Pathogenesis of Spondyloarthritis and Their Clinical Significance. Curr Rheumatol Rep 2024; 26:204-213. [PMID: 38492148 PMCID: PMC11116266 DOI: 10.1007/s11926-024-01144-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2024] [Indexed: 03/18/2024]
Abstract
PURPOSE OF REVIEW Janus kinase-signal transducers and activators of transcription cell signaling proteins (JAK-STATs) play a key regulatory role in functioning of several inflammatory cytokines. JAK-STAT signaling proteins are the key regulators of the cytokine/cytokine receptor system involved in the pathogenesis of various autoimmune disease including spondyloarthritis (SpA). This article mainly highlights the JAK-STAT signaling system, its association with the relevant cytokine/cytokine-receptor system, and its regulatory role in pathogenesis of SpA. Also, we have briefly addressed the principle for the use JAKi in SpA and the current status of use of JAK inhibitors (JAKi) in SpA. RECENT FINDINGS Recent developments with newer JAK molecules as well as other molecules beyond JAK inhibitors are now an exciting field for the development of novel therapies for autoimmune diseases and various malignant conditions. In this article, we have provided a special emphasis on how various cell signaling systems beyond JAK/STAT pathway are relevant to SpA and have provided a comprehensive review on this upcoming field in respect to the novel TYK2 inhibitors, RORγT inhibitors, mTOR inhibitors, NGF inhibitors, and various STAT kinase inhibitors. SpA are a group of autoimmune diseases with multifactorial etiologies. SpA is linked with genetic predisposition, environmental risk factors, and the immune system-mediated systemic inflammation. Here, we have provided the regulatory role of JAK/STAT pathway and other intracellular signaling system in the pathogenesis of SpA and its therapeutic relevance.
Collapse
Affiliation(s)
- Siba P Raychaudhuri
- Department of Rheumatology, UC Davis Medical Center, Sacramento, CA, USA.
- VA Sacramento Medical Center, Department of Veterans Affairs, Northern California Health Care System, Mather, CA, USA.
- UC Davis School of Medicine, Davis, CA, USA.
| | - Ruchi J Shah
- Department of Rheumatology, UC Davis Medical Center, Sacramento, CA, USA
| | - Sneha Banerjee
- VA Sacramento Medical Center, Department of Veterans Affairs, Northern California Health Care System, Mather, CA, USA
| | - Smriti K Raychaudhuri
- VA Sacramento Medical Center, Department of Veterans Affairs, Northern California Health Care System, Mather, CA, USA
| |
Collapse
|
4
|
Morand E, Merola JF, Tanaka Y, Gladman D, Fleischmann R. TYK2: an emerging therapeutic target in rheumatic disease. Nat Rev Rheumatol 2024; 20:232-240. [PMID: 38467779 DOI: 10.1038/s41584-024-01093-w] [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: 02/09/2024] [Indexed: 03/13/2024]
Abstract
Tyrosine kinase 2 (TYK2) is a member of the JAK kinase family of intracellular signalling molecules. By participating in signalling pathways downstream of type I interferons, IL-12, IL-23 and IL-10, TYK2 elicits a distinct set of immune events to JAK1, JAK2 and JAK3. TYK2 polymorphisms have been associated with susceptibility to various rheumatic diseases including systemic lupus erythematosus and dermatomyositis. In vitro and animal studies substantiate these findings, highlighting a role for TYK2 in diseases currently managed by antagonists of cytokines that signal through TYK2. Various inhibitors of TYK2 have now been studied in human disease, and one of these inhibitors, deucravacitinib, has now been approved for the treatment of psoriasis. Phase II trials of deucravacitinib have also reported positive results in the treatment of psoriatic arthritis and systemic lupus erythematosus, with a preliminary safety profile that seems to differ from that of the JAK1, JAK2 and JAK3 inhibitors. Two other inhibitors of TYK2, brepocitinib and ropsacitinib, are also in earlier stages of clinical trials. Overall, TYK2 inhibitors hold promise for the treatment of a distinct spectrum of autoimmune diseases and could potentially have a safety profile that differs from other JAK inhibitors.
Collapse
Affiliation(s)
- Eric Morand
- Centre for Inflammatory Diseases, Monash University, and Department of Rheumatology, Monash Health, Clayton, Victoria, Australia.
| | - Joseph F Merola
- Department of Medicine, Division of Rheumatology and Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Dafna Gladman
- Schroeder Arthritis Institute, Krembil Research Institute, Toronto Western Hospital, Division of Rheumatology University of Toronto, Toronto, Ontario, Canada
| | - Roy Fleischmann
- Metroplex Clinical Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
5
|
Fukui S, Kawaai S, Sawada H, Kishimoto M. Upadacitinib for the treatment of adults with active non-radiographic axial spondyloarthritis (nr-axSpA). Expert Rev Clin Immunol 2024; 20:141-153. [PMID: 37955181 DOI: 10.1080/1744666x.2023.2282696] [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: 07/11/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023]
Abstract
INTRODUCTION Non-radiographic axial spondyloarthritis (nr-axSpA) is a chronic inflammatory condition with axial and peripheral musculoskeletal involvement, fulfilling criteria of axSpA in the absence of advanced radiographic sacroiliitis. While appropriate treatment is required for chronic pain and disability resulting from disease progression, the limited availability of treatment options becomes evident. Upadacitinib, an oral selective Janus kinase 1 inhibitor, was approved in Europe, the United States, and other countries for management of nr-axSpA with inadequate response to existing therapies. AREA COVERED This review summarizes essential drug profiles, efficacy, and safety of upadacitinib for nr-axSpA in conjunction with data pertaining to radiographic axSpA. EXPERT OPINION In a phase 3 trial, upadacitinib exhibited efficacy for patients with nr-axSpA, irrespective of prior exposures to biological disease-modifying antirheumatic drugs (bDMARDs). The safety profiles of upadacitinib in nr-axSpA mirrored those in other indications, underscoring its potential as a promising treatment option for nr-axSpA. Concurrently, physicians should be aware of the absence of real-world data, longitudinal efficacy and safety, direct comparative studies between upadacitinib and bDMARDs in nr-axSpA, and evidence for precision medicine to identify patients who may optimally benefit from upadacitinib over bDMARDs. Future research is imperative to facilitate the effective utilization of upadacitinib in daily clinical practice.
Collapse
Affiliation(s)
- Sho Fukui
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Emergency and General Medicine, Kyorin University School of Medicine, Tokyo, Japan
- Immuno-Rheumatology Center, St. Luke's International Hospital, Tokyo, Japan
| | - Satoshi Kawaai
- Immuno-Rheumatology Center, St. Luke's International Hospital, Tokyo, Japan
| | - Haruki Sawada
- Department of Medicine, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI, USA
| | - Mitsumasa Kishimoto
- Department of Nephrology and Rheumatology, Kyorin University School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Bernal-Alferes B, Gómez-Mosqueira R, Ortega-Tapia GT, Burgos-Vargas R, García-Latorre E, Domínguez-López ML, Romero-López JP. The role of γδ T cells in the immunopathogenesis of inflammatory diseases: from basic biology to therapeutic targeting. J Leukoc Biol 2023; 114:557-570. [PMID: 37040589 DOI: 10.1093/jleuko/qiad046] [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: 07/15/2022] [Revised: 04/01/2023] [Accepted: 04/06/2023] [Indexed: 04/13/2023] Open
Abstract
The γδ T cells are lymphocytes with an innate-like phenotype that can distribute to different tissues to reside and participate in homeostatic functions such as pathogen defense, tissue modeling, and response to stress. These cells originate during fetal development and migrate to the tissues in a TCR chain-dependent manner. Their unique manner to respond to danger signals facilitates the initiation of cytokine-mediated diseases such as spondyloarthritis and psoriasis, which are immune-mediated diseases with a very strong link with mucosal disturbances, either in the skin or the gut. In spondyloarthritis, γδ T cells are one of the main sources of IL-17 and, therefore, the main drivers of inflammation and probably new bone formation. Remarkably, this population can be the bridge between gut and joint inflammation.
Collapse
Affiliation(s)
- Brian Bernal-Alferes
- Laboratorio de Inmunoquímica 1, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Lázaro Cárdenas, Prolongación de Carpio y Plan de Ayala s/n, Col. Santo Tomás C.P. 11340 Alcaldía Miguel Hidalgo, Ciudad de México, México
| | - Rafael Gómez-Mosqueira
- Laboratorio de Inmunoquímica 1, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Lázaro Cárdenas, Prolongación de Carpio y Plan de Ayala s/n, Col. Santo Tomás C.P. 11340 Alcaldía Miguel Hidalgo, Ciudad de México, México
| | - Graciela Teresa Ortega-Tapia
- Laboratorio de Inmunoquímica 1, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Lázaro Cárdenas, Prolongación de Carpio y Plan de Ayala s/n, Col. Santo Tomás C.P. 11340 Alcaldía Miguel Hidalgo, Ciudad de México, México
| | - Rubén Burgos-Vargas
- Departamento de Reumatología, Hospital General de México "Dr. Eduardo Liceaga", Dr. Balmis No. 148 Col. Doctores C.P. 06720, Alcaldía Cuauhtémoc Ciudad de México, México
| | - Ethel García-Latorre
- Laboratorio de Inmunoquímica 1, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Lázaro Cárdenas, Prolongación de Carpio y Plan de Ayala s/n, Col. Santo Tomás C.P. 11340 Alcaldía Miguel Hidalgo, Ciudad de México, México
| | - María Lilia Domínguez-López
- Laboratorio de Inmunoquímica 1, Posgrado en Ciencias Quimicobiológicas, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Lázaro Cárdenas, Prolongación de Carpio y Plan de Ayala s/n, Col. Santo Tomás C.P. 11340 Alcaldía Miguel Hidalgo, Ciudad de México, México
| | - José Pablo Romero-López
- Laboratorio de Patogénesis Molecular, Edificio A4, Red MEDICI, Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios Número 1, Colonia Los Reyes Ixtacala, C.P. 54090, Tlalnepantla, Estado de México, México
| |
Collapse
|
7
|
Yudhani RD, Pakha DN, Suyatmi S, Irham LM. Identifying pathogenic variants related to systemic lupus erythematosus by integrating genomic databases and a bioinformatic approach. Genomics Inform 2023; 21:e37. [PMID: 37813633 PMCID: PMC10584638 DOI: 10.5808/gi.23002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 06/15/2023] [Accepted: 08/09/2023] [Indexed: 10/11/2023] Open
Abstract
Systemic lupus erythematosus (SLE) is an inflammatory-autoimmune disease with a complex multi-organ pathogenesis, and it is known to be associated with significant morbidity and mortality. Various genetic, immunological, endocrine, and environmental factors contribute to SLE. Genomic variants have been identified as potential contributors to SLE susceptibility across multiple continents. However, the specific pathogenic variants that drive SLE remain largely undefined. In this study, we sought to identify these pathogenic variants across various continents using genomic and bioinformatic-based methodologies. We found that the variants rs35677470, rs34536443, rs17849502, and rs13306575 are likely damaging in SLE. Furthermore, these four variants appear to affect the gene expression of NCF2, TYK2, and DNASE1L3 in whole blood tissue. Our findings suggest that these genomic variants warrant further research for validation in functional studies and clinical trials involving SLE patients. We conclude that the integration of genomic and bioinformatic-based databases could enhance our understanding of disease susceptibility, including that of SLE.
Collapse
Affiliation(s)
- Ratih Dewi Yudhani
- Department of Pharmacology, Faculty of Medicine, Universitas Sebelas Maret, Surakarta 57126, Indonesia
| | - Dyonisa Nasirochmi Pakha
- Department of Pharmacology, Faculty of Medicine, Universitas Sebelas Maret, Surakarta 57126, Indonesia
| | - Suyatmi Suyatmi
- Department of Histology, Faculty of Medicine, Universitas Sebelas Maret, Surakarta 57126, Indonesia
| | | |
Collapse
|
8
|
Leit S, Greenwood J, Carriero S, Mondal S, Abel R, Ashwell M, Blanchette H, Boyles NA, Cartwright M, Collis A, Feng S, Ghanakota P, Harriman GC, Hosagrahara V, Kaila N, Kapeller R, Rafi SB, Romero DL, Tarantino PM, Timaniya J, Toms AV, Wester RT, Westlin W, Srivastava B, Miao W, Tummino P, McElwee JJ, Edmondson SD, Masse CE. Discovery of a Potent and Selective Tyrosine Kinase 2 Inhibitor: TAK-279. J Med Chem 2023; 66:10473-10496. [PMID: 37427891 DOI: 10.1021/acs.jmedchem.3c00600] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
TYK2 is a key mediator of IL12, IL23, and type I interferon signaling, and these cytokines have been implicated in the pathogenesis of multiple inflammatory and autoimmune diseases such as psoriasis, rheumatoid arthritis, lupus, and inflammatory bowel diseases. Supported by compelling data from human genome-wide association studies and clinical results, TYK2 inhibition through small molecules is an attractive therapeutic strategy to treat these diseases. Herein, we report the discovery of a series of highly selective pseudokinase (Janus homology 2, JH2) domain inhibitors of TYK2 enzymatic activity. A computationally enabled design strategy, including the use of FEP+, was instrumental in identifying a pyrazolo-pyrimidine core. We highlight the utility of computational physics-based predictions used to optimize this series of molecules to identify the development candidate 30, a potent, exquisitely selective cellular TYK2 inhibitor that is currently in Phase 2 clinical trials for the treatment of psoriasis and psoriatic arthritis.
Collapse
Affiliation(s)
- Silvana Leit
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Jeremy Greenwood
- Schrödinger, Inc., 1540 Broadway, New York, New York 10036, United States
| | - Samantha Carriero
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Sayan Mondal
- Schrödinger, Inc., 1540 Broadway, New York, New York 10036, United States
| | - Robert Abel
- Schrödinger, Inc., 1540 Broadway, New York, New York 10036, United States
| | - Mark Ashwell
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Heather Blanchette
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Nicholas A Boyles
- Schrödinger, Inc., 1540 Broadway, New York, New York 10036, United States
| | - Mark Cartwright
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Alan Collis
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Shulu Feng
- Schrödinger, Inc., 1540 Broadway, New York, New York 10036, United States
| | - Phani Ghanakota
- Schrödinger, Inc., 1540 Broadway, New York, New York 10036, United States
| | - Geraldine C Harriman
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Vinayak Hosagrahara
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Neelu Kaila
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Rosanna Kapeller
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Salma B Rafi
- Schrödinger, Inc., 1540 Broadway, New York, New York 10036, United States
| | - Donna L Romero
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Paul M Tarantino
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Jignesh Timaniya
- Piramal Pharma Solutions, Plot No. 18, Pharmez, Ahmedabad 382215, Gujarat, India
| | - Angela V Toms
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Ronald T Wester
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - William Westlin
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Bhaskar Srivastava
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Wenyan Miao
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Peter Tummino
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Joshua J McElwee
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Scott D Edmondson
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| | - Craig E Masse
- Nimbus Therapeutics, 22 Boston Wharf Road, Floor 9, Boston, Massachusetts 02210, United States
| |
Collapse
|
9
|
Abstract
Over the past two decades, advancements in understanding the pathogenesis of axial spondyloarthritis have led to discoveries of new therapeutic targets, particularly the interleukin-17, tumor necrosis factor axis, and Janus kinase-signal transducer and activator of transcription pathway. While many of the available agents have proven to be efficacious and safe for the treatment of axial spondyloarthritis, a remarkable percentage of patients either fail or cannot tolerate these medications. This has prompted researchers to look for new targets that would maximize efficacy and minimize toxicity. In this article, we review novel agents that were recently approved, in trials, and possible future targets or mechanisms. We also discuss their role as it pertains to the prevention of radiographic progression and the management of extra-musculoskeletal manifestations.
Collapse
Affiliation(s)
- Mohamad Bittar
- The University of Tennessee Health Science Center, Division of Connective Tissue Disease (Rheumatology), 956 Court Avenue, Coleman Building, Suite G326, Memphis, TN 38163, USA.
| | - Philip Mease
- Swedish Medical Center/Providence St. Joseph Health and University of Washington School of Medicine, Seattle Rheumatology Associates, 601 Broadway, Suite 600, Seattle, WA 98102, USA.
| |
Collapse
|
10
|
Sen R, Caplan L. Current treatment and molecular targets for axial spondyloarthritis: Evidence from randomized controlled trials. Curr Opin Pharmacol 2022; 67:102307. [PMID: 36335714 DOI: 10.1016/j.coph.2022.102307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022]
Abstract
Axial spondyloarthritis (axSpA) is a chronic inflammatory disease that predominantly affects the axial skeleton and is characterized by inflammatory back pain. While much has been published regarding non-steroidal anti-inflammatory drugs and tumor necrosis factor inhibitors, other classes of medications which leverage alternate molecular mechanisms receive less attention. In this review, we summarize a few of the novel targets in axSpA, review the putative mechanism of action of therapies that focus on these targets, and reference the germane recently completed, ongoing, or proposed randomized controlled clinical trials. The agents addressed include inhibitors of interleukin-23, interleukin-17, janus kinases, granulocyte-macrophage colony-stimulating factor, macrophage migration inhibitory factor, antibodies recognizing T cell receptor beta variable 9 gene positive clones, as well as inhibitors of mitogen-activated protein kinase-activated protein kinase-2.
Collapse
Affiliation(s)
- Rouhin Sen
- Rocky Mountain Regional Veterans Affairs Medical Center (VAMC), Denver, CO, USA; University of Colorado School of Medicine, Aurora, CO, USA
| | - Liron Caplan
- Rocky Mountain Regional Veterans Affairs Medical Center (VAMC), Denver, CO, USA; University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
11
|
Kenyon M, Maguire S, Rueda Pujol A, O'Shea F, McManus R. The genetic backbone of ankylosing spondylitis: how knowledge of genetic susceptibility informs our understanding and management of disease. Rheumatol Int 2022; 42:2085-2095. [PMID: 35939079 PMCID: PMC9548471 DOI: 10.1007/s00296-022-05174-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/20/2022] [Indexed: 11/25/2022]
Abstract
Ankylosing spondylitis (AS) is a seronegative, chronic inflammatory arthritis with high genetic burden. A strong association with HLA-B27 has long been established, but to date its contribution to disease aetiology remains unresolved. Recent insights through genome wide studies reveal an increasing array of immunogenetic risk variants extraneous to the HLA complex in AS cohorts. These genetic traits build a complex profile of disease causality, highlighting several molecular pathways associated with the condition. This and other evidence strongly implicates T-cell-driven pathology, revolving around the T helper 17 cell subset as an important contributor to disease. This prominence of the T helper 17 cell subset has presented the opportunity for therapeutic intervention through inhibition of interleukins 17 and 23 which drive T helper 17 activity. While targeting of interleukin 17 has proven effective, this success has not been replicated with interleukin 23 inhibition in AS patients. Evidence points to significant genetic diversity between AS patients which may, in part, explain the observed refractoriness among a proportion of patients. In this review we discuss the impact of genetics on our understanding of AS and its relationship with closely linked pathologies. We further explore how genetics can be used in the development of therapeutics and as a tool to assist in the diagnosis and management of patients. This evidence indicates that genetic profiling should play a role in the clinician’s choice of therapy as part of a precision medicine strategy towards disease management.
Collapse
Affiliation(s)
- Marcus Kenyon
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.
| | - Sinead Maguire
- Department of Rheumatology, St James' Hospital, Dublin, Ireland
| | - Anna Rueda Pujol
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Finbar O'Shea
- Department of Rheumatology, St James' Hospital, Dublin, Ireland
| | - Ross McManus
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
12
|
Leit S, Greenwood JR, Mondal S, Carriero S, Dahlgren M, Harriman GC, Kennedy-Smith JJ, Kapeller R, Lawson JP, Romero DL, Toms AV, Shelley M, Wester RT, Westlin W, McElwee JJ, Miao W, Edmondson SD, Masse CE. Potent and selective TYK2-JH1 inhibitors highly efficacious in rodent model of psoriasis. Bioorg Med Chem Lett 2022; 73:128891. [PMID: 35842205 DOI: 10.1016/j.bmcl.2022.128891] [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: 05/16/2022] [Revised: 07/02/2022] [Accepted: 07/09/2022] [Indexed: 11/17/2022]
Abstract
TYK2 is a member of the JAK family of kinases and a key mediator of IL-12, IL-23, and type I interferon signaling. These cytokines have been implicated in the pathogenesis of multiple inflammatory and autoimmune diseases such as psoriasis, rheumatoid arthritis, lupus, and inflammatory bowel diseases. Supported by compelling data from human genetic association studies, TYK2 inhibition is an attractive therapeutic strategy for these diseases. Herein, we report the discovery of a series of highly selective catalytic site TYK2 inhibitors designed using FEP+ and structurally enabled design starting from a virtual screen hit. We highlight the structure-based optimization to identify a lead candidate 30, a potent cellular TYK2 inhibitor with excellent selectivity, pharmacokinetic properties, and in vivo efficacy in a mouse psoriasis model.
Collapse
Affiliation(s)
- Silvana Leit
- Nimbus Therapeutics, 130 Prospect St., Cambridge, MA 02139, United States.
| | | | - Sayan Mondal
- Schrodinger, Inc., 1540 Broadway, New York, NY 10036, United States
| | - Samantha Carriero
- Nimbus Therapeutics, 130 Prospect St., Cambridge, MA 02139, United States
| | - Markus Dahlgren
- Schrodinger, Inc., 1540 Broadway, New York, NY 10036, United States
| | | | | | - Rosana Kapeller
- Schrodinger, Inc., 1540 Broadway, New York, NY 10036, United States
| | - Jon P Lawson
- Nimbus Therapeutics, 130 Prospect St., Cambridge, MA 02139, United States
| | - Donna L Romero
- Nimbus Therapeutics, 130 Prospect St., Cambridge, MA 02139, United States
| | - Angela V Toms
- Nimbus Therapeutics, 130 Prospect St., Cambridge, MA 02139, United States
| | - Mee Shelley
- Schrodinger, Inc., 1540 Broadway, New York, NY 10036, United States
| | - Ronald T Wester
- Nimbus Therapeutics, 130 Prospect St., Cambridge, MA 02139, United States
| | - William Westlin
- Nimbus Therapeutics, 130 Prospect St., Cambridge, MA 02139, United States
| | - Joshua J McElwee
- Nimbus Therapeutics, 130 Prospect St., Cambridge, MA 02139, United States
| | - Wenyan Miao
- Nimbus Therapeutics, 130 Prospect St., Cambridge, MA 02139, United States
| | - Scott D Edmondson
- Nimbus Therapeutics, 130 Prospect St., Cambridge, MA 02139, United States
| | - Craig E Masse
- Nimbus Therapeutics, 130 Prospect St., Cambridge, MA 02139, United States
| |
Collapse
|
13
|
Sun L, Wang B, Sun T, Zhou F, Zhu B, Li C, Wan H, Ding Z. Investigation on the mechanism of 2,3,4',5-Tetrahydroxystilbene 2-o-D-glucoside in the treatment of inflammation based on network pharmacology. Comput Biol Med 2022; 145:105448. [PMID: 35364310 DOI: 10.1016/j.compbiomed.2022.105448] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Inflammation is the pathogenesis of various chronic diseases plaguing clinic for years.Fallopia multiflora (Thunb.) is a traditional Chinese herbal medicine with a long history of application in detoxification and anti-inflammation. 2,3,4',5-Tetrahydroxystilbene 2-o-D-glucoside (TSG) is a main active compound of F. multiflora. However, the mechanism of TSG in the treatment of inflammation remains unknown. METHODS Network pharmacology and molecular docking were employed to explore the mechanism of anti-inflammatory effect of TSG. Potential targets of TSG and inflammation were obtained from Swiss Target Prediction, Pharm Mapper, and GeneCards database. Protein-protein interaction (PPI) networks, GO and KEGG pathway enrichment analysis were performed to elucidate the interaction of targets. Moreover, the anti-inflammatory effect of TSG was validated by in vitro experiments using flow cytometry, RT-qPCR, Western blot, and immunocytochemistry assays. RESULTS PPI network and gene enrichment analysis showed that TSG may exert a protein kinase binding activity, and IKBKB, MAPK1, NFKBIA, and RELA were predicted as the targets of anti-inflammation. Verified by molecular docking and Western blot, TSG may target NF-κB and ERK2 related signals to alleviate inflammatory damage. Furthermore, TSG effectively downregulated the expression of inflammatory cytokine, the nuclear translocation of NF-κB p65, and the production of reactive oxygen species (ROS). CONCLUSION TSG possesses significant anti-inflammatory effect. TSG may display a protein kinase binding activity and target NF-κB and ERK2 related signals to treat the inflammation. This work may enlighten the potential application of TSG in anti-inflammation and indicate network pharmacology was an effective tool for the further study of TCM.
Collapse
Affiliation(s)
- Ling Sun
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China.
| | - Bixu Wang
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China.
| | - Tong Sun
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China.
| | - Fangmei Zhou
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China.
| | - Bingqi Zhu
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China.
| | - Chang Li
- Institute of Cardio-cerebrovascular Disease, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China.
| | - Haitong Wan
- Institute of Cardio-cerebrovascular Disease, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China.
| | - Zhishan Ding
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, People's Republic of China.
| |
Collapse
|
14
|
Nielsen OH, Boye TL, Chakravarti D, Gubatan J. Selective tyrosine kinase 2 inhibitors in inflammatory bowel disease. Trends Pharmacol Sci 2022; 43:424-436. [PMID: 35277286 DOI: 10.1016/j.tips.2022.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 11/26/2022]
Abstract
Recent significant advances have been made in the treatment of chronic inflammatory diseases with initiation of the era of biologics. However, an unmet medical need still exists for novel targeted therapies. Compared with biologics, Janus kinase inhibitors (JAKis) are a new drug class of orally administered small molecules that have been shown to efficiently modulate complex cytokine-driven inflammation in preclinical models and human studies. Unfortunately, serious adverse effects have been reported with the first introduced pan-JAKi, tofacitinib. Here, we review tyrosine kinase 2 (TYK2) signaling in the pathophysiology of inflammatory bowel disease (IBD), examine mechanisms of action of selective TYK2 inhibitors (TYK2is), and discuss the potential for these inhibitors in efforts to balance benefits and harms.
Collapse
Affiliation(s)
- Ole Haagen Nielsen
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark.
| | - Theresa Louise Boye
- Department of Gastroenterology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Deepavali Chakravarti
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Gubatan
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
15
|
Toussirot E. The Use of Janus Kinase Inhibitors in Axial Spondyloarthritis: Current Insights. Pharmaceuticals (Basel) 2022; 15:ph15030270. [PMID: 35337068 PMCID: PMC8951918 DOI: 10.3390/ph15030270] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/17/2022] [Indexed: 01/24/2023] Open
Abstract
Current pharmacological treatments of axial spondyloarthritis (axSpA) are limited to non-steroidal anti-inflammatory drugs (NSAIDs) and biological agents, including TNFα inhibitors and IL-17 inhibitors. Despite the availability of these agents, many patients either fail to respond adequately, lose their initial therapeutic response over time, or develop undesirable side effects, thus highlighting the need for new treatment options. Janus kinase (JAK) and signal transducers and activators of transcription (STAT) are a group of intracellular kinases that play a role in the signaling pathway induced by cytokines and certain growth factors associated with the inflammatory process of axSpA. There are several lines of evidence implicating the JAK–STAT pathway in the pathophysiological process of axSpA, including genetic data, the use of certain JAK in the intracellular signal of specific cytokines involved in axSpA (IL-23, IL-22, and IL-6), and data from experimental models of SpA. This provides a rationale for the assessment of JAK inhibitors (JAKi) in clinical trials with patients with axSpA. In this review, we examine the role of JAK–STAT signaling in the pathogenesis of axSpA and summarize the results from recent clinical trials of JAKi (tofacitinib, upadacitinib, and filgotinib) in patients with axSpA.
Collapse
Affiliation(s)
- Eric Toussirot
- INSERM CIC-1431, Centre d’Investigation Clinique, Pôle Recherche, CHU de Besançon, 25000 Besançon, France;
- Rhumatologie, Pôle PACTE (Pathologies Aiguës Chroniques Transplantation Éducation), CHU de Besançon, 25000 Besançon, France
- Département Universitaire de Thérapeutique, Université de Bourgogne Franche-Comté, 25000 Besançon, France
- INSERM UMR1098 Right “Relations Hôte Greffon Tumeurs, Ingénierie Cellulaire et Génique”, Université de Bourgogne Franche-Comté, 25000 Besançon, France
| |
Collapse
|
16
|
Orozco G. Fine mapping with epigenetic information and 3D structure. Semin Immunopathol 2022; 44:115-125. [PMID: 35022890 PMCID: PMC8837508 DOI: 10.1007/s00281-021-00906-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022]
Abstract
Since 2005, thousands of genome-wide association studies (GWAS) have been published, identifying hundreds of thousands of genetic variants that increase risk of complex traits such as autoimmune diseases. This wealth of data has the potential to improve patient care, through personalized medicine and the identification of novel drug targets. However, the potential of GWAS for clinical translation has not been fully achieved yet, due to the fact that the functional interpretation of risk variants and the identification of causal variants and genes are challenging. The past decade has seen the development of great advances that are facilitating the overcoming of these limitations, by utilizing a plethora of genomics and epigenomics tools to map and characterize regulatory elements and chromatin interactions, which can be used to fine map GWAS loci, and advance our understanding of the biological mechanisms that cause disease.
Collapse
Affiliation(s)
- Gisela Orozco
- Centre for Genetics and Genomics Versus Arthritis, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9LJ, UK. .,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|