51
|
Zhang S, Song J, Yang Y, Miao H, Yang L, Liu Y, Zhang X, Liu Y, Wang T. Type I interferonopathies with novel compound heterozygous TREX1 mutations in two siblings with different symptoms responded to tofacitinib. Pediatr Rheumatol Online J 2021; 19:1. [PMID: 33407657 PMCID: PMC7789551 DOI: 10.1186/s12969-020-00490-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 12/09/2020] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Type I interferonopathies are a group of rare autoimmune diseases characterised by excessive activation of type I interferon that leads to disturbances in immune function. Three prime repair exonuclease 1 (TREX1) is an important exonuclease and plays an important role in DNA damage repair. TREX1 mutations are associated with many type I interferonopathies. Studies have been published on the effectiveness of tofacitinib in the treatment of type I interferonopathies. The aim of this study is to identify the pathogenic variation in a Chinese family with type I interferonopathies and to observe the therapeutic effects of tofacitinib. METHODS A Chinese family with two members with type I interferonopathies was investigated. Whole exome sequencing and Sanger sequencing were applied for mutation screening using peripheral blood DNA of the patient and her family members. Sequencing results were analysed using bioinformatics software tools including VarCards and PolyPhen-2. Close clinical follow-up and observation were used to record changes in the disease before and after treatment with tofacitinib. RESULTS Compound heterozygous variants of TREX1 were observed in the patient's genome. One was a missense variant (NM_016381; c.C227T; p.Ala76Val) from the patient's father, and the other was a frameshift variant (NM_016381; c.458dupA; p.Gln153Glnfs*3) from the patient's mother. One of the proband's elder brothers with similar skin lesions also carried these two variants. This brother of the proband had more serious cutaneous involvement with the comorbidity of cerebral palsy. These TREX1 variants have not been reported in previous studies and are predicted to be highly pathogenic. The proband was given tofacitinib that led to a marked improvement. CONCLUSIONS We identified two novel complex heterozygous variants in the TREX1 gene, which may underlie the molecular pathogenesis of the type I interferonopathies observed in members of this family. Tofacitinib could be an alternative treatment for this disease.
Collapse
Affiliation(s)
- Shiyu Zhang
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Jiaxing Song
- grid.506261.60000 0001 0706 7839Department of Medical Genetics and National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
| | - Yuyan Yang
- grid.506261.60000 0001 0706 7839Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Huilei Miao
- grid.506261.60000 0001 0706 7839Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Lu Yang
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Yuehua Liu
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 China
| | - Xue Zhang
- grid.506261.60000 0001 0706 7839Department of Medical Genetics and National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005 China
| | - Yaping Liu
- Department of Medical Genetics and National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China.
| | - Tao Wang
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
52
|
Nash P, Kerschbaumer A, Dörner T, Dougados M, Fleischmann RM, Geissler K, McInnes I, Pope JE, van der Heijde D, Stoffer-Marx M, Takeuchi T, Trauner M, Winthrop KL, de Wit M, Aletaha D, Baraliakos X, Boehncke WH, Emery P, Isaacs JD, Kremer J, Lee EB, Maksymowych WP, Voshaar M, Tam LS, Tanaka Y, van den Bosch F, Westhovens R, Xavier R, Smolen JS. Points to consider for the treatment of immune-mediated inflammatory diseases with Janus kinase inhibitors: a consensus statement. Ann Rheum Dis 2021; 80:71-87. [PMID: 33158881 PMCID: PMC7788060 DOI: 10.1136/annrheumdis-2020-218398] [Citation(s) in RCA: 158] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Janus kinase inhibitors (JAKi) have been approved for use in various immune-mediated inflammatory diseases. With five agents licensed, it was timely to summarise the current understanding of JAKi use based on a systematic literature review (SLR) on efficacy and safety. METHODS Existing data were evaluated by a steering committee and subsequently reviewed by a 29 person expert committee leading to the formulation of a consensus statement that may assist the clinicians, patients and other stakeholders once the decision is made to commence a JAKi. The committee included patients, rheumatologists, a gastroenterologist, a haematologist, a dermatologist, an infectious disease specialist and a health professional. The SLR informed the Task Force on controlled and open clinical trials, registry data, phase 4 trials and meta-analyses. In addition, approval of new compounds by, and warnings from regulators that were issued after the end of the SLR search date were taken into consideration. RESULTS The Task Force agreed on and developed four general principles and a total of 26 points for consideration which were grouped into six areas addressing indications, treatment dose and comedication, contraindications, pretreatment screening and risks, laboratory and clinical follow-up examinations, and adverse events. Levels of evidence and strengths of recommendations were determined based on the SLR and levels of agreement were voted on for every point, reaching a range between 8.8 and 9.9 on a 10-point scale. CONCLUSION The consensus provides an assessment of evidence for efficacy and safety of an important therapeutic class with guidance on issues of practical management.
Collapse
Affiliation(s)
- Peter Nash
- School of Medicine, Griffith University, Brisbane, Queensland, Australia
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Dörner
- Dept. Med./Rheumatology and Clinical Immunology, Charite Univ. Hospital, Berlin, Germany
| | - Maxime Dougados
- Hopital Cochin, Rheumatology, Université Paris Descartes, Paris, France
| | - Roy M Fleischmann
- Department of Medicine, Southwestern University of Texas, Dallas, Texas, USA
| | | | - Iain McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Janet E Pope
- Medicine, Division of Rheumatology, The University of Western Ontario, London, Ontario, Canada
| | | | - Michaela Stoffer-Marx
- Section for Outcomes Research, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | | | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | | | - Maarten de Wit
- Medical Humanities, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Daniel Aletaha
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria
| | | | | | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, UK
| | - John D Isaacs
- Musculoskeletal Research Group, Newcastle University, Newcastle upon Tyne, UK
| | - Joel Kremer
- Rheumatology, Albany Medical College, Albany, New York, USA
| | - Eun Bong Lee
- Internal Medicine, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Walter P Maksymowych
- Medicine, University of Alberta Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada
| | - Marieke Voshaar
- Medical Humanities, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Lai-Shan Tam
- Department of Medicine & Therapeutics, Chinese University of Hong Kong Shaw College, New Territories, Hong Kong
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | | | | | - Ricardo Xavier
- Division of Rheumatology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Josef S Smolen
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
53
|
Nash P, Kerschbaumer A, Dörner T, Dougados M, Fleischmann RM, Geissler K, McInnes I, Pope JE, van der Heijde D, Stoffer-Marx M, Takeuchi T, Trauner M, Winthrop KL, de Wit M, Aletaha D, Baraliakos X, Boehncke WH, Emery P, Isaacs JD, Kremer J, Lee EB, Maksymowych WP, Voshaar M, Tam LS, Tanaka Y, van den Bosch F, Westhovens R, Xavier R, Smolen JS. Points to consider for the treatment of immune-mediated inflammatory diseases with Janus kinase inhibitors: a consensus statement. Ann Rheum Dis 2021. [PMID: 33158881 DOI: 10.1136/annrheumdis2020-218580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
OBJECTIVES Janus kinase inhibitors (JAKi) have been approved for use in various immune-mediated inflammatory diseases. With five agents licensed, it was timely to summarise the current understanding of JAKi use based on a systematic literature review (SLR) on efficacy and safety. METHODS Existing data were evaluated by a steering committee and subsequently reviewed by a 29 person expert committee leading to the formulation of a consensus statement that may assist the clinicians, patients and other stakeholders once the decision is made to commence a JAKi. The committee included patients, rheumatologists, a gastroenterologist, a haematologist, a dermatologist, an infectious disease specialist and a health professional. The SLR informed the Task Force on controlled and open clinical trials, registry data, phase 4 trials and meta-analyses. In addition, approval of new compounds by, and warnings from regulators that were issued after the end of the SLR search date were taken into consideration. RESULTS The Task Force agreed on and developed four general principles and a total of 26 points for consideration which were grouped into six areas addressing indications, treatment dose and comedication, contraindications, pretreatment screening and risks, laboratory and clinical follow-up examinations, and adverse events. Levels of evidence and strengths of recommendations were determined based on the SLR and levels of agreement were voted on for every point, reaching a range between 8.8 and 9.9 on a 10-point scale. CONCLUSION The consensus provides an assessment of evidence for efficacy and safety of an important therapeutic class with guidance on issues of practical management.
Collapse
Affiliation(s)
- Peter Nash
- School of Medicine, Griffith University, Brisbane, Queensland, Australia
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria
| | - Thomas Dörner
- Dept. Med./Rheumatology and Clinical Immunology, Charite Univ. Hospital, Berlin, Germany
| | - Maxime Dougados
- Hopital Cochin, Rheumatology, Université Paris Descartes, Paris, France
| | - Roy M Fleischmann
- Department of Medicine, Southwestern University of Texas, Dallas, Texas, USA
| | | | - Iain McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Janet E Pope
- Medicine, Division of Rheumatology, The University of Western Ontario, London, Ontario, Canada
| | | | - Michaela Stoffer-Marx
- Section for Outcomes Research, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | | | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine 3, Medical University of Vienna, Vienna, Austria
| | | | - Maarten de Wit
- Medical Humanities, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Daniel Aletaha
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria
| | | | | | - Paul Emery
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, UK
| | - John D Isaacs
- Musculoskeletal Research Group, Newcastle University, Newcastle upon Tyne, UK
| | - Joel Kremer
- Rheumatology, Albany Medical College, Albany, New York, USA
| | - Eun Bong Lee
- Internal Medicine, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Walter P Maksymowych
- Medicine, University of Alberta Faculty of Medicine and Dentistry, Edmonton, Alberta, Canada
| | - Marieke Voshaar
- Medical Humanities, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Lai-Shan Tam
- Department of Medicine & Therapeutics, Chinese University of Hong Kong Shaw College, New Territories, Hong Kong
| | - Yoshiya Tanaka
- First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | | | | | - Ricardo Xavier
- Division of Rheumatology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Josef S Smolen
- Division of Rheumatology, Department of Medicine 3, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
54
|
Cazzato S, Omenetti A, Ravaglia C, Poletti V. Lung involvement in monogenic interferonopathies. Eur Respir Rev 2020; 29:200001. [PMID: 33328278 PMCID: PMC9489100 DOI: 10.1183/16000617.0001-2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 05/27/2020] [Indexed: 12/29/2022] Open
Abstract
Monogenic type I interferonopathies are inherited heterogeneous disorders characterised by early onset of systemic and organ specific inflammation, associated with constitutive activation of type I interferons (IFNs). In the last few years, several clinical reports identified the lung as one of the key target organs of IFN-mediated inflammation. The major pulmonary patterns described comprise children's interstitial lung diseases (including diffuse alveolar haemorrhages) and pulmonary arterial hypertension but diagnosis may be challenging. Respiratory symptoms may be either mild or absent at disease onset and variably associated with systemic or organ specific inflammation. In addition, associated extrapulmonary clinical features may precede lung function impairment by years, and patients may display severe/endstage lung involvement, although this may be clinically hidden during the long-term disease course. Conversely, a few cases of atypical severe lung involvement at onset have been reported without clinically manifested extrapulmonary signs. Hence, a multidisciplinary approach involving pulmonologists, paediatricians and rheumatologists should always be considered when a monogenic interferonopathy is suspected. Pulmonologists should also be aware of the main pattern of presentation to allow prompt diagnosis and a targeted therapeutic strategy. In this regard, promising therapeutic strategies rely on Janus kinase-1/2 (JAK-1/2) inhibitors blocking the type I IFN-mediated intracellular cascade.
Collapse
Affiliation(s)
- Salvatore Cazzato
- Pediatric Unit, Dept of Mother and Child Health, Salesi Children's Hospital, Ancona, Italy
- Joint first authors
| | - Alessia Omenetti
- Pediatric Unit, Dept of Mother and Child Health, Salesi Children's Hospital, Ancona, Italy
- Joint first authors
| | - Claudia Ravaglia
- Dept of Diseases of the Thorax, Ospedale GB Morgagni, Forlì, Italy
| | - Venerino Poletti
- Dept of Diseases of the Thorax, Ospedale GB Morgagni, Forlì, Italy
- Dept of Respiratory Diseases & Allergy, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
55
|
Gadina M, Chisolm DA, Philips RL, McInness IB, Changelian PS, O'Shea JJ. Translating JAKs to Jakinibs. THE JOURNAL OF IMMUNOLOGY 2020; 204:2011-2020. [PMID: 32253269 DOI: 10.4049/jimmunol.1901477] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/11/2020] [Indexed: 12/21/2022]
Abstract
The discovery of JAKs and STATs and their roles in cytokine and IFN action represented a significant basic advance and a new paradigm in cell signaling. This was quickly followed by discoveries pointing to their essential functions, including identification of JAK3 mutations as a cause of SCID. This and other findings predicted the use of therapeutically targeting JAKs as a new strategy for treating immune and inflammatory diseases. This now is a reality with seven approved jakinibs being used to treat multiple forms of arthritis, inflammatory bowel disease and myeloproliferative neoplasms, and numerous ongoing clinical trials in other settings. This story provides interesting insights into the process of translating basic discoveries and also reveals the need to return to basic work to fill gaps that now become apparent.
Collapse
Affiliation(s)
- Massimo Gadina
- Translational Immunology Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Danielle A Chisolm
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Rachael L Philips
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Iain B McInness
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, United Kingdom; and
| | | | - John J O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
56
|
Hasan MJ, Rabbani R, Anam AM, Huq SMR. Additional baricitinib loading dose improves clinical outcome in COVID-19. Open Med (Wars) 2020; 16:41-46. [PMID: 33364433 PMCID: PMC7729634 DOI: 10.1515/med-2021-0010] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/02/2020] [Accepted: 10/06/2020] [Indexed: 01/15/2023] Open
Abstract
Pneumonia associated with coronavirus disease 2019 (COVID-19) has been accounted for high mortality rate in severe COVID-19 worldwide, and additional serious scarcity of standard and effective anti-inflammatory drug in COVID-19 pneumonia management is a big challenge. Baricitinib, a Janus kinase (JAK) inhibitor, is a promising drug in COVID-19 pneumonia. This study aims to compare the clinical outcome of moderate-to-severe COVID-19 pneumonia treated with baricitinib with or without a loading dose. This prospective case-control study enrolled 37 adult patients where 17 patients (control) received baricitinib at 4 mg oral daily dose and 20 patients (case) received an additional single 8 mg oral loading dose. The median day to gain blood oxygen saturation level ≥95% (in room air) and return in normal breathing function were lower in case group than the control group. The requirement of intensive care unit and mechanical ventilation support was higher in the control group than in the case group [29.4% (N = 17)/10% (N = 20), P < 0.05; 11.8% (N = 17)/5% (N = 20), P > 0.05), respectively]. Thus, an additional loading dose of baricitinib revealed better clinical outcome of patients with COVID-19 pneumonia.
Collapse
Affiliation(s)
- Md Jahidul Hasan
- Clinical Pharmacy Services, Department of Pharmacy, Square Hospitals Ltd, 18/F Bir Uttam Qazi Nuruzzaman Sarak, West Panthapath, Dhaka-1205, Bangladesh
| | - Raihan Rabbani
- Department of Medical Services, Internal Medicine and ICU, Square Hospitals Ltd, 18/F Bir Uttam Qazi Nuruzzaman Sarak, West Panthapath, Dhaka-1205, Bangladesh
| | - Ahmad Mursel Anam
- Department of Medical Services, High Dependency Unit (HDU), Square Hospitals Ltd, 18/F Bir Uttam Qazi Nuruzzaman Sarak, West Panthapath, Dhaka-1205, Bangladesh
| | - Shihan Mahmud Redwanul Huq
- Department of Medical Services, Internal Medicine and ICU, Square Hospitals Ltd, 18/F Bir Uttam Qazi Nuruzzaman Sarak, West Panthapath, Dhaka-1205, Bangladesh
| |
Collapse
|
57
|
Hamilton CE, Craiglow BG. JAK Inhibitors for the Treatment of Pediatric Alopecia Areata. J Investig Dermatol Symp Proc 2020; 20:S31-S36. [PMID: 33099381 DOI: 10.1016/j.jisp.2020.04.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alopecia areata is a common autoimmune condition that disproportionately affects children and can significantly hinder quality of life. Few safe and effective therapies are available for the treatment of severely affected pediatric patients. JAK inhibitors have been recently established as an effective and well-tolerated therapy in adults, but there are limited data regarding the use of JAK inhibitors to treat alopecia areata in children. Here, we review the available literature regarding the use of JAK inhibitors in children in dermatology and across other medical disciplines.
Collapse
Affiliation(s)
- Claire E Hamilton
- Department of Dermatology, Yale University, New Haven, Connecticut, USA
| | - Brittany G Craiglow
- Department of Dermatology, Yale University, New Haven, Connecticut, USA; Dermatology Physicians of Connecticut, Fairfield, Connecticut, USA.
| |
Collapse
|
58
|
Bieber T. Novel therapies based on the pathophysiology of atopic dermatitis. J Dtsch Dermatol Ges 2020; 17:1150-1162. [PMID: 31765104 DOI: 10.1111/ddg.13965] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 09/17/2019] [Indexed: 12/26/2022]
Abstract
Our understanding of the mechanisms underlying atopic dermatitis has been instrumental in the development of novel pharmacological agents. Worldwide, more than seventy companies are currently developing at least one new drug for the treatment of atopic dermatitis. Based on our current understanding of the pathophysiology of the disease, the present review article highlights the most important novel therapeutic strategies.
Collapse
Affiliation(s)
- Thomas Bieber
- Department of Dermatology and Allergology, University Medical Center, Rheinische Friedrich Wilhelms University, Bonn, Germany
| |
Collapse
|
59
|
Zhao X, Sheng XY, Payne CD, Zhang X, Wang F, Cui YM. Pharmacokinetics, Safety, and Tolerability of Single- and Multiple-Dose Once-Daily Baricitinib in Healthy Chinese Subjects: A Randomized Placebo-Controlled Study. Clin Pharmacol Drug Dev 2020; 9:952-960. [PMID: 32945153 PMCID: PMC9292889 DOI: 10.1002/cpdd.868] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 08/09/2020] [Indexed: 01/25/2023]
Abstract
The objective of this phase 1 study was to evaluate the pharmacokinetics, safety, and tolerability of baricitinib after single and multiple doses in healthy Chinese adults. Eligible subjects received a once‐daily dose of baricitinib 2, 4, or 10 mg or placebo on day 1 (single dose) and days 4 through 10 for 7 consecutive days (multiple doses). Plasma pharmacokinetic samples were collected up to 48 hours after dosing on days 1 and 10, with predose samples collected before dosing on day 1 and days 4 through 10. Safety and tolerability were also assessed. Baricitinib was rapidly absorbed, reaching peak plasma concentrations within 0.5 to 1 hour (median). Plasma concentrations declined rapidly following the attainment of peak concentrations, with a mean terminal half‐life of 5.7 to 7.3 hours. Steady‐state plasma concentrations of baricitinib were achieved after the second day of once‐daily dosing, with minimal accumulation of baricitinib in plasma (up to 10% increase in area under the plasma concentration–time curve). Single‐ and multiple‐dose mean values for area under the plasma concentration–time curve from time zero to infinity and maximum plasma concentration appeared to increase in an approximately dose‐proportional manner across the dose range. Single and multiple oral doses of once‐daily baricitinib up to 10 mg were well tolerated by healthy Chinese subjects.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| | - Xiao Yan Sheng
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| | | | - Xin Zhang
- Medical Department, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Feng Wang
- Medical Department, Lilly Suzhou Pharmaceutical Co. Ltd., Shanghai, China
| | - Yi Min Cui
- Department of Pharmacy, Peking University First Hospital, Beijing, China
| |
Collapse
|
60
|
Kim H, Dill S, O'Brien M, Vian L, Li X, Manukyan M, Jain M, Adeojo LW, George J, Perez M, Grom AA, Sutter M, Feldman BM, Yao L, Millwood M, Brundidge A, Pichard DC, Cowen EW, Shi Y, Lu S, Tsai WL, Gadina M, Rider LG, Colbert RA. Janus kinase (JAK) inhibition with baricitinib in refractory juvenile dermatomyositis. Ann Rheum Dis 2020; 80:406-408. [PMID: 32843325 DOI: 10.1136/annrheumdis-2020-218690] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Hanna Kim
- Juvenile Myositis Pathogenesis and Therapeutics Unit, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA .,Pediatric Clinical Trials Unit, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Samantha Dill
- Pediatric Clinical Trials Unit, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA.,Office of the Clinical Director, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Michelle O'Brien
- Pediatric Clinical Trials Unit, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA.,Office of the Clinical Director, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Laura Vian
- Translational Immunology Section, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Xiaobai Li
- Biostatistics and Clinical Epidemiology Service, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Manuk Manukyan
- Pediatric Clinical Trials Unit, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA.,Office of the Clinical Director, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Minal Jain
- Rehabilitation Medicine Department, Physical Therapy Section, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Lilian W Adeojo
- Clinical Pharmacokinetics Research Unit, Pharmacy Department, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Jomy George
- Clinical Pharmacokinetics Research Unit, Pharmacy Department, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Maria Perez
- Pediatric Rheumatology, Cook Children's Medical Center, Fort Worth, Texas, USA
| | - Alexei A Grom
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Michelle Sutter
- Department of Rheumatology, Children's Hospital Colorado, Aurora, Colorado, USA.,Pediatrics Rheumatology, University of Colorado Denver, Denver, Colorado, USA
| | - Brian M Feldman
- Division of Rheumatology, Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Lawrence Yao
- Radiology and Imaging Sciences, National Institutes of Health Clinical Center, Bethesda, Maryland, USA
| | - Michelle Millwood
- Pediatric Clinical Trials Unit, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA.,Office of the Clinical Director, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - April Brundidge
- Pediatric Clinical Trials Unit, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA.,Office of the Clinical Director, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Dominique C Pichard
- Dermatology Branch, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Edward W Cowen
- Dermatology Branch, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Yinghui Shi
- Office of the Clinical Director, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Shajia Lu
- Translational Immunology Section, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Wanxia Li Tsai
- Translational Immunology Section, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Massimo Gadina
- Translational Immunology Section, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| | - Lisa G Rider
- Environmental Autoimmunity Group, NIEHS/NIH/DHHS, Bethesda, Maryland, USA
| | - Robert A Colbert
- Pediatric Clinical Trials Unit, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA.,Office of the Clinical Director, NIH, National Institute of Arthritis and Musculoskeletal and Skin Diseases, Bethesda, Maryland, USA
| |
Collapse
|
61
|
Jorgensen SCJ, Tse CLY, Burry L, Dresser LD. Baricitinib: A Review of Pharmacology, Safety, and Emerging Clinical Experience in COVID-19. Pharmacotherapy 2020; 40:843-856. [PMID: 32542785 PMCID: PMC7323235 DOI: 10.1002/phar.2438] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A hyperinflammatory response to severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) infection, reminiscent of cytokine release syndrome, has been implicated in the pathophysiology of acute respiratory distress syndrome and organ damage in patients with coronavirus disease 2019 (COVID-19). Agents that inhibit components of the pro-inflammatory cascade have garnered interest as potential treatment options with hopes that dampening the proinflammatory process may improve clinical outcomes. Baricitinib is a reversible Janus-associated kinase (JAK)-inhibitor that interrupts the signaling of multiple cytokines implicated in COVID-19 immunopathology. It may also have antiviral effects by targeting host factors that viruses rely for cell entry and by suppressing type I interferon driven angiotensin-converting-enzyme-2 upregulation. However, baricitinib's immunosuppressive effects may be detrimental during acute viral infections by delaying viral clearance and increasing vulnerability to secondary opportunistic infections. The lack of reliable biomarkers to monitor patients' immune status as illness evolves complicates deployment of immunosuppressive drugs like baricitinib. Furthermore, baricitinib carries the risk of increased thromboembolic events, which is concerning given the proclivity towards a hypercoagulable state in patients with COVID-19. In this article, we review available data on baricitinib with an emphasis on immunosuppressive and antiviral pharmacology, pharmacokinetics, safety, and current progress in COVID-19 clinical trials.
Collapse
Affiliation(s)
| | - Christopher L Y Tse
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Lisa Burry
- Department of Pharmacy, Sinai Health System, Toronto, Ontario, Canada.,Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Linda D Dresser
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.,Antimicrobial Stewardship Program, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
62
|
Wolf C, Brück N, Koss S, Griep C, Kirschfink M, Palm-Beden K, Fang M, Röber N, Winkler S, Berner R, Latz E, Günther C, Lee-Kirsch MA. Janus kinase inhibition in complement component 1 deficiency. J Allergy Clin Immunol 2020; 146:1439-1442.e5. [PMID: 32325142 DOI: 10.1016/j.jaci.2020.04.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/26/2020] [Accepted: 04/03/2020] [Indexed: 12/31/2022]
Affiliation(s)
- Christine Wolf
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Normi Brück
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Sarah Koss
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Constanze Griep
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Michael Kirschfink
- Institute for Immunology, Universitätsklinikum Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Katharina Palm-Beden
- Klinik für Kinder- und Jugendrheumatologie, St Josef-Stift, Sendenhorst, Germany
| | - Mingyan Fang
- BGI Shenzhen and China National Genebank, Shenzhen, China
| | - Nadja Röber
- Institute for Immunology, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefan Winkler
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Reinhard Berner
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Eike Latz
- Institute of Innate Immunity, University of Bonn, Bonn, Germany
| | - Claudia Günther
- Department of Dermatology, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Min Ae Lee-Kirsch
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
63
|
Ezzeldin E, Iqbal M, Asiri YA, Ali AA, Alam P, El-Nahhas T. A Hydrophilic Interaction Liquid Chromatography-Tandem Mass Spectrometry Quantitative Method for Determination of Baricitinib in Plasma, and Its Application in a Pharmacokinetic Study in Rats. Molecules 2020; 25:E1600. [PMID: 32244454 PMCID: PMC7180984 DOI: 10.3390/molecules25071600] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 03/27/2020] [Accepted: 03/28/2020] [Indexed: 12/19/2022] Open
Abstract
Baricitinib, is a selective and reversible Janus kinase inhibitor, is commonly used to treat adult patients with moderately to severely active rheumatoid arthritis (RA). A fast, reproducible and sensitive method of liquid chromatography-tandem mass spectrometry (LC-MS/MS) for the quantification of baricitinib in rat plasma has been developed. Irbersartan was used as the internal standard (IS). Baracitinib and IS were extracted from plasma by liquid-liquid extraction using a mixture of n-hexane and dichloromethane (1:1) as extracting agent. Chromatographic separation was performed using Acquity UPLC HILIC BEH 1.7 µm 2.1 × 50 mm column with the mobile phase consisting of 0.1% formic acid in acetonitrile and 20 mM ammonium acetate (pH 3) (97:3). The electrospray ionization in the positive-mode was used for sample ionization in the multiple reaction monitoring mode. Baricitinib and the IS were quantified using precursor-to-production transitions of m/z 372.15 > 251.24 and 429.69 > 207.35 for baricitinib and IS, respectively. The method was validated according to the recent FDA and EMA guidelines for bioanalytical method validation. The lower limit of quantification was 0.2 ng/mL, whereas the intra-day and inter-day accuracies of quality control (QCs) samples were ranged between 85.31% to 89.97% and 87.50% to 88.33%, respectively. Linearity, recovery, precision, and stability parameters were found to be within the acceptable range. The method was applied successfully applied in pilot pharmacokinetic studies.
Collapse
Affiliation(s)
- Essam Ezzeldin
- Department of Pharmaceutical Chemistry and Drug Bioavailability Unit, Central Laboratory, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia or (M.I.)
- National Organization for Drug Control and Research, Cairo 12611, Egypt
| | - Muzaffar Iqbal
- Department of Pharmaceutical Chemistry and Drug Bioavailability Unit, Central Laboratory, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia or (M.I.)
| | - Yousif A. Asiri
- Clinical Pharmacy Department, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Azza A Ali
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls) Al-Azhar University, Cairo 11754, Egypt;
| | - Prawez Alam
- Pharmacognosy Department, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Toqa El-Nahhas
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls) Al-Azhar University, Cairo 11754, Egypt;
| |
Collapse
|
64
|
Sönmez HE, Karaaslan C, de Jesus AA, Batu ED, Anlar B, Sözeri B, Bilginer Y, Karaguzel D, Ayvaz DC, Tezcan I, Goldbach-Mansky R, Ozen S. A clinical score to guide in decision making for monogenic type I IFNopathies. Pediatr Res 2020; 87:745-752. [PMID: 31641281 PMCID: PMC8425764 DOI: 10.1038/s41390-019-0614-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 08/13/2019] [Accepted: 10/01/2019] [Indexed: 01/09/2023]
Abstract
OBJECTIVE To develop a set of clinical criteria that identifies patients with a potential autoinflammatory IFNopathy. METHODS Based on a literature review, a set of clinical criteria identifying genetically confirmed monogenic IFNopathies was selected. For validation, the clinical score was assessed in healthy controls (HCs) and 18 disease controls, including 2 known autoimmune IFNopathies, juvenile systemic lupus erythematosus (JSLE, n = 4) and dermatomyositis (JDM, n = 4); adenosine deaminase 2 deficiency (DADA2, n = 4); and oligoarticular juvenile idiopathic arthritis (oJIA, n = 6). We assessed an IFN score (IRG-S) in whole blood by NanoString using a previously published 28-gene-IRG-S and a reduced 6-gene-IRG-S. RESULTS The 12 patients with a possible IFNopathy had higher clinical scores (3-5) than the patients with sJLE, JDM, DADA2, and oJIA and in HCs. Both the 28-IRG-S and 6-IRG-S were significantly higher in the autoinflammatory IFNopathy patients compared to HCs and oJIA and DADA2 patients but not different from patients with JSLE and JDM. Subsequently, genetic analysis revealed mutations in genes previously reported in genes related to the IFN pathway in 9 of the 12 patients. CONCLUSION We developed a clinical score to identify patients with possible autoinflammatory IFNopathies. A clinical score was associated with a high IRG-S and may serve to identify patients with an autoinflammatory IFNopathy.
Collapse
Affiliation(s)
- Hafize Emine Sönmez
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Cagatay Karaaslan
- Department of Biology, Molecular Biology Section, Hacettepe University Faculty of Science, Ankara, Turkey
| | - Adriana A. de Jesus
- Translational Autoinflammatory Diseases Section (TADS), Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ezgi Deniz Batu
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Banu Anlar
- Division of Neurology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Betül Sözeri
- Division of Rheumatology, Department of Pediatrics, Umraniye Research and Training Hospital, Istanbul, Turkey
| | - Yelda Bilginer
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Dilara Karaguzel
- Department of Biology, Molecular Biology Section, Hacettepe University Faculty of Science, Ankara, Turkey
| | - Deniz Cagdas Ayvaz
- Division of Immunology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Ilhan Tezcan
- Division of Immunology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Raphaela Goldbach-Mansky
- Translational Autoinflammatory Diseases Section (TADS), Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Seza Ozen
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey.
| |
Collapse
|
65
|
Krutzke S, Rietschel C, Horneff G. Baricitinib in therapy of COPA syndrome in a 15-year-old girl. Eur J Rheumatol 2020; 7:S78-S81. [PMID: 31449490 PMCID: PMC7004263 DOI: 10.5152/eurjrheum.2019.18177] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 03/24/2019] [Indexed: 12/18/2022] Open
Abstract
COPA syndrome is a newly discovered hereditary immunodeficiency affecting the lung, kidneys, and joints. The mutated gene encodes the α subunit of the coatomer complex I, a protein transporter from the Golgi back to the endoplasmic reticulum. The impaired return of proteins leads to intracellular stress. The syndrome is an autoimmune and autoinflammatory disease that can be grouped among the interferonopathies. The knowledge about COPA syndrome and its treatment is still limited. In this paper, we describe an additional patient, a 15-year-old girl with rheumatoid factor-positive polyarthritis and rheumatoid nodules since the age of 2, who developed interstitial lung disease. The detected mutation c.698G>A was causing the disease. The patient presented with symmetric polyarthritis on wrists, fingers, and hip and ankle joints, with significant functional impairment, and high disease activity. Laboratory parameters demonstrated chronic inflammation, hypergammaglobulinemia, high titre ANA (antinuclear antibodies) and CCP (anti-citrullinated protein) antibodies, and rheumatoid factors. Therapies with various DMARDs (Disease Modifying Anti-Rheumatic Drugs) and biologicals failed. Upon baricitinib application, the clinical activity decreased dramatically with disappearance of joint pain and morning stiffness and significant decrease of joint swelling. A low disease activity was reached after 12 months, with complete disappearance of rheumatoid nodules. In contrast to IL-1 (interleukin-1), IL-6, and TNF (tumor necrosis factor) inhibitors, baricitinib was very successful, probably because baricitinib acts as a JAK-1/2 (janus kinase-1/2) inhibitor in the IFNα/β (inteferone α/β) pathway. A relatively higher dose in children is necessary. COPA syndrome represents a novel disorder of intracellular transport. Reviewing published literature on COPA syndrome, in addition to our patient, there were 31 cases further described.
Collapse
Affiliation(s)
- Sophia Krutzke
- Department of General Paediatrics, Centre for Paediatric Rheumatology, Clinic Sankt Augustin, Sankt Augustin, Germany
| | - Christoph Rietschel
- Department for Pediatric Rheumatology, Clementine Kinderhospital, Frankfurt, Germany
| | - Gerd Horneff
- Department of Pediatric and Adolescents medicine, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
66
|
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: 212] [Impact Index Per Article: 53.0] [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
|
67
|
Tüngler V, Doebler-Neumann M, Salandin M, Kaufmann P, Wolf C, Lucas N, Harmuth F, Reichbauer J, Krägeloh-Mann I, Schüle R, Lee-Kirsch MA. Aicardi-Goutières syndrome due to a paternal mosaic IFIH1 mutation. NEUROLOGY-GENETICS 2019; 6:e384. [PMID: 32042913 PMCID: PMC6936312 DOI: 10.1212/nxg.0000000000000384] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 10/31/2019] [Indexed: 11/15/2022]
Affiliation(s)
- Victoria Tüngler
- Department of Pediatrics (V.T., C.W., N.L., M.A.L.-K.), Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden; Department of Neuropediatrics (M.D.-N., I.K.-M.), University of Tübingen, Germany; Child Neurology and Psychiatry Unit (M.S.), Paediatric Department, Bolzano Regional Hospital; Child Haematology and Oncology Unit (P.K.), Paediatric Department, Bolzano Regional Hospital, Italy; Institute of Medical Genetics and Applied Genomics (F.H.), University of Tübingen; and Center for Neurology and Hertie-Institute for Clinical Brain Research (J.R., R.S.), University of Tübingen and German Center of Neurodegenerative Diseases, Germany
| | - Marion Doebler-Neumann
- Department of Pediatrics (V.T., C.W., N.L., M.A.L.-K.), Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden; Department of Neuropediatrics (M.D.-N., I.K.-M.), University of Tübingen, Germany; Child Neurology and Psychiatry Unit (M.S.), Paediatric Department, Bolzano Regional Hospital; Child Haematology and Oncology Unit (P.K.), Paediatric Department, Bolzano Regional Hospital, Italy; Institute of Medical Genetics and Applied Genomics (F.H.), University of Tübingen; and Center for Neurology and Hertie-Institute for Clinical Brain Research (J.R., R.S.), University of Tübingen and German Center of Neurodegenerative Diseases, Germany
| | - Michaela Salandin
- Department of Pediatrics (V.T., C.W., N.L., M.A.L.-K.), Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden; Department of Neuropediatrics (M.D.-N., I.K.-M.), University of Tübingen, Germany; Child Neurology and Psychiatry Unit (M.S.), Paediatric Department, Bolzano Regional Hospital; Child Haematology and Oncology Unit (P.K.), Paediatric Department, Bolzano Regional Hospital, Italy; Institute of Medical Genetics and Applied Genomics (F.H.), University of Tübingen; and Center for Neurology and Hertie-Institute for Clinical Brain Research (J.R., R.S.), University of Tübingen and German Center of Neurodegenerative Diseases, Germany
| | - Peter Kaufmann
- Department of Pediatrics (V.T., C.W., N.L., M.A.L.-K.), Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden; Department of Neuropediatrics (M.D.-N., I.K.-M.), University of Tübingen, Germany; Child Neurology and Psychiatry Unit (M.S.), Paediatric Department, Bolzano Regional Hospital; Child Haematology and Oncology Unit (P.K.), Paediatric Department, Bolzano Regional Hospital, Italy; Institute of Medical Genetics and Applied Genomics (F.H.), University of Tübingen; and Center for Neurology and Hertie-Institute for Clinical Brain Research (J.R., R.S.), University of Tübingen and German Center of Neurodegenerative Diseases, Germany
| | - Christine Wolf
- Department of Pediatrics (V.T., C.W., N.L., M.A.L.-K.), Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden; Department of Neuropediatrics (M.D.-N., I.K.-M.), University of Tübingen, Germany; Child Neurology and Psychiatry Unit (M.S.), Paediatric Department, Bolzano Regional Hospital; Child Haematology and Oncology Unit (P.K.), Paediatric Department, Bolzano Regional Hospital, Italy; Institute of Medical Genetics and Applied Genomics (F.H.), University of Tübingen; and Center for Neurology and Hertie-Institute for Clinical Brain Research (J.R., R.S.), University of Tübingen and German Center of Neurodegenerative Diseases, Germany
| | - Nadja Lucas
- Department of Pediatrics (V.T., C.W., N.L., M.A.L.-K.), Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden; Department of Neuropediatrics (M.D.-N., I.K.-M.), University of Tübingen, Germany; Child Neurology and Psychiatry Unit (M.S.), Paediatric Department, Bolzano Regional Hospital; Child Haematology and Oncology Unit (P.K.), Paediatric Department, Bolzano Regional Hospital, Italy; Institute of Medical Genetics and Applied Genomics (F.H.), University of Tübingen; and Center for Neurology and Hertie-Institute for Clinical Brain Research (J.R., R.S.), University of Tübingen and German Center of Neurodegenerative Diseases, Germany
| | - Florian Harmuth
- Department of Pediatrics (V.T., C.W., N.L., M.A.L.-K.), Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden; Department of Neuropediatrics (M.D.-N., I.K.-M.), University of Tübingen, Germany; Child Neurology and Psychiatry Unit (M.S.), Paediatric Department, Bolzano Regional Hospital; Child Haematology and Oncology Unit (P.K.), Paediatric Department, Bolzano Regional Hospital, Italy; Institute of Medical Genetics and Applied Genomics (F.H.), University of Tübingen; and Center for Neurology and Hertie-Institute for Clinical Brain Research (J.R., R.S.), University of Tübingen and German Center of Neurodegenerative Diseases, Germany
| | - Jennifer Reichbauer
- Department of Pediatrics (V.T., C.W., N.L., M.A.L.-K.), Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden; Department of Neuropediatrics (M.D.-N., I.K.-M.), University of Tübingen, Germany; Child Neurology and Psychiatry Unit (M.S.), Paediatric Department, Bolzano Regional Hospital; Child Haematology and Oncology Unit (P.K.), Paediatric Department, Bolzano Regional Hospital, Italy; Institute of Medical Genetics and Applied Genomics (F.H.), University of Tübingen; and Center for Neurology and Hertie-Institute for Clinical Brain Research (J.R., R.S.), University of Tübingen and German Center of Neurodegenerative Diseases, Germany
| | - Ingeborg Krägeloh-Mann
- Department of Pediatrics (V.T., C.W., N.L., M.A.L.-K.), Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden; Department of Neuropediatrics (M.D.-N., I.K.-M.), University of Tübingen, Germany; Child Neurology and Psychiatry Unit (M.S.), Paediatric Department, Bolzano Regional Hospital; Child Haematology and Oncology Unit (P.K.), Paediatric Department, Bolzano Regional Hospital, Italy; Institute of Medical Genetics and Applied Genomics (F.H.), University of Tübingen; and Center for Neurology and Hertie-Institute for Clinical Brain Research (J.R., R.S.), University of Tübingen and German Center of Neurodegenerative Diseases, Germany
| | - Rebecca Schüle
- Department of Pediatrics (V.T., C.W., N.L., M.A.L.-K.), Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden; Department of Neuropediatrics (M.D.-N., I.K.-M.), University of Tübingen, Germany; Child Neurology and Psychiatry Unit (M.S.), Paediatric Department, Bolzano Regional Hospital; Child Haematology and Oncology Unit (P.K.), Paediatric Department, Bolzano Regional Hospital, Italy; Institute of Medical Genetics and Applied Genomics (F.H.), University of Tübingen; and Center for Neurology and Hertie-Institute for Clinical Brain Research (J.R., R.S.), University of Tübingen and German Center of Neurodegenerative Diseases, Germany
| | - Min Ae Lee-Kirsch
- Department of Pediatrics (V.T., C.W., N.L., M.A.L.-K.), Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden; Department of Neuropediatrics (M.D.-N., I.K.-M.), University of Tübingen, Germany; Child Neurology and Psychiatry Unit (M.S.), Paediatric Department, Bolzano Regional Hospital; Child Haematology and Oncology Unit (P.K.), Paediatric Department, Bolzano Regional Hospital, Italy; Institute of Medical Genetics and Applied Genomics (F.H.), University of Tübingen; and Center for Neurology and Hertie-Institute for Clinical Brain Research (J.R., R.S.), University of Tübingen and German Center of Neurodegenerative Diseases, Germany
| |
Collapse
|
68
|
Marino A, Tirelli F, Giani T, Cimaz R. Periodic fever syndromes and the autoinflammatory diseases (AIDs). J Transl Autoimmun 2019; 3:100031. [PMID: 32743516 PMCID: PMC7388371 DOI: 10.1016/j.jtauto.2019.100031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/13/2019] [Accepted: 12/13/2019] [Indexed: 12/14/2022] Open
Abstract
Innate immune system represents the ancestral defense against infectious agents preserved along the evolution and species; it is phylogenetically older than the adaptive immune system, which exists only in the vertebrates. Cells with phagocytic activity such as neutrophils, macrophages, and natural killer (NK) cells play a key role in innate immunity. In 1999 Kastner et al. first introduced the term “autoinflammation” describing two diseases characterized by recurrent episodes of systemic inflammation without any identifiable infectious trigger: Familial Mediterranean Fever (FMF) and TNF Receptor Associated Periodic Syndrome (TRAPS). Autoinflammatory diseases (AIDs) are caused by self-directed inflammation due to an alteration of innate immunity leading to systemic inflammatory attacks typically in an on/off mode. In addition to inflammasomopathies, nuclear factor (NF)-κB-mediated disorders (also known as Rhelopathies) and type 1 interferonopathies are subjects of more recent studies. This review aims to provide an overview of the field with the most recent updates (see “Most recent developments in..” paragraphs) and a description of the newly identified AIDs. Autoinflammatory diseases are caused by self-directed inflammation. Alteration of innate immunity leads to systemic inflammation attacks. The autoinflammatory field is exponentially expanding. The advances in AIDs have led to new insights into immune system understanding. Autoimmunity and autoinflammation features may be simultaneously present.
Collapse
Affiliation(s)
- Achille Marino
- Department of Pediatrics, Desio Hospital, ASST Monza, Desio, MB, Italy.,Biomedical Sciences, University of Florence, Florence, Italy
| | - Francesca Tirelli
- Rheumatology Unit, Meyer Children's Hospital, University of Florence, Florence, Italy
| | - Teresa Giani
- Rheumatology Unit, Meyer Children's Hospital, University of Florence, Florence, Italy.,Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Rolando Cimaz
- Department of Clinical Sciences and Community Health, University of Milano, Milan, Italy
| |
Collapse
|
69
|
Papa R, Picco P, Gattorno M. The expanding pathways of autoinflammation: a lesson from the first 100 genes related to autoinflammatory manifestations. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 120:1-44. [PMID: 32085880 DOI: 10.1016/bs.apcsb.2019.11.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AutoInflammatory Diseases (AIDs) are a group of innate immune system disorders characterized by sterile inflammation without evidence of pathogenic autoantibodies or auto-reactive T lymphocytes. An expanding spectrum of genes and molecular pathways are associated with AIDs. Inflammasomopathies are secondary to dysregulation of multi-protein complexes, called inflammasomes, leading to an excessive maturation and secretion of IL1β and IL18. Patients present with persistent or recurrent systemic inflammation, abdominal and chest pain, skin rashes and are sensible to IL1 inhibitors. Unfolded proteins response causes a small number of AIDs that we propose to call immuno-proteinopathies, characterized by recurrent fevers and deep tissues inflammation. Other inflammatory conditions can occur in case of abnormalities of actin polymerization and the term of immuno-actinopathies is proposed. Generalized pustular psoriasis is a marker of autoinflammation mainly affecting the keratinocytes. Specific treatment targeting the p40 subunit of IL12 and IL23 or IL-17 are usually effective. Granulomatous inflammation characterizes AIDs related to NOD2 signaling defects. Defects in the ubiquitin-proteasome system cause a group of relopathies and some interferonopathies related to defect of the proteasome function (CANDLE syndrome). Gain of function of proteins regulating the production of type I interferons lead to severe inflammatory conditions, called interferonopathies. The JAK/STAT inhibitors are usually effective in these latter conditions. In conclusions, the identification of the main intracellular pathways involved in rare monogenic AIDs allows not only the proper classification of different conditions, but also highlight a pivotal role of possible novel therapeutic targets for the future.
Collapse
Affiliation(s)
- Riccardo Papa
- Autoinflammatory Diseases and Immunodeficiencies Centre, IRCCS Istituto Giannina Gaslini, Genova, GE, Italy
| | - Paolo Picco
- Autoinflammatory Diseases and Immunodeficiencies Centre, IRCCS Istituto Giannina Gaslini, Genova, GE, Italy
| | - Marco Gattorno
- Autoinflammatory Diseases and Immunodeficiencies Centre, IRCCS Istituto Giannina Gaslini, Genova, GE, Italy
| |
Collapse
|
70
|
Solimani F, Meier K, Ghoreschi K. Emerging Topical and Systemic JAK Inhibitors in Dermatology. Front Immunol 2019; 10:2847. [PMID: 31849996 PMCID: PMC6901833 DOI: 10.3389/fimmu.2019.02847] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022] Open
Abstract
Accumulating data on cellular and molecular pathways help to develop novel therapeutic strategies in skin inflammation and autoimmunity. Examples are psoriasis and atopic dermatitis, two clinically and immunologically well-defined disorders. Here, the elucidation of key pathogenic factors such as IL-17A/IL-23 on the one hand and IL-4/IL-13 on the other hand profoundly changed our therapeutic practice. The knowledge on intracellular pathways and governing factors is shifting our attention to new druggable molecules. Multiple cytokine receptors signal through Janus kinases (JAKs) and associated signal transducer and activators of transcription (STATs). Inhibition of JAKs can simultaneously block the function of multiple cytokines. Therefore, JAK inhibitors (JAKi) are emerging as a new class of drugs, which in dermatology can either be used systemically as oral drugs or locally in topical formulations. Inhibition of JAKs has been shown to be effective in various skin disorders. The first oral JAKi have been recently approved for the treatment of rheumatoid arthritis and psoriatic arthritis. Currently, multiple inhibitors of the JAK/STAT pathway are being investigated for skin diseases like alopecia areata, atopic dermatitis, dermatomyositis, graft-versus-host-disease, hidradenitis suppurativa, lichen planus, lupus erythematosus, psoriasis, and vitiligo. Here, we aim to discuss the immunological basis and current stage of development of JAKi in dermatology.
Collapse
Affiliation(s)
- Farzan Solimani
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Katharina Meier
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
71
|
Bieber T. Neuartige Therapien auf der Grundlage der Pathophysiologie der atopischen Dermatitis. J Dtsch Dermatol Ges 2019; 17:1150-1163. [PMID: 31765109 DOI: 10.1111/ddg.13965_g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 09/17/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Thomas Bieber
- Klinik und Poliklinik für Dermatologie und Allergologie, Universitätsklinikum, Rheinische Friedrich-Wilhelms-Universität Bonn, Erklärung zu nicht-finanziellen Interesse: Organisation: Klinik und Poliklinik für Dermatologie und Allergologie Position in dieser Organisation: Direktor Mitgliedschaft und Position in wissenschaftlichen Gesellschaften/Berufsverbänden/Vereinigungen: Direktor bei Christine-Kühne Center for Allergy Research and Education (CK-CARE) der Kühne-Stiftung; Sprecher beim Zentrum für Translationale Medizin (Med. Fakultät Bonn und Bundes -Institut für Arzneimittel und Medizinprodukte (BfArM), Bonn; Vorsitzender des Wissenschaftlichen Beirates des BfArM, Bonn; Stellvertretender Vorsitzender des gemeinsamen Wissenschaftlichen Beirates der Bundesoberbehörden am Bundesministerium für Gesundheit (BMG), Berlin; Mitglied der Nationalen Akademie der Naturforscher, Leopoldina Halle; Ehrenamtliches Mitglied folgender Aufsichtsräte: Hochgebirgsklinik (Davos) sowie Davos Biosciences (Davos, Schweiz). Mitglied und Ehrenmitglied zahlreicher nationaler und internationaler wissenschaftlicher Gesellschaften
| |
Collapse
|
72
|
Balci S, Ekinci RMK, de Jesus AA, Goldbach-Mansky R, Yilmaz M. Baricitinib experience on STING-associated vasculopathy with onset in infancy: A representative case from Turkey. Clin Immunol 2019; 212:108273. [PMID: 31626957 DOI: 10.1016/j.clim.2019.108273] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/10/2019] [Accepted: 10/10/2019] [Indexed: 11/26/2022]
Abstract
Stimulator of interferon genes associated vasculopathy with onset in infancy (SAVI), caused by heterozygote gain-of-function mutations in TMEM173, is characterized by fever attacks with ulcerating cutaneous manifestations on cold-sensitive areas and interstitial lung disease. A six-month-old boy was admitted to our hospital with fever, cough, and rash on the external surface of both upper and lower extremities. Respiratory symptoms consistent with ILD developed and skin lesions evolved to eschar formation particularly on acral regions. Ultimately, diagnosis of SAVI was confirmed at the age of 10 months due to the high level of interferon-score and a heterozygous N154S mutation in TMEM173. Since systemic corticosteroid and ruxolitinib were not effective, baricitinib was initiated at the age of 15 months, resulting in alleviation of fever attacks, cutaneous manifestations and respiratory symptoms within 2 months. In conclusion, we reported an infant diagnosed with SAVI at the age of 10 months and treated with baricitinib.
Collapse
Affiliation(s)
- Sibel Balci
- Department of Pediatric Rheumatology, Cukurova University Faculty of Medicine, Adana, Turkey.
| | | | - Adriana Almeida de Jesus
- Translational Autoinflammatory Disease Studies (TADS), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA.
| | - Raphaela Goldbach-Mansky
- Translational Autoinflammatory Disease Studies (TADS), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA.
| | - Mustafa Yilmaz
- Department of Pediatric Rheumatology, Cukurova University Faculty of Medicine, Adana, Turkey; Department of Pediatric Allergy and Immunology, Cukurova University Faculty of Medicine, Adana, Turkey
| |
Collapse
|
73
|
Gavegnano C, Haile WB, Hurwitz S, Tao S, Jiang Y, Schinazi RF, Tyor WR. Baricitinib reverses HIV-associated neurocognitive disorders in a SCID mouse model and reservoir seeding in vitro. J Neuroinflammation 2019; 16:182. [PMID: 31561750 PMCID: PMC6764124 DOI: 10.1186/s12974-019-1565-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 08/28/2019] [Indexed: 12/12/2022] Open
Abstract
Background Since HIV-associated neurocognitive disorders (HANDs) occur in up to half of HIV-positive individuals, even with combined antiretroviral therapy (cART), adjunctive therapies are needed. Chronic CNS inflammation contributes to HAND and HIV encephalitis (HIVE). Baricitinib is a JAK 1/2 inhibitor approved in the USA, EU, and Japan for rheumatoid arthritis, demonstrating potent inhibition of IL-6, D-dimer, CRP, TNF-α, IFN-α/β, and other pro-inflammatory cytokines. Methods Our modified murine HAND model was used to evaluate the ability of baricitinib to cross the blood-brain barrier (BBB) and modulate monocyte/macrophage-driven HAND. Severity of HAND was measured by assessing cognitive performance of low- and high-dose baricitinib treated versus untreated HAND mice. The severity of brain neuroinflammation was evaluated in these mouse groups after flow cytometric analyses. We also assessed the ability of baricitinib to block events in myeloid and lymphoid cells in vitro that may undergird the persistence of HIV in the central nervous system (CNS) in primary human macrophages (Mϕ) and lymphocytes including HIV replication, HIV-induced activation, reservoir expansion, and reservoir maintenance. Results In vivo, both doses of 10 and 50 mg/kg qd baricitinib crossed the BBB and reversed behavioral abnormalities conferred by HIV infection. Moreover, baricitinib significantly reduced HIV-induced neuroinflammation marked by glial activation: activated microglia (MHCII+/CD45+) and astrogliosis (GFAP). Baricitinib also significantly reduced the percentage of p24+ human macrophages in mouse brains (p < 0.05 versus HAND mice; t test). In vitro, baricitinib significantly reduced markers of persistence, reservoir size, and reseeding in Mϕ. Conclusion These results show that blocking the JAK/STAT pathway reverses cognitive deficits and curtails inflammatory markers in HAND in mice. Our group recently reported safety and tolerability of ruxolitinib in HIV-infected individuals (Marconi et al., Safety, tolerability and immunologic activity of ruxolitinib added to suppressive ART, 2019), underscoring potential safety and utility of JAK inhibitors for additional human trials. The data reported herein coupled with our recent human trial with JAK inhibitors provide compelling preclinical data and impetus for considering a trial of baricitinib in HAND individuals treated with cART to reverse cognitive deficits and key events driving viral persistence. Electronic supplementary material The online version of this article (10.1186/s12974-019-1565-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christina Gavegnano
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA.,Emory Center for AIDS Research (CFAR), Emory University, Atlanta, GA, 30322, USA
| | - Woldeab B Haile
- Emory Center for AIDS Research (CFAR), Emory University, Atlanta, GA, 30322, USA.,Department of Neurology, School of Medicine, Emory University, Atlanta, GA, 30209, USA.,Atlanta Veterans Affairs Medical Center, Decatur, GA, 30033, USA
| | - Selwyn Hurwitz
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA.,Emory Center for AIDS Research (CFAR), Emory University, Atlanta, GA, 30322, USA
| | - Sijia Tao
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA.,Emory Center for AIDS Research (CFAR), Emory University, Atlanta, GA, 30322, USA
| | - Yong Jiang
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA.,Emory Center for AIDS Research (CFAR), Emory University, Atlanta, GA, 30322, USA
| | - Raymond F Schinazi
- Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA. .,Emory Center for AIDS Research (CFAR), Emory University, Atlanta, GA, 30322, USA.
| | - William R Tyor
- Emory Center for AIDS Research (CFAR), Emory University, Atlanta, GA, 30322, USA. .,Department of Neurology, School of Medicine, Emory University, Atlanta, GA, 30209, USA. .,Atlanta Veterans Affairs Medical Center, Decatur, GA, 30033, USA.
| |
Collapse
|
74
|
Ohmura K. Nakajo-Nishimura syndrome and related proteasome-associated autoinflammatory syndromes. J Inflamm Res 2019; 12:259-265. [PMID: 31576159 PMCID: PMC6765212 DOI: 10.2147/jir.s194098] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/28/2019] [Indexed: 12/22/2022] Open
Abstract
Nakajo-Nishimura syndrome (NNS) is a rare hereditary autoinflammatory disorder with lipodystrophy. This disease is caused by a homozygous mutation of PSMB8 gene, which encodes immunoproteasome subunit β5i. Phenotypes of NNS patients are periodic fever, pernio-like rash, nodular erythema-like eruptions, and lipomuscular dystrophy, especially in the upper body, leading to the characteristic long, clubbed fingers. NNS was considered to be endemic to the Kansai area of Japan, but patients with similar phenotypes and the mutation of PSMB8 gene were reported in other countries, and named Chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (CANDLE) syndrome and joint contractures, muscular atrophy, microcytic anemia, and panniculitis-associated lipodystrophy (JMP) syndrome. These syndromes are now called proteasome-associated autoinflammatory syndromes (PRAASs), and their main pathophysiological mechanism seems to be interferonopathy. In this review, the history, characteristics, and the pathophysiological mechanism of PRAASs will be discussed, focusing mainly on NNS.
Collapse
Affiliation(s)
- Koichiro Ohmura
- Department of Rheumatology and Clinical Immunology, Kyoto University Graduate School of Medicine, Kyoto606-8507, Japan
| |
Collapse
|
75
|
Jamilloux Y, El Jammal T, Vuitton L, Gerfaud-Valentin M, Kerever S, Sève P. JAK inhibitors for the treatment of autoimmune and inflammatory diseases. Autoimmun Rev 2019; 18:102390. [PMID: 31520803 DOI: 10.1016/j.autrev.2019.102390] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 02/07/2023]
Abstract
Cytokines play a central role in the pathophysiology of autoimmune and inflammatory diseases. Several cytokines signal through the JAK-STAT pathway, which is now recognized as a major target to inhibit the effect of a wide array of cytokines. JAK inhibitors are increasingly used in the setting of inflammatory and autoimmune diseases. While the currently approved drugs are panJAK inhibitors, more selective small molecules are being developed and tested in various rheumatic disorders. In this extensive review, we present evidence- or hypothesis-based perspectives for these drugs in various rheumatologic conditions, such as rheumatoid arthritis, systemic lupus erythematosus, giant cell arteritis, and autoinflammatory diseases.
Collapse
Affiliation(s)
- Yvan Jamilloux
- Department of Internal Medicine, Lyon University Hospital, Lyon, France.
| | - Thomas El Jammal
- Department of Internal Medicine, Lyon University Hospital, Lyon, France
| | - Lucine Vuitton
- Department of Gastroenterology, Besancon University Hospital, Besancon, France
| | | | - Sébastien Kerever
- Department of Anesthesiology and Critical Care, Lariboisière University Hospital, AP-HP, ECSTRA Team, CRESS, Epidemiology and Statistics Center, Sorbonne Paris Cité, UMR 1153, INSERM, University Denis Diderot - Paris VII, Paris, France
| | - Pascal Sève
- Department of Internal Medicine, Lyon University Hospital, Lyon, France
| |
Collapse
|
76
|
Abstract
The introduction of biologics 20 years ago has significantly changed the pharmacotherapy of juvenile idiopathic arthritis. Biologics are among the most successful innovations, not only in rheumatology. In addition to their strong efficacy the rapid onset of improvement , biologics offer an option for prevention of long-term damage and a realistic prospect of remission. This article discusses innovations in biologics and the importance of the latest group for targeted therapy with small molecules in pediatric rheumatology.
Collapse
Affiliation(s)
- Gerd Horneff
- Zentrum für Allgemein Pädiatrie und Neonatologie, Asklepios Klinik Sankt Augustin, 53757, Sankt Augustin, Deutschland.
| |
Collapse
|
77
|
Targeting IFNGR/IL6R or downstream JAK1/JAK2 to control GvHD. Oncotarget 2018; 9:35721-35722. [PMID: 30515263 PMCID: PMC6254668 DOI: 10.18632/oncotarget.26291] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 10/26/2018] [Indexed: 12/30/2022] Open
|
78
|
The classification, genetic diagnosis and modelling of monogenic autoinflammatory disorders. Clin Sci (Lond) 2018; 132:1901-1924. [PMID: 30185613 PMCID: PMC6123071 DOI: 10.1042/cs20171498] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/30/2018] [Accepted: 08/07/2018] [Indexed: 12/13/2022]
Abstract
Monogenic autoinflammatory disorders are an increasingly heterogeneous group of conditions characterised by innate immune dysregulation. Improved genetic sequencing in recent years has led not only to the discovery of a plethora of conditions considered to be 'autoinflammatory', but also the broadening of the clinical and immunological phenotypic spectra seen in these disorders. This review outlines the classification strategies that have been employed for monogenic autoinflammatory disorders to date, including the primary innate immune pathway or the dominant cytokine implicated in disease pathogenesis, and highlights some of the advantages of these models. Furthermore, the use of the term 'autoinflammatory' is discussed in relation to disorders that cross the innate and adaptive immune divide. The utilisation of next-generation sequencing (NGS) in this population is examined, as are potential in vivo and in vitro methods of modelling to determine pathogenicity of novel genetic findings. Finally, areas where our understanding can be improved are highlighted, such as phenotypic variability and genotype-phenotype correlations, with the aim of identifying areas of future research.
Collapse
|
79
|
Sanchez GAM, Reinhardt A, Ramsey S, Wittkowski H, Hashkes PJ, Berkun Y, Schalm S, Murias S, Dare JA, Brown D, Stone DL, Gao L, Klausmeier T, Foell D, de Jesus AA, Chapelle DC, Kim H, Dill S, Colbert RA, Failla L, Kost B, O'Brien M, Reynolds JC, Folio LR, Calvo KR, Paul SM, Weir N, Brofferio A, Soldatos A, Biancotto A, Cowen EW, Digiovanna JJ, Gadina M, Lipton AJ, Hadigan C, Holland SM, Fontana J, Alawad AS, Brown RJ, Rother KI, Heller T, Brooks KM, Kumar P, Brooks SR, Waldman M, Singh HK, Nickeleit V, Silk M, Prakash A, Janes JM, Ozen S, Wakim PG, Brogan PA, Macias WL, Goldbach-Mansky R. JAK1/2 inhibition with baricitinib in the treatment of autoinflammatory interferonopathies. J Clin Invest 2018. [PMID: 29649002 DOI: 10.1172/jci98814)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Monogenic IFN-mediated autoinflammatory diseases present in infancy with systemic inflammation, an IFN response gene signature, inflammatory organ damage, and high mortality. We used the JAK inhibitor baricitinib, with IFN-blocking activity in vitro, to ameliorate disease. METHODS Between October 2011 and February 2017, 10 patients with CANDLE (chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperatures), 4 patients with SAVI (stimulator of IFN genes-associated [STING-associated] vasculopathy with onset in infancy), and 4 patients with other interferonopathies were enrolled in an expanded access program. The patients underwent dose escalation, and the benefit was assessed by reductions in daily disease symptoms and corticosteroid requirement. Quality of life, organ inflammation, changes in IFN-induced biomarkers, and safety were longitudinally assessed. RESULTS Eighteen patients were treated for a mean duration of 3.0 years (1.5-4.9 years). The median daily symptom score decreased from 1.3 (interquartile range [IQR], 0.93-1.78) to 0.25 (IQR, 0.1-0.63) (P < 0.0001). In 14 patients receiving corticosteroids at baseline, daily prednisone doses decreased from 0.44 mg/kg/day (IQR, 0.31-1.09) to 0.11 mg/kg/day (IQR, 0.02-0.24) (P < 0.01), and 5 of 10 patients with CANDLE achieved lasting clinical remission. The patients' quality of life and height and bone mineral density Z-scores significantly improved, and their IFN biomarkers decreased. Three patients, two of whom had genetically undefined conditions, discontinued treatment because of lack of efficacy, and one CANDLE patient discontinued treatment because of BK viremia and azotemia. The most common adverse events were upper respiratory infections, gastroenteritis, and BK viruria and viremia. CONCLUSION Upon baricitinib treatment, clinical manifestations and inflammatory and IFN biomarkers improved in patients with the monogenic interferonopathies CANDLE, SAVI, and other interferonopathies. Monitoring safety and efficacy is important in benefit-risk assessment. TRIAL REGISTRATION ClinicalTrials.gov NCT01724580 and NCT02974595. FUNDING This research was supported by the Intramural Research Program of the NIH, NIAID, and NIAMS. Baricitinib was provided by Eli Lilly and Company, which is the sponsor of the expanded access program for this drug.
Collapse
Affiliation(s)
- Gina A Montealegre Sanchez
- Translational Autoinflammatory Disease Section, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Adam Reinhardt
- Faculty of Physicians of the University of Nebraska Medical Center, College of Medicine, Omaha, Nebraska, USA
| | | | - Helmut Wittkowski
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | | | - Yackov Berkun
- Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | | | - Jason A Dare
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Diane Brown
- Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Deborah L Stone
- National Human Genome Research Institute, NIH, Bethesda, Maryland, USA
| | - Ling Gao
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Adriana A de Jesus
- Translational Autoinflammatory Disease Section, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Dawn C Chapelle
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Hanna Kim
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Samantha Dill
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Robert A Colbert
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Laura Failla
- Translational Autoinflammatory Disease Section, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Bahar Kost
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Michelle O'Brien
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | | | - Les R Folio
- Clinical Center, NIH, Bethesda, Maryland, USA
| | | | | | - Nargues Weir
- National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | | | - Ariane Soldatos
- National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, Maryland, USA
| | - Angelique Biancotto
- National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Edward W Cowen
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | | | - Massimo Gadina
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Andrew J Lipton
- Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | | | | | - Joseph Fontana
- National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Ahmad S Alawad
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Rebecca J Brown
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Kristina I Rother
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Theo Heller
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | | | - Parag Kumar
- Clinical Center, NIH, Bethesda, Maryland, USA
| | - Stephen R Brooks
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Meryl Waldman
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Harsharan K Singh
- University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Volker Nickeleit
- University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Maria Silk
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | | | - Seza Ozen
- Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Paul G Wakim
- Biostatistics and Clinical Epidemiology Service, NIH Clinical Center, Bethesda, Maryland, USA
| | - Paul A Brogan
- University College London (UCL) Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Foundation, London, United Kingdom
| | | | - Raphaela Goldbach-Mansky
- Translational Autoinflammatory Disease Section, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| |
Collapse
|
80
|
Sanchez GAM, Reinhardt A, Ramsey S, Wittkowski H, Hashkes PJ, Berkun Y, Schalm S, Murias S, Dare JA, Brown D, Stone DL, Gao L, Klausmeier T, Foell D, de Jesus AA, Chapelle DC, Kim H, Dill S, Colbert RA, Failla L, Kost B, O'Brien M, Reynolds JC, Folio LR, Calvo KR, Paul SM, Weir N, Brofferio A, Soldatos A, Biancotto A, Cowen EW, Digiovanna JJ, Gadina M, Lipton AJ, Hadigan C, Holland SM, Fontana J, Alawad AS, Brown RJ, Rother KI, Heller T, Brooks KM, Kumar P, Brooks SR, Waldman M, Singh HK, Nickeleit V, Silk M, Prakash A, Janes JM, Ozen S, Wakim PG, Brogan PA, Macias WL, Goldbach-Mansky R. JAK1/2 inhibition with baricitinib in the treatment of autoinflammatory interferonopathies. J Clin Invest 2018; 128:3041-3052. [PMID: 29649002 PMCID: PMC6026004 DOI: 10.1172/jci98814] [Citation(s) in RCA: 348] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 04/04/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND. Monogenic IFN–mediated autoinflammatory diseases present in infancy with systemic inflammation, an IFN response gene signature, inflammatory organ damage, and high mortality. We used the JAK inhibitor baricitinib, with IFN-blocking activity in vitro, to ameliorate disease. METHODS. Between October 2011 and February 2017, 10 patients with CANDLE (chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperatures), 4 patients with SAVI (stimulator of IFN genes–associated [STING-associated] vasculopathy with onset in infancy), and 4 patients with other interferonopathies were enrolled in an expanded access program. The patients underwent dose escalation, and the benefit was assessed by reductions in daily disease symptoms and corticosteroid requirement. Quality of life, organ inflammation, changes in IFN-induced biomarkers, and safety were longitudinally assessed. RESULTS. Eighteen patients were treated for a mean duration of 3.0 years (1.5–4.9 years). The median daily symptom score decreased from 1.3 (interquartile range [IQR], 0.93–1.78) to 0.25 (IQR, 0.1–0.63) (P < 0.0001). In 14 patients receiving corticosteroids at baseline, daily prednisone doses decreased from 0.44 mg/kg/day (IQR, 0.31–1.09) to 0.11 mg/kg/day (IQR, 0.02–0.24) (P < 0.01), and 5 of 10 patients with CANDLE achieved lasting clinical remission. The patients’ quality of life and height and bone mineral density Z-scores significantly improved, and their IFN biomarkers decreased. Three patients, two of whom had genetically undefined conditions, discontinued treatment because of lack of efficacy, and one CANDLE patient discontinued treatment because of BK viremia and azotemia. The most common adverse events were upper respiratory infections, gastroenteritis, and BK viruria and viremia. CONCLUSION. Upon baricitinib treatment, clinical manifestations and inflammatory and IFN biomarkers improved in patients with the monogenic interferonopathies CANDLE, SAVI, and other interferonopathies. Monitoring safety and efficacy is important in benefit-risk assessment. TRIAL REGISTRATION. ClinicalTrials.gov NCT01724580 and NCT02974595. FUNDING. This research was supported by the Intramural Research Program of the NIH, NIAID, and NIAMS. Baricitinib was provided by Eli Lilly and Company, which is the sponsor of the expanded access program for this drug.
Collapse
Affiliation(s)
- Gina A Montealegre Sanchez
- Translational Autoinflammatory Disease Section, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Adam Reinhardt
- Faculty of Physicians of the University of Nebraska Medical Center, College of Medicine, Omaha, Nebraska, USA
| | | | - Helmut Wittkowski
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | | | - Yackov Berkun
- Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | | | - Jason A Dare
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Diane Brown
- Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Deborah L Stone
- National Human Genome Research Institute, NIH, Bethesda, Maryland, USA
| | - Ling Gao
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | - Dirk Foell
- Department of Pediatric Rheumatology and Immunology, University Children's Hospital, Muenster, Germany
| | - Adriana A de Jesus
- Translational Autoinflammatory Disease Section, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Dawn C Chapelle
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Hanna Kim
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Samantha Dill
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Robert A Colbert
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Laura Failla
- Translational Autoinflammatory Disease Section, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| | - Bahar Kost
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Michelle O'Brien
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | | | - Les R Folio
- Clinical Center, NIH, Bethesda, Maryland, USA
| | | | | | - Nargues Weir
- National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | | | - Ariane Soldatos
- National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, Maryland, USA
| | - Angelique Biancotto
- National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Edward W Cowen
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | | | - Massimo Gadina
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Andrew J Lipton
- Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | | | | | - Joseph Fontana
- National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Ahmad S Alawad
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Rebecca J Brown
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Kristina I Rother
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Theo Heller
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | | | - Parag Kumar
- Clinical Center, NIH, Bethesda, Maryland, USA
| | - Stephen R Brooks
- National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Meryl Waldman
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Harsharan K Singh
- University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Volker Nickeleit
- University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | - Maria Silk
- Eli Lilly and Company, Indianapolis, Indiana, USA
| | | | | | - Seza Ozen
- Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Paul G Wakim
- Biostatistics and Clinical Epidemiology Service, NIH Clinical Center, Bethesda, Maryland, USA
| | - Paul A Brogan
- University College London (UCL) Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Foundation, London, United Kingdom
| | | | - Raphaela Goldbach-Mansky
- Translational Autoinflammatory Disease Section, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, Maryland, USA
| |
Collapse
|
81
|
Abstract
Abstract: Autoinflammatory diseases are disorders of the innate immune system characterized by recurrent systematic inflammation and serious complications. Dysregulation of inflammasome and overproduction of interleukin-1 play a major role in the pathogenesis of autoinflammatory diseases. The diagnosis of these rare conditions rely on recognising the pattern of presentation and differential diagnosis. Manifestations may include fever, rash, serositis (pleuritis and peritonitis), arthritis, meningitis and uveitis. Secondary amyloidosis may complicate longstanding disease. Advances in our understanding of the molecular and pathophysiological basis of the autoinflammatory diseases have resulted in new treatment strategies. Early diagnosis and effective therapy are critical to prevent irreversible organ damage. The purpose of this review is to describe the major clinical, genetic, and therapeutic features of the most common autoinflammatory syndromes. Orv Hetil. 2018; 159(23): 898–907.
Collapse
Affiliation(s)
- Bernadett Mosdósi
- Klinikai Központ, Gyermekgyógyászati Klinika, Pécsi Tudományegyetem, Általános Orvostudományi Kar Pécs, József A. u. 7., 7622
| | - Beáta Tóth
- Laboratóriumi Medicina Intézet, Debreceni Egyetem, Általános Orvostudományi Kar Debrecen
| |
Collapse
|
82
|
Davidson S, Steiner A, Harapas CR, Masters SL. An Update on Autoinflammatory Diseases: Interferonopathies. Curr Rheumatol Rep 2018; 20:38. [PMID: 29846818 DOI: 10.1007/s11926-018-0748-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Type I interferons (IFNαβ) induce the expression of hundreds of genes; thus, it is unsurprising that the initiation, transmission, and resolution of the IFNαβ-mediated immune response is tightly controlled. Mutations that alter nucleic acid processing and recognition, ablate IFNαβ-specific negative feedback mechanisms, or result in dysfunction of the proteasome system can all induce pathogenic IFNαβ signalling and are the focus of this review. RECENT FINDINGS Recent advances have delineated the precise cytoplasmic mechanisms that facilitate self-DNA to be recognised by cGAS and self-RNA to be recognised by RIG-I or MDA-5. This helps clarify interferonopathies associated with mutations in genes which code for DNase-II and ADAR1, among others. Similarly, loss of function mutations in Pol α, which lowers the presence of antagonistic ligands in the cytosol, or gain of function mutations in RIG-I and MDA-5, result in increased propensity for receptor activation and therefore IFNαβ induction. As the aetiology of monogenic autoinflammatory diseases are uncovered, novel and sometimes unsuspected molecular interactions and signalling pathways are being defined. This review covers developments that have come to light over the past 3 years, with reference to the study of interferonopathies.
Collapse
Affiliation(s)
- Sophia Davidson
- Inflammation division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia.
| | - Annemarie Steiner
- Inflammation division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Cassandra R Harapas
- Inflammation division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia
| | - Seth L Masters
- Inflammation division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia. .,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
83
|
Davidson S, Steiner A, Harapas CR, Masters SL. An Update on Autoinflammatory Diseases: Interferonopathies. Curr Rheumatol Rep 2018. [PMID: 29846818 DOI: 10.1007/s11926-018-0748-y)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
PURPOSE OF REVIEW Type I interferons (IFNαβ) induce the expression of hundreds of genes; thus, it is unsurprising that the initiation, transmission, and resolution of the IFNαβ-mediated immune response is tightly controlled. Mutations that alter nucleic acid processing and recognition, ablate IFNαβ-specific negative feedback mechanisms, or result in dysfunction of the proteasome system can all induce pathogenic IFNαβ signalling and are the focus of this review. RECENT FINDINGS Recent advances have delineated the precise cytoplasmic mechanisms that facilitate self-DNA to be recognised by cGAS and self-RNA to be recognised by RIG-I or MDA-5. This helps clarify interferonopathies associated with mutations in genes which code for DNase-II and ADAR1, among others. Similarly, loss of function mutations in Pol α, which lowers the presence of antagonistic ligands in the cytosol, or gain of function mutations in RIG-I and MDA-5, result in increased propensity for receptor activation and therefore IFNαβ induction. As the aetiology of monogenic autoinflammatory diseases are uncovered, novel and sometimes unsuspected molecular interactions and signalling pathways are being defined. This review covers developments that have come to light over the past 3 years, with reference to the study of interferonopathies.
Collapse
Affiliation(s)
- Sophia Davidson
- Inflammation division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia.
| | - Annemarie Steiner
- Inflammation division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Cassandra R Harapas
- Inflammation division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia
| | - Seth L Masters
- Inflammation division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052, Australia. .,Department of Medical Biology, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
84
|
Abstract
PURPOSE OF REVIEW To review recent scientific advances and therapeutic approaches in the expanding field of type I interferonopathies. Type I interferonopathies represent a genetically and phenotypically heterogenous group of disorders of the innate immune system caused by constitutive activation of antiviral type I interferon (IFN). Clinically, type I interferonopathies are characterized by autoinflammation and varying degrees of autoimmunity or immunodeficiency. The elucidation of the underlying genetic causes has revealed novel cell-intrinsic mechanisms that protect the organism against inappropriate immune recognition of self nucleic acids by cytosolic nucleic acid sensors. The type I IFN system is subject to a tight and complex regulation. Disturbances of its checks and balances can spark an unwanted immune response causing uncontrolled type I IFN signaling. Novel mechanistic insight into pathways that control the type I IFN system is providing opportunities for targeted therapeutic approaches by repurposing drugs such as Janus kinase inhibitors or reverse transcriptase inhibitors.
Collapse
|
85
|
Affiliation(s)
- Dae Chul Jeong
- Division of Pediatric Rheumatology and Clinical Immunology, Department of Pediatrics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|