1
|
Konzett V, Smolen JS, Nash P, Winthrop K, Aletaha D, Dörner T, Fleischmann R, Tanaka Y, Primdahl J, Baraliakos X, McInnes IB, Trauner M, Sattar N, de Wit M, Schoones JW, Kerschbaumer A. Safety of Janus kinase inhibitors in immune-mediated inflammatory diseases-a systematic literature review informing the 2024 update of an international expert consensus statement. Ann Rheum Dis 2025:S0003-4967(25)00080-9. [PMID: 39934016 DOI: 10.1016/j.ard.2025.01.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 02/13/2025]
Abstract
OBJECTIVES This systematic literature review (SLR) on safety outcomes was performed to inform the 2024 update of the expert consensus statement on the treatment of immune-mediated inflammatory diseases (IMIDs) with Janus kinase inhibitors (JAKi). METHODS An update of the 2019 SLR was performed in MEDLINE, Embase, and the Cochrane Library. For safety, randomised, placebo-controlled or active-controlled trials on all JAKi investigated in IMIDs, long-term extension (LTE) studies, pooled trial data analyses, and cohort and claims studies were included. RESULTS We screened 13,905 records, of which 209 were finally included. Three safety trials and 13 post hoc analyses, 83 efficacy randomised controlled trials (RCTs) with adequate safety reporting, 56 integrated safety analyses and LTE of RCTs, 20 additional conference abstracts on RCT data, as well as 37 real-world cohort studies were presented to the task force. Safety profiles of JAKi were overall consistent across compounds and indications, but impacts of patient profiles, treatment dosing, and other cofactors like background medications on drug safety could be observed. Furthermore, differential effects of variously selective JAKi on distinct adverse events of special interest (AESI) and laboratory outcomes were discerned. CONCLUSION A substantial amount of literature was published on JAKi safety since 2019. A comprehensive overview of these data supports the optimal use of JAKi in patients with IMIDs, by consideration and balance of their benefits as well as risks in every patient.
Collapse
Affiliation(s)
- Victoria Konzett
- Department of Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria.
| | - Josef S Smolen
- Department of Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Peter Nash
- Griffith University School of Medicine, Gold Coast, QLD, Australia
| | | | - Daniel Aletaha
- Department of Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - Thomas Dörner
- Rheumatology, Charite Medical Faculty Berlin, Berlin, Germany
| | - Roy Fleischmann
- Metroplex Clinical Research Center, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Jette Primdahl
- Danish Hospital for Rheumatic Diseases, University Hospital of Southern Denmark, Sønderborg, Denmark
| | | | - Iain B McInnes
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Michael Trauner
- Department of Medicine III, Division of Gastroenterology and Hepatology, Medical University of Vienna, Vienna, Austria
| | - Naveed Sattar
- Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Maarten de Wit
- Stichting Tools, Patient Research Partner, Amsterdam, the Netherlands
| | - Jan W Schoones
- Directorate of Research Policy, Leiden University Medical Center, Leiden, the Netherlands
| | - Andreas Kerschbaumer
- Department of Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Taylor PC, Balsa A, Mongey AB, Filková M, Chebbah M, Le Clanche S, Verhagen LAW, Witte T, Opris-Belinski D, Marotte H, Avouac J. Does Concomitant Use of Methotrexate with JAK Inhibition Confer Benefit for Cardiovascular Outcomes? A Commentary. Rheumatol Ther 2024; 11:1425-1435. [PMID: 39397239 PMCID: PMC11557820 DOI: 10.1007/s40744-024-00721-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/27/2024] [Indexed: 10/15/2024] Open
Abstract
This commentary explores the potential cardiovascular (CV) benefits of combining methotrexate (MTX) and Janus kinase inhibitors (JAKis) in the treatment of rheumatoid arthritis (RA). While European guidelines recommend MTX as first-line treatment, concerns about the CV risks associated with JAKis have emerged. This article reviews the existing literature to assess the role of concomitant MTX in reducing CV risk when used with JAKis. Clinical trials confirm the efficacy of JAKis in combination with MTX in terms of treatment outcomes in RA. However, the number of major adverse cardiovascular events (MACEs) reported is too low to draw conclusions on adverse CV outcomes. Indirect evidence does, however, suggest potential protective effects of MTX on CV outcomes, as several mechanisms may contribute to MTX's cardioprotective effects, including reduced inflammation, adenosine monophosphate-activated protein kinase (AMPK) activation, increased cholesterol efflux, and adenosine accumulation. These mechanisms and the available data may support the case for CV benefits of concomitant MTX when JAKis are used in the treatment of patients with RA, although further research is needed. In particular, the lipid paradox associated with RA highlights the complex relationship between RA treatments (MTX, JAKis, tumor necrosis factor (TNF) inhibitors, and interleukin (IL)-6 receptor inhibitors), inflammation, different lipid profiles, and CV risk. In the absence of contraindications and when MTX is tolerated, this commentary suggests the concomitant use of MTX and JAKis as a preferred option for optimizing CV protection in patients with RA.
Collapse
Affiliation(s)
- Peter C Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Windmill Road, Oxford, OX3 7LD, UK.
| | - Alejandro Balsa
- Rheumatology Unit, University Hospital La Paz, Institute for Health Research-IdiPAZ, Universidad Autonoma de Madrid, 28046, Madrid, Spain
| | - Anne-Barbara Mongey
- Department of Rheumatology, School of Medicine and Medical Sciences, St. Vincent's University Hospital, University College Dublin, Dublin 4, Ireland
| | - Mária Filková
- Institute of Rheumatology, Department of Rheumatology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Myriam Chebbah
- Département Affaires Médicales, Public Health Expertise, 10 Boulevard de Sébastopol, 75004, Paris, France
| | - Solenn Le Clanche
- Département Affaires Médicales, Public Health Expertise, 10 Boulevard de Sébastopol, 75004, Paris, France
| | | | - Torsten Witte
- Klinik Für Rheumatologie und Immunologie, Medizinische Hochschule Hannover Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Daniela Opris-Belinski
- Internal Medicine and Rheumatology, Sfanta Maria Hospital, Carol Davila University of Medicine, Bucharest, Romania
| | - Hubert Marotte
- Université Jean Monnet, Service de Rhumatologie, CHU Saint-Etienne, Mines Saint-Etienne, INSERM U1059, Saint-Etienne, France
| | - Jérôme Avouac
- Service de Rhumatologie, Hôpital Cochin, AP-HP Centre-Université Paris Cité, 75014, Paris, France
| |
Collapse
|
3
|
Kirchhof MG, Prajapati VH, Gooderham M, Hong CH, Lynde CW, Maari C, Turchin I, Papp KA. Practical Recommendations on Laboratory Monitoring in Patients with Atopic Dermatitis on Oral JAK Inhibitors. Dermatol Ther (Heidelb) 2024; 14:2653-2668. [PMID: 39115712 PMCID: PMC11393246 DOI: 10.1007/s13555-024-01243-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 07/19/2024] [Indexed: 09/13/2024] Open
Abstract
Oral Janus kinase inhibitors (JAKi), a class of advanced targeted systemic therapy, have demonstrated efficacy and safety in the treatment of moderate-to-severe atopic dermatitis (AD). Like other small molecules, oral JAKi have the potential for off-target effects including laboratory-related adverse events (AEs). Product labels for oral JAKi recommend an initial laboratory assessment and follow-up 4-12 weeks later to monitor for potential changes, based on evidence from clinical trials across therapeutic indications for oral JAKi, which may not reflect a population of moderate-to-severe AD patients typically seen in routine clinical practice. To address this gap, a panel of eight dermatologists with clinical and research experience with oral JAKi for the management of AD conducted a targeted review of the literature focused on key laboratory-related AEs associated with oral JAKi in the moderate-to-severe AD population. Based on the synthesis of evidence and informed opinion, a set of best practice statements related to fundamental standards of care and consensus recommendations on laboratory monitoring were suggested, and level of agreement was ascertained using a Likert scale from 0 to 100. There was a high level of agreement on three of the four suggested recommendations related to assessment and monitoring of key laboratory parameters and to dose reduction or switching in response to laboratory changes; there was a lower level of agreement related to the frequency of ongoing laboratory monitoring. Appropriate patient selection and laboratory assessment is an important strategy to mitigate the potential risks associated with oral JAKi when treating AD.
Collapse
Affiliation(s)
- Mark G Kirchhof
- Dermatology Division, Department of Medicine, University of Ottawa and The Ottawa Hospital, Ottawa, ON, Canada
- Probity Medical Research Inc., 135 Union St. E., Waterloo, ON, N2J 1C3, Canada
| | - Vimal H Prajapati
- Probity Medical Research Inc., 135 Union St. E., Waterloo, ON, N2J 1C3, Canada
- Division of Dermatology, Department of Medicine, Section of Community Pediatrics, Department of Pediatrics; and Section of Pediatric Rheumatology, Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Skin Health and Wellness Centre, Calgary, AB, Canada
- Dermatology Research Institute, Calgary, AB, Canada
| | - Melinda Gooderham
- Probity Medical Research Inc., 135 Union St. E., Waterloo, ON, N2J 1C3, Canada
- Department of Dermatology, Queen's University, Kingston, ON, Canada
- SKiN Centre for Dermatology, Peterborough, ON, Canada
| | - Chih-Ho Hong
- Probity Medical Research Inc., 135 Union St. E., Waterloo, ON, N2J 1C3, Canada
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC, Canada
- Dr. Chih-Ho Hong Medical Inc., Surrey, BC, Canada
| | - Charles W Lynde
- Probity Medical Research Inc., 135 Union St. E., Waterloo, ON, N2J 1C3, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Lynde Dermatology, Toronto, ON, Canada
| | - Catherine Maari
- Division of Dermatology, Department of Medicine, Montreal University Hospital Center, Montreal, QC, Canada
- Innovaderm Research Inc., Montreal, QC, Canada
| | - Irina Turchin
- Probity Medical Research Inc., 135 Union St. E., Waterloo, ON, N2J 1C3, Canada
- Department of Medicine, Dalhousie University, Halifax, NS, Canada
- Brunswick Dermatology Center, Fredericton, NB, Canada
| | - Kim A Papp
- Probity Medical Research Inc., 135 Union St. E., Waterloo, ON, N2J 1C3, Canada.
- K Papp Clinical Research, Waterloo, ON, Canada.
| |
Collapse
|
4
|
Szekanecz Z, Buch MH, Charles-Schoeman C, Galloway J, Karpouzas GA, Kristensen LE, Ytterberg SR, Hamar A, Fleischmann R. Efficacy and safety of JAK inhibitors in rheumatoid arthritis: update for the practising clinician. Nat Rev Rheumatol 2024; 20:101-115. [PMID: 38216757 DOI: 10.1038/s41584-023-01062-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 01/14/2024]
Abstract
Janus kinase (JAK) inhibitors, including tofacitinib, baricitinib, upadacitinib and filgotinib, are increasingly used in the treatment of rheumatoid arthritis (RA). There has been debate about their safety, particularly following the issuance of guidance by regulatory agencies advising caution in their use in certain patients. The registrational clinical trials and registry data of JAK inhibitors did not identify a difference in the risk of major adverse cardiovascular events (MACEs), venous thromboembolism, malignancies or infections (other than herpes zoster) with a JAK inhibitor versus a biologic DMARD. In the ORAL Surveillance trial, which enrolled patients >50 years of age with ≥1 cardiovascular risk factor, tofacitinib was statistically inferior to TNF inhibitors for the occurrence of MACEs and malignancy. Further post hoc analysis of the data revealed that an age of ≥65 years, a high baseline cardiovascular risk, a history of smoking, sustained inflammation, disease activity and suboptimal treatment of cardiovascular comorbidities all increase the risk of these outcomes. The guidance issued by regulatory agencies should be carefully considered to ensure appropriate and safe treatment of patients with RA without undertreatment of patients who might benefit from JAK inhibitor, as well as biologic, treatment. As always, the risks associated with the use of these agents, treatment goals, costs and patient preferences should be discussed with the patient.
Collapse
Affiliation(s)
- Zoltán Szekanecz
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Maya H Buch
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- NIHR Manchester Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Christina Charles-Schoeman
- Division of Rheumatology, Department of Medicine, Harbour-University of California Los Angeles Medical Centre, Los Angeles, CA, USA
| | - James Galloway
- Department of Inflammation Biology and Centre for Rheumatic Diseases, King's College London, London, UK
| | - George A Karpouzas
- Division of Rheumatology, Department of Medicine, Harbour-University of California Los Angeles Medical Centre, Los Angeles, CA, USA
| | - Lars Erik Kristensen
- The Parker Institute, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| | | | - Attila Hamar
- Department of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Roy Fleischmann
- Metroplex Clinical Research Center and University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
5
|
邹 雪, 白 小, 张 丽. [Effectiveness of tofacitinib combined with iguratimod in the treatment of difficult-to-treat moderate-to-severe rheumatoid arthritis]. BEIJING DA XUE XUE BAO. YI XUE BAN = JOURNAL OF PEKING UNIVERSITY. HEALTH SCIENCES 2023; 55:1013-1021. [PMID: 38101782 PMCID: PMC10723989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Indexed: 12/17/2023]
Abstract
OBJECTIVE To investigate the efficacy and safety of iguratimod combined with tofacitinib in patients with difficult-to-treat moderate-to-severe rheumatoid arthritis (RA). METHODS In this prospective clinical study, 30 patients with difficult-to-treat moderate-to-severe RA who attended the Department of Rheumatology and Immunology of Shanxi Province Fenyang Hospital from September 2021 to June 2022 were selected. Twenty-three patients enrollment had been treated with 2 or more conventional synthetic disease modifying anti-rheumatic drugs (DMARDs) for more than 6 months. At least, methotrexate or leflunomide was included. Seven patients were treated with conventional synthetic DMARDs combined with tumor necrosis factor antagonists. Because all the patients had not reached the target of treatment, the combination treatment regimen of DMARDs was changed to iguratimod and tofacitinib. The observation period was 12 weeks. Clinical data were collected before and after treatment. At the end of 4 weeks, 8 weeks and 12 weeks, the clinical data were collected such as swollen joints count (SJC), tender joints count (TJC), time of morning stiffness, clinical disease activity index (CDAI), health status assessment questionnaire (HAQ), and 28-joint disease activity score (DAS28) were included. We collected laboratory indicators, recorded the patient's medication, and observed some changes to see if any adverse drug reactions occurred during the treatment. RESULTS There were significant differences in erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), rheumatoid factor (RF), platelet (PLT), SJC, TJC, DAS28 based on ESR(DAS28-ESR), time of morning stiffness, HAQ, CDAI, and anti-cyclic citrullinated peptide antibody before and after treatment. The differences had statistical significance (P < 0.05). There was no statistical differences in globulin before and after treatment (P>0.05). During the treatment of iguratimod combined with tofacitinib, there was no serious adverse reactions such as leukopenia, significant elevation of liver enzymes, allergy or thromboemblolic events that occurred in all the patients. CONCLUSION Iguratimod combined with tofacitinib in the treatment of difficult-to-treat moderate-to-severe RA may have efficacy. The machanism was improving the patients' recent clinical symptoms by reducing inflammatory indexes. This combination treatment regimen with iguratimod and tofacitinib has a good safety profile.
Collapse
Affiliation(s)
- 雪 邹
- 山西医科大学附属汾阳医院, 山西省汾阳医院风湿免疫科, 山西汾阳 032200Department of Rheumatology and Immunology, Fenyang Hospital Affiliated to Shanxi Medical University, Shanxi Province Fenyang Hospital, Fenyang 032200, Shanxi, China
- 苏州永鼎医院消化内科, 江苏苏州 215100Department of Gastroenterology, Suzhou Yongding Hospital, Suzhou 215100, Jiangsu, China
| | - 小娟 白
- 山西医科大学附属汾阳医院, 山西省汾阳医院风湿免疫科, 山西汾阳 032200Department of Rheumatology and Immunology, Fenyang Hospital Affiliated to Shanxi Medical University, Shanxi Province Fenyang Hospital, Fenyang 032200, Shanxi, China
| | - 丽卿 张
- 山西医科大学附属汾阳医院, 山西省汾阳医院风湿免疫科, 山西汾阳 032200Department of Rheumatology and Immunology, Fenyang Hospital Affiliated to Shanxi Medical University, Shanxi Province Fenyang Hospital, Fenyang 032200, Shanxi, China
| |
Collapse
|
6
|
Yan J, Yang S, Han L, Ba X, Shen P, Lin W, Li T, Zhang R, Huang Y, Huang Y, Qin K, Wang Y, Tu S, Chen Z. Dyslipidemia in rheumatoid arthritis: the possible mechanisms. Front Immunol 2023; 14:1254753. [PMID: 37954591 PMCID: PMC10634280 DOI: 10.3389/fimmu.2023.1254753] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease, of which the leading cause of death is cardiovascular disease (CVD). The levels of total cholesterol (TC), low-density lipoprotein cholesterol (LDL-c), and high-density lipoprotein cholesterol (HDL-c) in RA decrease especially under hyperinflammatory conditions. It is conflictive with the increased risk of CVD in RA, which is called "lipid paradox". The systemic inflammation may explain this apparent contradiction. The increased systemic proinflammatory cytokines in RA mainly include interleukin-6(IL-6)、interleukin-1(IL-1)and tumor necrosis factor alpha(TNF-α). The inflammation of RA cause changes in the subcomponents and structure of HDL particles, leading to a weakened anti-atherosclerosis function and promoting LDL oxidation and plaque formation. Dysfunctional HDL can further worsen the abnormalities of LDL metabolism, increasing the risk of cardiovascular disease. However, the specific mechanisms underlying lipid changes in RA and increased CVD risk remain unclear. Therefore, this article comprehensively integrates the latest existing literature to describe the unique lipid profile of RA, explore the mechanisms of lipid changes, and investigate the impact of lipid changes on cardiovascular disease.
Collapse
Affiliation(s)
- Jiahui Yan
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Sisi Yang
- Department of Geriatrics, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Liang Han
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xin Ba
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Pan Shen
- Department of Rheumatology and Immunology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weiji Lin
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Li
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Ruiyuan Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Ying Huang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yao Huang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Kai Qin
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yu Wang
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Shenghao Tu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Zhe Chen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Ertus C, Scailteux LM, Lescoat A, Berthe P, Auffret V, Dupuy A, Oger E, Droitcourt C. Major adverse cardiovascular events in patients with atopic dermatitis treated with oral Janus kinase inhibitors: a systematic review and meta-analysis. Br J Dermatol 2023; 189:368-380. [PMID: 37410552 DOI: 10.1093/bjd/ljad229] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND On the basis of safety data for patients with inflammatory rheumatism or inflammatory bowel disease, treatment with Janus kinase (JAK) inhibitors (JAKi) has been linked to the occurrence of major adverse cardiovascular events (MACE). However, these inflammatory diseases are proatherogenic; in contrast, patients with atopic dermatitis (AD) do not usually have a high cardiovascular (CV) comorbidity burden. OBJECTIVES To perform a systematic review and meta-analysis of MACE in patients with AD treated with JAKi. METHODS We systematically searched PubMed, Embase, Cochrane Library and Google Scholar from their inception to 2 September 2022. Cohort studies, randomized controlled trials and pooled safety analyses providing CV safety data on patients taking JAKi for AD were selected. We included patients aged ≥ 12 years. We built a 'controlled-period' cohort (n = 9309; 6000 exposed to JAKi and 3309 exposed to comparators) and an 'all-JAKi' cohort (n = 9118 patients exposed to a JAKi in any of the included studies). The primary outcome was a composite of acute coronary syndrome (ACS), ischaemic stroke and CV death. The broader secondary MACE outcome encompassed ACS, stroke (whether ischaemic or haemorrhagic), transient ischaemic attack and CV death. The frequency of primary and secondary MACE was assessed in both cohorts. A fixed-effects meta-analysis using the Peto method was used to calculate the odds ratio (OR) for MACE in the 'controlled-period' cohort. Evaluation of the risk of bias was done using the Cochrane risk-of-bias tool (version 2). Certainty of evidence was assessed using the Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach. RESULTS Eight per cent of the records identified initially met the selection criteria, corresponding to 23 records included in the 'all-JAKi' cohort. Patients had been exposed to baricitinib, upadacitinib, abrocitinib, ivarmacitinib, placebo or dupilumab. Four primary events (three with JAKi and one with placebo) and five secondary events (four with JAKi and one with placebo) occurred among 9309 patients in the 'controlled-period' cohort (MACE frequency 0.04% and 0.05%, respectively). Eight primary events and 13 secondary events occurred among 9118 patients in the 'all-JAKi' cohort (MACE frequency 0.08% and 0.14%, respectively). The OR for primary MACE in patients with AD treated with JAKi vs. placebo or dupilumab was 1.35 (95% confidence interval 0.15-12.21; I2 = 12%, very low certainty of evidence). CONCLUSIONS Our review highlights rare cases of MACE among JAKi users for AD. JAKi may have little-to-no effect on the occurrence of MACE in patients with AD vs. comparators, but the evidence is uncertain. Real-life long-term population-level safety studies are needed.
Collapse
Affiliation(s)
| | - Lucie-Marie Scailteux
- University of Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
- Pharmacovigilance, Pharmacoepidemiology and Drug Information Center, Department of Clinical Pharmacology, Rennes University Hospital, Rennes, France
| | - Alain Lescoat
- University of Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
- Department of Internal Medicine and Clinical Immunology, Rennes University Hospital, Rennes, France
| | | | - Vincent Auffret
- University of Rennes, CHU Rennes Service de Cardiologie, Inserm LTSI U1099, Rennes, France
| | - Alain Dupuy
- University of Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
- Pharmacovigilance, Pharmacoepidemiology and Drug Information Center, Department of Clinical Pharmacology, Rennes University Hospital, Rennes, France
| | - Emmanuel Oger
- University of Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
- Pharmacovigilance, Pharmacoepidemiology and Drug Information Center, Department of Clinical Pharmacology, Rennes University Hospital, Rennes, France
| | - Catherine Droitcourt
- Department of Dermatology
- University of Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| |
Collapse
|
8
|
Bostanghadiri N, Ziaeefar P, Mofrad MG, Yousefzadeh P, Hashemi A, Darban-Sarokhalil D. COVID-19: An Overview of SARS-CoV-2 Variants-The Current Vaccines and Drug Development. BIOMED RESEARCH INTERNATIONAL 2023; 2023:1879554. [PMID: 37674935 PMCID: PMC10480030 DOI: 10.1155/2023/1879554] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/07/2023] [Accepted: 08/04/2023] [Indexed: 09/08/2023]
Abstract
The world is presently in crisis facing an outbreak of a health-threatening microorganism known as COVID-19, responsible for causing uncommon viral pneumonia in humans. The virus was first reported in Wuhan, China, in early December 2019, and it quickly became a global concern due to the pandemic. Challenges in this regard have been compounded by the emergence of several variants such as B.1.1.7, B.1.351, P1, and B.1.617, which show an increase in transmission power and resistance to therapies and vaccines. Ongoing researches are focused on developing and manufacturing standard treatment strategies and effective vaccines to control the pandemic. Despite developing several vaccines such as Pfizer/BioNTech and Moderna approved by the U.S. Food and Drug Administration (FDA) and other vaccines in phase 4 clinical trials, preventive measures are mandatory to control the COVID-19 pandemic. In this review, based on the latest findings, we will discuss different types of drugs as therapeutic options and confirmed or developing vaccine candidates against SARS-CoV-2. We also discuss in detail the challenges posed by the variants and their effect on therapeutic and preventive interventions.
Collapse
Affiliation(s)
- Narjess Bostanghadiri
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Pardis Ziaeefar
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morvarid Golrokh Mofrad
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Parsa Yousefzadeh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Hashemi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Darban-Sarokhalil
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Chang CK, Chiang EPI, Chang KH, Tang KT, Chen PK, Yip HT, Chen CH, Chen DY. The Sizes and Composition of HDL-Cholesterol Are Significantly Associated with Inflammation in Rheumatoid Arthritis Patients. Int J Mol Sci 2023; 24:10645. [PMID: 37445823 DOI: 10.3390/ijms241310645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Rheumatoid arthritis (RA), a chronic inflammatory disease, carries a significant burden of atherosclerotic cardiovascular diseases (ASCVD). With their heterogeneous composition, high-density lipoprotein (HDL) particles have varied athero-protective properties, and some may even increase ASCVD risk. In this prospective and cross-sectional study, we aimed to examine the relationship between HDL sizes/metabolites and inflammation in RA. Using 1H-NMR-based lipid/metabolomics, differential HDL-related metabolites were identified between RA patients and healthy control (HC) subjects and between RA patients with and without anti-citrullinated peptide antibodies (ACPA). The correlation between the discriminative HDL-related metabolites and C-reactive protein (CRP) was evaluated in RA patients. RA patients demonstrated higher particle number, lipids, cholesterol, cholesterol ester, free cholesterol, and phospholipids in large/very large-sized HDLs. ACPA-positive patients had higher L-HDL-C and L-HDL-CE but lower small-/medium-sized HDL-TG levels than ACPA-negative patients. An inverse correlation was found between CRP levels and small-sized HDLs. Janus kinase inhibitor treatment was associated with increased serum small-sized HDL-related metabolites and decreased CRP levels. We are the first to reveal the significant associations between RA inflammation and HDL sizes/metabolites. A potential link between ACPA positivity and changes in serum levels of HDL-related metabolites was also observed in RA patients.
Collapse
Affiliation(s)
- Ching-Kun Chang
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung 404, Taiwan
- Translational Medicine Laboratory, Rheumatology Research Center, China Medical University Hospital, Taichung 404, Taiwan
- College of Medicine, China Medical University, Taichung 404, Taiwan
| | - En-Pei Isabel Chiang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 402, Taiwan
- Innovation and Development Center of Sustainable Agriculture, National Chung Hsing University, Taichung 402, Taiwan
| | - Kuang-Hsi Chang
- Department of Medical Research, Tungs' Taichung Metroharbor Hospital, Taichung 435, Taiwan
- Center for General Education, China Medical University, Taichung 404, Taiwan
- General Education Center, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 356, Taiwan
| | - Kuo-Tung Tang
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung 407, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Po-Ku Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung 404, Taiwan
- Translational Medicine Laboratory, Rheumatology Research Center, China Medical University Hospital, Taichung 404, Taiwan
- College of Medicine, China Medical University, Taichung 404, Taiwan
| | - Hei-Tung Yip
- College of Medicine, China Medical University, Taichung 404, Taiwan
- Management Office for Health Data, China Medical University Hospital, Taichung 404, Taiwan
| | - Chu-Huang Chen
- Vascular and Medicinal Research, Texas Heart Institute, Houston, TX 77030, USA
- Institute for Biomedical Sciences, Shinshu University, Nagano 390-8621, Japan
| | - Der-Yuan Chen
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung 404, Taiwan
- Translational Medicine Laboratory, Rheumatology Research Center, China Medical University Hospital, Taichung 404, Taiwan
- College of Medicine, China Medical University, Taichung 404, Taiwan
- College of Medicine, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
10
|
Xiong P, Zhang F, Liu F, Zhao J, Huang X, Luo D, Guo J. Metaflammation in glucolipid metabolic disorders: Pathogenesis and treatment. Biomed Pharmacother 2023; 161:114545. [PMID: 36948135 DOI: 10.1016/j.biopha.2023.114545] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 03/22/2023] Open
Abstract
The public health issue of glucolipid metabolic disorders (GLMD) has grown significantly, posing a grave threat to human wellness. Its prevalence is rising yearly and tends to affect younger people. Metaflammation is an important mechanism regulating body metabolism. Through a complicated multi-organ crosstalk network involving numerous signaling pathways such as NLRP3/caspase-1/IL-1, NF-B, p38 MAPK, IL-6/STAT3, and PI3K/AKT, it influences systemic metabolic regulation. Numerous inflammatory mediators are essential for preserving metabolic balance, but more research is needed to determine how they contribute to the co-morbidities of numerous metabolic diseases. Whether controlling the inflammatory response can influence the progression of GLMD determines the therapeutic strategy for such diseases. This review thoroughly examines the role of metaflammation in GLMD and combs the research progress of related therapeutic approaches, including inflammatory factor-targeting drugs, traditional Chinese medicine (TCM), and exercise therapy. Multiple metabolic diseases, including diabetes, non-alcoholic fatty liver disease (NAFLD), cardiovascular disease, and others, respond therapeutically to anti-inflammatory therapy on the whole. Moreover, we emphasize the value and open question of anti-inflammatory-based means for treating GLMD.
Collapse
Affiliation(s)
- Pingjie Xiong
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China.
| | - Fan Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China.
| | - Fang Liu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China.
| | - Jiayu Zhao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China.
| | - Xiaoqiang Huang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China.
| | - Duosheng Luo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China.
| | - Jiao Guo
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University; Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou 510006, China.
| |
Collapse
|
11
|
Qin X, Wang Y, Pedersen NL, Tang B, Hägg S. Dynamic patterns of blood lipids and DNA methylation in response to statin therapy. Clin Epigenetics 2022; 14:153. [PMID: 36443870 PMCID: PMC9706978 DOI: 10.1186/s13148-022-01375-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 11/15/2022] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Statins are lipid-lowering drugs and starting treatment has been associated with DNA methylation changes at genes related to lipid metabolism. However, the longitudinal pattern of how statins affect DNA methylation in relation to lipid levels has not been well investigated. METHODS We conducted an epigenetic association study in a longitudinal Swedish twin sample in previously reported lipid-related CpGs (cg10177197, cg17901584 and cg27243685). First, we applied a mixed-effect model to assess the association between blood lipids (total cholesterol (TC), low-density lipoprotein cholesterol (LDL), high-density lipoprotein cholesterol (HDL), total triglyceride (TG)) and DNA methylation. Then, we performed a piecewise latent linear-linear growth curve model (LGCM) to explore the long-term changing pattern of lipids and methylation in response to statin treatment. Finally, we used a bivariate autoregressive latent trajectory model with structured residuals (ALT-SR) to analyze the cross-lagged effects in different lipid-CpG pairs in statin users and non-users. RESULTS We replicated the associations between TC, LDL, HDL and DNA methylation level in cg17901584 and cg27243685 (P values ranged from 4.70E-12 to 1.84E-04). From the piecewise LGCM, we showed that TC and LDL significantly decreased in statin users before treatment started and then remained stable. For non-statin users, we only found a slightly significant decreasing trend for TC and TG. We observed a similar dynamic pattern for methylation levels at cg27243685 and cg17901584. Before statin initiation, cg27243685 showed a significantly increasing trend and cg17901584 a decreasing trend, but post-treatment, there were no additional changes. From the ALT-SR model, we found TG levels to be significantly associated with the DNA methylation level of cg27243685 at the next measurement in statin users (estimate = 0.383, 95% CI: 0.173, 0.594, P value < 0.001). CONCLUSIONS Longitudinal blood lipid and DNA methylation levels change after statin treatment initiation, where the latter is mostly a response to alterations in lipid levels and not vice versa.
Collapse
Affiliation(s)
- Xueying Qin
- grid.11135.370000 0001 2256 9319Department of Epidemiology and Biostatistics, School of Public Health, Peking University, 38# Xueyuan Road, Beijing, 100191 China ,grid.4714.60000 0004 1937 0626Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Nobels Väg 12A, 17177 Stockholm, Sweden
| | - Yunzhang Wang
- grid.4714.60000 0004 1937 0626Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Nobels Väg 12A, 17177 Stockholm, Sweden
| | - Nancy L. Pedersen
- grid.4714.60000 0004 1937 0626Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Nobels Väg 12A, 17177 Stockholm, Sweden
| | - Bowen Tang
- grid.4714.60000 0004 1937 0626Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Nobels Väg 12A, 17177 Stockholm, Sweden
| | - Sara Hägg
- grid.4714.60000 0004 1937 0626Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Nobels Väg 12A, 17177 Stockholm, Sweden
| |
Collapse
|
12
|
GlycA, hsCRP differentially associated with MI, ischemic stroke: In the Dallas Heart Study and Multi-Ethnic Study of Atherosclerosis: GlycA, hsCRP Differentially Associated MI, Stroke. Am J Prev Cardiol 2022; 12:100373. [PMID: 36061365 PMCID: PMC9428838 DOI: 10.1016/j.ajpc.2022.100373] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/07/2022] [Accepted: 08/17/2022] [Indexed: 11/23/2022] Open
Abstract
Objective Inflammatory markers are associated with cardiovascular disease (CVD); however, the ability to specifically predict myocardial infarction (MI) as well as ischemic stroke remains unknown. There has not been a direct comparison of the associations between GlycA and hsCRP and MI and ischemic stroke in a multi-ethnic pooled cohort. Methods Multi-center, multi-ethnic, population-based community prospective pooled cohort of the Dallas Heart Study (DHS) and Multi-Ethnic Study of Atherosclerosis (MESA). 9,785 participants without baseline CVD enrolled with median follow-up of 13.4 years. Fatal/nonfatal MI and fatal/nonfatal ischemic stroke were assessed separately and then combined. Results GlycA was moderately associated with hsCRP (R=0.58 in DHS and R=0.55 in MESA). In adjusted Cox proportional hazards models with competing risk adjusted for both inflammatory markers, GlycA was directly associated with MI (HR Q4 vs. Q1 1.90, 95% CI 1.39 to 2.58), whereas hsCRP was not (HR Q4 vs. Q1 0.92, 95% CI 0.70 to 1.21). Conversely, hsCRP was directly associated with ischemic stroke (HR Q4 vs. Q1 1.73, 95% CI 1.15 to 2.59), but GlycA was not (HR Q4 vs. Q1 1.21, 95% CI 0.77 to 1.90). GlycA improved net reclassification for MI and hsCRP did so for ischemic stroke. Conclusions Although both GlycA and hsCRP were associated with incident CVD, GlycA more strongly predicted incident MI, and hsCRP more strongly predicted ischemic stroke.
Collapse
|
13
|
Ferguson LD, Sattar N, McInnes IB. Managing Cardiovascular Risk in Patients with Rheumatic Disease. Rheum Dis Clin North Am 2022; 48:429-444. [PMID: 35400369 DOI: 10.1016/j.rdc.2022.02.003] [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: 10/18/2022]
Abstract
Individuals with rheumatoid arthritis, systemic lupus erythematosus, or gout have increased risk of cardiovascular disease (CVD) compared with the general population. This risk relates to a combination of traditional cardiovascular risk factors and disease-specific factors. Screening for CVD is important because CVD contributes to significant morbidity and mortality. Management includes tight control of disease activity to reduce inflammation, but with care to minimize use of nonsteroidal anti-inflammatory drugs and prolonged courses of high-dose corticosteroids. Traditional cardiovascular risk factors should be managed with a combination of lifestyle interventions and pharmacotherapy. The decision to start antihypertensive and lipid-lowering therapy should be based on individual CVD risk.
Collapse
Affiliation(s)
- Lyn D Ferguson
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK.
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
14
|
Jin X, Yang S, Lu J, Wu M. Small, Dense Low-Density Lipoprotein-Cholesterol and Atherosclerosis: Relationship and Therapeutic Strategies. Front Cardiovasc Med 2022; 8:804214. [PMID: 35224026 PMCID: PMC8866335 DOI: 10.3389/fcvm.2021.804214] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/02/2021] [Indexed: 12/14/2022] Open
Abstract
Low-density lipoprotein cholesterol (LDL-C) plays an important role in the formation, incidence, and development of atherosclerosis (AS). Low-density lipoproteins can be divided into two categories: large and light LDL-C and small, dense low-density lipoprotein cholesterol (sdLDL-C). In recent years, an increasing number of studies have shown that sdLDL-C has a strong ability to cause AS because of its unique characteristics, such as having small-sized particles and low density. Therefore, this has become the focus of further research. However, the specific mechanisms regarding the involvement of sdLDL-C in AS have not been fully explained. This paper reviews the possible mechanisms of sdLDL-C in AS by reviewing relevant literature in recent years. It was found that sdLDL-C can increase the atherogenic effect by regulating the activity of gene networks, monocytes, and enzymes. This article also reviews the research progress on the effects of sdLDL-C on endothelial function, lipid metabolism, and inflammation; it also discusses its intervention effect. Diet, exercise, and other non-drug interventions can improve sdLDL-C levels. Further, drug interventions such as statins, fibrates, ezetimibe, and niacin have also been found to improve sdLDL-C levels.
Collapse
Affiliation(s)
- Xiao Jin
- General Department of Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- General Department of Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Lu
- Beijing University of Chinese Medicine, Beijing, China
| | - Min Wu
- General Department of Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
15
|
A Review of Safety Outcomes from Clinical Trials of Baricitinib in Rheumatology, Dermatology and COVID-19. Adv Ther 2022; 39:4910-4960. [PMID: 36063279 PMCID: PMC9443639 DOI: 10.1007/s12325-022-02281-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/21/2022] [Indexed: 01/30/2023]
Abstract
Baricitinib is an oral, selective inhibitor of Janus kinase (JAK)1/JAK2 that transiently and reversibly inhibits many proinflammatory cytokines. This mechanism is a key mediator in a number of chronic inflammatory diseases; accordingly, baricitinib has been studied and approved for the treatment of several rheumatological and dermatological disorders, as well as COVID-19. This narrative review summarises and discusses the safety profile of baricitinib across these diseases, with special focus on adverse events of special interest (AESI) for JAK inhibitors, using integrated safety data sets of clinical trial data, and puts findings into context with the underlying risk in the respective disease populations, using supporting literature. We show that rates of infection with baricitinib generally reflected the inherent risk of the disease populations being treated, with serious infections and herpes zoster being more frequent in rheumatic diseases than in dermatological disorders, and herpes simplex being reported particularly in atopic dermatitis. Similarly, rates of major adverse cardiovascular events (MACE), venous thromboembolism (VTE) and malignancies were generally within or below the ranges reported for the respective disease populations, thereby reflecting the underlying risk; these events were therefore more frequent in patients with rheumatic diseases than in those with dermatological disorders, the latter of whom generally had low absolute risk. AESI were usually more common in patients with risk factors specific for each event. When a population similar to that of ORAL Surveillance was considered, the incidence rate of MACE with baricitinib was numerically lower than that reported with tofacitinib and similar to that of tumour necrosis factor inhibitors. No safety concerns were observed in hospitalised patients with COVID-19 who received baricitinib for up to 14 days. Identifying the patterns and likelihoods of AEs that occur during treatment in large groups of patients with different diseases can help the physician and patient better contextualise the benefit-to-risk ratio for the individual patient.
Collapse
|
16
|
Glycosylation and Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1325:307-319. [PMID: 34495542 DOI: 10.1007/978-3-030-70115-4_15] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, accounting for approximately 18 million deaths in 2017. Coronary artery disease is the predominant cause of death from CVD, followed by stroke. Owing to recent technological advancements, glycans and glycosylation patterns of proteins have been investigated in association with CVD risk factors and clinical events. These studies have found significant associations of glycans as biomarkers of systemic inflammation and major CVD risk factors and events. While more limited, studies have also shown that glycans may be useful for monitoring response to anti-inflammatory therapies and may be responsive to changes in lifestyle, particularly in patients with chronic inflammatory diseases. Glycans capture summative risk information related to inflammatory, immune, and signaling pathways and are promising biomarkers for CVD risk prediction and therapeutic monitoring.
Collapse
|
17
|
Le M, Berman-Rosa M, Ghazawi FM, Bourcier M, Fiorillo L, Gooderham M, Guenther L, Hanna S, Hong HCH, Landells I, Lansang P, Marcoux D, Wiseman MC, Yeung J, Lynde C, Litvinov IV. Systematic Review on the Efficacy and Safety of Oral Janus Kinase Inhibitors for the Treatment of Atopic Dermatitis. Front Med (Lausanne) 2021; 8:682547. [PMID: 34540860 PMCID: PMC8440866 DOI: 10.3389/fmed.2021.682547] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Atopic dermatitis is a chronic, relapsing and remitting disease that can be difficult to treat despite a recently approved biologic therapy targeting IL-4/IL-13 receptor. Oral janus kinase inhibitors (JAKi) represent a novel therapeutic class of targeted therapy to treat moderate-to-severe atopic dermatitis (AD). Objective: To review the efficacy, safety, and pharmacokinetic characteristics of oral JAKi in the treatment of AD. Methods: A PRISMA systematic review was conducted using MEDLINE, EMBASE (Ovid), and PubMed databases for studies assessing the efficacy, safety, and/or pharmacokinetic properties of oral forms of JAKi in the treatment of AD in pediatric or adult populations from inception to June 2021. Results: 496 papers were reviewed. Of 28 articles that underwent full text screening, 11 met our inclusion criteria for final qualitative review. Four studies examined abrocitinib; three studies examined baricitinib; three examined upadacitinib and one examined gusacitinib (ASN002). Significant clinical efficacy and a reassuring safety profile was reported for all JAKi agents reviewed. Rapid symptom control was reported for abrocitinib, baricitinib and upadacitinib. Limitations: Given the relatively limited evidence for each JAKi and the differences in patient eligibility criteria between studies, the data was not deemed suitable for a meta-analysis at this time. Conclusion: Given their ability to achieve rapid symptom control with a reassuring safety profile, we recommend considering the use of JAKi as a reliable systemic treatment option for adult patients with moderate-to-severe AD, who are unresponsive to topical or skin directed treatments.
Collapse
Affiliation(s)
- Michelle Le
- Division of Dermatology, McGill University Health Centre, Montreal, QC, Canada
| | - Melissa Berman-Rosa
- Division of Dermatology, McGill University Health Centre, Montreal, QC, Canada
| | - Feras M. Ghazawi
- Division of Dermatology, University of Ottawa, Ottawa, ON, Canada
| | - Marc Bourcier
- Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC, Canada
| | - Loretta Fiorillo
- Division of Pediatric Dermatology, University of Alberta, Edmonton, AB, Canada
| | - Melinda Gooderham
- SKiN Centre for Dermatology, Probity Medical Research, and Queens University, Peterborough, ON, Canada
| | - Lyn Guenther
- Division of Dermatology, University of Western Ontario, London, ON, Canada
| | | | - H. Chih-Ho Hong
- Department of Dermatology and Skin Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Ian Landells
- Division of Dermatology, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Perla Lansang
- Division of Dermatology, University of Toronto, Toronto, ON, Canada
| | - Danielle Marcoux
- Division of Pediatric Dermatology, University of Montreal, Montreal, QC, Canada
| | - Marni C. Wiseman
- Section of Dermatology, Department of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Jensen Yeung
- Division of Dermatology, University of Toronto, Toronto, ON, Canada
- Probity Medical Research, Waterloo, ON, Canada
| | | | - Ivan V. Litvinov
- Division of Dermatology, McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
18
|
Kerola AM, Rollefstad S, Semb AG. Atherosclerotic Cardiovascular Disease in Rheumatoid Arthritis: Impact of Inflammation and Antirheumatic Treatment. Eur Cardiol 2021; 16:e18. [PMID: 34040652 PMCID: PMC8145075 DOI: 10.15420/ecr.2020.44] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/19/2021] [Indexed: 11/16/2022] Open
Abstract
Patients with rheumatoid arthritis (RA) are at approximately 1.5-fold risk of atherosclerotic cardiovascular disease (CVD) compared with the general population, a phenomenon resulting from combined effects of traditional CVD risk factors and systemic inflammation. Rheumatoid synovitis and unstable atherosclerotic plaques share common inflammatory mechanisms, such as expression of proinflammatory cytokines interleukin (IL)-1, tumour necrosis factor (TNF)-α and IL-6. RA patients are undertreated in terms of CVD prevention, and structured CVD prevention programmes are warranted. Alongside management of traditional risk factors, suppressing systemic inflammation with antirheumatic medication is fundamental for the reduction of CVD risk among this high-risk patient group. Many antirheumatic drugs, especially methotrexate, TNF-α-inhibitors and IL-6-inhibitors are associated with reduced risk of CVD in observational studies among RA patients, but randomised controlled trials with hard CVD endpoints are lacking. In patients without rheumatic disease, anti-inflammatory therapies targeting nucleotide-binding oligomerisation domain, leucine-rich repeat and pyrin domain-containing protein 3 inflammasome and the IL-1/IL-6 pathway arise as potential therapies after an atherosclerotic CVD event.
Collapse
Affiliation(s)
- Anne Mirjam Kerola
- Preventive Cardio-Rheuma Clinic, Division of Rheumatology and Research, Diakonhjemmet Hospital Oslo, Norway.,Department of Rheumatology, Päijät-Häme Joint Authority for Health and Wellbeing Lahti, Finland
| | - Silvia Rollefstad
- Preventive Cardio-Rheuma Clinic, Division of Rheumatology and Research, Diakonhjemmet Hospital Oslo, Norway
| | - Anne Grete Semb
- Preventive Cardio-Rheuma Clinic, Division of Rheumatology and Research, Diakonhjemmet Hospital Oslo, Norway
| |
Collapse
|
19
|
Atzeni F, Rodríguez-Carrio J, Popa CD, Nurmohamed MT, Szűcs G, Szekanecz Z. Cardiovascular effects of approved drugs for rheumatoid arthritis. Nat Rev Rheumatol 2021; 17:270-290. [PMID: 33833437 DOI: 10.1038/s41584-021-00593-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2021] [Indexed: 02/07/2023]
Abstract
The risk of cardiovascular disease is increased in patients with rheumatoid arthritis compared with the general population owing to the influence of traditional and non-traditional risk factors. Inflammation has a pivotal contribution and can accelerate the atherosclerotic process. Although dampening inflammation with DMARDs should theoretically abrogate this process, evidence suggests that these drugs can also promote atherosclerosis directly and indirectly, hence adding to an increased cardiovascular burden. However, the extent and direction of the effects largely differ across drugs. Understanding how these drugs influence endothelial damage and vascular repair mechanisms is key to understanding these outcomes. NSAIDs and glucocorticoids can increase the cardiovascular risk. Conversely, conventional, biologic and targeted DMARDs control inflammation and reduce this risk, although some of these drugs can also aggravate traditional factors or thrombotic events. Given these data, the fundamental objective for clinicians should be disease control, in an individualized approach that considers the most appropriate drug for each patient, taking into account joint and cardiovascular outcomes. This Review provides a comprehensive analysis of the effects of DMARDs and other approved drugs on cardiovascular involvement in rheumatoid arthritis, from a clinical and mechanistic perspective, with a roadmap to inform the research agenda.
Collapse
Affiliation(s)
- Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, Messina, Italy.
| | - Javier Rodríguez-Carrio
- Department of Functional Biology, Immunology Area, Faculty of Medicine, University of Oviedo, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Călin D Popa
- Department of Rheumatology, Sint Maartenskliniek Nijmegen, Nijmegen, The Netherlands
| | - Michael T Nurmohamed
- Deptartment of Rheumatology, Amsterdam University Medical Center & Reade, Amsterdam, The Netherlands
| | - Gabriella Szűcs
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Szekanecz
- Division of Rheumatology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
20
|
Ferguson LD, Sattar N, McInnes IB. Managing Cardiovascular Risk in Patients with Rheumatic Disease. Med Clin North Am 2021; 105:247-262. [PMID: 33589100 DOI: 10.1016/j.mcna.2020.09.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Individuals with rheumatoid arthritis, systemic lupus erythematosus, or gout have increased risk of cardiovascular disease (CVD) compared with the general population. This risk relates to a combination of traditional cardiovascular risk factors and disease-specific factors. Screening for CVD is important because CVD contributes to significant morbidity and mortality. Management includes tight control of disease activity to reduce inflammation, but with care to minimize use of nonsteroidal anti-inflammatory drugs and prolonged courses of high-dose corticosteroids. Traditional cardiovascular risk factors should be managed with a combination of lifestyle interventions and pharmacotherapy. The decision to start antihypertensive and lipid-lowering therapy should be based on individual CVD risk.
Collapse
Affiliation(s)
- Lyn D Ferguson
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK.
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
21
|
Pope JE, Choy EH. C-reactive protein and implications in rheumatoid arthritis and associated comorbidities. Semin Arthritis Rheum 2020; 51:219-229. [PMID: 33385862 DOI: 10.1016/j.semarthrit.2020.11.005] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/19/2020] [Accepted: 11/05/2020] [Indexed: 02/07/2023]
Abstract
C-reactive protein (CRP) is routinely assessed as a marker of systemic inflammation in rheumatoid arthritis (RA). However, it is also an immune regulator that plays an important role in inflammatory pathways associated with RA and promotes atherogenic effects. Comorbidities linked to systemic inflammation are common in RA, and CRP has been associated with the risk for cardiovascular disease, diabetes, metabolic syndrome, pulmonary diseases, and depression. The relationship between systemic inflammation, CRP, and comorbidities in RA is complex, and it is challenging to determine how changing CRP levels may affect the risk or progression of these comorbidities. We review the biological role of CRP in RA and its implications for disease activity and treatment response. We also discuss the impact of treatment on CRP levels and whether reducing systemic inflammation and inhibiting CRP-mediated inflammatory pathways may have an impact on conditions commonly comorbid with RA.
Collapse
Affiliation(s)
- Janet E Pope
- Janet E. Pope: Schulich School of Medicine, University of Western Ontario, St. Joseph's Health Care, London, ON, Canada
| | - Ernest H Choy
- Ernest H. Choy: Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom.
| |
Collapse
|
22
|
GlycA measured by NMR spectroscopy is associated with disease activity and cardiovascular disease risk in chronic inflammatory diseases. Am J Prev Cardiol 2020; 4:100120. [PMID: 34327480 PMCID: PMC8315361 DOI: 10.1016/j.ajpc.2020.100120] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 02/08/2023] Open
Abstract
GlycA is a biomarker of systemic inflammation, quantifying both the protein concentrations and glycosylation states of several acute phase proteins. GlycA has been shown to be associated with both subclinical atherosclerosis and with cardiovascular disease (CVD). GlycA levels are higher in acute and chronic inflammation. During ongoing systemic inflammatory processes, GlycA specific acute phase reactants and proteins undergo circulating concentration and glycosylation pattern changes, and these alterations are reflected in the GlycA NMR signal. Additionally, levels associate with ongoing disease severity in individuals with rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and psoriasis thus capturing active inflammation. Furthermore, in these disease states, GlycA is associated with cardiovascular disease (CVD) independent of traditional risk factors including C-reactive protein (CRP). Finally, GlycA levels decrease with exercise, weight loss, and systemic anti-inflammatory agents. Therefore, GlycA appears to be a promising new composite biomarker of active systemic inflammation including assessing CVD risk in patients with inflammatory diseases. Patients with chronic inflammatory disorders are at increased risk for cardiovascular diseasenot captured by traditional risk factors. GlycA is a biomarker of acute phase reactants by NMR spectroscopy which captures disease activity in human inflammatory diseases. GlycA associates with cardiovascular disease and offers a tool to monitor primary disease activity and assess CVD risk in inflammatory diseases.
Collapse
|
23
|
El Jammal T, Sève P, Gerfaud-Valentin M, Jamilloux Y. State of the art: approved and emerging JAK inhibitors for rheumatoid arthritis. Expert Opin Pharmacother 2020; 22:205-218. [PMID: 32967471 DOI: 10.1080/14656566.2020.1822325] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is the most common autoimmune inflammatory arthritis in adults. In the past decade, many treatments have emerged to expand the therapeutic armamentarium of rheumatologists. Among emerging treatments, Janus Kinase inhibitors (JAKi) are promising in treating RA and several other inflammatory conditions, such as psoriatic arthritis (PsA). The JAK/STAT signaling pathway is located downstream certain cytokines receptors that are known to be involved in RA pathogenesis. So far, three JAKi are approved for the treatment of RA, while other JAKi, are under investigation. AREAS COVERED Herein, the authors review those JAKi approved and emerging for the treatment of RA and provide their expert perspectives on the subject area. EXPERT OPINION JAKi represent an interesting alternative to other DMARDs when MTX has failed. Long-term extension studies are still ongoing, but one can assume that most of the major safety concerns have already come out. Switching from one JAKi to another DMARD has been little studied, but in such cases, preferring a treatment which does not interfere with the JAK/STAT pathway seems to be a reasonable choice.
Collapse
Affiliation(s)
- Thomas El Jammal
- Department of Internal Medicine, Lyon University Hospital , Lyon, France
| | - Pascal Sève
- Department of Internal Medicine, Lyon University Hospital , Lyon, France
| | | | - Yvan Jamilloux
- Department of Internal Medicine, Lyon University Hospital , Lyon, France
| |
Collapse
|
24
|
Sands BE, Colombel JF, Ha C, Farnier M, Armuzzi A, Quirk D, Friedman GS, Kwok K, Salese L, Su C, Taub PR. Lipid Profiles in Patients With Ulcerative Colitis Receiving Tofacitinib-Implications for Cardiovascular Risk and Patient Management. Inflamm Bowel Dis 2020; 27:797-808. [PMID: 32870265 PMCID: PMC8128390 DOI: 10.1093/ibd/izaa227] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Patients with ulcerative colitis (UC) are at elevated risk of cardiovascular disease vs the general population, despite a lower prevalence of traditional risk factors, including hyperlipidemia. Mechanistic studies in patients with rheumatoid arthritis and psoriasis suggest that tofacitinib restores serum lipids to preinflammation levels by reversing inflammation-induced cholesterol metabolism changes. We reviewed data on lipid levels and cardiovascular events, alongside recommendations for managing lipid levels during tofacitinib treatment in patients with UC, based on up-to-date expert guidelines. METHODS Data were identified from a phase 3/open-label, long-term extension (OLE) tofacitinib UC clinical program (cutoff May 27, 2019). Literature was identified from PubMed (search terms "lipid," "cholesterol," "lipoprotein," "cardiovascular," "inflammation," "atherosclerosis," "tofacitinib," "rheumatoid arthritis," "psoriasis," "inflammatory bowel disease," "ulcerative colitis," "hyperlipidemia," and "guidelines") and author knowledge. Data were available from 4 phase 3 clinical trials of 1124 patients with moderately to severely active UC who received ≥1 dose of tofacitinib 5 or 10 mg twice daily in induction (two identical trials), maintenance, and OLE studies (treatment duration ≤6.8 years; 2576.4 patient-years of drug exposure). RESULTS In the OLE study, tofacitinib treatment was not associated with major changes from baseline in total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, triglycerides, total cholesterol/high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol/high-density lipoprotein cholesterol, with lipid levels and ratios generally remaining stable over time. The major adverse cardiovascular events incidence rate was 0.26/100 patient-years (95% confidence interval, 0.11-0.54). CONCLUSIONS Lipid levels and ratios remained generally unchanged from baseline in the OLE study after tofacitinib treatment, and major adverse cardiovascular events were infrequent. Long-term studies are ongoing. CLINICALTRIALS.GOV IDENTIFIERS NCT01465763, NCT01458951, NCT01458574, NCT01470612.
Collapse
Affiliation(s)
- Bruce E Sands
- Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York, USA,Address correspondence to: Bruce E. Sands, MD, Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1069, New York, NY 10029 ()
| | - Jean-Frédéric Colombel
- Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christina Ha
- Inflammatory Bowel Disease Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Michel Farnier
- PEC2, EA 7460, University of Bourgogne—Franche Comté and Department of Cardiology, CHU Dijon-Bourgogne, Dijon, France
| | - Alessandro Armuzzi
- IBD Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Daniel Quirk
- Inflammation & Immunology, Pfizer Inc, Collegeville, Pennsylvania, USA
| | - Gary S Friedman
- Inflammation & Immunology, Pfizer Inc, Collegeville, Pennsylvania, USA
| | - Kenneth Kwok
- Inflammation & Immunology, Pfizer Inc, New York, New York, USA
| | - Leonardo Salese
- Inflammation & Immunology, Pfizer Inc, Collegeville, Pennsylvania, USA
| | - Chinyu Su
- Inflammation & Immunology, Pfizer Inc, Collegeville, Pennsylvania, USA
| | - Pam R Taub
- Division of Cardiovascular Medicine, University of California, San Diego, California, USA
| |
Collapse
|
25
|
Efficacy of baricitinib in treating rheumatoid arthritis: Modulatory effects on fibrotic and inflammatory biomarkers in a real-life setting. Int Immunopharmacol 2020; 86:106748. [PMID: 32645631 DOI: 10.1016/j.intimp.2020.106748] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/03/2020] [Accepted: 06/25/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Baricitinib is a JAK inhibitor that blocks intracellular signalling pathways of inflammatory cytokines recommended for Rheumatoid arthritis (RA) patients not responding to initial treatment. Among RA extrareticular features, interstitial lung involvement is primarly characterized by fibrotic evolution. The aim of the present study was to analyse the effects of baricitinib in a population of RA and RA-ILD patients in a real-life setting, describing any changes in lung function parameters, serum inflammatory biomarkers and fibrotic biomarkers after 6 months of treatment. MATERIALS AND METHODS 15 patients (median (IQR) 65 (55-66); 13% males and 74% smokers) treated with baricitinib were enrolled. 4 patients (27%) were classified as RA-ILD before baricitinib therapy. Our study is the first to evaluate adipokine levels in RA patients (including a small population with RA-ILD) after six months of baricitinib treatment with a novel multiplex method. RESULTS The modulatory effects of baricitinib on lipid mediators were associated with clinical and functional improvement, demonstrated by the significant increase in DLco and KCO percentages after six months of treatment. Baricitinib decreased the systemic inflammation by lowering expression of IL-6 and CRP and reducing ESR and serum concentrations of adiponectin. A significant reduction of KL-6 levels in RA-ILD patients after six months of baricitinib therapy reflects the stability of interstitial lung involvement in these patients. CONCLUSION Baricitinib was demonstrated to be a safe immune modulator that reduces the concentrations biomarkers of lung fibrosis and inflammation in RA patients, including a subgroup with interstitial lung involvement.
Collapse
|
26
|
Nasonov EL, Lila AM. BARICITINIB: NEW PHARMACOTHERAPY OPTIONS FOR RHEUMATOID ARTHRITIS AND OTHER IMMUNE-MEDIATED INFLAMMATORY RHEUMATIC DISEASES. RHEUMATOLOGY SCIENCE AND PRACTICE 2020. [DOI: 10.14412/1995-4484-2020-304-316] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Deciphering the mechanisms of the pathogenesis of immune-mediated inflammatory rheumatic diseases (IMIRDs) in conjunction with designing a wide range of biological agents is one of the major medical advances in the 21st century. A new promising area of pharmacotherapy for IMIRDs is associated with the design of the so-called targeted oral medications that primarily include Janus kinase (JAK) inhibitors. The review presents new data on the efficacy and safety of the new JAK inhibitor baricitinib in treating rheumatoid arthritis and other IMIRDs.
Collapse
Affiliation(s)
- E. L. Nasonov
- V.A. Nasonova Research Institute of Rheumatology;
I.M. Sechenov First Moscow State Medical University (Sechenov University), Ministry of Health of Russia
| | - A. M. Lila
- V.A. Nasonova Research Institute of Rheumatology; Russian Medical Academy of
Continuing Professional Education, Ministry of Health of Russia
| |
Collapse
|
27
|
Singh S, Singh S. JAK-STAT inhibitors: Immersing therapeutic approach for management of rheumatoid arthritis. Int Immunopharmacol 2020; 86:106731. [PMID: 32590315 DOI: 10.1016/j.intimp.2020.106731] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 12/16/2022]
Abstract
Rheumatoid arthritis is a world leading cause of musculoskeletal disease. With the introduction of biological agents as treatment alternatives the clinical possibilities have grown exponentially. Currently most common Disease-modifying anti-rheumatic drugs (DMARDs) treatment option involves intravenous or subcutaneous injection, and some patients struggle to respond to DMARDs or lose their primary reaction. An oral drug formulation with lowered costs of manufacturing and flexibility for healthcare workers to preferably perform treatment will result in decreased healthcare expenditures and increased medication compliance. The JAK-STAT inhibitors, a new class of small molecules drugs, fulfills these criteria and has recently shown efficacy in rheumatoid arthritis. Here we give a summary of how JAK-STAT inhibitors function and a detailed review of current clinical trials. Convincing clinical results suggest that therapeutic inhibition of the JAK proteins can effectively modulate a complex cytokine-driven inflammation.
Collapse
Affiliation(s)
- Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, Hajipur, Bihar, India.
| | - Shantanu Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Export Promotion Industrial Park (EPIP) Zandaha Road, Hajipur, Bihar, India
| |
Collapse
|
28
|
Atherosclerotic cardiovascular disease prevention in rheumatoid arthritis. Nat Rev Rheumatol 2020; 16:361-379. [PMID: 32494054 DOI: 10.1038/s41584-020-0428-y] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2020] [Indexed: 12/18/2022]
Abstract
Patients with rheumatoid arthritis (RA) are at high risk of developing cardiovascular disease (CVD). Inflammation has a pivotal role in the pathogenesis of CVD. RA is an inflammatory joint disease and, compared with the general population, patients with RA have approximately double the risk of atherosclerotic CVD, stroke, heart failure and atrial fibrillation. Although this high risk of CVD has been known for decades, patients with RA receive poorer primary and secondary CVD preventive care than other high-risk patients, and an unmet need exists for improved CVD preventive measures for patients with RA. This Review summarizes the evidence for atherosclerotic CVD in patients with RA and provides a contemporary analysis of what is known and what needs to be further clarified about recommendations for CVD prevention in patients with RA compared with the general population. The management of traditional CVD risk factors, including blood pressure, lipids, diabetes mellitus and lifestyle-related risk factors, as well as the effects of inflammation and the use of antirheumatic medication on CVD risk and risk management in patients with RA are discussed. The main aim is to provide a roadmap of atherosclerotic CVD risk management and prevention for patients with RA.
Collapse
|
29
|
Ballout RA, Remaley AT. GlycA: A New Biomarker for Systemic Inflammation and Cardiovascular Disease (CVD) Risk Assessment. ACTA ACUST UNITED AC 2020; 5. [PMID: 32363327 DOI: 10.21037/jlpm.2020.03.03] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The GlycA test is a recently developed proton nuclear magnetic resonance (1H-NMR) spectroscopy-based assay that has been gaining increased interest as a serum biomarker for systemic inflammation, and consequently, as a potential biomarker for cardiovascular disease (CVD) risk assessment. The test has undergone investigation in several large cohort studies, since its development, to assess its predictive value for incident CVD events, CVD-associated mortality, and all-cause mortality. Despite variation in the generated estimates by these studies, they have all consistently demonstrated moderate-strength positive correlations between baseline GlycA levels, and incident CVD event rates and associated mortality. These correlations withheld testing even after adjusting for several other established CVD risk factors, including notable inflammatory biomarkers such as high-sensitivity C-reactive protein (hsCRP) and interleukin-6 (IL-6). Compared with hsCRP, which is a well-known inflammatory biomarker for CVD risk assessment, GlycA has a comparable predictive value for future CVD-related events. However, the indications to pursue GlycA testing, and its clinical utility in patient care management, are yet to be determined. In this review, we define the GlycA test and what it "measures", and provide a brief summary of the findings of studies showing its association with incident CVD rates, and CVD-related mortality, as well as its correlation with other inflammatory biomarkers, namely hsCRP. Finally, we highlight the analytical advantages of the GlycA test, compared with "traditional" inflammatory biomarkers, while also mentioning its current limitations.
Collapse
Affiliation(s)
- Rami A Ballout
- Lipoprotein Metabolism Section, Translational and Vascular Medicine Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD, USA
| | - Alan T Remaley
- Lipoprotein Metabolism Section, Translational and Vascular Medicine Branch, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
30
|
Hosseini A, Gharibi T, Marofi F, Javadian M, Babaloo Z, Baradaran B. Janus kinase inhibitors: A therapeutic strategy for cancer and autoimmune diseases. J Cell Physiol 2020; 235:5903-5924. [DOI: 10.1002/jcp.29593] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 01/08/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Arezoo Hosseini
- Immunology Research CenterTabriz University of Medical SciencesTabriz Iran
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
- Student Research CommitteeTabriz University of Medical SciencesTabriz Iran
- Aging Research InstituteTabriz University of Medical SciencesTabriz Iran
| | - Tohid Gharibi
- Immunology Research CenterTabriz University of Medical SciencesTabriz Iran
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
- Student Research CommitteeTabriz University of Medical SciencesTabriz Iran
- Aging Research InstituteTabriz University of Medical SciencesTabriz Iran
| | - Faroogh Marofi
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
| | - Mahsa Javadian
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
| | - Zohreh Babaloo
- Immunology Research CenterTabriz University of Medical SciencesTabriz Iran
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
| | - Behzad Baradaran
- Immunology Research CenterTabriz University of Medical SciencesTabriz Iran
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
| |
Collapse
|
31
|
Fragoulis GE, McInnes IB, Siebert S. JAK-inhibitors. New players in the field of immune-mediated diseases, beyond rheumatoid arthritis. Rheumatology (Oxford) 2020; 58:i43-i54. [PMID: 30806709 PMCID: PMC6390879 DOI: 10.1093/rheumatology/key276] [Citation(s) in RCA: 231] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/03/2018] [Indexed: 02/06/2023] Open
Abstract
Janus kinase (JAK)/signal transducers and activators of transcription (STATs) are a group of molecules associated with one of the major pathways through which many cytokines exert and integrate their function, and as such they are increasingly recognized as playing critical role in the pathogenesis subserving various immune-mediated diseases, including RA, PsA, SpAs, IBD, skin disorders (e.g. alopecia areata, atopic dermatitis), single-gene disorders like interferonopathies, and others. JAKs are the key initiating players of the JAK/STAT pathway. Upon binding of their respective effector molecules (cytokines, IFNs, growth factors and others) to type I and type II receptors, JAKs are activated, and through phosphorylation of themselves and of other molecules (including STATs), they mediate signal transduction to the nucleus. A class of drugs—called JAK inhibitors or JAKinibs—that block one or more JAKs has been developed in the last decade, and now numbers >20 members. Although, so far, JAK inhibitors have been marketed only for RA and PsA, these drugs have been tested in phase 2 and phase 3 clinical trials for other inflammatory conditions and beyond. In this review, we summarize the clinical data, including efficacy and safety, available for JAK inhibitors used in some immune-mediated conditions other than RA.
Collapse
Affiliation(s)
- George E Fragoulis
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Stefan Siebert
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
32
|
Porter Starr KN, Connelly MA, Orenduff MC, McDonald SR, Sloane R, Huffman KM, Kraus WE, Bales CW. Impact on cardiometabolic risk of a weight loss intervention with higher protein from lean red meat: Combined results of 2 randomized controlled trials in obese middle-aged and older adults. J Clin Lipidol 2019; 13:920-931. [PMID: 31771921 DOI: 10.1016/j.jacl.2019.09.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 09/09/2019] [Accepted: 09/26/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND The recognized benefits of a higher protein diet on muscle mass and strength in older adults are tempered by concerns of the potentially negative cardiometabolic impact of dietary sources of animal protein. OBJECTIVE The aim of this study was to explore the cardiometabolic impact of 2 weight reduction diets: a higher protein diet, providing balanced portions of lean beef and pork throughout the day, vs. a diet following the Recommended Daily Allowance level of protein in obese middle-aged and older adults. METHODS Data from Measuring Eating, Activity and Strength: Understanding the Response-Using Protein and Protein Optimization in Women Enables Results-Using Protein were combined for the present analysis. Subjects were randomly assigned to a 6-month weight loss diet (500 kcal deficit) and prescribed a Recommended Daily Allowance level of protein (0.8 g protein/kg BW), control group, or a higher level of protein (1.2 g protein/kg BW), protein group. For the protein group, lean, high-quality protein was evenly distributed between meals or balanced throughout the day (30 g protein/meal). The following cardiometabolic markers were quantified by nuclear magnetic resonance spectroscopy: lipids, lipoproteins, GlycA, trimethylamine-N-oxide, betaine, branched-chain amino acids, and lipoprotein insulin resistance index scores. RESULTS In both groups (control [n = 27] and protein [n = 53]), there were significant (P ≤ .05) changes from baseline in weight loss (-6.2% and -7.2%), distance walked (+53.1 and +75.0 meters), and fasting plasma glucose (-7.5 and -6.2 mg/dL), respectively. At endpoint, protein group had significantly (P ≤ .05) lower triglycerides (-17.3 mg/dL), large very-low-density lipoprotein particle concentration (VLDL-P; -1.2 nmol/L), total low-density lipoprotein particle concentration (LDL-P; -67.8 nmol/L), small LDL-P (-59.4 nmol/L) and lipoprotein insulin resistance index (-5.9), whereas control group had significantly (P ≤ .05) lower GlycA (-13.1 μmol/L), total VLDL-P (-7.9 nmol/L), and small VLDL-P (-7.0 nmol/L). Differences between groups were observed for small VLDL-P (P = .02) and protein intake (P < .0001). CONCLUSIONS These findings suggest that a hypocaloric diet with either traditional (0.8 g/kg BW/d) or higher protein (1.2 g/kg BW/d; predominantly from lean red meat) content improves risk markers of cardiovascular disease and type II diabetes in obese middle-aged and older adults. Both diets were also associated with improved physical function, and neither had an adverse impact on cardiometabolic outcomes.
Collapse
Affiliation(s)
- Kathryn N Porter Starr
- Center for the Study of Aging, Duke University School of Medicine, Durham, NC, USA; Department of Medicine, Duke University School of Medicine, Durham, NC, USA; Geriatric Research, Education, and Clinical Center, Durham VA Medical Center, Durham, NC, USA.
| | - Margery A Connelly
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, NC, USA
| | - Melissa C Orenduff
- Center for the Study of Aging, Duke University School of Medicine, Durham, NC, USA
| | - Shelley R McDonald
- Center for the Study of Aging, Duke University School of Medicine, Durham, NC, USA; Department of Medicine, Duke University School of Medicine, Durham, NC, USA; Geriatric Research, Education, and Clinical Center, Durham VA Medical Center, Durham, NC, USA
| | - Richard Sloane
- Center for the Study of Aging, Duke University School of Medicine, Durham, NC, USA; Geriatric Research, Education, and Clinical Center, Durham VA Medical Center, Durham, NC, USA
| | - Kim M Huffman
- Department of Medicine and Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - William E Kraus
- Department of Medicine and Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Connie W Bales
- Center for the Study of Aging, Duke University School of Medicine, Durham, NC, USA; Department of Medicine, Duke University School of Medicine, Durham, NC, USA; Geriatric Research, Education, and Clinical Center, Durham VA Medical Center, Durham, NC, USA
| |
Collapse
|
33
|
Wu J, Zhu Z, Yu Q, Ding C. Tyrosine kinase inhibitors for the treatment of rheumatoid arthritis: phase I to Ⅱ clinical trials. Expert Opin Investig Drugs 2019; 28:1113-1123. [PMID: 31738612 DOI: 10.1080/13543784.2019.1692812] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Introduction: Rheumatoid arthritis (RA) is a chronic, refractory disorder caused by autoimmunity in the synovial joints. Disease-modifying anti-rheumatic drugs (DMARDs) and biologicals offer remission in only two-thirds of RA patients within 3 months, hence new therapeutic approaches are necessary. Tyrosine kinase inhibitors (TKIs) are newly developed small molecule drugs which have demonstrated encouraging results in this disease.Areas covered: The key findings from phase I and II clinical trials that have investigated the use of novel TKIs in the treatment of RA are discussed. We examined the literature published between January 2014 to January 2019 using electronic databases including PubMed, Web of Science, Medline, Embase, and Google Scholar. Additional information about phase I and II trials on the ClinicalTrial.gov website up to January 2019 was also retrieved.Expert opinion: JAK inhibitors are promising drugs with sound efficacy and acceptable safety and may be beneficial to patients who do not respond to DMARDs and biologicals. The response rates among RA patients to TKIs are diverse; genetic and environmental factors may be involved in the varying responses which are closely related to the pathogenesis of RA. Future studies may reveal the underlying mechanisms of resistance and non-response.
Collapse
Affiliation(s)
- Jing Wu
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhaohua Zhu
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qinghong Yu
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Changhai Ding
- Department of Rheumatology and Clinical Immunology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.,Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| |
Collapse
|
34
|
Richez C, Morel J, Cornec D, Daïen C, Goupille P, Lazaro E, Lequerré T, Nocturne G, de Lédinghen V, Le Goff B, Pourcher V, Prati C, Seror R, Tournadre A, Truchetet ME, Sibilia J, Pham T. Practical management of patients on Janus kinase inhibitor (JAKi) therapy: Practical fact sheets drawn up by the Rheumatism and Inflammation Club (CRI), a group endorsed by the French Society for Rheumatology (SFR). Joint Bone Spine 2019; 86 Suppl 1:eS2-eS103. [PMID: 31791545 DOI: 10.1016/s1297-319x(19)30154-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
| | | | | | - Claire Daïen
- Service de Rhumatologie, CHU Montpellier, Montpellier
| | | | | | | | | | - Victor de Lédinghen
- Unité d'Hépatologie et transplantation hépatique, Hôpital Haut-Lévêque, CHU Bordeaux, Pessac
| | | | - Valérie Pourcher
- Service de Maladies Infectieuses et Tropicales, CHU Pitié-Salpétrière, Paris
| | | | - Raphaèle Seror
- Service de Rhumatologie, CHU Bicêtre, Le Kremlin-Bicêtre
| | - Anne Tournadre
- Service de Rhumatologie, CHU Clermont-Ferrand, Clermont-Ferrand
| | | | - Jean Sibilia
- Service de Rhumatologie, CHU Hautepierre, Strasbourg
| | - Thao Pham
- Service de Rhumatologie, CHU Sainte-Marguerite, Marseille
| |
Collapse
|
35
|
Bechman K, Yates M, Galloway JB. The new entries in the therapeutic armamentarium: The small molecule JAK inhibitors. Pharmacol Res 2019; 147:104392. [PMID: 31401212 PMCID: PMC6876279 DOI: 10.1016/j.phrs.2019.104392] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023]
Abstract
The past decade has witnessed an explosion in trial data on JAK inhibitors (JAKi). These small molecules target the Janus kinase - signal transducer and activator of transcription (JAK-STAT) pathway, blocking crucial cytokines across a septum of rheumatic diseases. As a class, JAKi are beginning to demonstrate efficacy on par, if not superior to biologics. Two first generation JAKi are licensed for use in inflammatory arthritis; tofacitinib and baricitinib. Next-generation JAKi have been designed with selective affinity for one JAK enzymes, the aim to reduce unwanted adverse effects without declining clinical efficacy. Emerging data with selective JAK1 inhibitors upadacitinib and filgotinib looks very promising. Despite differences in selectivity between JAKi, an overlap exists in their safety profiles. Across the class, a characteristic safety signal is emerging with viral opportunistic infections, particularly herpes zoster. Post marketing drug surveillance will be essential in evaluating the long-term risk with these agents.
Collapse
Affiliation(s)
- Katie Bechman
- Centre for Rheumatic Diseases, Kings College London, United Kingdom.
| | - Mark Yates
- Centre for Rheumatic Diseases, Kings College London, United Kingdom
| | - James B Galloway
- Centre for Rheumatic Diseases, Kings College London, United Kingdom
| |
Collapse
|
36
|
Fleischmann R, Pangan AL, Song I, Mysler E, Bessette L, Peterfy C, Durez P, Ostor AJ, Li Y, Zhou Y, Othman AA, Genovese MC. Upadacitinib Versus Placebo or Adalimumab in Patients With Rheumatoid Arthritis and an Inadequate Response to Methotrexate: Results of a Phase
III
, Double‐Blind, Randomized Controlled Trial. Arthritis Rheumatol 2019; 71:1788-1800. [DOI: 10.1002/art.41032] [Citation(s) in RCA: 194] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 06/28/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Roy Fleischmann
- University of Texas Southwestern Medical Center and Metroplex Clinical Research Center Dallas
| | | | | | - Eduardo Mysler
- Organización Medica de Investigación Buenos Aires Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Xie W, Huang Y, Xiao S, Sun X, Fan Y, Zhang Z. Impact of Janus kinase inhibitors on risk of cardiovascular events in patients with rheumatoid arthritis: systematic review and meta-analysis of randomised controlled trials. Ann Rheum Dis 2019; 78:1048-1054. [PMID: 31088790 DOI: 10.1136/annrheumdis-2018-214846] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVES To investigate the effect of Janus kinase inhibitors (Jakinibs) on cardiovascular risk in adult patients with rheumatoid arthritis (RA) via a meta-analysis of randomised controlled trials (RCTs). METHODS PubMed, Embase and Cochrane library were thoroughly searched for RCTs reporting safety issues in patients with RA receiving Jakinibs, from inception to October 2018. The primary and secondary outcomes were all cardiovascular events (CVEs) and major adverse cardiovascular events (MACEs)/venous thromboembolism events (VTEs). OR and 95% CI were calculated using the Mantel-Haenszel fixed-effect method. RESULTS 26 RCTs randomising 11 799 patients were included. No significant difference was observed regarding all CVEs risk following Jakinibs usage in general (OR 1.04 (0.61 to 1.76), p = 0.89), tofacitinib (OR 0.63 (0.26 to 1.54), p = 0.31), baricitinib (OR 1.21 (0.51 to 2.83), p = 0.66), upadacitinib (OR 3.29 (0.59 to 18.44), p = 0.18), peficitinib (OR 0.43 (0.07 to 2.54), p = 0.35) or decernotinib (OR 1.12 (0.13 to 10.11), p = 0.92). Likewise, there was no significant difference for Jakinibs treatment overall regarding occurrence of MACEs (OR 0.80 (0.36 to 1.75), p = 0.57) or VTEs (OR 1.16 (0.48 to 2.81), p = 0.74). Dose-dependent impact of Jakinibs on the risks of all CVEs, MACEs and VTEs was not observed in tofacitinib (5 mg vs 10 mg), upadacitinib (15 mg vs 30 mg), whereas baricitinib at 2 mg was found to be safer than 4 mg in all CVEs incidence (OR 0.19 (0.04 to 0.88), p = 0.03). CONCLUSION The existing evidence from RCTs indicated no significant change in cardiovascular risk for Jakinib-treated patients with RA in a short-term perspective, but postmarketing data are sorely needed to ascertain their cardiovascular safety, especially at the higher dose, due to increased risk of thromboembolism events for both tofacitinib and baricitinib at higher dosage.
Collapse
Affiliation(s)
- Wenhui Xie
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China
| | - Yanrong Huang
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China
| | - Shiyu Xiao
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Xiaoying Sun
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China
| | - Yong Fan
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China
| | - Zhuoli Zhang
- Department of Rheumatology and Clinical Immunology, Peking University First Hospital, Beijing, China
| |
Collapse
|
38
|
Mackey RH, Kuller LH, Moreland LW. Inflammatory joint diseases and atherosclerosis: time to look beyond the 'lipid paradox'. Curr Opin Lipidol 2019; 30:342-349. [PMID: 31145122 DOI: 10.1097/mol.0000000000000620] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Uncertainty persists about the contribution of lipids to the increased risk of cardiovascular disease (CVD) among rheumatoid arthritis and other inflammatory joint disease (IJD) patients. In reviewing recent research, we consider potential insights gained by quantifying lipoprotein particles directly, rather than by their lipid content. RECENT FINDINGS Although inflammation often decreases LDL cholesterol (LDL-C), and anti-inflammatory medications often increase LDL-C, both inflammation and anti-inflammatory medications can increase atherogenic Apolipoprotein B (ApoB)-containing lipoprotein particles, attenuated by statins. CVD risk factors, that is, smoking, obesity, ApoB, may increase years prior to IJD diagnosis. Increased risks of nonatherosclerotic myocardial and pulmonary disease, heart failure and mortality may be directly related to disease activity, inflammation, and possibly to HDL particles and function. SUMMARY For IJD patients, higher cumulative lifetime exposure to CVD risk factors accelerates atherosclerosis and subsequent CVD risk that is underestimated by current risk factor levels. CVD risk reduction in IJD requires aggressive and earlier reduction in CVD risk factors (ApoB lipoproteins, smoking, hypertension, diabetes, lack of physical activity), in addition to control of disease activity and inflammation. Lipid-lowering medications can attenuate anti-inflammatory medication-induced increases in ApoB and LDL-C, but can also reduce CVD risk due to cumulative lifetime exposure.
Collapse
Affiliation(s)
- Rachel H Mackey
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh
| | - Lewis H Kuller
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, University of Pittsburgh
| | - Larry W Moreland
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
39
|
Cardiometabolic comorbidities in RA and PsA: lessons learned and future directions. Nat Rev Rheumatol 2019; 15:461-474. [PMID: 31292564 DOI: 10.1038/s41584-019-0256-0] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2019] [Indexed: 02/07/2023]
Abstract
Cardiometabolic comorbidities present a considerable burden for patients with rheumatoid arthritis (RA) or psoriatic arthritis (PsA). Both RA and PsA are associated with an increased risk of cardiovascular disease (CVD). PsA more often exhibits an increased risk of metabolically linked comorbidities such as obesity, insulin resistance, type 2 diabetes mellitus and non-alcoholic fatty liver disease. Although both RA and PsA are characterized by a state of chronic inflammation, the mechanisms that contribute to CVD risk in these conditions might not be identical. In RA, systemic inflammation is thought to directly contribute to CVD risk, whereas in PsA, adiposity is thought to contribute to a notable metabolic phenotype that, in turn, contributes to CVD risk. Hence, appropriate management strategies that consider the increased risk of cardiometabolic comorbidities in patients with inflammatory arthropathy are important. In RA, such strategies should focus on the prediction of CVD risk and its management through targeting chronic inflammation and traditional CVD risk factors. In PsA, management strategies should additionally focus on targeting metabolic components, including weight management, which might not only help improve disease activity in the joints, entheses and skin, but also reduce the risk of metabolic comorbidities and improve the quality of life of patients.
Collapse
|
40
|
Taylor PC, Weinblatt ME, Burmester GR, Rooney TP, Witt S, Walls CD, Issa M, Salinas CA, Saifan C, Zhang X, Cardoso A, González‐Gay MA, Takeuchi T. Cardiovascular Safety During Treatment With Baricitinib in Rheumatoid Arthritis. Arthritis Rheumatol 2019; 71:1042-1055. [PMID: 30663869 PMCID: PMC6618316 DOI: 10.1002/art.40841] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 01/15/2019] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To assess the frequency of cardiovascular and venous thromboembolic events in clinical studies of baricitinib, an oral, selective JAK1 and JAK2 inhibitor approved in more than 50 countries for the treatment of moderately-to-severely active rheumatoid arthritis (RA). METHODS Data were pooled from 9 RA studies. Placebo comparison up to 24 weeks included data from 6 studies. Randomized dose comparison between baricitinib doses of 2 mg and 4 mg used data from 4 studies and from the associated long-term extension study. The data analysis set designated "All-bari-RA" included all baricitinib exposures at any dose. RESULTS Overall, 3,492 RA patients received baricitinib (7,860 patient-years of exposure). No imbalance compared to the placebo group was seen in the incidence of major adverse cardiovascular events (MACE) (incidence rates [IRs] of 0.5 per 100 patient-years for placebo and 0.8 per 100 patient-years for 4 mg baricitinib), arterial thrombotic events (ATE) (IRs of 0.5 per 100 patient-years for placebo and 0.5 per 100 patient-years for 4 mg baricitinib), or congestive heart failure (CHF) broad term (IRs of 4.3 per 100 patient-years for placebo and 2.4 per 100 patient-years for 4 mg baricitinib). Deep vein thrombosis (DVT)/pulmonary embolism (PE) were reported in 0 of 1,070 patients treated with placebo and 6 of 997 patients treated with 4 mg baricitinib during the placebo-controlled period; these events were serious in 2 of 6 patients, while all 6 had risk factors and 1 patient developed DVT/PE after discontinuation of the study drug. In the 2 mg-4 mg-extended data analysis set, IRs of DVT/PE were comparable between the doses across event types (IRs of 0.5 per 100 patient-years in those receiving 2 mg baricitinib and 0.6 per 100 patient-years in those receiving 4 mg baricitinib). In the All-bari-RA data analysis set, the rates were stable over time, with an IR of DVT/PE of 0.5 per 100 patient-years. CONCLUSION In RA clinical trials, no association was found between baricitinib treatment and the incidence of MACE, ATE, or CHF. With regard to incidence of DVT/PE, 6 events occurred in patients treated with 4 mg baricitinib, but no cases of DVT/PE were reported in the placebo group. During longer-term evaluation, the incidence of DVT/PE was similar between the baricitinib dose groups, with consistent IR values over time, and this was similar to the rates previously reported in patients with RA.
Collapse
Affiliation(s)
| | | | | | | | - Sarah Witt
- Eli Lilly and CompanyIndianapolisIndiana
| | | | - Maher Issa
- Eli Lilly and CompanyIndianapolisIndiana
| | | | | | - Xin Zhang
- Eli Lilly and CompanyIndianapolisIndiana
| | | | - Miguel A. González‐Gay
- Hospital Universitario Marqués de ValdecillaIDIVAL and University of Cantabria, Santander, Spainand University of the WitwatersrandJohannesburgSouth Africa
| | | |
Collapse
|
41
|
Taylor PC. Clinical efficacy of launched JAK inhibitors in rheumatoid arthritis. Rheumatology (Oxford) 2019; 58:i17-i26. [PMID: 30806707 PMCID: PMC6390878 DOI: 10.1093/rheumatology/key225] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 06/20/2018] [Indexed: 12/15/2022] Open
Abstract
Tofacitinib and baricitinib are the first orally available, small-molecule inhibitors of Janus kinase (JAK) enzymes to be approved for the treatment of RA. Tofacitinib is a selective JAK1, 3 inhibitor with less activity against JAK2 and TYK2 and baricitinib is a selective, oral JAK1, 2 inhibitor with moderate activity against TYK2 and significantly less activity against JAK3. Both drugs have undergone extensive phase III clinical trials in RA and demonstrated rapid improvements in disease activity, function and patient-reported outcomes as well as disease modification. Tofacitinib 5 mg bd, was approved by the Federal Drug Administration in 2012 for the treatment of RA in patients who are intolerant or unresponsive to MTX. An extended release formulation for the treatment of RA was approved by Federal Drug Administration in 2016. In 2017 the European Medicines Agency approved tofacitinib 5 mg bd in combination with MTX and baricitinib 4 mg and 2 mg once daily for the treatment of moderate to severe active RA in adult patients who are intolerant or unresponsive to one or more conventional synthetic DMARDs.
Collapse
Affiliation(s)
- Peter C Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK
| |
Collapse
|
42
|
Genovese MC, Fleischmann R, Combe B, Hall S, Rubbert-Roth A, Zhang Y, Zhou Y, Mohamed MEF, Meerwein S, Pangan AL. Safety and efficacy of upadacitinib in patients with active rheumatoid arthritis refractory to biologic disease-modifying anti-rheumatic drugs (SELECT-BEYOND): a double-blind, randomised controlled phase 3 trial. Lancet 2018; 391:2513-2524. [PMID: 29908670 DOI: 10.1016/s0140-6736(18)31116-4] [Citation(s) in RCA: 325] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 05/07/2018] [Accepted: 05/10/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Phase 2 studies with upadacitinib, a selective Janus kinase 1 (JAK1) inhibitor, have shown safety and efficacy in the treatment of patients with active rheumatoid arthritis. We did this study to further assess the safety and efficacy of upadacitinib in patients with an inadequate response to biologic disease-modifying anti-rheumatic drugs (bDMARDs). METHODS We did this double-blind, randomised controlled phase 3 trial at 153 sites in 26 countries. Patients were aged 18 years or older, had active rheumatoid arthritis and previous inadequate response or intolerance to bDMARDs, and were receiving concomitant background conventional synthetic DMARDS (csDMARDs). We randomly assigned patients (2:2:1:1) by interactive response technology to receive once-daily oral extended-release upadacitinib 15 mg or 30 mg or placebo for 12 weeks, followed by upadacitinib 15 mg or 30 mg from week 12 onwards. The two separate primary endpoints were the proportions of patients achieving a 20% improvement in American College of Rheumatology criteria (ACR20) at week 12 and the proportion of patients achieving a 28-joint disease activity score using C-reactive protein (DAS28[CRP]) of 3·2 or less at week 12. Efficacy and safety analyses were done in the modified intention-to-treat population of all patients who received at least one dose of study drug. Data are presented up to week 24 of this ongoing study. The trial is registered with ClinicalTrials.gov (NCT02706847). FINDINGS Between March 15, 2016, and Jan 10, 2017, 499 patients were randomly assigned (n=165 upadacitinib 15 mg; n=165 upadacitinib 30 mg; n=85 placebo then upadacitinib 15 mg; and n=84 placebo then upadacitinib 30 mg) and one patient was withdrawn from the 15 mg upadacitinib group before the start of study treatment. Mean disease duration was 13·2 years (SD 9·5); 235 (47%) of 498 patients had received one previous bDMARD, 137 (28%) had received two, and 125 (25%) had received at least three; 451 (91%) patients completed treatment up to week 12 and 419 (84%) patients completed treatment up to week 24. At week 12, ACR20 was achieved by 106 (65%; 95% CI 57-72) of 164 patients receiving upadacitinib 15 mg and 93 (56%; 49-64) of 165 patients receiving upadacitinib 30 mg compared with 48 (28%; 22-35) of 169 patients receiving placebo (p<0·0001 for each dose vs placebo). DAS28(CRP) of 3·2 or less was achieved by 71 (43%; 95% CI 36-51) of 164 patients receiving upadacitinib 15 mg and 70 (42%; 35-50) of 165 patients receiving upadacitinib 30 mg versus 24 (14%; 9-20) of 169 patients receiving placebo (p<0·0001 for each dose vs placebo). Up to week 12, overall numbers of patients with adverse events were similar for the placebo group (95 [56%] of 169) and the upadacitinib 15 mg group (91 [55%] of 164), but higher in the upadacitinib 30 mg group (111 [67%] of 165). At week 12, the most common adverse events occurring in at least 5% of patients in any treatment group were upper respiratory tract infection (13 [8%] of 169 in the placebo group; 13 [8%] of 164 in the upadacitinib 15 mg group; ten [6%] of 165 in the upadacitinib 30 mg group), nasopharyngitis (11 [7%]; seven [4%]; nine [5%]), urinary tract infection (ten [6%]; 15 [9%]; nine [5%]), and worsening of rheumatoid arthritis (ten [6%]; four [2%]; six [4%]). The number of patients with serious adverse events was higher in the upadacitinib 30 mg group (12 [7%]) than in the upadacitinib 15 mg group (eight [5%]); no serious adverse events were reported in patients receiving placebo. More patients in the upadacitinib 30 mg group had serious infections, herpes zoster, and adverse events leading to discontinuation than in the upadacitinib 15 mg and placebo groups. During the placebo-controlled phase of the study, one case of pulmonary embolism, three malignancies, one major adverse cardiovascular event, and one death were reported in patients receiving upadacitinib; none were reported in patients receiving placebo. INTERPRETATION Both doses of upadacitinib led to rapid and significant improvements compared with placebo over 12 weeks in patients with refractory rheumatoid arthritis. FUNDING AbbVie Inc.
Collapse
Affiliation(s)
- Mark C Genovese
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Palo Alto, CA, USA.
| | - Roy Fleischmann
- University of Texas Southwestern, Metroplex Clinical Research Center, Dallas, Texas, USA
| | - Bernard Combe
- Department of Rheumatology, CHU Montpellier, Université de Montpellier, Montpellier, France
| | - Stephen Hall
- Department of Medicine, Monash University, Cabrini Health and Emeritus Research, Malvern, VIC, Australia
| | | | | | | | | | | | | |
Collapse
|
43
|
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
|
44
|
Cardiovascular Safety of Biologics and JAK Inhibitors in Patients with Rheumatoid Arthritis. Curr Rheumatol Rep 2018; 20:42. [DOI: 10.1007/s11926-018-0752-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|