1
|
van der Werf J, Fleming NI. Are single nucleotide polymorphisms underutilized for guiding treatment of inflammatory bowel disease? Immunol Cell Biol 2025. [PMID: 40313162 DOI: 10.1111/imcb.70029] [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: 11/25/2024] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 05/03/2025]
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD), ulcerative colitis (UC) and IBD unclassified (IBDU), significantly impacts quality of life. Despite significant advances in the management of the conditions, responses to treatments vary greatly, and this is due partly to our natural genetic variation. Here we will review the evidence for whether single nucleotide polymorphisms (SNPs) have the potential to guide treatment decisions for people with IBD. We will first consider SNPs that exhibit strong associations with IBD pathogenesis and their relevance to epithelial barrier integrity, cytokine production, and immune system function. Then, we will cover those SNPs implicated in altering response to our various current IBD therapeutics, including the recently implemented drugs ustekinumab and tofacitinib. Finally, we will explore lesser-known SNPs that exhibit complex relationships with the disease and which may be undervalued as pharmacogenetic tools. Overall, it will be demonstrated that SNPs associated with IBD pathology are largely distinct from those predicting response to treatments and that new discoveries of clinically useful tools can be expected from therapy-focused investigations. Given the growing list of treatments available, we argue that beneficial personalization of treatments based on SNPs is still underutilized.
Collapse
Affiliation(s)
| | - Nicholas Ian Fleming
- Department of Pathology, University of Otago, Dunedin, New Zealand
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
2
|
Tanaka Y, Yamanaka H, Toyoizumi S, Hirose T, Takeuchi T. Laboratory markers predicting tofacitinib efficacy in Japanese patients with rheumatoid arthritis: A pooled analysis of Phase 2/3 randomised controlled clinical trials. Mod Rheumatol 2025; 35:417-424. [PMID: 39657989 DOI: 10.1093/mr/roae109] [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: 09/03/2024] [Revised: 11/10/2024] [Accepted: 11/24/2024] [Indexed: 12/12/2024]
Abstract
OBJECTIVES We characterised early changes in laboratory parameters in Japanese patients with rheumatoid arthritis and assessed whether these changes at Month (M)1 were predictive of tofacitinib efficacy at M3. METHODS This post hoc analysis included pooled data from Japanese patients receiving tofacitinib or placebo in three Phase 2/Phase 3 studies (NCT00603512; NCT00687193; NCT00847613). Outcomes included changes from baseline in laboratory parameters (Week 2 and M1/3); efficacy end points were Disease Activity Score in 28 joints, based on C-reactive protein; Disease Activity Score in 28 joints, based on erythrocyte sedimentation rate; and American College of Rheumatology-N index at M3. Univariate/multivariable analyses assessed whether changes at M1 were predictive of efficacy at M3. RESULTS Overall, 467 patients receiving tofacitinib and 104 receiving placebo were included. Tofacitinib treatment was associated with decreases from baseline in C-reactive protein, erythrocyte sedimentation rate, neutrophils, and platelets and increases in lymphocytes, haemoglobin, and lipids up to M3. Decreased platelet count and increased low-density lipoprotein cholesterol and haemoglobin at M1 were predictive of changes in efficacy outcomes at M3. CONCLUSIONS Changes in laboratory parameters at M1 could potentially be used to assess whether tofacitinib therapy will be effective at M3; however, further investigation is needed.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | | | | | | | - Tsutomu Takeuchi
- Division of Rheumatology, Department of Internal Medicine, Keio University, Tokyo, Japan
- Department of Rheumatology and Applied Immunology, Saitama Medical University, Iruma, Saitama, Japan
| |
Collapse
|
3
|
Park S, Kim MK, Park SB, Kim DH, Byun YJ, Choi SA. Neurological Adverse Events Associated with the Use of Janus Kinase Inhibitors: A Pharmacovigilance Study Based on Vigibase. Pharmaceuticals (Basel) 2025; 18:394. [PMID: 40143170 PMCID: PMC11944788 DOI: 10.3390/ph18030394] [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: 02/11/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Janus kinase (JAK) inhibitors are a new class of targeted therapies that block cytokines and the signal transduction and activators of transcription (STAT) pathway. However, post-marketing surveillance studies have led to revised recommendations, highlighting potential serious heart-related events and cancer risk of JAK inhibitors. Here, we aimed to determine the neurological adverse events (AEs) of JAK inhibitors (tofacitinib, ruxolitinib, and baricitinib) based on a global real-world database. Methods: We analyzed individual case safety reports from the Uppsala Monitoring Center from January 1968 to 4 April 2022. A disproportionality analysis was performed using the proportional reporting ratio (PRR), reporting odds ratio (ROR), and information component (IC) to detect signals. Signals were classified according to the hierarchy of the Medical Dictionary for Regulatory Activities (MedDRA). Additionally, a stratified disproportionality analysis by age group and sex was performed for major AEs. Results: A total of 30,051,159 reports for all drugs were analyzed in this study. Among 105,798 reports of tofacitinib, 14.1% (14,863 reports) were neurological AEs. For ruxolitinib and baricitinib, 14.5% (6317 reports) and 10.2% (1216 reports) were neurological AEs, respectively. Various neurological AE signals were detected for tofacitinib and ruxolitinib, with memory impairment exhibiting the highest number of reports and a positive signal in the stratified disproportionality analysis by age group. Baricitinib did not reach the signal detection threshold. Conclusions: This study suggests the potential for neurological AEs, including memory impairment, associated with tofacitinib and ruxolitinib use based on a real-world database.
Collapse
Affiliation(s)
- Sunny Park
- Research Institute of Pharmaceutical Sciences, Korea University, Sejong 339-770, Republic of Korea; (S.P.)
| | - Min Kyu Kim
- College of Pharmacy, Korea University, Sejong 339-770, Republic of Korea; (M.K.K.)
| | - Sung Bin Park
- College of Pharmacy, Korea University, Sejong 339-770, Republic of Korea; (M.K.K.)
| | - Dong Hyeok Kim
- College of Pharmacy, Korea University, Sejong 339-770, Republic of Korea; (M.K.K.)
| | - Young Joo Byun
- Research Institute of Pharmaceutical Sciences, Korea University, Sejong 339-770, Republic of Korea; (S.P.)
- College of Pharmacy, Korea University, Sejong 339-770, Republic of Korea; (M.K.K.)
| | - Soo An Choi
- Research Institute of Pharmaceutical Sciences, Korea University, Sejong 339-770, Republic of Korea; (S.P.)
- College of Pharmacy, Korea University, Sejong 339-770, Republic of Korea; (M.K.K.)
| |
Collapse
|
4
|
Sanchez-Alvarez C, Bond M, Soowamber M, Camellino D, Anderson M, Langford CA, Dejaco C, Touma Z, Ramiro S. Measuring treatment outcomes and change in disease activity in giant cell arteritis: a systematic literature review informing the development of the EULAR-ACR response criteria on behalf of the EULAR-ACR response criteria in giant cell arteritis task force. RMD Open 2023; 9:e003233. [PMID: 37349123 PMCID: PMC10314653 DOI: 10.1136/rmdopen-2023-003233] [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: 04/15/2023] [Accepted: 05/12/2023] [Indexed: 06/24/2023] Open
Abstract
OBJECTIVES To identify criteria and descriptors used to measure response to treatment and change in disease activity in giant cell arteritis (GCA). METHODS A systematic literature review (SLR) to retrieve randomised controlled trials (RCTs) and longitudinal observational studies (LOS). Criteria and descriptors of active disease, remission, response, improvement, worsening and relapse were extracted. RCTs, LOS with >20 subjects, and qualitative research studies were included. RESULTS 10 593 studies were retrieved, of which 116 were included (11 RCTs, 104 LOS, 1 qualitative study). No unified definition of response to therapy was found. Most RCTs used composite endpoints to assess treatment outcomes. Active disease was described in all RCTs and 19% of LOS; and was largely defined by a combination of clinical and laboratory components. Remission was reported in 73% of RCTs and 42% of LOS; It was predominantly defined as the combination of clinical and laboratory components. One LOS reported response with a definition resembling the definition of remission from other studies. Improvement was rarely used as an endpoint and it was mostly a surrogate of remission. No study specifically defined worsening. Relapse was reported in all RCTs and 86% of LOS. It was predominantly defined as the combination of clinical, laboratory and treatment components. CONCLUSIONS The results of this SLR demonstrate that definitions of response used in clinical studies of GCA are scant and heterogeneous. RCTs and LOS mainly used remission and relapse as treatment outcomes. The descriptors identified will inform the development of the future European Alliance of Associations for Rheumatology-American College of Rheumatology response criteria for GCA.
Collapse
Affiliation(s)
- Catalina Sanchez-Alvarez
- Division of Rheumatology & Clinical Immunology, Department of Internal Medicine, University of Florida, Gainesville, Florida, USA
| | - Milena Bond
- Department of Rheumatology, Hospital of Bruneck, (ASAA-SABES), Teaching hospital of the Paracelsus University, Bruneck, Italy
| | - Medha Soowamber
- Department of Rheumatology, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Dario Camellino
- Department of Rheumatology, Local Health Trust, Genoa, Italy
| | - Melanie Anderson
- Department of Library and Information Services, University Health Network, Toronto, Ontario, Canada
| | - Carol A Langford
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, Ohio, USA
| | - Christian Dejaco
- Department of Rheumatology, Hospital of Bruneck, (ASAA-SABES), Teaching hospital of the Paracelsus University, Bruneck, Italy
- Rheumatology, Medical University Graz, Graz, Austria
| | - Zahi Touma
- Department of Medicine, Division of Rheumatology, University of Toronto, Toronto, Ontario, Canada
| | - Sofia Ramiro
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Rheumatology, Zuyderland Medical Center, Heerlen, The Netherlands
| |
Collapse
|
5
|
Bieberich AA, Asquith CRM. Utilization of Supervised Machine Learning to Understand Kinase Inhibitor Toxophore Profiles. Int J Mol Sci 2023; 24:ijms24065088. [PMID: 36982163 PMCID: PMC10049021 DOI: 10.3390/ijms24065088] [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: 01/17/2023] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
There have been more than 70 FDA-approved drugs to target the ATP binding site of kinases, mainly in the field of oncology. These compounds are usually developed to target specific kinases, but in practice, most of these drugs are multi-kinase inhibitors that leverage the conserved nature of the ATP pocket across multiple kinases to increase their clinical efficacy. To utilize kinase inhibitors in targeted therapy and outside of oncology, a narrower kinome profile and an understanding of the toxicity profile is imperative. This is essential when considering treating chronic diseases with kinase targets, including neurodegeneration and inflammation. This will require the exploration of inhibitor chemical space and an in-depth understanding of off-target interactions. We have developed an early pipeline toxicity screening platform that uses supervised machine learning (ML) to classify test compounds’ cell stress phenotypes relative to a training set of on-market and withdrawn drugs. Here, we apply it to better understand the toxophores of some literature kinase inhibitor scaffolds, looking specifically at a series of 4-anilinoquinoline and 4-anilinoquinazoline model libraries.
Collapse
Affiliation(s)
- Andrew A. Bieberich
- AsedaSciences Inc., 1281 Win Hentschel Boulevard, West Lafayette, IN 47906, USA
| | - Christopher R. M. Asquith
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland
- Structural Genomics Consortium and Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Correspondence: ; Tel.: +358-50-400-3138; Fax: +358-82-944-4091
| |
Collapse
|
6
|
Comparison of the effects of peficitinib and tofacitinib in the adjuvant-induced arthritis rat model. Eur J Pharmacol 2023; 941:175490. [PMID: 36608862 DOI: 10.1016/j.ejphar.2023.175490] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/22/2022] [Accepted: 01/03/2023] [Indexed: 01/05/2023]
Abstract
We investigated and compared the pharmacologic properties of two Janus kinase (JAK) inhibitors, peficitinib and tofacitinib, in an adjuvant-induced arthritis rat model. Repeated administration of peficitinib (3 - 30 mg/kg) or tofacitinib (1 - 10 mg/kg) exhibited a dose-related and significant attenuation of arthritis score, paw swelling, pain threshold, grip strength and histopathologic injuries in the model; peficitinib 10 mg/kg and tofacitinib 3 mg/kg demonstrated comparable efficacy. Equivalent Cmax and AUC0-12h values were observed with peficitinib 10 mg/kg and tofacitinib 3 mg/kg, suggesting that the two drugs may demonstrate comparable efficacy on arthritis-associated symptoms at comparable plasma concentration levels. However, peficitinib 10 mg/kg had greater efficacy than tofacitinib 3 mg/kg on some inflammation- and bone destruction-associated parameters in the paw fluid, including the production of vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF), receptor activator of nuclear factor kappa-B ligand, and matrix metalloproteinase-3, which are associated with arthritis exacerbation. Peficitinib 10 mg/kg also showed significantly greater inhibitory effects than tofacitinib 3 mg/kg on loss of bone mineral density and synovial thickening score, which might be a result of the VEGF and PDGF receptor kinase inhibitory effects of peficitinib, in addition to JAK inhibition. In conclusion, both tofacitinib and peficitinib potently improved arthritis and associated symptoms in adjuvant-induced arthritis rats; moreover, owing to possible differences in the mechanism of action of the two drugs, peficitinib may have exerted its effects through JAK inhibition and additional unique off-target properties.
Collapse
|
7
|
Schneeberger EE, Citera G, Nash P, Smolen JS, Mease PJ, Soriano ER, Helling C, Szumski AE, Mundayat R, de León DP. Comparison of disease activity index for psoriatic arthritis (DAPSA) and minimal disease activity (MDA) targets for patients with psoriatic arthritis: A post hoc analysis of data from phase 3 tofacitinib studies. Semin Arthritis Rheum 2023; 58:152134. [PMID: 36476498 DOI: 10.1016/j.semarthrit.2022.152134] [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: 07/20/2022] [Revised: 10/10/2022] [Accepted: 11/07/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To compare achievement of Disease Activity Index in Psoriatic Arthritis (DAPSA) remission (REM)/low disease activity (LDA) with very low disease activity (VLDA)/minimal disease activity (MDA) targets in tofacitinib-treated patients with psoriatic arthritis (PsA). METHODS In this post hoc analysis, data were pooled from two phase 3 studies (6 months' [NCT01882439] and 12 months' [NCT01877668] duration) of patients with PsA receiving tofacitinib 5 or 10 mg twice daily. Cut-offs for DAPSA targets: ≤4 for clinical REM and >4-≤14 for LDA. VLDA and MDA were defined as meeting 7 or ≥5, respectively, of 7 criteria. An ordered logistic regression model was performed to evaluate associations between baseline characteristics and achievement of DAPSA targets as well as VLDA/MDA at month 3. Agreement between achieving DAPSA and VLDA/MDA targets at months 1-6 was assessed via kappa tests. Change from baseline in Health Assessment Questionnaire-Disability Index (HAQ-DI) and Short Form-36 Health Survey (SF-36) Physical Component Summary (PCS) scores (month 6), modified Total Sharp Score (mTSS) and proportion of radiographic non-progressors (mTSS ≤0.5) at month 12 (NCT01877668 only) were compared across DAPSA and VLDA/MDA targets. RESULTS Increased disease activity at baseline was associated with reduced likelihood of achieving DAPSA-REM/DAPSA-LDA or VLDA/MDA at month 3. There was moderate agreement (kappa values 0.41-0.60) between DAPSA-REM and VLDA, and DAPSA-LDA and MDA, from months 1 to 6, although over half of patients achieving DAPSA-REM and over two thirds of patients achieving DAPSA-LDA, respectively, were not captured by VLDA and MDA. Achieving DAPSA-REM/DAPSA-LDA or VLDA/MDA was associated with improved HAQ-DI and SF-36 PCS scores at month 6, and slightly reduced radiographic progression at month 12. CONCLUSION This analysis of data from tofacitinib-treated patients with PsA demonstrated moderate agreement between the DAPSA and VLDA/MDA composite instruments. In agreement with previous studies, VLDA and MDA may be more difficult to achieve than DAPSA-REM and DAPSA-LDA, respectively. However, the clinical and prognostic relevance of this finding should be determined. These data support DAPSA and VLDA/MDA as useful tools for evaluating disease activity and treatment response in PsA. CLINICALTRIALS GOV: NCT01882439; NCT01877668.
Collapse
Affiliation(s)
| | - Gustavo Citera
- Instituto de Rehabilitación Psicofísica, Buenos Aires, Argentina
| | - Peter Nash
- Department of Medicine, Griffith University, Brisbane, Queensland, Australia
| | - Josef S Smolen
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Philip J Mease
- Swedish Medical Center/Providence St. Joseph Health and University of Washington, Seattle, WA, United States
| | - Enrique R Soriano
- Rheumatology Unit, Hospital Italiano de Buenos Aires and Instituto Universitario Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
8
|
Avdeeva AS. Inflammatory markers in rheumatic diseases. RHEUMATOLOGY SCIENCE AND PRACTICE 2022. [DOI: 10.47360/1995-4484-2022-561-569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Immune-mediated rheumatic diseases (IMRDs) are a broad group of pathological conditions based on impaired immunological tolerance to one’s own tissues leading to inflammation and irreversible organ damage. Laboratory diagnosis of IMRDs includes a wide range of biomarkers (autoantibodies, acute phase proteins, cytokines, markers of endothelial damage, components of the complement system, immunoglobulins, cryoglobulins, lymphocyte subpopulations, indicators of bone metabolism, apoptosis markers, genetic markers, etc). One of the leading aspects of laboratory diagnosis of IMRDs is the study of the level of inflammation markers in the blood (erythrocyte sedimentation rate, C-reactive protein (CRP), serum amyloid protein (CAA), ferritin, procalcitonin, apolipoprotein AI, calprotectin, etc). The analysis of inflammation markers makes it possible to assess the disease activity, the nature of the progression and the prognosis of the outcomes of a chronic inflammatory process, as well as the effectiveness of the therapy. The review presents the latest data on the role of the most frequently studied inflammatory markers such as CRP, CAA and ferritin.
Collapse
|
9
|
Gillard L, Pouchot J, Cohen-Aubart F, Koné-Paut I, Mouterde G, Michaud M, Reumaux H, Savey L, Belot A, Fautrel B, Mitrovic S. JAK inhibitors in difficult-to-treat adult-onset Still's disease and systemic-onset juvenile idiopathic arthritis. Rheumatology (Oxford) 2022; 62:1594-1604. [PMID: 35920788 DOI: 10.1093/rheumatology/keac440] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES Excessive and inappropriate production of pro-inflammatory cytokines plays a key role in Still's disease. JAK inhibitor (JAKi) agents mainly block pro-inflammatory cytokine pathways, notably IL-6 and IFN. The objective was to assess the efficacy and safety of JAKi agents in difficult-to-treat systemic juvenile idiopathic arthritis (SJIA) or adult-onset Still's disease (AOSD). METHODS This retrospective study was based on a national survey conducted in the departments of rheumatology, paediatric rheumatology and internal medicine of French hospitals regarding SJIA and AOSD patients who received JAKi agents. The data were collected with a standardised questionnaire and analysed at different times (treatment initiation, months 1, 3, and 6 and the end of follow-up). RESULTS Nine patients (7 adults) were included. All patients showed inadequate response to corticosteroids or conventional synthetic or biologic disease-modifying anti-rheumatic drugs. Baricitinib was used in 5 patients, ruxolitinib in 2, tofacitinib in 2, and upadacitinib in 1. A JAKi was used combined with corticosteroids in all but 2 patients. A JAKi was associated with anakinra and corticosteroids in one patient, and with methotrexate, anakinra and corticosteroids in another. The median follow-up was 16 [1-33] months. Two cases out 9 showed complete remission, 3/9 partial response and 4/9 treatment failure. At the last visit, corticosteroids could be decreased but not stopped. Tolerance of the JAKi was acceptable (no severe adverse events). CONCLUSION JAKi agents may be a therapeutic option for some patients with difficult-to-treat Still's disease, especially those with partial response to medium- or high-dose corticosteroids or biologics.
Collapse
Affiliation(s)
- Louise Gillard
- Service de Rhumatologie, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, AP-HP, France
| | - Jacques Pouchot
- Service de Médecine Interne, Hôpital Européen Georges Pompidou, Université Paris Cité, AP-HP, Paris, France
| | - Fleur Cohen-Aubart
- Service de Médecine Interne 2, Hôpital Pitié-Salpêtrière, Sorbonne Université, AP-HP, Paris, France
| | - Isabelle Koné-Paut
- Service de Rhumatologie Pédiatrique, Hôpital de Bicêtre, Université de Paris Saclay, AP-HP, Le Kremlin-Bicêtre, France.,Centre de Référence des Maladies Autoinflammatoires et des Amyloses (CéRéMAIA), Paris, France
| | - Gaël Mouterde
- Service de Rhumatologie, CHU de Montpellier, Université de Montpellier, Montpellier, France
| | - Martin Michaud
- Service de Médecine Interne, Clinique Ambroise Paré, Toulouse, France
| | - Héloïse Reumaux
- Service de Rhumatologie Pédiatrique, Hôpital Jeanne de Flandres, Université de Lille, CHU de Lille, Lille, France
| | - Lea Savey
- Service de Médecine Interne, Hôpital Tenon, Sorbonne Université, AP-HP, Paris, France
| | - Alexandre Belot
- Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard, Lyon 1, Centre National de la Recherche Scientifique, UMR5308, ENS de Lyon, Lyon, France.,Centre de référence pour les maladies rhumatologiques et inflammatoires pédiatriques (RAISE), Hopital Femme Mère Enfant, Lyon, France.,Service de Néphrologie, Rhumatologie et Néphrologie Pédiatriques, Unité de rhumatologie, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France
| | - Bruno Fautrel
- Service de Rhumatologie, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, AP-HP, France.,Centre de Référence des Maladies Autoinflammatoires et des Amyloses (CéRéMAIA), Paris, France.,Institut d'Epidémiologie et de Santé Publique Pierre Louis, UMR S1136, Equipe PEPITES, Paris, France
| | - Stéphane Mitrovic
- Service de Rhumatologie, Hôpital Pitié-Salpêtrière, Sorbonne Université, Paris, AP-HP, France.,Centre de Référence des Maladies Autoinflammatoires et des Amyloses (CéRéMAIA), Paris, France.,Département de Médecine Interne, Unité de Rhumatologie, Institut Mutualiste Montsouris, Paris, France
| | | |
Collapse
|
10
|
Wang CR, Tsai HW. Immediate-release tofacitinib reduces insulin resistance in non-diabetic active rheumatoid arthritis patients: A single-center retrospective study. World J Diabetes 2022; 13:454-465. [PMID: 35800413 PMCID: PMC9210542 DOI: 10.4239/wjd.v13.i6.454] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/18/2022] [Accepted: 05/28/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND An increased risk of insulin resistance (IR) has been identified in rheumatoid arthritis (RA), a chronic inflammatory disorder with elevated levels of pathogenic cytokines. Biologics targeting proinflammatory cytokines can control the disease and improve insulin sensitivity in RA. Although Janus kinase (JAK) signaling can regulate cytokine receptors and participate in RA pathogenesis, it remains to be elucidated whether there is a reduction of IR in such patients under JAK inhibitor (JAKi) therapy.
AIM To study the effect of JAKi treatment on the reduction of IR in RA patients with active disease.
METHODS A retrospective study was carried out from April 1, 2017 to March 31, 2021 in a population of non-diabetic patients with active RA who were undergoing tofacitinib (TOF) therapy with 5 mg twice-daily immediate-release formulation.
RESULTS Fifty-six RA patients, aged 30 years to 75 years (mean ± SD: 52.3 ± 11.1) with disease activity score 28 values ranging from 4.54 to 7.37 (5.82 ± 0.74), were classified into high-IR (> 2.0) and low-IR (≤ 2.0) groups based on their baseline homeostatic model assessment (HOMA)-IR levels. They had no previous exposure to JAKi, and received TOF therapy for no less than 6 mo. In 30 patients who were naïve to biologics, after a 24-week therapeutic period, the high-IR group showed reduced HOMA-IR levels (3.331 ± 1.036 vs 2.292 ± 0.707, P < 0.001). In another 26 patients who were exposed to tumor necrosis factor-α or interleukin-6 blockers, the high-IR group, despite having achieved a decrease but with lower magnitude than in naïve patients, showed reduced HOMA-IR levels (2.924 ± 0.790 vs 2.545 ± 1.080, P = 0.018).
CONCLUSION In this retrospective study, reduced IR was achieved in non-diabetic active RA patients following 24 wk of TOF therapy.
Collapse
Affiliation(s)
- Chrong-Reen Wang
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan 70403, Taiwan
| | - Hung-Wen Tsai
- Department of Pathology, National Cheng Kung University Hospital, Tainan 70403, Taiwan
| |
Collapse
|
11
|
Lees CW, Deuring JJ, Chiorean M, Daperno M, Bonfanti G, Germino R, Brown PB, Modesto I, Edwards RA. Prediction of early clinical response in patients receiving tofacitinib in the OCTAVE Induction 1 and 2 studies. Therap Adv Gastroenterol 2021; 14:17562848211054710. [PMID: 35154388 PMCID: PMC8832332 DOI: 10.1177/17562848211054710] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 10/04/2021] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Tofacitinib is an oral, small molecule Janus kinase inhibitor for the treatment of ulcerative colitis (UC). Outcome prediction based on early treatment response, along with clinical and laboratory variables, would be very useful for clinical practice. The aim of this study was to determine early variables predictive of responder status in patients with UC treated with tofacitinib. METHODS Data were collected from patients treated with tofacitinib 10 mg twice daily in the OCTAVE Induction 1 and 2 studies (NCT01465763 and NCT01458951). Logistic regression and random forest analyses were performed to determine the power of clinical and/or laboratory variables to predict 2- and 3-point partial Mayo score responder status of patients at Weeks 4 or 8 after baseline. RESULTS From a complete list of variables measured in OCTAVE Induction 1 and 2, analyses identified partial Mayo score, partial Mayo subscore (stool frequency, rectal bleeding, and Physician Global Assessment), cholesterol level, and C-reactive protein level as sufficient variables to predict responder status. Using these variables at baseline and Week 2 predicted responder status at Week 4 with 84-87% accuracy and Week 8 with 74-79% accuracy. Variables at baseline, Weeks 2 and 4 could predict responder status at Week 8 with 85-87% accuracy. CONCLUSION Using a limited set of time-dependent variables, statistical and machine learning models enabled early and clinically meaningful predictions of tofacitinib treatment outcomes in patients with moderately to severely active UC.
Collapse
Affiliation(s)
| | | | - Michael Chiorean
- IBD Center of Excellence, Digestive Disease
Institute, Virginia Mason, Seattle, WA, USA
| | - Marco Daperno
- SC Gastroenterologia, AO Ordine Mauriziano di
Torino, Torino, Italy
| | | | | | | | | | | |
Collapse
|
12
|
Srivastava S, Samarpita S, Ganesan R, Rasool M. CYT387 Inhibits the Hyperproliferative Potential of Fibroblast-like Synoviocytes via Modulation of IL-6/JAK1/STAT3 Signaling in Rheumatoid Arthritis. Immunol Invest 2021; 51:1582-1597. [PMID: 34704880 DOI: 10.1080/08820139.2021.1994589] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Fibroblast-like synoviocytes (FLS) are the critical effector cells primarily involved in rheumatoid arthritis (RA) disease pathogenesis. Interleukin (IL)-6, a proinflammatory cytokine most abundantly expressed in the rheumatoid synovium, promotes Janus kinase (JAK)/signal transducer and transcriptional activator (STAT) signaling cascade activation in RA-FLS, thus leading to its aggressive phenotype, invasiveness, and joint destruction. Momelotinib (CYT387) is a selective small-molecule inhibitor of JAK1/2 and is clinically approved to treat myelofibrosis. However, the therapeutic efficacy of CYT387 in FLS mediated RA pathogenesis is less known. In the present study, we investigated the modulatory effect of CYT387 on IL6/JAK/STAT signaling cascade in FLS induced RA pathogenesis. CYT387 treatment inhibited IL-6 induced high proliferative and migratory potential of FLS cells isolated from adjuvant-induced arthritic (AA) rats. CYT387 reduced the expression of PRMT5, survivin, and HIF-1α mediated by IL-6/sIL-6R in AA-FLS in a dose-dependent manner. The IL-6/sIL-6R induced expression of angiogenic factors such as VEGF and PIGF in AA-FLS cells was found downregulated by CYT387 treatment. Importantly, CYT387 significantly reduced IL-6/sIL-6R dependent activation of JAK1 and STAT3 and increased SOCS3 expression in AA-FLS cells. Next, the S3I-201 mediated blockade of STAT3 activation supported the inhibitory effect of CYT387 on IL-6/JAK1/STAT3 signaling cascade in AA-FLS. Overall, this study proves that CYT387 inhibits proliferation, migration, and pathogenic disease potential of FLS isolated from adjuvant-induced arthritic (AA) rats via targeting IL-6/JAK1/STAT3 signaling cascade.
Collapse
Affiliation(s)
- Susmita Srivastava
- Immunopathology Lab, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, India
| | - Snigdha Samarpita
- Immunopathology Lab, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, India
| | - Ramamoorthi Ganesan
- Immunology Program, Department of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Mahaboobkhan Rasool
- Immunopathology Lab, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, India
| |
Collapse
|
13
|
Yu N, Peng C, Chen W, Sun Z, Zheng J, Zhang S, Ding Y, Shi Y. Circulating Metabolomic Signature in Generalized Pustular Psoriasis Blunts Monocyte Hyperinflammation by Triggering Amino Acid Response. Front Immunol 2021; 12:739514. [PMID: 34567002 PMCID: PMC8455999 DOI: 10.3389/fimmu.2021.739514] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Generalized pustular psoriasis (GPP), the most grievous variant of psoriasis, is featured by dysregulated systemic inflammatory response. The cellular and molecular basis of GPP is poorly understood. Blood monocytes are key players of host defense and producers of inflammatory cytokines including IL-1β. How the immune response of monocytes is affected by metabolic internal environment in GPP remains unclear. Here, we performed a metabolomic and functional investigation of GPP serum and monocytes. We demonstrated a significant increase in IL-1β production from GPP monocytes. In GPP circulation, serum amyloid A (SAA), an acute-phase reactant, was dramatically increased, which induced the release of IL-1β from monocytes in a NLRP3-dependent manner. Using metabolomic analysis, we showed that GPP serum exhibited an amino acid starvation signature, with glycine, histidine, asparagine, methionine, threonine, lysine, valine, isoleucine, tryptophan, tyrosine, alanine, proline, taurine and cystathionine being markedly downregulated. In functional assay, under amino acid starvation condition, SAA-stimulated mature IL-1β secretion was suppressed. Mechanistically, at post-transcriptional level, amino acid starvation inhibited the SAA-mediated reactive oxygen species (ROS) formation and NLRP3 inflammasome activation. Moreover, the immune-modulatory effect of amino acid starvation was blocked by silencing general control nonderepressible 2 kinase (GCN2), suggesting the involvement of amino acid response (AAR) pathway. Collectively, our results suggested that decreased serum amino acids in GPP blunted the innate immune response in blood monocytes through AAR pathway, serving as a feedback mechanism preventing excessive inflammation in GPP.
Collapse
Affiliation(s)
- Ning Yu
- Department of Dermatology, Shanghai Skin Disease Hospital, Institute of Psoriasis, Tongji University, Shanghai, China
| | - Chen Peng
- Department of Dermatology, Shanghai Skin Disease Hospital, Institute of Psoriasis, Tongji University, Shanghai, China
| | - Wenjuan Chen
- Department of Dermatology, Shanghai Skin Disease Hospital, Institute of Psoriasis, Tongji University, Shanghai, China
| | - Ziwen Sun
- Department of Dermatology, Shanghai Skin Disease Hospital, Institute of Psoriasis, Tongji University, Shanghai, China
| | - Jianfeng Zheng
- Department of Dermatology, Shanghai Skin Disease Hospital, Institute of Psoriasis, Tongji University, Shanghai, China
| | - Shujie Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Yangfeng Ding
- Department of Dermatology, Shanghai Skin Disease Hospital, Institute of Psoriasis, Tongji University, Shanghai, China
| | - Yuling Shi
- Department of Dermatology, Shanghai Skin Disease Hospital, Institute of Psoriasis, Tongji University, Shanghai, China
| |
Collapse
|
14
|
Uchio A, Matsumoto T, Maenohara Y, Omata Y, Takahashi H, Iwasawa M, Juji T, Nakamura I, Tanaka S. Systemic inflammatory responses after orthopedic surgery in patients with rheumatoid arthritis treated with tofacitinib. Clin Rheumatol 2021; 40:5077-5083. [PMID: 34545450 DOI: 10.1007/s10067-021-05914-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 08/20/2021] [Accepted: 09/05/2021] [Indexed: 12/01/2022]
Abstract
OBJECTIVE To investigate the acute phase response to surgical stress in patients with rheumatoid arthritis (RA) treated with tofacitinib, a Janus kinase (JAK) inhibitor. METHODS A retrospective matched pair analysis of 34 patients treated with tofacitinib and 34 patients treated with conventional disease-modifying anti-rheumatic drugs (csDMARDs) was performed. Patients were matched for age, sex, and type of surgery; body temperature, C-reactive protein (CRP) level, and white blood cell (WBC) count, neutrophil count, and lymphocyte count were compared between the tofacitinib and csDMARDs groups within 2 weeks after orthopedic surgery. Postoperative complications within 90 days were also assessed. RESULTS No surgical site infection or delayed wound healing was observed in the tofacitinib group; whereas, one case of superficial infection was noted in the csDMARDs group. A similar postoperative increase in body temperature and CRP level was observed in both the groups. Postoperatively, the tofacitinib group showed an increase in WBC and neutrophils counts and a decrease in lymphocyte count, unlike the csDMARDs group. In contrast to two patients (2.6%) in the csDMARDs group, seven patients (20.6%) in the tofacitinib group had lymphocyte counts below 500 cells/μL within 2 weeks postoperatively. CONCLUSION Tofacitinib did not suppress postoperative increase in body temperature and CRP level. Because of the postoperative decrease in lymphocyte count in patients treated with tofacitinib, the timing for resuming tofacitinib treatment after surgery should be carefully considered. Key Points • This study is the first to report the complications and systemic inflammatory responses after orthopedic surgery in patients treated with tofacitinib in comparison with matched pairs treated with conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs) • While tofacitinib does not suppress postoperative increase in body temperature and CRP level, the postoperative decrease in lymphocyte count in patients treated with tofacitinib is significant compared with patients treated with csDMARDs • Attention should be paid to a reduced lymphocyte count when to resume tofacitinib after surgery.
Collapse
Affiliation(s)
- Akihiro Uchio
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.,Department of Orthopaedic Surgery, National Hospital Organization, Sagamihara Hospital, 18-1 Sakuradai, Minami-ku, Sagamihara City, Kanagawa, 252-0314, Japan
| | - Takumi Matsumoto
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Yuji Maenohara
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yasunori Omata
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hiroshi Takahashi
- Department of Rheumatology, JCHO Yugawara Hospital, 2-21-6 Chuo, Yugawara, Ashigara-shimo, Kanagawa, 259-0396, Japan
| | - Mitsuyasu Iwasawa
- Department of Orthopaedic Surgery, National Hospital Organization, Sagamihara Hospital, 18-1 Sakuradai, Minami-ku, Sagamihara City, Kanagawa, 252-0314, Japan
| | - Takuo Juji
- Department of Rheumatology, JCHO Yugawara Hospital, 2-21-6 Chuo, Yugawara, Ashigara-shimo, Kanagawa, 259-0396, Japan
| | - Ichiro Nakamura
- Department of Rheumatology, JCHO Yugawara Hospital, 2-21-6 Chuo, Yugawara, Ashigara-shimo, Kanagawa, 259-0396, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
15
|
Zhou YW, Xie Y, Tang LS, Pu D, Zhu YJ, Liu JY, Ma XL. Therapeutic targets and interventional strategies in COVID-19: mechanisms and clinical studies. Signal Transduct Target Ther 2021; 6:317. [PMID: 34446699 PMCID: PMC8390046 DOI: 10.1038/s41392-021-00733-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/27/2021] [Accepted: 07/14/2021] [Indexed: 02/06/2023] Open
Abstract
Owing to the limitations of the present efforts on drug discovery against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the lack of the understanding of the biological regulation mechanisms underlying COVID-19, alternative or novel therapeutic targets for COVID-19 treatment are still urgently required. SARS-CoV-2 infection and immunity dysfunction are the two main courses driving the pathogenesis of COVID-19. Both the virus and host factors are potential targets for antiviral therapy. Hence, in this study, the current therapeutic strategies of COVID-19 have been classified into "target virus" and "target host" categories. Repurposing drugs, emerging approaches, and promising potential targets are the implementations of the above two strategies. First, a comprehensive review of the highly acclaimed old drugs was performed according to evidence-based medicine to provide recommendations for clinicians. Additionally, their unavailability in the fight against COVID-19 was analyzed. Next, a profound analysis of the emerging approaches was conducted, particularly all licensed vaccines and monoclonal antibodies (mAbs) enrolled in clinical trials against primary SARS-CoV-2 and mutant strains. Furthermore, the pros and cons of the present licensed vaccines were compared from different perspectives. Finally, the most promising potential targets were reviewed, and the update of the progress of treatments has been summarized based on these reviews.
Collapse
Affiliation(s)
- Yu-Wen Zhou
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yao Xie
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Department of Dermatovenerology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Lian-Sha Tang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Dan Pu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ya-Juan Zhu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ji-Yan Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Xue-Lei Ma
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
- Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
16
|
Morris G, Bortolasci CC, Puri BK, Marx W, O'Neil A, Athan E, Walder K, Berk M, Olive L, Carvalho AF, Maes M. The cytokine storms of COVID-19, H1N1 influenza, CRS and MAS compared. Can one sized treatment fit all? Cytokine 2021; 144:155593. [PMID: 34074585 PMCID: PMC8149193 DOI: 10.1016/j.cyto.2021.155593] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/03/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023]
Abstract
An analysis of published data appertaining to the cytokine storms of COVID-19, H1N1 influenza, cytokine release syndrome (CRS), and macrophage activation syndrome (MAS) reveals many common immunological and biochemical abnormalities. These include evidence of a hyperactive coagulation system with elevated D-dimer and ferritin levels, disseminated intravascular coagulopathy (DIC) and microthrombi coupled with an activated and highly permeable vascular endothelium. Common immune abnormalities include progressive hypercytokinemia with elevated levels of TNF-α, interleukin (IL)-6, and IL-1β, proinflammatory chemokines, activated macrophages and increased levels of nuclear factor kappa beta (NFκB). Inflammasome activation and release of damage associated molecular patterns (DAMPs) is common to COVID-19, H1N1, and MAS but does not appear to be a feature of CRS. Elevated levels of IL-18 are detected in patients with COVID-19 and MAS but have not been reported in patients with H1N1 influenza and CRS. Elevated interferon-γ is common to H1N1, MAS, and CRS but levels of this molecule appear to be depressed in patients with COVID-19. CD4+ T, CD8+ and NK lymphocytes are involved in the pathophysiology of CRS, MAS, and possibly H1N1 but are reduced in number and dysfunctional in COVID-19. Additional elements underpinning the pathophysiology of cytokine storms include Inflammasome activity and DAMPs. Treatment with anakinra may theoretically offer an avenue to positively manipulate the range of biochemical and immune abnormalities reported in COVID-19 and thought to underpin the pathophysiology of cytokine storms beyond those manipulated via the use of, canakinumab, Jak inhibitors or tocilizumab. Thus, despite the relative success of tocilizumab in reducing mortality in COVID-19 patients already on dexamethasone and promising results with Baricitinib, the combination of anakinra in combination with dexamethasone offers the theoretical prospect of further improvements in patient survival. However, there is currently an absence of trial of evidence in favour or contravening this proposition. Accordingly, a large well powered blinded prospective randomised controlled trial (RCT) to test this hypothesis is recommended.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | | | - Wolfgang Marx
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Adrienne O'Neil
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Melbourne School of Population and Global Health, Melbourne, Australi
| | - Eugene Athan
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, The University of Melbourne, Melbourne, Australia
| | - Lisa Olive
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, School of Psychology, Geelong, Australia
| | - Andre F Carvalho
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, University of Toronto, Toronto, Canada, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
| |
Collapse
|
17
|
Roszkowski L, Ciechomska M. Tuning Monocytes and Macrophages for Personalized Therapy and Diagnostic Challenge in Rheumatoid Arthritis. Cells 2021; 10:cells10081860. [PMID: 34440629 PMCID: PMC8392289 DOI: 10.3390/cells10081860] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/21/2022] Open
Abstract
Monocytes/macrophages play a central role in chronic inflammatory disorders, including rheumatoid arthritis (RA). Activation of these cells results in the production of various mediators responsible for inflammation and RA pathogenesis. On the other hand, the depletion of macrophages using specific antibodies or chemical agents can prevent their synovial tissue infiltration and subsequently attenuates inflammation. Their plasticity is a major feature that helps the switch from a pro-inflammatory phenotype (M1) to an anti-inflammatory state (M2). Therefore, understanding the precise strategy targeting pro-inflammatory monocytes/macrophages should be a powerful way of inhibiting chronic inflammation and bone erosion. In this review, we demonstrate potential consequences of different epigenetic regulations on inflammatory cytokines production by monocytes. In addition, we present unique profiles of monocytes/macrophages contributing to identification of new biomarkers of disease activity or predicting treatment response in RA. We also outline novel approaches of tuning monocytes/macrophages by biologic drugs, small molecules or by other therapeutic modalities to reduce arthritis. Finally, the importance of cellular heterogeneity of monocytes/macrophages is highlighted by single-cell technologies, which leads to the design of cell-specific therapeutic protocols for personalized medicine in RA in the future.
Collapse
|
18
|
Huang TTM, Chien YC, Wang CH, Chang SY, Wang JT, Hsieh SC, Yeh YC, Ku SC, Yu CJ, Chiang BL, Chang SC, Tolwani A. Successful Treatment of a Critically Ill COVID-19 Patient Using Continuous Renal Replacement Therapy With Enhanced Cytokine Removal and Tocilizumab: A Case Report. Front Med (Lausanne) 2021; 8:649583. [PMID: 34164411 PMCID: PMC8215202 DOI: 10.3389/fmed.2021.649583] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/19/2021] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic has caused multiple deaths worldwide. Since no specific therapies are currently available, treatment for critically ill patients with COVID-19 is supportive. The most severe patients need sustained life support for recovery. We herein describe the course of a critically ill COVID-19 patient with multi-organ failure, including acute respiratory failure, acute kidney injury, and fulminant cytokine release syndrome (CRS), who required mechanical ventilation and extracorporeal membrane oxygenation support. This patient with a predicted high mortality risk was successfully managed with a careful strategy of oxygenation, uremic toxin removal, hemodynamic support, and most importantly, cytokine-targeted intervention for CRS, including cytokine/endotoxin removal, anti-cytokine therapy, and immune modulation. Comprehensive cytokine data, CRS parameters, and biochemical data of extracorporeal removal were provided to strengthen the rationale of this strategy. In this report, we demonstrate that timely combined hemoperfusion with cytokine adsorptive capacity and anti-cytokine therapy can successfully treat COVID-19 patients with fulminant CRS. It also highlights the importance of implementing cytokine-targeted therapy for severe COVID-19 guided by the precise measurement of disease activity.
Collapse
Affiliation(s)
- Thomas Tao-Min Huang
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Ying-Chun Chien
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Chih-Hsien Wang
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jann-Tay Wang
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Song-Chou Hsieh
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Yu-Chang Yeh
- Department of Anesthesiology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Shih-Chi Ku
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Chong-Jen Yu
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Bor-Luen Chiang
- Department of Pediatrics, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shan-Chwen Chang
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Ashita Tolwani
- Division of Nephrology, University of Alabama, Birmingham, AL, United States
| |
Collapse
|
19
|
Isailovic N, Ceribelli A, Cincinelli G, Vecellio M, Guidelli G, Caprioli M, Luciano N, Motta F, Selmi C, De Santis M. Lymphocyte modulation by tofacitinib in patients with rheumatoid arthritis. Clin Exp Immunol 2021; 205:142-149. [PMID: 33899926 DOI: 10.1111/cei.13609] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 12/15/2022] Open
Abstract
Tofacitinib is an oral small molecule targeting the intracellular Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathways approved for the treatment of active rheumatoid arthritis (RA). We investigated the effects of tofacitinib on the response of RA lymphocytes to B and T cell collagen epitopes in their native and post-translationally modified forms. In particular, peripheral blood mononuclear cells (PBMCs) from patients with RA and healthy subjects were cultured with type II collagen peptides (T261-273, B359-369, carT261-273, citB359-369) or with phorbol myristate acetate (PMA)/ionomycin/CD40L in the presence or absence of 100 nM tofacitinib for 20 h and analyzed by fluorescence activated cell sorter (FACS). Cultures without brefeldin A were used for cytokine supernatant enzyme-linked immunosorbent assay (ELISA) analysis. Tofacitinib down-regulated inflammatory cytokines by stimulated B [interleukin (IL)-6 and tumor necrosis factor (TNF)-α] and T [interferon (IFN)-γ, IL-17 or TNF-α] cells in the short term, while a significant reduction of IL-17 and IL-6 levels in peripheral blood mononuclear cell (PBMC) supernatant was also observed. IL-10 was significantly reduced in collagen-stimulated B cells from patients with RA and increased in controls, thus mirroring an altered response to collagen self-epitopes in RA. Tofacitinib partially prevented the IL-10 down-modulation in RA B cells stimulated with collagen epitopes. In conclusion, the use of tofacitinib exerts a rapid regulatory effect on B cells from patients with RA following stimulation with collagen epitopes while not reducing inflammatory cytokine production by lymphocytes.
Collapse
Affiliation(s)
- Natasa Isailovic
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy
| | - Angela Ceribelli
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Gilberto Cincinelli
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy
| | - Matteo Vecellio
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy.,Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Giacomo Guidelli
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy
| | - Marta Caprioli
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy
| | - Nicoletta Luciano
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy
| | - Francesca Motta
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Maria De Santis
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital IRCCS, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| |
Collapse
|
20
|
Magrì S, Chessa L, Demurtas M, Cabras F, Mocci G. Review article: safety of new biologic agents for inflammatory bowel disease in the liver. Eur J Gastroenterol Hepatol 2021; 33:623-630. [PMID: 33470695 DOI: 10.1097/meg.0000000000002076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
New biologic agents (vedolizumab, ustekinumab and tofacitinib) represent an effective treatment for inflammatory bowel diseases and have been recently approved. However, with a rapidly evolving complement of advanced targeted therapies, new concerns about their potentially undesirable effects on liver function emerge. In particular, little is known about safety data in patients with hepatitis B virus, hepatitis C virus chronic infections, cirrhosis and in transplanted patients who are accumulating. In addition, these new agents have also been associated with drug-induced liver injury. Limited data on the efficacy of vedolizumab in patients with primary sclerosing cholangitis are also available. This article reviews available data about hepatic safety concerns in patients receiving vedolizumab, ustekinumab and tofacitinib with and without preexistent hepatic diseases.
Collapse
Affiliation(s)
- Salvatore Magrì
- Department of Endoscopic Surgery, 'G.Brotzu' Hospital, Piazzale Alessandro Ricchi, Cagliari
| | - Luchino Chessa
- Department of Medical Sciences and Public Health, University of Cagliari, Presidio Policlinico of Monserrato, Monserrato
| | - Mauro Demurtas
- Endoscopic Unit, Department of Surgery, San Martino Hospital, Viale Fondazione Rockfeller, Oristano
| | - Francesco Cabras
- Gastroenterology Unit, Department of Surgery, 'G. Brotzu' Hospital, Piazzale Alessandro Ricchi, Cagliari, Italy
| | - Giammarco Mocci
- Gastroenterology Unit, Department of Surgery, 'G. Brotzu' Hospital, Piazzale Alessandro Ricchi, Cagliari, Italy
| |
Collapse
|
21
|
Eriksson P, Mölne J, Wirestam L, Sjöwall C. Successful Treatment of AA Amyloidosis in Ankylosing Spondylitis Using Tocilizumab: Report of Two Cases and Review of the Literature. Front Med (Lausanne) 2021; 8:661101. [PMID: 33981717 PMCID: PMC8107218 DOI: 10.3389/fmed.2021.661101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/01/2021] [Indexed: 12/02/2022] Open
Abstract
Historically, secondary amyloidosis has been a feared complication of chronic inflammatory conditions. The fibril protein AA derives from the acute phase reactant serum amyloid A (SAA). Long-term elevation of SAA levels remains a major risk factor for the development of AA amyloidosis in rheumatic diseases, and the prognosis may be unpredictable. Nowadays, with increased availability of effective biological agents, the incidence of AA amyloidosis seems to be declining. Still, genetically predisposed subjects with slowly progressive disease and mild symptoms combined with ongoing systemic inflammation may be at risk. Interleukin-6 (IL-6) is one of the drivers of SAA release and effectiveness of the humanized anti-IL-6 receptor antibody tocilizumab (TCZ) for the treatment of AA amyloidosis has been observed in some rheumatic conditions. Herein, we report two male subjects with longstanding ankylosing spondylitis (AS) complicated by renal amyloidosis who received TCZ with rapid and beneficial effects regarding inflammation and proteinuria. To the best of our knowledge, the use of TCZ in AS patients with this extra-articular manifestation has not previously been described. The paper includes histopathology, clinical follow-up, and longitudinal data of the two cases along with a comprehensive review of relevant literature. Mechanisms behind amyloid-mediated tissue damage and organ dysfunction are discussed. Altogether, our data highlight that blocking IL-6 signaling may represent a promising therapeutic option in patients with renal AA amyloidosis.
Collapse
Affiliation(s)
- Per Eriksson
- Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection, Linköping University, Linköping, Sweden
| | - Johan Mölne
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Sahlgrenska Academy, Gothenburg, Sweden
| | - Lina Wirestam
- Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection, Linköping University, Linköping, Sweden
| | - Christopher Sjöwall
- Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection, Linköping University, Linköping, Sweden
| |
Collapse
|
22
|
Sebba A. Pain: A Review of Interleukin-6 and Its Roles in the Pain of Rheumatoid Arthritis. Open Access Rheumatol 2021; 13:31-43. [PMID: 33707975 PMCID: PMC7943546 DOI: 10.2147/oarrr.s291388] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 01/22/2021] [Indexed: 12/29/2022] Open
Abstract
Pain is a major and common symptom reported as a top priority in patients with rheumatoid arthritis (RA). Intuitively, RA-related pain is often considered to be a natural consequence of peripheral inflammation, so treatment of RA is expected to manage pain concurrently as part of inflammation control. However, pain in patients with RA can be poorly correlated with objective measures of inflammation, for example, in patients who are otherwise in remission. Joint damage appears to account for only a fraction of this residual pain. Emerging evidence suggests that alteration of peripheral and central pain processing contributes to RA-related pain; this is parallel to, but somewhat independent of, joint inflammation. Interleukin (IL)-6 is a proinflammatory cytokine that contributes to the pathogenesis of RA. It exerts systemic effects via signaling through soluble forms of the IL-6 receptor (“trans-signaling”). Evidence from preclinical studies demonstrates that intra-articular IL-6 can produce long-lasting peripheral sensitization to mechanical stimulation and suggests an important role for IL-6 in central pain sensitization. This may be partly explained by its ability to activate neurons through trans-signaling, affecting nociceptive plasticity and nerve fiber regrowth. Local activity at neuron endings may culminate in altered pain processing in the central nervous system because of persistent signaling from sensitized peripheral neurons. Peripheral and central sensitization can promote the development of chronic pain, which can have a significant impact on patients’ health and quality of life. A proportion of pain in RA may be more appropriately managed as an entity separate from inflammation. Both the peripheral and central nervous systems should be recognized as important potential systems targeted by RA. The substantial burden of RA-related chronic pain suggests that pain should be a key focus in RA management and should be assessed and addressed early and separately from the inflammatory component.
Collapse
Affiliation(s)
- Anthony Sebba
- Division of Rheumatology, University of South Florida, Tampa, FL, USA
| |
Collapse
|
23
|
Sorić Hosman I, Kos I, Lamot L. Serum Amyloid A in Inflammatory Rheumatic Diseases: A Compendious Review of a Renowned Biomarker. Front Immunol 2021; 11:631299. [PMID: 33679725 PMCID: PMC7933664 DOI: 10.3389/fimmu.2020.631299] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Serum amyloid A (SAA) is an acute phase protein with a significant importance for patients with inflammatory rheumatic diseases (IRD). The central role of SAA in pathogenesis of IRD has been confirmed by recent discoveries, including its involvement in the activation of the inflammasome cascade and recruitment of interleukin 17 producing T helper cells. Clinical utility of SAA in IRD was originally evaluated nearly half a century ago. From the first findings, it was clear that SAA could be used for evaluating disease severity and monitoring disease activity in patients with rheumatoid arthritis and secondary amyloidosis. However, cost-effective and more easily applicable markers, such as C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR), overwhelmed its use in clinical practice. In the light of emerging evidences, SAA has been discerned as a more sensitive biomarker in a wide spectrum of IRD, especially in case of subclinical inflammation. Furthermore, a growing number of studies are confirming the advantages of SAA over many other biomarkers in predicting and monitoring response to biological immunotherapy in IRD patients. Arising scientific discoveries regarding the role of SAA, as well as delineating SAA and its isoforms as the most sensitive biomarkers in various IRD by recently developing proteomic techniques are encouraging the revival of its clinical use. Finally, the most recent findings have shown that SAA is a biomarker of severe Coronavirus disease 2019 (COVID-19). The aim of this review is to discuss the SAA-involving immune system network with emphasis on mechanisms relevant for IRD, as well as usefulness of SAA as a biomarker in various IRD. Therefore, over a hundred original papers were collected through an extensive PubMed and Scopus databases search. These recently arising insights will hopefully lead to a better management of IRD patients and might even inspire the development of new therapeutic strategies with SAA as a target.
Collapse
Affiliation(s)
- Iva Sorić Hosman
- Department of Pediatrics, Zadar General Hospital, Zadar, Croatia
| | - Ivanka Kos
- Division of Nephrology, Dialysis and Transplantation, Department of Pediatrics, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Lovro Lamot
- Division of Nephrology, Dialysis and Transplantation, Department of Pediatrics, University Hospital Centre Zagreb, Zagreb, Croatia.,Department of Pediatrics, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
24
|
Berg EL, Denker SP, O'Mahony A. Development and Validation of Disease Assays for Phenotypic Screening. PHENOTYPIC DRUG DISCOVERY 2020. [DOI: 10.1039/9781839160721-00020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Screening in phenotypic assays is an important strategy for the discovery of innovative drugs and novel targets. Here we present key strategies for developing successful phenotypic screens and prosecuting phenotypic drug discovery (PDD) programs. Successful screens incorporate physiological relevance through the use of human cell types and assay designs that have (1) strong mechanistic connection to clinical outcomes and (2) strong biological justification for both efficacy and safety. In addition to guidance for designing successful screens, we also propose incorporation of specific counterscreens at an early point in the program. The suggested counterscreens are based on analysis of 1000s of drugs and drug candidates profiled through a large set of human-based phenotypic assays. These assays include cytotoxicity in human primary vascular endothelial cells, proliferation of endothelial cells, and proliferation of lymphocytes, all under specific activation conditions. These counterscreens form a generic screening funnel to triage a large fraction of early-stage hits, binning compounds into those with undesirable mechanisms (associated with acute toxicity), mechanisms with utility for oncology indications, and mechanisms useful for autoimmune indications. The application of this screening funnel offers a standardized and more predictable path for prosecuting PDD programs, reducing the risk of failure, and improving program timelines.
Collapse
Affiliation(s)
- Ellen L. Berg
- Eurofins Discovery 111 Anza Blvd, Suite 414 Burlingame CA 94010 USA
| | - Sheryl P. Denker
- Eurofins Discovery 111 Anza Blvd, Suite 414 Burlingame CA 94010 USA
| | - Alison O'Mahony
- Eurofins Discovery 111 Anza Blvd, Suite 414 Burlingame CA 94010 USA
| |
Collapse
|
25
|
Borcherding N, Jethava Y, Vikas P. Repurposing Anti-Cancer Drugs for COVID-19 Treatment. Drug Des Devel Ther 2020; 14:5045-5058. [PMID: 33239864 PMCID: PMC7680713 DOI: 10.2147/dddt.s282252] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/29/2020] [Indexed: 12/24/2022] Open
Abstract
The novel coronavirus disease 2019 (COVID-19) pandemic has caused catastrophic damage to human life across the globe along with social and financial hardships. According to the Johns Hopkins University Coronavirus Resource Center, more than 41.3 million people worldwide have been infected, and more than 1,133,000 people have died as of October 22, 2020. At present, there is no available vaccine and a scarcity of efficacious therapies. However, there is tremendous ongoing effort towards identifying effective drugs and developing novel vaccines. Early data from Adaptive COVID-19 Treatment Trials (ACTT) sponsored by the National Institute of Allergy and Infectious Diseases (NIAID) and compassionate use study have shown promise for remdesivir, leading to emergency authorization by the Food and Drug Administration (FDA) for treatment of hospitalized COVID-19 patients. However, several randomized studies have now shown no benefit or increased adverse events associated with remdesivir treatment. Drug development is a time-intensive process and requires extensive safety and efficacy evaluations. In contrast, drug repurposing is a time-saving and cost-effective drug discovery strategy geared towards using existing drugs instead of de novo drug discovery. Treatments for cancer and COVID-19 often have similar goals of controlling inflammation, inhibiting cell division, and modulating the host microenvironment to control the disease. In this review, we focus on anti-cancer drugs that can potentially be repurposed for COVID-19 and are currently being tested in clinical trials.
Collapse
Affiliation(s)
- Nicholas Borcherding
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA, USA
- Department of Pathology, University of Iowa, College of Medicine, Iowa City, IA, USA
- Cancer Biology Graduate Program, University of Iowa, College of Medicine, Iowa City, IA, USA
- Medical Scientist Training Program, University of Iowa, College of Medicine, Iowa City, IA, USA
| | - Yogesh Jethava
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA, USA
- Department of Internal Medicine, University of Iowa, College of Medicine, Iowa City, IA, USA
| | - Praveen Vikas
- Holden Comprehensive Cancer Center, University of Iowa, College of Medicine, Iowa City, IA, USA
- Department of Internal Medicine, University of Iowa, College of Medicine, Iowa City, IA, USA
| |
Collapse
|
26
|
Luo W, Ige OO, Beacon TH, Su RC, Huang S, Davie JR, Lakowski TM. The treatment of SARS-CoV2 with antivirals and mitigation of the cytokine storm syndrome: the role of gene expression. Genome 2020; 64:400-415. [PMID: 33197212 DOI: 10.1139/gen-2020-0130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the absence of a vaccine, the treatment of SARS-CoV2 has focused on eliminating the virus with antivirals or mitigating the cytokine storm syndrome (CSS) that leads to the most common cause of death: respiratory failure. Herein we discuss the mechanisms of antiviral treatments for SARS-CoV2 and treatment strategies for the CSS. Antivirals that have shown in vitro activity against SARS-CoV2, or the closely related SARS-CoV1 and MERS-CoV, are compared on the enzymatic level and by potency in cells. For treatment of the CSS, we discuss medications that reduce the effects or expression of cytokines involved in the CSS with an emphasis on those that reduce IL-6 because of its central role in the development of the CSS. We show that some of the medications covered influence the activity or expression of enzymes involved in epigenetic processes and specifically those that add or remove modifications to histones or DNA. Where available, the latest clinical data showing the efficacy of the medications is presented. With respect to their mechanisms, we explain why some medications are successful, why others have failed, and why some untested medications may yet prove useful.
Collapse
Affiliation(s)
- Wenxia Luo
- Pharmaceutical Analysis Laboratory, College of Pharmacy, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Olufola O Ige
- Pharmaceutical Analysis Laboratory, College of Pharmacy, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Tasnim H Beacon
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Ruey-Chyi Su
- National HIV and Retrovirology Laboratory, JC Wilt Infectious Disease Research Centre, Winnipeg, MB R3E 3R2, Canada
| | - Shujun Huang
- Pharmaceutical Analysis Laboratory, College of Pharmacy, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - James R Davie
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Ted M Lakowski
- Pharmaceutical Analysis Laboratory, College of Pharmacy, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| |
Collapse
|
27
|
Bao Q, Zhang L, Wang N, Gabet B, Yang W, Gao X, You Q, Jiang Z. Hydrogen Peroxide Inducible JAK3 Covalent Inhibitor: Prodrug for the Treatment of RA with Enhanced Safety Profile. ACS Med Chem Lett 2020; 11:2182-2189. [PMID: 33214827 DOI: 10.1021/acsmedchemlett.0c00323] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022] Open
Abstract
Selective inhibition of Janus kinases (JAKs) is an arising strategy in drug discovery. Covalent inhibitors targeting a unique cysteine in JAK3 exhibit ultraselectivity among JAK family members. However, safety and tissue specific concerns still remain. A prodrug of a known JAK3 covalent inhibitor sensitive to H2O2 was designed and synthesized and its therapeutic effect was evaluated in the CIA (collagen-induced arthritis) mice model of RA (rheumatoid arthritis). The prodrug strategy relied on the introduction of a hydrogen peroxide-sensitive borate trigger group to avoid random covalent binding to thiol functionalities in biomacromolecules. The results show that the prodrug can be activated and released under pathophysiological concentration of H2O2. In addition, the prodrug demonstrated stability to the physiological environment. In comparison to the parent compound, the prodrug showed a similar therapeutic effect in the CIA model but notably exhibited lower toxicity and a larger therapeutic window.
Collapse
Affiliation(s)
- Qichao Bao
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Liangying Zhang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Nan Wang
- Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, College of Pharmacy, University of Minnesota, 717 Delaware Street, SE, Minneapolis, Minnesota 55414, United States
| | - Brian Gabet
- Department of Medicinal Chemistry and Institute for Therapeutics Discovery and Development, College of Pharmacy, University of Minnesota, 717 Delaware Street, SE, Minneapolis, Minnesota 55414, United States
| | - Weikang Yang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xingyang Gao
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Qidong You
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhengyu Jiang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
28
|
Delcuve GP, Lakowski TM, Su RC, Beacon TH, Davie JR. SARS-CoV-2 multifaceted interaction with human host. Part I: What we have learnt and done so far, and the still unknown realities. IUBMB Life 2020; 72:2313-2330. [PMID: 32918855 DOI: 10.1002/iub.2380] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/07/2020] [Accepted: 08/18/2020] [Indexed: 12/16/2022]
Abstract
SARS-CoV-2, the causing agent of the ongoing COVID-19 pandemic, is a beta-coronavirus which has 80% genetic homology with SARS-CoV, but displays increased virulence and transmissibility. Initially, SARS-CoV-2 was considered a respiratory virus generally causing a mild disease, only severe and fatal in the elderly and individuals with underlying conditions. Severe illnesses and fatalities were attributed to a cytokine storm, an excessive response from the host immune system. However, with the number of infections over 10 millions and still soaring, the insidious and stealthy nature of the virus has emerged, as it causes a vast array of diverse unexpected symptoms among infected individuals, including the young and healthy. It has become evident that besides infecting the respiratory tract, SARS-CoV-2 can affect many organs, possibly through the infection of the endothelium. This review presents an overview of our learning curve with the novel virus emergence, transmission, pathology, biological properties and host-interactions. It also briefly describes remedial measures taken until an effective vaccine is available, that is non-pharmaceutical interventions to reduce the viral spread and the repurposing of existing drugs, approved or in development for other conditions to eliminate the virus or mitigate the cytokine storm.
Collapse
Affiliation(s)
- Geneviève P Delcuve
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ted M Lakowski
- College of Pharmacy, Pharmaceutical Analysis Laboratory, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ruey-Chyi Su
- National HIV and Retrovirology Laboratory, JC Wilt Infectious Disease Research Centre, Winnipeg, Manitoba, Canada
| | - Tasnim H Beacon
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - James R Davie
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
29
|
Blesl A, Rainer F, Pollheimer M, Kump P, Durchschein F, Wenzl H, Eller K, Krisper P, Petritsch W, Högenauer C. Successful Pregnancies After Regression of AA Amyloidosis by Anti-inflammatory Therapy in Chronic Active Crohn's Disease. Dig Dis Sci 2020; 65:2730-2734. [PMID: 31865490 DOI: 10.1007/s10620-019-05988-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/02/2019] [Indexed: 12/09/2022]
Affiliation(s)
- Andreas Blesl
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.
| | - Florian Rainer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | | | - Patrizia Kump
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Franziska Durchschein
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Heimo Wenzl
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Kathrin Eller
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Peter Krisper
- Division of Nephrology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Wolfgang Petritsch
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Christoph Högenauer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| |
Collapse
|
30
|
Cantini F, Blandizzi C, Niccoli L, Petrone L, Goletti D. Systematic review on tuberculosis risk in patients with rheumatoid arthritis receiving inhibitors of Janus Kinases. Expert Opin Drug Saf 2020; 19:861-872. [PMID: 32552289 DOI: 10.1080/14740338.2020.1774550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Janus kinases inhibitors (anti-JAKs), including tofacitinib, baricitinib, upadacitinib, and filgotinib, represent a new class of synthetic targeted drugs for the treatment of rheumatoid arthritis (RA). In this review, the risk of active tuberculosis (TB) occurrence in patients receiving anti-JAKs was assessed. The literature on this topic, updated to 29 February 2020 was reviewed. Overall, 40 reports (22 tofacitinib, 10 baricitinib, 5 upadacitinib, 3 filgotinib) were examined. A low frequency, not exceeding 0.25%, of active TB cases in patients were exposed to anti-JAKs. Only 1 of 89 recorded cases in tofactinib and baricitinib exposure occurred in countries at intermediate or high TB risk, and most of the cases probably were due to first mycobacterium tuberculosis (Mtb) exposure. Although no cases were observed in patients receiving upadacitinib and filgotinib, long-term trials and data from real-life are required to more precisely address the TB risk associated with the two drugs. AREAS COVERED Discussion on the TB risk associated with anti-JAKs, and on the need for accurate evaluation of host-related risk factors in high risk countries. EXPERT OPINION Available data on anti-JAKs suggest a negligible risk of active TB occurrence in low endemic areas.
Collapse
Affiliation(s)
| | - Corrado Blandizzi
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa , Pisa, Italy
| | - Laura Niccoli
- Rheumatology Department, Hospital of Prato , Prato, Italy
| | - Linda Petrone
- Translational Research Unit, Department of Epidemiology and Preclinical Research, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS , Rome, Italy
| | - Delia Goletti
- Translational Research Unit, Department of Epidemiology and Preclinical Research, "L. Spallanzani" National Institute for Infectious Diseases (INMI), IRCCS , Rome, Italy
| |
Collapse
|
31
|
Song LT, Lai W, Li JS, Mu YZ, Li CY, Jiang SY. The interaction between serum amyloid A and Toll-like receptor 2 pathway regulates inflammatory cytokine secretion in human gingival fibroblasts. J Periodontol 2019; 91:129-137. [PMID: 31347700 DOI: 10.1002/jper.19-0050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 05/19/2019] [Accepted: 05/29/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Serum amyloid A (SAA) has been identified to trigger inflammation response, and play a crucial role in chronic inflammatory diseases. However, the regulatory mechanism of SAA still remains unclear during the development of periodontitis METHODS: SAA mRNA and protein expression were detected in healthy and inflammatory gingival tissues using real-time polymerase chain reaction (PCR) and immunohistochemistry. Human recombinant SAA (Apo-SAA), Pam3CSK4 (a Toll-like receptor (TLR) 2 ligand), siRNA-SAA, or TLR2 neutralizing antibody was applied to treat human gingival fibroblasts, respectively, or combined. SAA, TLRs, and inflammatory cytokines interleukin (IL)-6 and IL-8 were analyzed by real-time PCR, western blotting, or enzyme-linked immunosorbent assay. RESULTS SAA expression increased in human inflammatory gingival tissues from patients with periodontitis (P <0.05). Apo-SAA could increase not only the mRNA expression of TLR2 (P <0.05), but also IL-6 and IL-8 mRNA and protein levels (P <0.05) which was suppressed by TLR2 antibody in human gingival fibroblasts. Pam3CSK4 increased SAA, IL-6, and IL-8 levels (P <0.05). However, the expression of SAA, IL-6, and IL-8 decreased after transfection of siRNA-SAA (P <0.05). CONCLUSION SAA not only increases in inflammatory gingiva, but also triggers inflammatory cytokine secretion via interacting with TLR2 pathway in human gingival fibroblasts, which indicates that SAA is involved in periodontal inflammation.
Collapse
Affiliation(s)
- Li-Ting Song
- Hospital of Stomatology, School of Dentistry, Tianjin Medical University, Tianjin, P. R. China
| | - Wen Lai
- Hospital of Stomatology, School of Dentistry, Tianjin Medical University, Tianjin, P. R. China
| | - Jia-Shan Li
- Hospital of Stomatology, School of Dentistry, Tianjin Medical University, Tianjin, P. R. China
| | - Yu-Zhu Mu
- Hospital of Stomatology, School of Dentistry, Tianjin Medical University, Tianjin, P. R. China
| | - Chang-Yi Li
- Hospital of Stomatology, School of Dentistry, Tianjin Medical University, Tianjin, P. R. China
| | - Shao-Yun Jiang
- Hospital of Stomatology, School of Dentistry, Tianjin Medical University, Tianjin, P. R. China.,Center of Stomatology, Shenzhen Hospital, Peking University, Shenzhen, Guangdong, P. R. China
| |
Collapse
|
32
|
Machavaram KK, Endo-Tsukude C, Terao K, Gill KL, Hatley OJ, Gardner I, Parrott N, Ducray PS. Simulating the Impact of Elevated Levels of Interleukin-6 on the Pharmacokinetics of Various CYP450 Substrates in Patients with Neuromyelitis Optica or Neuromyelitis Optica Spectrum Disorders in Different Ethnic Populations. AAPS JOURNAL 2019; 21:42. [PMID: 30887238 DOI: 10.1208/s12248-019-0309-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 02/18/2019] [Indexed: 01/05/2023]
Abstract
A physiologically based pharmacokinetic (PBPK) model was used to simulate the impact of elevated levels of interleukin (IL)-6 on the exposure of several orally administered cytochrome P450 (CYP) probe substrates (caffeine, S-warfarin, omeprazole, dextromethorphan, midazolam, and simvastatin). The changes in exposure of these substrates in subjects with rheumatoid arthritis (and hence elevated IL-6 levels) compared with healthy subjects were predicted with a reasonable degree of accuracy. The PBPK model was then used to simulate the change in oral exposure of the probe substrates in North European Caucasian, Chinese, and Japanese population of patients with neuromyelitis optica (NMO) or NMO spectrum disorder with elevated plasma IL-6 levels (up to 100 pg/mL). Moderate interactions [mean AUC fold change, ≤ 2.08 (midazolam) or 2.36 (simvastatin)] was predicted for CYP3A4 probe substrates and weak interactions (mean AUC fold change, ≤ 1.29-1.97) were predicted for CYP2C19, CYP2C9, and CYP2D6 substrates. No notable interaction was predicted with CYP1A2. Although ethnic differences led to differences in simulated exposure for some of the probe substrates, there were no marked differences in the predicted magnitude of the change in exposure following IL-6-mediated suppression of CYPs. Decreased levels of serum albumin (as reported in NMO patients) had little impact on the magnitude of the simulated IL-6-mediated drug interactions. This PBPK modeling approach allowed us to leverage knowledge from different disease and ethnic populations to make predictions of cytokine-related DDIs in a rare disease population where actual clinical studies would otherwise be difficult to conduct.
Collapse
Affiliation(s)
| | | | - Kimio Terao
- Clinical Pharmacology Department, Chugai Pharmaceutical Co., Ltd, Tokyo, Japan
| | | | | | - Iain Gardner
- Certara UK Limited, Simcyp Division, Sheffield, UK
| | - Neil Parrott
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | | |
Collapse
|
33
|
Simon N, Antignani A, Hewitt SM, Gadina M, Alewine C, FitzGerald D. Tofacitinib enhances delivery of antibody-based therapeutics to tumor cells through modulation of inflammatory cells. JCI Insight 2019; 4:123281. [PMID: 30720466 DOI: 10.1172/jci.insight.123281] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 01/29/2019] [Indexed: 12/14/2022] Open
Abstract
The routes by which antibody-based therapeutics reach malignant cells are poorly defined. Tofacitinib, an FDA-approved JAK inhibitor, reduced tumor-associated inflammatory cells and allowed increased delivery of antibody-based agents to malignant cells. Alone, tofacitinib exhibited no antitumor activity, but combinations with immunotoxins or an antibody-drug conjugate resulted in increased antitumor responses. Quantification using flow cytometry revealed that antibody-based agents accumulated in malignant cells at higher percentages following tofacitinib treatment. Profiling of tofacitinib-treated tumor-bearing mice indicated that cytokine transcripts and various proteins involved in chemotaxis were reduced compared with vehicle-treated mice. Histological analysis revealed significant changes to the composition of the tumor microenvironment, with reductions in monocytes, macrophages, and neutrophils. Tumor-associated inflammatory cells contributed to non-target uptake of antibody-based therapeutics, with mice treated with tofacitinib showing decreased accumulation of therapeutics in intratumoral inflammatory cells and increased delivery to malignant cells. The present findings serve as a rationale for conducting trials where short-term treatments with tofacitinib could be administered in combination with antibody-based therapies.
Collapse
Affiliation(s)
- Nathan Simon
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Antonella Antignani
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Stephen M Hewitt
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Massimo Gadina
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Christine Alewine
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - David FitzGerald
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| |
Collapse
|
34
|
Dieter BP, Meek RL, Anderberg RJ, Cooney SK, Bergin JL, Zhang H, Nair V, Kretzler M, Brosius FC, Tuttle KR. Serum amyloid A and Janus kinase 2 in a mouse model of diabetic kidney disease. PLoS One 2019; 14:e0211555. [PMID: 30763329 PMCID: PMC6375550 DOI: 10.1371/journal.pone.0211555] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 01/16/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Serum amyloid A (SAA), a potent inflammatory mediator, and Janus kinase 2 (JAK2), an intracellular signaling kinase, are increased by diabetes. The aims were to elucidate: 1) a JAK2-mediated pathway for increased SAA in the kidneys of diabetic mice; 2) a JAK2-SAA pathway for inflammation in podocytes. METHODS Akita diabetic mice (129S6) with podocyte JAK2 overexpression and angiotensin II infusion (4 weeks) were given a JAK1,2 inhibitor (LY03103801, 3 mg/kg/day orally for the last two weeks). Kidneys were immunostained for SAA isoform 3 (SAA3). SAA3 knockout and control mouse podocytes were exposed to advanced glycation end products (AGE) or exogenous SAA with JAK2 inhibition (Tyrphostin AG 490, 50μM). JAK2 activity (phosphorylation, Western blot, 1 hour) and mRNA for SAA3 and associated inflammatory genes (Cxcl5, Ccl2, and Ccl5) were measured by RT-PCR (20 hours). RESULTS SAA3 protein was present throughout the diabetic kidney, and podocyte JAK2 overexpression increased tubulointerstitial SAA3 compared to wild type diabetic controls, 43% versus 14% (p = 0.007); JAK1,2 inhibition attenuated the increase in SAA3 to 15% (p = 0.003). Urine albumin-to-creatinine ratio (r = 0.49, p = 0.03), mesangial index (r = 0.64, p = 0.001), and glomerulosclerosis score (r = 0.51, p = 0.02) were associated with SAA3 immunostaining scores across mouse groups. Exposing podocytes to AGE or exogenous SAA increased JAK2 activity within one hour and mRNA for associated inflammatory genes after 20 hours. JAK2 inhibition reduced SAA3 mRNA expression in podocytes exposed to AGE or SAA. SAA3 knockout podocytes had >85% lower AGE-induced inflammatory genes. CONCLUSION JAK1,2 inhibition reduced SAA and histological features of DKD in podocyte JAK2-overexpressing mice. In podocytes exposed to a diabetes-like condition, JAK2 inhibition reduced expression of SAA, while SAA knockout blocked expression of associated pro-inflammatory mediators. SAA may promote JAK2-dependent inflammation in the diabetic kidney.
Collapse
Affiliation(s)
- Brad P. Dieter
- Providence Medical Research Center, Providence Health Care, Spokane, Washington, United States of America
| | - Rick L. Meek
- Providence Medical Research Center, Providence Health Care, Spokane, Washington, United States of America
| | - Robert J. Anderberg
- Providence Medical Research Center, Providence Health Care, Spokane, Washington, United States of America
| | - Sheryl K. Cooney
- Providence Medical Research Center, Providence Health Care, Spokane, Washington, United States of America
| | - Jen L. Bergin
- Providence Medical Research Center, Providence Health Care, Spokane, Washington, United States of America
| | - Hongyu Zhang
- Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Viji Nair
- Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Matthias Kretzler
- Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Frank C. Brosius
- Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Katherine R. Tuttle
- Providence Medical Research Center, Providence Health Care, Spokane, Washington, United States of America
- Institute of Translational Health Sciences, Kidney Research Institute, Nephrology Division University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
35
|
Cao Y, Xu S, Kong W, Cai H, Xu Y. Identification and validation of differentially expressed proteins in serum of CSU patients with different duration of wheals using an iTRAQ labeling, 2D-LC-MS/MS. Exp Ther Med 2018; 16:4527-4536. [PMID: 30542401 PMCID: PMC6257644 DOI: 10.3892/etm.2018.6818] [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: 03/23/2018] [Accepted: 09/14/2018] [Indexed: 11/28/2022] Open
Abstract
Chronic spontaneous urticaria (CSU) is one of the most common types of chronic urticaria (CU), with symptoms that recur easily, migrate and are refractory. It is unclear whether association between the differentiation of protein expression levels in the serum of CSU patients and the different duration of wheals exists. In the present study the samples were divided according to the duration of the wheals into group A (wheal duration <2 h) and group B (wheal duration 12–24 h). Differentially expressed proteins in sera of CSU patients with different durations of wheals were identified and validated with isobaric tags for relative and absolute quantitation (iTRAQ) in combination with two-dimensional liquid chromatography/tandem mass spectrometry (2D-LC-MS/MS). Three hundred and seventy CSU serum-related proteins were initially identified. Among these proteins, ~30 had significant differences between the groups. According to the classification of biological functions and upregulated/downregulated values, serum amyloid A (SAA), CFL1, TPM4 and monocyte differentiation antigen (CD14) were chosen and validated by enzyme-linked immunosorbent assay (ELISA). The expression levels of CD14 in sera were not significantly different among the groups. SAA, CFL1 and TPM4 were associated with the wheal duration in CSU patients and therefore could be considered as new potential inflammatory biomarkers associated with CSU.
Collapse
Affiliation(s)
- Yanyun Cao
- Department of Dermatology, Pudong New Area People's Hospital, Shanghai 201299, P.R. China
| | - Shunming Xu
- Department of Dermatology, Pudong New Area People's Hospital, Shanghai 201299, P.R. China
| | - Wei Kong
- Department of Dermatology, Pudong New Area People's Hospital, Shanghai 201299, P.R. China
| | - Haibin Cai
- Department of Dermatology, Pudong New Area People's Hospital, Shanghai 201299, P.R. China
| | - Yang Xu
- Department of Dermatology, Pudong New Area People's Hospital, Shanghai 201299, P.R. China
| |
Collapse
|
36
|
Development of a Novel Diagnostic Biomarker Set for Rheumatoid Arthritis Using a Proteomics Approach. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7490723. [PMID: 30662913 PMCID: PMC6312602 DOI: 10.1155/2018/7490723] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/28/2018] [Accepted: 11/05/2018] [Indexed: 12/13/2022]
Abstract
Background Rheumatoid arthritis (RA) is an autoimmune disease that starts with inflammation of the synovial membrane. Studies have been conducted to develop methods for efficient diagnosis of RA and to identify the mechanisms underlying RA development. Blood samples can be useful for detecting disturbance of homeostasis in patients with RA. Nanoliquid chromatography-tandem mass spectrometry (LC-MS/MS) is an efficient proteomics approach to analyze blood sample and quantify serum proteins. Methods Serum samples of 18 healthy controls and 18 patients with RA were analyzed by LC-MS/MS. Selected candidate biomarkers were validated by enzyme-linked immunosorbent assay (ELISA) using sera from 43 healthy controls and 44 patients with RA. Results Thirty-eight proteins were significantly differentially expressed by more than 2-fold in healthy controls and patients with RA. Based on a literature survey, we selected six candidate RA biomarkers. ELISA was used to evaluate whether these proteins effectively allow distinguishing patients with RA from healthy controls and monitoring drug efficacy. SAA4, gelsolin, and vitamin D-binding protein were validated as potential biomarkers of RA for screening and drug efficacy monitoring of RA. Conclusions We identified a panel of three biomarkers for RA which has potential for application in RA diagnosis and drug efficacy monitoring. Further, our findings will aid in understanding the pathogenesis of RA.
Collapse
|
37
|
Gastrointestinal Perforations with Biologics in Patients with Rheumatoid Arthritis: Implications for Clinicians. Drug Saf 2018; 41:545-553. [PMID: 29392593 DOI: 10.1007/s40264-018-0639-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Gastrointestinal (GI) perforations are rare events in rheumatoid arthritis (RA) patients, but cause significant morbidity and mortality. Several studies indicate that RA patients may be at higher risk of GI perforation. Traditional RA treatments such as glucocorticoids and non-steroidal anti-inflammatory drugs increase the risk of perforation. In the past two decades, a new class of therapeutic agents called biologics has been added to the RA treatment armamentarium. Biologics are effective in controlling disease activity and are generally well tolerated; however, reports of GI perforations in association with biologics have arisen. In particular, drugs that inhibit the interleukin (IL)-6 cytokine receptor have demonstrated a higher risk of perforation compared with other therapies. Recent reports also suggest that janus kinase inhibitors may increase the risk of perforation, perhaps via downstream effects on IL-6 signaling. In this review, we discuss current data on the risk of GI perforations among RA patients receiving targeted therapies and its clinical relevance.
Collapse
|
38
|
Ko EK, Chorich LP, Sullivan ME, Cameron RS, Layman LC. JAK/STAT signaling pathway gene expression is reduced following Nelf knockdown in GnRH neurons. Mol Cell Endocrinol 2018; 470:151-159. [PMID: 29050862 DOI: 10.1016/j.mce.2017.10.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 10/13/2017] [Accepted: 10/14/2017] [Indexed: 12/27/2022]
Abstract
Hypothalamic gonadotropin releasing hormone (GnRH) is crucial for the proper function of the hypothalamic-pituitary-gonadal (HPG) axis, subsequent puberty, and reproduction. When GnRH neuron migration or GnRH regulation is impaired, hypogonadotropic hypogonadism results. Mutations in the gene for nasal embryonic luteinizing hormone-releasing factor (NELF) have been identified in GnRH-deficient humans. NELF is a predominantly nuclear protein that may participate in gene transcription, but the genes NELF regulates are unknown. To address this question, RNA was extracted from NLT GnRH neuronal cells following either stable Nelf knockdown or scrambled control and subjected to cDNA arrays. Transcription factors and cell migration gene expression was altered most commonly. Members of the Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathway, including Stat1, Stat2, Stat5a, Jak2, Irf7 and Irf9, were significantly down regulated as assessed by RT-qPCR. Protein levels of STAT1, phospho-STAT1, and JAK2 were reduced, but the protein level of phospho-JAK2 was not. These findings suggest a role for NELF in the regulation of the JAK/STAT signaling pathway, which have important functions in GnRH neurons.
Collapse
Affiliation(s)
- Eun Kyung Ko
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, United States
| | - Lynn P Chorich
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, United States
| | - Megan E Sullivan
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, United States
| | - Richard S Cameron
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, United States
| | - Lawrence C Layman
- Section of Reproductive Endocrinology, Infertility, & Genetics, Department of Obstetrics & Gynecology, Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
39
|
Giollo A, Bissell LA, Buch MH. Cardiovascular outcomes of patients with rheumatoid arthritis prescribed disease modifying anti-rheumatic drugs: a review. Expert Opin Drug Saf 2018; 17:697-708. [PMID: 29871535 DOI: 10.1080/14740338.2018.1483331] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is associated with a heightened risk of cardiovascular disease (CVD), with both traditional CV risk factors and inflammation contributing to this risk. AREAS COVERED This review highlights the burden of CVD in RA and associated traditional CV risk factors, including the complexity of dyslipidemia in RA and the so-called 'lipid paradox.' Furthermore, the recognized RA-disease-specific factors associated with higher risk of CVD and the role of systemic inflammation in the pathogenesis of CVD in RA will be addressed. With the advent of biologic and targeted synthetic therapies in the treatment of RA, the effect of conventional and newer generation disease modifying anti-rheumatic therapies (DMARDs) on CV risk and associated risk factors will also be discussed. EXPERT OPINION Identifying the RA phenotype at greatest risk of CVD, understanding the interplay of increased traditional risk factors, common inflammatory processes and RA-specific factors, and personalized use of DMARDs according to disease phenotype and comorbidity to reduce this risk are key areas for future research.
Collapse
Affiliation(s)
- Alessandro Giollo
- a Leeds Institute of Rheumatic and Musculoskeletal Medicine , University of Leeds, Chapel Allerton Hospital , Leeds , UK.,b NIHR Leeds Biomedical Research Centre , Leeds Teaching Hospitals NHS Trust , Leeds , LS7 4SA , UK.,c Department of Medicine, Rheumatology Unit , University of Verona , Verona , Italy
| | - Lesley-Anne Bissell
- a Leeds Institute of Rheumatic and Musculoskeletal Medicine , University of Leeds, Chapel Allerton Hospital , Leeds , UK.,b NIHR Leeds Biomedical Research Centre , Leeds Teaching Hospitals NHS Trust , Leeds , LS7 4SA , UK
| | - Maya H Buch
- a Leeds Institute of Rheumatic and Musculoskeletal Medicine , University of Leeds, Chapel Allerton Hospital , Leeds , UK.,b NIHR Leeds Biomedical Research Centre , Leeds Teaching Hospitals NHS Trust , Leeds , LS7 4SA , UK
| |
Collapse
|
40
|
Febvre-James M, Bruyère A, Le Vée M, Fardel O. The JAK1/2 Inhibitor Ruxolitinib Reverses Interleukin-6-Mediated Suppression of Drug-Detoxifying Proteins in Cultured Human Hepatocytes. Drug Metab Dispos 2018; 46:131-140. [PMID: 29162613 DOI: 10.1124/dmd.117.078048] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/17/2017] [Indexed: 02/13/2025] Open
Abstract
The inflammatory cytokine interleukin (IL)-6, which basically activates the Janus kinase (JAK)/ signal transducer and activator of transcription (STAT) signaling pathway, is well known to repress expression of hepatic cytochromes P-450 (P450s) and transporters. Therapeutic proteins, like monoclonal antibodies targeting IL-6 or its receptor, have consequently been demonstrated to restore full hepatic detoxification capacity, which results in inflammatory disease-related drug-drug interactions (idDDIs). In the present study, we investigated whether ruxolitinib, a small drug acting as a JAK1/2 inhibitor and currently used in the treatment of myeloproliferative neoplasms, may also counteract the repressing effects of IL-6 toward hepatic detoxifying systems. Ruxolitinib was found to fully inhibit IL-6-mediated repression of P450 (CYP1A2, CYP2B6, and CYP3A4) and transporter (NTCP, OATP1B1, and OCT1) mRNA levels in primary human hepatocytes and differentiated hepatoma HepaRG cells. Such effects were dose-dependent, with ruxolitinib EC50 values around 1.0-1.2 μM and thus close to ruxolitinib plasma levels that can be reached in patients. Moreover, they were associated with concomitant restoration of P450 and drug transporter activities in IL-6-exposed HepaRG cells. By contrast, ruxolitinib failed to suppress the repression of drug-detoxifying protein mRNA levels caused by IL-1β The JAK inhibitor and anti-rheumatoid arthritis compound tofacitinib was additionally found to reverse IL-6-mediated suppression of P450 and transporter mRNA expressions. Taken together, our results demonstrated that small drugs acting as JAK inhibitors, like ruxolitinib, counteract IL-6-mediated repression of drug-metabolizing enzymes and drug transporters in cultured human hepatocytes. These JAK inhibitors may consequently be hypothesized to restore hepatic detoxification capacity for patients suffering from inflammatory diseases, which may in turn cause idDDIs.
Collapse
Affiliation(s)
- Marie Febvre-James
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, Université de Rennes 1, Rennes, France (M.F.-J., A.B., M.L.V., O.F.) and Pôle Biologie, Centre Hospitalier Universitaire, Rennes, France (O.F.)
| | - Arnaud Bruyère
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, Université de Rennes 1, Rennes, France (M.F.-J., A.B., M.L.V., O.F.) and Pôle Biologie, Centre Hospitalier Universitaire, Rennes, France (O.F.)
| | - Marc Le Vée
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, Université de Rennes 1, Rennes, France (M.F.-J., A.B., M.L.V., O.F.) and Pôle Biologie, Centre Hospitalier Universitaire, Rennes, France (O.F.)
| | - Olivier Fardel
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, Université de Rennes 1, Rennes, France (M.F.-J., A.B., M.L.V., O.F.) and Pôle Biologie, Centre Hospitalier Universitaire, Rennes, France (O.F.)
| |
Collapse
|
41
|
Rogier R, Evans-Marin H, Manasson J, van der Kraan PM, Walgreen B, Helsen MM, van den Bersselaar LA, van de Loo FA, van Lent PL, Abramson SB, van den Berg WB, Koenders MI, Scher JU, Abdollahi-Roodsaz S. Alteration of the intestinal microbiome characterizes preclinical inflammatory arthritis in mice and its modulation attenuates established arthritis. Sci Rep 2017; 7:15613. [PMID: 29142301 PMCID: PMC5688157 DOI: 10.1038/s41598-017-15802-x] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 11/02/2017] [Indexed: 12/12/2022] Open
Abstract
Perturbations of the intestinal microbiome have been observed in patients with new-onset and chronic autoimmune inflammatory arthritis. However, it is currently unknown whether these alterations precede the development of arthritis or are rather a consequence of disease. Modulation of intestinal microbiota by oral antibiotics or germ-free condition can prevent arthritis in mice. Yet, the therapeutic potential of modulation of the microbiota after the onset of arthritis is not well characterized. We here show that the intestinal microbial community undergoes marked changes in the preclinical phase of collagen induced arthritis (CIA). The abundance of the phylum Bacteroidetes, specifically families S24-7 and Bacteroidaceae was reduced, whereas Firmicutes and Proteobacteria, such as Ruminococcaceae, Lachnospiraceae and Desulfovibrinocaceae, were expanded during the immune-priming phase of arthritis. In addition, we found that the abundance of lamina propria Th17, but not Th1, cells is highly correlated with the severity of arthritis. Elimination of the intestinal microbiota during established arthritis specifically reduced intestinal Th17 cells and attenuated arthritis. These effects were associated with reduced serum amyloid A expression in ileum and synovial tissue. Our observations suggest that intestinal microbiota perturbations precede arthritis, and that modulation of the intestinal microbiota after the onset of arthritis may offer therapeutic opportunities.
Collapse
Affiliation(s)
- Rebecca Rogier
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Heather Evans-Marin
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, United States
| | - Julia Manasson
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, United States
| | - Peter M van der Kraan
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Birgitte Walgreen
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Monique M Helsen
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Fons A van de Loo
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Peter L van Lent
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Steven B Abramson
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, United States
| | - Wim B van den Berg
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marije I Koenders
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jose U Scher
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, United States
| | - Shahla Abdollahi-Roodsaz
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands.
- Department of Medicine, Division of Rheumatology, New York University School of Medicine, New York, United States.
| |
Collapse
|
42
|
Prevention of chronic renal allograft rejection by AS2553627, a novel JAK inhibitor, in a rat transplantation model. Transpl Immunol 2017; 46:14-20. [PMID: 28988984 DOI: 10.1016/j.trim.2017.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 10/02/2017] [Accepted: 10/04/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Janus kinase (JAK) inhibitors are thought to be promising candidates to aid renal transplantation. However, the effectiveness of JAK inhibitors against features of chronic rejection, including interstitial fibrosis/tubular atrophy (IF/TA) and glomerulosclerosis, has not been elucidated. Here, we investigated the effect of AS2553627, a novel JAK inhibitor, on the development of chronic rejection in rat renal transplantation. METHODS Lewis (LEW) to Brown Norway (BN) rat renal transplantation was performed. Tacrolimus (TAC) at 0.1mg/kg was administered intramuscularly once a day for 10 consecutive days starting on the day of transplantation (days 0 to 9) to prevent initial acute rejection. After discontinuation of TAC treatment from days 10 to 28, AS2553627 (1 and 10mg/kg) was orally administered with TAC. At 13weeks after renal transplantation, grafts were harvested for histopathological and mRNA analysis. Creatinine and donor-specific antibodies were measured from plasma samples. Urinary protein and kidney injury markers were also evaluated. RESULTS AS2553627 in combination with TAC exhibited low plasma creatinine and a marked decrease in urinary protein and kidney injury markers, such as tissue inhibitor of metalloproteinase-1 and kidney injury molecule-1. At 13weeks, histopathological analysis revealed that AS2553627 treatment inhibited glomerulosclerosis and IF/TA. In addition, upregulation of cell surface markers, fibrosis/epithelial-mesenchymal transition and inflammation-related genes were reduced by the combination of AS2553672 and TAC, particularly CD8 and IL-6 mRNAs, indicating that AS2553627 prevented cell infiltration and inflammation in renal allografts. CONCLUSIONS These results indicate the therapeutic potential of JAK inhibitors in chronic rejection progression, and suggest that AS2553627 is a promising agent to improve long-term graft survival after renal transplantation.
Collapse
|
43
|
Efficacy of topical tofacitinib in promoting hair growth in non-scarring alopecia: possible mechanism via VEGF induction. Arch Dermatol Res 2017; 309:729-738. [DOI: 10.1007/s00403-017-1777-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 08/12/2017] [Accepted: 08/31/2017] [Indexed: 01/15/2023]
|
44
|
Aguilar-Pimentel A, Graessel A, Alessandrini F, Fuchs H, Gailus-Durner V, Hrabě de Angelis M, Russkamp D, Chaker A, Ollert M, Blank S, Gutermuth J, Schmidt-Weber CB. Improved efficacy of allergen-specific immunotherapy by JAK inhibition in a murine model of allergic asthma. PLoS One 2017; 12:e0178563. [PMID: 28570653 PMCID: PMC5453633 DOI: 10.1371/journal.pone.0178563] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/15/2017] [Indexed: 12/22/2022] Open
Abstract
Background Allergen-specific immunotherapy (AIT) is the only curative treatment for type-1 allergies, but sometimes shows limited therapeutic response as well as local and systemic side effects. Limited control of local inflammation and patient symptoms hampers its widespread use in severe allergic asthma. Objective Our aim was to evaluate whether AIT is more effective in suppression of local inflammation if performed under the umbrella of short-term non-specific immunomodulation using a small molecule inhibitor of JAK pathways. Methods In C57BL/6J mice, a model of ovalbumin (OVA)-induced allergic airway inflammation and allergen-specific immunotherapy was combined with the administration of Tofacitinib (TOFA, a FDA-approved JAK inhibitor) from 48 hours prior to 48 hours after therapeutic OVA-injection. The effect of TOFA on human FOXP3+CD4+ T cells was studied in vitro. Results AIT combined with short-term TOFA administration was significantly more effective in suppressing total cell and eosinophil infiltration into the lung, local cytokine production including IL-1β and CXCL1 and showed a trend for the reduction of IL-4, IL-13, TNF-α and IL-6 compared to AIT alone. Furthermore, TOFA co-administration significantly reduced systemic IL-6, IL-1β and OVA-specific IgE levels and induced IgG1 to the same extent as AIT alone. Additionally, TOFA enhanced the induction of human FOXP3+CD4+ T cells. Conclusions This proof of concept study shows that JAK inhibition did not inhibit tolerance induction, but improved experimental AIT at the level of local inflammation. The improved control of local inflammation might extend the use of AIT in more severe conditions such as polyallergy, asthma and high-risk patients suffering from mastocytosis or anaphylaxis.
Collapse
Affiliation(s)
- Antonio Aguilar-Pimentel
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Center Munich, Neuherberg, Germany
| | - Anke Graessel
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Member of the German Center for Lung research (DZL), Munich, Germany
| | - Francesca Alessandrini
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Member of the German Center for Lung research (DZL), Munich, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Center Munich, Neuherberg, Germany
| | - Valerie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Center Munich, Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Center Munich, Neuherberg, Germany.,Experimental Genetics, School of Life Science Weihenstephan, Technical University of Munich, Freising, Germany
| | - Dennis Russkamp
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Member of the German Center for Lung research (DZL), Munich, Germany
| | - Adam Chaker
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Member of the German Center for Lung research (DZL), Munich, Germany.,Department of Otolaryngology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg.,Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis, University of Southern Denmark, Odense, Denmark
| | - Simon Blank
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Member of the German Center for Lung research (DZL), Munich, Germany
| | - Jan Gutermuth
- Department of Dermatology, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Member of the German Center for Lung research (DZL), Munich, Germany
| |
Collapse
|
45
|
Bryleva EY, Keaton SA, Grit J, Madaj Z, Sauro-Nagendra A, Smart L, Halstead S, Achtyes E, Brundin L. The acute-phase mediator serum amyloid A is associated with symptoms of depression and fatigue. Acta Psychiatr Scand 2017; 135:409-418. [PMID: 28374419 DOI: 10.1111/acps.12730] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Establish whether inflammatory biomarkers-serum amyloid A (SAA), C-reactive protein (CRP), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α)-are related to key symptoms of depression, including anxiety and fatigue, in a cross-sectional, out-patient setting to identify biomarkers that reflect psychiatric symptomatology in a naturalistic, real-life population. METHODS We measured SAA, CRP, IL-6, and TNF-α in plasma samples from 89 adult psychiatric out-patients by multiplex, high-sensitivity electrochemiluminescent assays. Psychiatric symptoms were evaluated using the Hamilton Depression Rating Scale (HAMD-17), the Patient Health Questionnaire (PHQ-9), and the Center for Epidemiological Studies Depression Scale (CES-D). RESULTS Plasma SAA was most robustly associated with depressive symptoms across diagnostic boundaries in this cohort of out-patients. Elevated SAA was significantly associated with higher total scores on the HAMD-17 scale and correlated with multiple scale items that rated symptoms of fatigue and depressed mood, but not with anxiety-related items. CONCLUSIONS SAA might constitute a cross-diagnostic marker indicative of depressed mood and fatigue in a naturalistic patient setting. Because SAA activates Toll-like receptors 2 and 4, present on macrophages and glial cells, its association with depression severity could also implicate this inflammatory mediator in the pathogenesis of mood disorders.
Collapse
Affiliation(s)
- E Y Bryleva
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - S A Keaton
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA.,Division of Psychiatry and Behavioral Medicine, Michigan State University, Grand Rapids, MI, USA.,Department of Physiology, Michigan State University, East Lansing, MI, USA
| | - J Grit
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Z Madaj
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, MI, USA
| | - A Sauro-Nagendra
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - L Smart
- Pine Rest Christian Mental Health Services, Grand Rapids, MI, USA
| | - S Halstead
- Pine Rest Christian Mental Health Services, Grand Rapids, MI, USA
| | - E Achtyes
- Division of Psychiatry and Behavioral Medicine, Michigan State University, Grand Rapids, MI, USA.,Pine Rest Christian Mental Health Services, Grand Rapids, MI, USA
| | - L Brundin
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA.,Pine Rest Christian Mental Health Services, Grand Rapids, MI, USA
| |
Collapse
|
46
|
Yu N, Zhang S, Lu J, Li Y, Yi X, Tang L, Su L, Ding Y. Serum amyloid A, an acute phase protein, stimulates proliferative and proinflammatory responses of keratinocytes. Cell Prolif 2016; 50. [PMID: 27910163 DOI: 10.1111/cpr.12320] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/27/2016] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVES Serum amyloid A (SAA), an acute phase protein, is highly expressed in psoriatic lesions but its function is not fully understood. The aim of this study was to explore its role in activation of keratinocytes. MATERIALS AND METHODS Real-time PCR and immunofluorescence were performed to examine SAA expression in imiquimod (IMQ)-induced psoriasis-like mice. In vivo function of SAA was examined by treating psoriasis-like mice with SAA neutralising antibody. Cell viability was monitored using the CCK-8 assay. Real-time PCR was performed to determine expression of genes associated with differentiation and inflammation. Ki67+ percentage and immunological markers were analysed by flow cytometry. Involvement of formyl peptide receptor-like 1 (FPRL1) in SAA signal transduction was determined by RNA interference. Binding of SAA and FPRL1 was examined by co-immunoprecipitaion. Western blotting was conducted to assess phosphorylation of downstream signalling molecules. RESULTS SAA was highly expressed in skin lesions of IMQ-treated psoriasis-like mice and neutralising SAA attenuated epidermal hyperplasia and inflammation. SAA in vitro promoted keratinocyte proliferation and expression of immunological mediators, while inhibiting differentiation. Effects of SAA on keratinocyte proliferation and inflammation were mediated by FPRL1, as well as activation of the PI3K/Akt pathway. CONCLUSIONS These observations indicate that SAA/FPRL1 contributed to pathogenesis of psoriasis by promoting keratinocyte proliferation and inflammation, thus providing a potential therapeutic target for disease therapy.
Collapse
Affiliation(s)
- Ning Yu
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| | - Shujie Zhang
- Experimental Research Center, Eye and ENT Hospital of Fudan University, Shanghai, China
| | - Jiajing Lu
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| | - Ying Li
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| | - Xuemei Yi
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| | - Li Tang
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| | - Lina Su
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| | - Yangfeng Ding
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| |
Collapse
|
47
|
Hwang YG, Balasubramani GK, Metes ID, Levesque MC, Bridges SL, Moreland LW. Differential response of serum amyloid A to different therapies in early rheumatoid arthritis and its potential value as a disease activity biomarker. Arthritis Res Ther 2016; 18:108. [PMID: 27188329 PMCID: PMC4869396 DOI: 10.1186/s13075-016-1009-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/29/2016] [Indexed: 12/23/2022] Open
Abstract
Background The aim was to compare the effect of etanercept (ETN) and conventional synthetic disease-modifying anti-rheumatic drug (DMARD) therapy on serum amyloid A (SAA) levels and to determine whether SAA reflects rheumatoid arthritis (RA) disease activity better than C-reactive protein (CRP). Methods We measured SAA and CRP at baseline, 24, 48, and 102 week follow-up visits in 594 patients participating in the Treatment of early RA (TEAR) study. We used Spearman correlation coefficients (rho) to evaluate the relationship between SAA and CRP and mixed effects models to determine whether ETN and methotrexate (MTX) treatment compared to triple DMARD therapy differentially lowered SAA. Akaike information criteria (AIC) were used to determine model fits. Results SAA levels were only moderately correlated with CRP levels (rho = 0.58, p < 0.0001). There were significant differences in SAA by both visit (p = 0.0197) and treatment arm (p = 0.0130). RA patients treated with ETN plus MTX had a larger reduction in SAA than patients treated with traditional DMARD therapy. Similar results were found for serum CRP by visit (p = 0.0254) and by treatment (p < 0.0001), with a more pronounced difference than for SAA. Across all patients and time points, models of the disease activity score of 28 joints (DAS28)-erythrocyte sedimentation rate (ESR) using SAA levels were better than models using CRP; the ΔAIC between the SAA and CRP models was 305. Conclusions SAA may be a better biomarker of RA disease activity than CRP, especially during treatment with tumor necrosis factor (TNF) antagonists. This warrants additional studies in other cohorts of patients on treatment for RA. Trial registration (ClinicalTrials.gov identifier: NCT00259610, Date of registration: 28 November 2005)
Collapse
Affiliation(s)
- Yong Gil Hwang
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, PA, 15261, USA.
| | - Goundappa K Balasubramani
- Department of Epidemiology, School of Public Health, University of Pittsburgh, 130 DeSoto Street, 127 Parran Hall, Pittsburgh, PA, 15261, USA
| | - Ilinca D Metes
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Marc C Levesque
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, PA, 15261, USA.,AbbVie Inc, 100 Research Dr, Worcester, MA, 01605, USA
| | - S Louis Bridges
- Department of Medicine, Division of Clinical Immunology and Rheumatology Birmingham, University of Alabama at Birmingham, Shelby Building, Room 178B, 1825 University Blvd., Birmingham, AL, 35294-2182, USA
| | - Larry W Moreland
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, PA, 15261, USA
| |
Collapse
|
48
|
Schwartz DM, Bonelli M, Gadina M, O'Shea JJ. Type I/II cytokines, JAKs, and new strategies for treating autoimmune diseases. Nat Rev Rheumatol 2016; 12:25-36. [PMID: 26633291 PMCID: PMC4688091 DOI: 10.1038/nrrheum.2015.167] [Citation(s) in RCA: 452] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cytokines are major drivers of autoimmunity, and biologic agents targeting cytokines have revolutionized the treatment of immune-mediated diseases. Despite the effectiveness of these drugs, they do not induce complete remission in all patients, prompting the development of alternative strategies - including targeting of intracellular signal transduction pathways downstream of cytokines. Many cytokines that bind type I and type II cytokine receptors are critical regulators of immune-mediated diseases and employ the Janus kinase (JAK) and signal transducer and activator of transcription (STAT) pathway to exert their effect. Pharmacological inhibition of JAKs blocks the actions of type I/II cytokines, and within the past 3 years therapeutic JAK inhibitors, or Jakinibs, have become available to rheumatologists. Jakinibs have proven effective for the treatment of rheumatoid arthritis and other inflammatory diseases. Adverse effects of these agents are largely related to their mode of action and include infections and hyperlipidemia. Jakinibs are currently being investigated for a number of new indications, and second-generation selective Jakinibs are being developed and tested. Targeting STATs could be a future avenue for the treatment of rheumatologic diseases, although substantial challenges remain. Nonetheless, the ability to therapeutically target intracellular signalling pathways has already created a new paradigm for the treatment of rheumatologic disease.
Collapse
Affiliation(s)
- Daniella M Schwartz
- Molecular Immunology &Inflammation Branch, and Translational Immunology Section, National Institute of Arthritis &Musculoskeletal and Skin Diseases, NIH, Building 10, 9000 Rockville Pike, Bethesda, Maryland 20892-1616, USA
| | - Michael Bonelli
- Medical University of Vienna, Department of Rheumatology, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Massimo Gadina
- Molecular Immunology &Inflammation Branch, and Translational Immunology Section, National Institute of Arthritis &Musculoskeletal and Skin Diseases, NIH, Building 10, 9000 Rockville Pike, Bethesda, Maryland 20892-1616, USA
| | - John J O'Shea
- Molecular Immunology &Inflammation Branch, and Translational Immunology Section, National Institute of Arthritis &Musculoskeletal and Skin Diseases, NIH, Building 10, 9000 Rockville Pike, Bethesda, Maryland 20892-1616, USA
| |
Collapse
|
49
|
Yu N, Liu S, Yi X, Zhang S, Ding Y. Serum amyloid A induces interleukin-1β secretion from keratinocytes via the NACHT, LRR and PYD domains-containing protein 3 inflammasome. Clin Exp Immunol 2015; 179:344-53. [PMID: 25231464 DOI: 10.1111/cei.12458] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2014] [Indexed: 01/10/2023] Open
Abstract
Interleukin (IL)-1β is now emerging as a critical cytokine in the pathogenesis of T helper type 17 (Th17)-mediated skin diseases, including psoriasis. Psoriatic keratinocytes are a major source of IL-1β; however, the mechanisms triggering IL-1β processing remain unknown. Recently, an acute-phase protein serum amyloid A (SAA) has been identified as a danger signal that triggers inflammasome activation and IL-1β secretion. In this study, we detected increased SAA mRNA and protein expression in psoriatic epidermis. In cultured keratinocytes, SAA up-regulated the expression of pro-IL-1β and secretion of mature IL-1β. On the transcriptional level, blocking Toll-like receptor-2 (TLR-2), TLR-4 or nuclear factor kappa B (NF-κB) attenuated SAA-induced expression of IL-1β mRNA. SAA up-regulated caspase-1 and NACHT, LRR and PYD domains-containing protein 3 (NLRP3) expression in keratinocytes. Inhibiting caspase-1 activity and silencing NLRP3 decreased IL-1β secretion, confirming NLRP3 as the SAA-responsive inflammasome on the post-transcriptional level. The mechanism of SAA-triggered NLRP3 activation and subsequent IL-1β secretion was found to involve the generation of reactive oxygen species. Finally, the expression of SAA by keratinocytes was up-regulated by IL-17A. Taken together, our results indicate that keratinocyte-derived SAA triggers a key inflammatory mediator, IL-1β, via NLRP3 inflammasome activation, providing new potential targets for the treatment of this chronic skin disease.
Collapse
Affiliation(s)
- N Yu
- Department of Dermatology, Shanghai Skin Disease Hospital, Shanghai, China
| | | | | | | | | |
Collapse
|