1
|
Freedman MS, Wojcik J, Holmberg KH, Fluck M, D'Antonio M, Hyvert Y, Stinchi S, D'Urso V, Dangond F. Pharmacodynamic biomarkers of long-term interferon beta-1a therapy in REFLEX and REFLEXION. J Neuroimmunol 2021; 360:577715. [PMID: 34536787 DOI: 10.1016/j.jneuroim.2021.577715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/16/2021] [Accepted: 09/04/2021] [Indexed: 01/04/2023]
Abstract
This post-hoc analysis evaluated candidate biomarkers of long-term efficacy of subcutaneous interferon beta-1a (sc IFN β-1a) in REFLEX/REFLEXION studies of clinically isolated syndrome. Samples from 507 REFLEX and 287 REFLEXION study participants were analyzed. All investigated biomarkers were significantly upregulated 1.5-4-fold in response to sc IFN β-1a treatment versus baseline (p ≤ 0.008). The validity of MX1, 2'5'OAS, and IL-1RA as biomarkers of response to sc IFN β-1a was confirmed in this large patient cohort, with biomarkers consistently upregulated in a dose-dependent manner. Neopterin, TRAIL, and IP-10 were confirmed as biomarkers associated with long-term sc IFN β-1a treatment efficacy over 5 years.
Collapse
Affiliation(s)
- Mark S Freedman
- University of Ottawa and the Ottawa Hospital Research Institute, Ottawa, ON, Canada.
| | | | | | | | - Mauro D'Antonio
- Merck R.B.M S.p.A, Instituto Di Ricerche Biomediche 'Antoine Marxer', Colleretto Giacosa, Turin, Italy
| | | | | | | | - Fernando Dangond
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA
| |
Collapse
|
2
|
Jakimovski D, Kolb C, Ramanathan M, Zivadinov R, Weinstock-Guttman B. Interferon β for Multiple Sclerosis. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a032003. [PMID: 29311124 DOI: 10.1101/cshperspect.a032003] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite that the availability of new therapeutic options has expanded the multiple sclerosis (MS) disease-modifying therapy arsenal, interferon β (IFN-β) remains an important therapy option in the current decision-making process. This review will summarize the present knowledge of IFN-β mechanism of action, the overall safety, and the short- and long-term efficacy of its use in relapsing remitting MS and clinically isolated syndromes. Data on secondary progressive MS is also provided, although no clear benefit was identified.
Collapse
Affiliation(s)
- Dejan Jakimovski
- Buffalo Neuroimaging Analysis Center, Department of Neurology, University at Buffalo, State University of New York, Buffalo, New York 14203
| | - Channa Kolb
- Jacobs MS Center, Department of Neurology, University at Buffalo, State University of New York, Buffalo, New York 14202
| | - Murali Ramanathan
- Jacobs MS Center, Department of Neurology, University at Buffalo, State University of New York, Buffalo, New York 14202.,Department of Pharmaceutical Sciences, School of Medicine and Biomedical Sciences, State University of New York, Buffalo, New York 14214
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, University at Buffalo, State University of New York, Buffalo, New York 14203.,MR Imaging Clinical Translational Research Center, School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, New York 14203
| | - Bianca Weinstock-Guttman
- Jacobs MS Center, Department of Neurology, University at Buffalo, State University of New York, Buffalo, New York 14202
| |
Collapse
|
3
|
Zettl UK, Hecker M, Aktas O, Wagner T, Rommer PS. Interferon β-1a and β-1b for patients with multiple sclerosis: updates to current knowledge. Expert Rev Clin Immunol 2018; 14:137-153. [DOI: 10.1080/1744666x.2018.1426462] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Uwe Klaus Zettl
- Department of Neurology, Neuroimmunology Section, University of Rostock, Rostock, Germany
| | - Michael Hecker
- Department of Neurology, Neuroimmunology Section, University of Rostock, Rostock, Germany
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Torsten Wagner
- Department of Medical Affairs, Merck KGaA, Darmstadt, Germany
| | - Paulus S. Rommer
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4
|
Dwyer MG, Silva D, Bergsland N, Horakova D, Ramasamy D, Durfee J, Vaneckova M, Havrdova E, Zivadinov R. Neurological software tool for reliable atrophy measurement (NeuroSTREAM) of the lateral ventricles on clinical-quality T2-FLAIR MRI scans in multiple sclerosis. NEUROIMAGE-CLINICAL 2017; 15:769-779. [PMID: 28706852 PMCID: PMC5496213 DOI: 10.1016/j.nicl.2017.06.022] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/19/2017] [Accepted: 06/16/2017] [Indexed: 11/18/2022]
Abstract
Background There is a need for a brain volume measure applicable to the clinical routine scans. Nearly every multiple sclerosis (MS) protocol includes low-resolution 2D T2-FLAIR imaging. Objectives To develop and validate cross-sectional and longitudinal brain atrophy measures on clinical-quality T2-FLAIR images in MS patients. Methods A real-world dataset from 109 MS patients from 62 MRI scanners was used to develop a lateral ventricular volume (LVV) algorithm with a longitudinal Jacobian-based extension, called NeuroSTREAM. Gold-standard LVV was calculated on high-resolution T1 1 mm, while NeuroSTREAM LVV was obtained on low-resolution T2-FLAIR 3 mm thick images. Scan-rescan reliability was assessed in 5 subjects. The variability of LVV measurement at different field strengths was tested in 76 healthy controls and 125 MS patients who obtained both 1.5T and 3T scans in 72 hours. Clinical validation of algorithm was performed in 176 MS patients who obtained serial yearly MRI 1.5T scans for 10 years. Results Correlation between gold-standard high-resolution T1 LVV and low-resolution T2-FLAIR LVV was r = 0.99, p < 0.001 and the scan-rescan coefficient of variation was 0.84%. Correlation between low-resolution T2-FLAIR LVV on 1.5T and 3T was r = 0.99, p < 0.001 and the scan-rescan coefficient of variation was 2.69% cross-sectionally and 2.08% via Jacobian integration. NeuroSTREAM showed comparable effect size (d = 0.39–0.71) in separating MS patients with and without confirmed disability progression, compared to SIENA and VIENA. Conclusions Brain atrophy measurement on clinical quality T2-FLAIR scans is feasible, accurate, reliable, and relates to clinical outcomes. A robust algorithm for measuring lateral ventricular volume on clinical FLAIR scans is proposed. The algorithm combines multi-atlas joint fusion labeling with level-set smoothness-constraining refinement. Results show a similar relationship to disability progression as with established metrics on high-resolution scans.
Collapse
Affiliation(s)
- Michael G Dwyer
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.
| | | | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; Magnetic Resonance Laboratory, IRCCS Don Gnocchi Foundation, Milan, Italy
| | - Dana Horakova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Deepa Ramasamy
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Jaqueline Durfee
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Manuela Vaneckova
- Department of Radiology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Eva Havrdova
- Department of Neurology and Center of Clinical Neuroscience, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA; MR Imaging Clinical Translational Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
5
|
Uher T, Fellows K, Horakova D, Zivadinov R, Vaneckova M, Sobisek L, Tyblova M, Seidl Z, Krasensky J, Bergsland N, Weinstock-Guttman B, Havrdova E, Ramanathan M. Serum lipid profile changes predict neurodegeneration in interferon-β1a-treated multiple sclerosis patients. J Lipid Res 2016; 58:403-411. [PMID: 27923871 DOI: 10.1194/jlr.m072751] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/01/2016] [Indexed: 12/20/2022] Open
Abstract
The purpose of this work was to determine whether changes in cholesterol profiles after interferon-β (IFN-β)1a treatment initiation following the first demyelinating event suggestive of multiple sclerosis are associated with clinical and MRI outcomes over 4 years. A group of 131 patients (age: 27.9 ± 7.8 years, 63% female) with serial 3-monthly clinical and 12-monthly MRI follow-ups over 4 years were investigated. Serum cholesterol profiles, including total cholesterol (TC), HDL cholesterol (HDL-C), and LDL cholesterol (LDL-C) were obtained at baseline, 1 month, 3 months, and every 6 months thereafter. IFN-β1a initiation caused rapid decreases in serum HDL-C, LDL-C, and TC within 1 month of IFN-β1a initiation (all P < 0.001) that returned slowly toward baseline. In predictive mixed model analyses, greater percent decreases in HDL-C after 3 months of IFN-β1a treatment initiation were associated with less brain atrophy over the 4 year time course, as assessed by percent brain volume change (P < 0.001), percent gray matter volume change (P < 0.001), and percent lateral ventricle volume change (P = 0.005). Decreases in cholesterol biomarkers following IFN-β1a treatment are associated with brain atrophy outcomes over 4 years. Pharmacological interventions targeting lipid homeostasis may be clinically beneficial for disrupting neurodegenerative processes.
Collapse
Affiliation(s)
- Tomas Uher
- Department of Neurology and Center of Clinical Neuroscience Charles University in Prague, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Kelly Fellows
- Department of Pharmaceutical Sciences, State University of New York, Buffalo, NY
| | - Dana Horakova
- Department of Neurology and Center of Clinical Neuroscience Charles University in Prague, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Robert Zivadinov
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY.,MR Imaging Clinical Translational Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| | - Manuela Vaneckova
- Department of Radiology, Charles University in Prague, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Lukas Sobisek
- Department of Statistics and Probability, University of Economics in Prague, Czech Republic
| | - Michaela Tyblova
- Department of Neurology and Center of Clinical Neuroscience Charles University in Prague, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Zdenek Seidl
- Department of Radiology, Charles University in Prague, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Jan Krasensky
- Department of Radiology, Charles University in Prague, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) "S.Maria Nascente", Don Gnocchi Foundation, Milan, Italy
| | - Bianca Weinstock-Guttman
- Jacobs Multiple Sclerosis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| | - Eva Havrdova
- Department of Neurology and Center of Clinical Neuroscience Charles University in Prague, First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Murali Ramanathan
- Department of Pharmaceutical Sciences, State University of New York, Buffalo, NY .,Jacobs Multiple Sclerosis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| |
Collapse
|
6
|
Ruparelia N, Chai JT, Fisher EA, Choudhury RP. Inflammatory processes in cardiovascular disease: a route to targeted therapies. Nat Rev Cardiol 2016; 14:133-144. [PMID: 27905474 DOI: 10.1038/nrcardio.2016.185] [Citation(s) in RCA: 345] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Inflammatory processes are firmly established as central to the development and complications of cardiovascular diseases. Elevated levels of inflammatory markers have been shown to be predictive of future cardiovascular events. The specific targeting of these processes in experimental models has been shown to attenuate myocardial and arterial injury, reduce disease progression, and promote healing. However, the translation of these observations and the demonstration of clear efficacy in clinical practice have been disappointing. A major limitation might be that tools currently used to measure 'inflammation' are insufficiently precise and do not provide information about disease site and activity, or discriminate between functionally important activation pathways. The challenge, therefore, is to make measures of inflammation that are more meaningful, and which can guide specific targeted therapies. In this Review, we consider the roles of inflammatory processes in the related pathologies of atherosclerosis and acute myocardial infarction, by providing an evaluation of the known and emerging inflammatory pathways. We highlight contemporary techniques to characterize and quantify inflammation, and consider how they might be used to guide specific treatments. Finally, we discuss emerging opportunities in the field, including their current limitations and challenges that are the focus of ongoing study.
Collapse
Affiliation(s)
- Neil Ruparelia
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, OX3 9DU, UK
| | - Joshua T Chai
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, OX3 9DU, UK.,Acute Vascular Imaging Centre, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK
| | - Edward A Fisher
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, OX3 9DU, UK.,The Center for the Prevention of Cardiovascular Disease and the Division of Cardiology, Department of Medicine, New York University School of Medicine, New York, New York 10016, USA
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford, OX3 9DU, UK.,Acute Vascular Imaging Centre, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford, OX3 9DU, UK
| |
Collapse
|
7
|
Ruparelia N, Godec J, Lee R, Chai JT, Dall'Armellina E, McAndrew D, Digby JE, Forfar JC, Prendergast BD, Kharbanda RK, Banning AP, Neubauer S, Lygate CA, Channon KM, Haining NW, Choudhury RP. Acute myocardial infarction activates distinct inflammation and proliferation pathways in circulating monocytes, prior to recruitment, and identified through conserved transcriptional responses in mice and humans. Eur Heart J 2015; 36:1923-34. [PMID: 25982896 PMCID: PMC4571177 DOI: 10.1093/eurheartj/ehv195] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 04/14/2015] [Accepted: 04/29/2015] [Indexed: 11/13/2022] Open
Abstract
AIMS Monocytes play critical roles in tissue injury and repair following acute myocardial infarction (AMI). Specifically targeting inflammatory monocytes in experimental models leads to reduced infarct size and improved healing. However, data from humans are sparse, and it remains unclear whether monocytes play an equally important role in humans. The aim of this study was to investigate whether the monocyte response following AMI is conserved between humans and mice and interrogate patterns of gene expression to identify regulated functions. METHODS AND RESULTS Thirty patients (AMI) and 24 control patients (stable coronary atherosclerosis) were enrolled. Female C57BL/6J mice (n = 6/group) underwent AMI by surgical coronary ligation. Myocardial injury was quantified by magnetic resonance imaging (human) and echocardiography (mice). Peripheral monocytes were isolated at presentation and at 48 h. RNA from separated monocytes was hybridized to Illumina beadchips. Acute myocardial infarction resulted in a significant peripheral monocytosis in both species that positively correlated with the extent of myocardial injury. Analysis of the monocyte transcriptome following AMI demonstrated significant conservation and identified inflammation and mitosis as central processes to this response. These findings were validated in both species. CONCLUSIONS Our findings show that the monocyte transcriptome is conserved between mice and humans following AMI. Patterns of gene expression associated with inflammation and proliferation appear to be switched on prior to their infiltration of injured myocardium suggesting that the specific targeting of inflammatory and proliferative processes in these immune cells in humans are possible therapeutic strategies. Importantly, they could be effective in the hours after AMI.
Collapse
Affiliation(s)
- Neil Ruparelia
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford OX3 9DU, UK Oxford Heart Centre, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK
| | - Jernej Godec
- Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| | - Regent Lee
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford OX3 9DU, UK
| | - Joshua T Chai
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford OX3 9DU, UK
| | - Erica Dall'Armellina
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford OX3 9DU, UK Acute Vascular Imaging Centre, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford OX3 9DU, UK
| | - Debra McAndrew
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford OX3 9DU, UK
| | - Janet E Digby
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford OX3 9DU, UK
| | - J Colin Forfar
- Oxford Heart Centre, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK
| | | | - Rajesh K Kharbanda
- Oxford Heart Centre, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK
| | - Adrian P Banning
- Oxford Heart Centre, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford OX3 9DU, UK
| | - Craig A Lygate
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford OX3 9DU, UK
| | - Keith M Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford OX3 9DU, UK Oxford Heart Centre, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK
| | - Nicholas W Haining
- Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| | - Robin P Choudhury
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford OX3 9DU, UK Oxford Heart Centre, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK Acute Vascular Imaging Centre, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Headley Way, Oxford OX3 9DU, UK
| |
Collapse
|
8
|
Interferon-β1a modulates glutamate neurotransmission in the CNS through CaMKII and GluN2A-containing NMDA receptors. Neuropharmacology 2015; 100:98-105. [PMID: 26116817 DOI: 10.1016/j.neuropharm.2015.06.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 05/28/2015] [Accepted: 06/17/2015] [Indexed: 12/16/2022]
Abstract
Interferons (IFNs) are widely expressed cytokines with antiviral and immune-modulating effects and have been utilised for the treatment of several human pathological conditions. In particular, the immune-modulatory drug IFN-β is utilized in the treatment of multiple sclerosis (MS), a chronic autoimmune and neurodegenerative disorder of the central nervous system (CNS). Although the effects of IFN-β on immune cells functions have been widely investigated, information about the ability of the drug to modulate neuronal transmission in the CNS is still largely lacking. The aim of this study was to investigate the ability of IFN-β1a to modulate excitatory synaptic transmission in the CNS. Whole cell patch-clamp electrophysiological recordings were performed in the nucleus striatum, one of the CNS grey matter structures that is prone to degenerate during the course of MS. We demonstrate that the drug IFN-β1a, independently from its known peripheral immune-modulating action, is able to directly modulate synaptic transmission. In particular, we demonstrated that IFN-β1a reduces the amplitude of striatal excitatory post-synaptic currents, indicating an inhibitory effect on glutamate neurotransmission, and in particular on its NMDA component. The inhibitory effect of IFN-β1a on striatal glutamate neurotransmission was found to be mediated by a novel post-synaptic mechanism requiring Ca(2+), CaMKII and the GluN2A subunit of the NMDA receptor, without the involvement of the classic STAT1 pathway. The evidence of a novel neuro-modulating effect of IFN-β shed light on the mechanisms of action of the drug and on the complex bidirectional interaction occurring between the immune and the nervous system. This article is part of the Special Issue entitled 'Synaptopathy--from Biology to Therapy'.
Collapse
|
9
|
Giedraitiene N, Kaubrys G, Kizlaitiene R, Bagdonatė L, Griskevicius L, Valceckiene V, Stoskus M. Therapeutic Plasma Exchange in Multiple Sclerosis Patients with Abolished Interferon-beta Bioavailability. Med Sci Monit 2015; 21:1512-9. [PMID: 26009248 PMCID: PMC4456985 DOI: 10.12659/msm.894119] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 05/13/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Neutralizing antibodies (NAb) to interferon-beta (IFN-β) are associated with reduced bioactivity and efficacy of IFN-β in multiple sclerosis (MS). The myxovirus resistance protein A (MxA) gene expression is one of the most appropriate markers of biological activity of exogenous IFN-β. We hypothesized that therapeutic plasma exchange (TPE) can restore the ability of IFN-β to induce the MxA mRNA expression and that maintenance plasmapheresis can sustain the bioavailability of IFN-β. MATERIAL AND METHODS Eligible patients underwent 4 primary separate plasma exchange sessions. After the induction TPE sessions, they were transferred to maintenance plasmapheresis. Bioactivity of IFN-β was expressed as in vivo MxA mRNA induction in whole blood using RT-qPCR. RESULTS Six patients with low IFN-β bioavailability detected by the MxA mRNA response were included. Four patients became biological responders after induction plasmapheresis. In 2 patients an increase of MxA mRNA expression was found, but the values persisted below the cut-off and the patients remained as "poor biological responders". The effect of maintenance plasmapheresis was transient: MxA mRNA expression values reverted to the baseline levels after 1-2 months. CONCLUSIONS Therapeutic plasma exchange is able to restore the bioavailability of IFN-β in the majority of studied patients, but the effect of TPE on the IFN-β bioavailability was transient.
Collapse
Affiliation(s)
- Natasa Giedraitiene
- Clinic of Neurology and Neurosurgery, Faculty of Medicine, Vilnius University, Center of Neurology, Vilnius University Hospital Santariskiu Clinics, Vilnius, Lithuania
| | - Gintaras Kaubrys
- Clinic of Neurology and Neurosurgery, Faculty of Medicine, Vilnius University, Center of Neurology, Vilnius University Hospital Santariskiu Clinics, Vilnius, Lithuania
| | - Rasa Kizlaitiene
- Clinic of Neurology and Neurosurgery, Faculty of Medicine, Vilnius University, Center of Neurology, Vilnius University Hospital Santariskiu Clinics, Vilnius, Lithuania
| | - Loreta Bagdonatė
- Center for Laboratory Medicine, Vilnius University Hospital Santariskiu Clinics, Vilnius, Lithuania
| | - Laimonas Griskevicius
- Internal Medicine Clinic, Family Medicine and Oncology, Faculty of Medicine, Vilnius University, Center of Hematology, Oncology and Transfusion Medicine, Vilnius University Hospital Santariskiu Clinics, Vilnius, Lithuania
| | - Vilma Valceckiene
- Center for Hematology, Oncology and Transfusion Medicine, Vilnius University Hospital Santariskiu Clinics, Vilnius, Lithuania
| | - Mindaugas Stoskus
- Center for Hematology, Oncology and Transfusion Medicine, Vilnius University Hospital Santariskiu Clinics, Vilnius, Lithuania
| |
Collapse
|
10
|
Bertolotto A, Granieri L, Marnetto F, Valentino P, Sala A, Capobianco M, Malucchi S, Di Sapio A, Malentacchi M, Matta M, Caldano M. Biological monitoring of IFN-β therapy in Multiple Sclerosis. Cytokine Growth Factor Rev 2015; 26:241-8. [DOI: 10.1016/j.cytogfr.2014.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 12/09/2014] [Indexed: 11/26/2022]
|
11
|
Bertolotto A. Evaluation of the impact of neutralizing antibodies on IFNβ response. Clin Chim Acta 2015; 449:31-6. [PMID: 25769291 DOI: 10.1016/j.cca.2015.02.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 10/23/2022]
Abstract
IFNβ therapeutic action depends on a sequence of biological steps: i) the interaction between interferon beta (IFNβ) and its receptor (IFNAR) located at the cell surface of peripheral blood mononuclear cells; ii) activation of second messengers; iii) transcription of several genes containing specific ISRE regions (Interferon Stimulated Response Elements); and iv) synthesis of specific proteins. Although IFNβ therapy has improved treatment options of patients with multiple sclerosis (MS), the long-term efficacy of IFNβs can be compromised due to the development of neutralizing antibodies (NAbs). High titer NAbs develop in about 15% of patients; they abolish IFNβ biological activity and consequently the therapeutic action of IFNβ. Different IFNβ preparations carry different risks of developing NAbs, ranging from 3 to 28%. The risk of inducing NAbs must be considered in the selection of treatment. Guidelines for NAbs testing and the therapeutic decision in case of NAbs positivity have been established. NAbs positivity predicts MRI and clinical activity. Precocious identification of Nabs-positive patients and switch to alternative treatments can improve the percentage of responders and allow a better allocation of relevant economical resources.
Collapse
Affiliation(s)
- Antonio Bertolotto
- Neurologia 2-CRESM (Centro Riferimento Regionale Sclerosi Multipla), AOU San Luigi, Orbassano, Italy.
| |
Collapse
|
12
|
Interferon-β1a protects neurons against mitochondrial toxicity via modulation of STAT1 signaling: electrophysiological evidence. Neurobiol Dis 2013; 62:387-93. [PMID: 24135008 DOI: 10.1016/j.nbd.2013.09.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 09/09/2013] [Accepted: 09/24/2013] [Indexed: 11/22/2022] Open
Abstract
Multiple sclerosis, one of the main causes of non-traumatic neurological disability in young adults, is an inflammatory and neurodegenerative disorder of the central nervous system. Although the pathogenesis of neuroaxonal damage occurring during the course of the disease is still largely unknown, there is accumulating evidence highlighting the potential role of mitochondria in multiple sclerosis-associated neuronal degeneration. The aim of the present study was to investigate, by utilizing electrophysiological techniques in brain striatal slices, the potential protective effects of interferon-β1a, one of the most widely used medication for multiple sclerosis, against acute neuronal dysfunction induced by mitochondrial toxins. Interferon-β1a was found to exert a dose-dependent protective effect against the progressive loss of striatal field potential amplitude induced by the mitochondrial complex I inhibitor rotenone. Interferon-β1a also reduced the generation of the rotenone-induced inward current in striatal spiny neurons. Conversely, interferon-β1a did not influence the electrophysiological effects of the mitochondrial complex II inhibitor 3-nitropropionic acid. The protective effect of interferon-β1a against mitochondrial complex I inhibition was found to be dependent on the activation of STAT1 signaling. Conversely, endogenous dopamine depletion and the modulation of the p38 MAPK and mTOR pathways did not influence the effects of interferon-β1a. During experimental autoimmune encephalomyelitis (EAE) striatal rotenone toxicity was enhanced but the protective effect of interferon-β1a was still evident. These results support future studies investigating the role played by specific intracellular signaling pathways in mediating the potential link among inflammation, mitochondrial impairment and neuroaxonal degeneration in multiple sclerosis.
Collapse
|
13
|
Interferon-beta therapy in multiple sclerosis: the short-term and long-term effects on the patients' individual gene expression in peripheral blood. Mol Neurobiol 2013; 48:737-56. [PMID: 23636981 DOI: 10.1007/s12035-013-8463-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 04/16/2013] [Indexed: 01/17/2023]
Abstract
Therapy with interferon-beta (IFN-beta) is a mainstay in the management of relapsing-remitting multiple sclerosis (MS), with proven long-term effectiveness and safety. Much has been learned about the molecular mechanisms of action of IFN-beta in the past years. Previous studies described more than a hundred genes to be modulated in expression in blood cells in response to the therapy. However, for many of these genes, the precise temporal expression pattern and the therapeutic relevance are unclear. We used Affymetrix microarrays to investigate in more detail the gene expression changes in peripheral blood mononuclear cells from MS patients receiving subcutaneous IFN-beta-1a. The blood samples were obtained longitudinally at five different time points up to 2 years after the start of therapy, and the patients were clinically followed up for 5 years. We examined the functions of the genes that were upregulated or downregulated at the transcript level after short-term or long-term treatment. Moreover, we analyzed their mutual interactions and their regulation by transcription factors. Compared to pretreatment levels, 96 genes were identified as highly differentially expressed, many of them already after the first IFN-beta injection. The interactions between these genes form a large network with multiple feedback loops, indicating the complex crosstalk between innate and adaptive immune responses during therapy. We discuss the genes and biological processes that might be important to reduce disease activity by attenuating the proliferation of autoreactive immune cells and their migration into the central nervous system. In summary, we present novel insights that extend the current knowledge on the early and late pharmacodynamic effects of IFN-beta therapy and describe gene expression differences between the individual patients that reflect clinical heterogeneity.
Collapse
|
14
|
Bates S. The role of gene expression profiling in drug discovery. Curr Opin Pharmacol 2012; 11:549-56. [PMID: 21752712 DOI: 10.1016/j.coph.2011.06.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Revised: 06/19/2011] [Accepted: 06/21/2011] [Indexed: 12/23/2022]
Abstract
Monitoring gene expression through the dual approaches of transcriptomics (RNA profiling) and proteomics (protein profiling) has become a key component in our efforts to understand complex biological processes. From the molecular stratification of disease states and the selection of potential drug targets, to patient selection and the confirmation of engagement of pharmacology in clinical studies, we are seeing the impact of gene expression profiling across all phases of the drug discovery process. Ongoing technological advances have driven an expansion in the use of these techniques, demonstrated utility in preclinical and clinical settings and increased regulatory and clinical acceptance. As technologies continue to advance apace, gene expression profiling is likely to play an increasingly important role in the future development of drug discovery.
Collapse
Affiliation(s)
- Stewart Bates
- Biomarker Discovery, Biopharmaceutical R&D Unit, GlaxoSmithKline, Stevenage, SG1 2NY, Hertfordshire, UK.
| |
Collapse
|
15
|
Abstract
The development of neutralizing antibodies (NAbs) is a major problem in multiple sclerosis (MS) patients treated with interferon-beta (IFN-ß). Whereas binding antibodies (BAbs) can be demonstrated in the vast majority of patients, only a smaller proportion of patients develop NAbs. The principle in NAb in vitro assays is the utilization of cultured cell lines that are responsive to IFN-ß. The cytopathic effect (CPE) assay measures the capacity of NAbs to neutralize IFN- ß's protective effect on cells challenged with virus and the MxA induction assay measures the ability of NAbs to reduce the IFN-ß-induced expression of MxA, either at the mRNA or the protein level. A titer of >20 neutralizing units/ml traditionally defines NAb posi-tivity. NAbs in high titers completely abrogate the in vivo response to IFN-ß, whereas the effect of low and intermediate titers is unpredictable. As clinically important NAbs appear only after 9-18 months IFN- ß0 therapy, short-term studies of two years or less are unsuitable for evaluation of clinical NAb effects. All long-term trials of three years or more concordantly show evidence of a detrimental effect of NAbs on relapses, disease activity on MRI, or on disease progression. Persistent high titers of NAbs indicate an abrogation of the biological response and, hence, absence of therapeutic efficacy, and this observation should lead to a change of therapy. As low and medium titers are ambiguous treatment decisions in patients with low NAb titres should be guided by determination of in vivo mRNA MxA induction and clinical disease activity.
Collapse
Affiliation(s)
- Per Soelberg Sorensen
- Danish Multiple Sclerosis Research Center Department of Neurology 2082, Copenhagen University Hospital Rigshospitalet, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
16
|
Goertsches RH, Zettl UK, Hecker M. Sieving treatment biomarkers from blood gene-expression profiles: a pharmacogenomic update on two types of multiple sclerosis therapy. Pharmacogenomics 2011; 12:423-32. [PMID: 21449680 DOI: 10.2217/pgs.10.190] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Interferon-β (IFN-β) and glatiramer acetate are routinely used to inhibit disease activity in multiple sclerosis, but their mechanisms of action are incompletely understood. Individual treatment responses vary and candidate molecular markers that predict them have yet to be established. Why some patients respond poorly to a certain treatment while others respond well is addressed by the pharmacogenomic approach, which postulates that the molecular response to treatment correlates with the clinical effects, and thus seeks biological markers to estimate prognosis, guide therapy, comprehend the drugs' mechanisms of action and offer insights into disease pathogenesis. A poor clinical response can be owing to genetic variants in drug receptors or signaling components, or the appearance of neutralizing antibodies that interfere with the drug's binding efficacy. Independently, such mechanisms could lead to inadequate, that is to say unchanged, molecular responses, or exceedingly increased or decreased changes. By means of DNA microarray studies, various research groups endeavour to establish a clinically relevant relationship between the biological response to these drugs and treatment effects. Molecular profiles obtained in this way differ in the pattern and number of modulated genes, suggesting the existence of an individual 'drug-response fingerprint'. To further unravel the underlying regulatory interaction structure of the genes responsive to these immunotherapies represents a daunting but inevitable task. In this article, we focus on longitudinal ex vivo transcriptomic studies in multiple sclerosis and its therapy. We will discuss recurrently reported biomarker candidates, emphasizing those of immunologically meaning, and review studies with network module outputs.
Collapse
Affiliation(s)
- Robert H Goertsches
- University of Rostock, Department of Neurology, Gehlsheimer Strasse 20, 18147 Rostock, Germany.
| | | | | |
Collapse
|
17
|
Abstract
A thorough understanding of the structure and biology of a biotherapeutic is crucial to defining a suitable strategy for pharmacokinetic characterization in proof-of-concept disease models, toxicology species as well as the healthy and disease indication patient populations. This manuscript summarizes parameters that impact bioanalytical strategy for over 50 biotherapeutics indicated for the treatment of oncology, rheumatoid arthritis, allergy, multiple sclerosis, hematology, metabolism and infectious disease. We have addressed numerous therapeutic modalities including chimeric, humanized and fully human monoclonal antibodies, replacement proteins, peptides and fusion proteins, including polyethylene glycol and Fc fusions, as well as antibody–drug conjugates. With the rapid evolution of biotherapeutics over the last 20 years and the contraction of the pharmaceutical and biotechnology labor force, efficient workflow management becomes a crucial bioanalytical component. Thus, we have also addressed new technologies that have demonstrated either increased throughput or enhanced characterization, including Meso Scale Discovery, Gyrolab and affinity MS.
Collapse
|
18
|
Killestein J, Polman CH. Determinants of interferon β efficacy in patients with multiple sclerosis. Nat Rev Neurol 2011; 7:221-8. [DOI: 10.1038/nrneurol.2011.22] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
19
|
One-year evaluation of factors affecting the biological activity of interferon beta in multiple sclerosis patients. J Neurol 2010; 258:895-903. [PMID: 21153733 DOI: 10.1007/s00415-010-5844-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 10/06/2010] [Accepted: 11/17/2010] [Indexed: 01/01/2023]
Abstract
MxA is an antiviral protein induced by type I interferons (IFN) and some viruses; MxA gene expression is an appropriate marker for measuring biologic activity of exogenous IFNβ, as its induction indicates IFNAR receptor stimulation. A recent study has shown that measurement of MxA mRNA, after 1 year of treatment, predicts clinical responsiveness to IFNβ therapy. Loss of IFNβ bioactivity is mostly due to anti-IFNβ antibodies (both neutralizing and binding), non-compliance and receptor saturation. The aim of this study was to evaluate all possible causes of loss of IFNβ bioactivity after 1 year in treated patients. One hundred sixty-seven multiple sclerosis (MS) patients were included. One year after beginning IFNβ therapy, each patient underwent a blood test; MxA gene expression was measured by real time PCR, antiviral CPE assay to detect neutralizing antibodies (NAbs), and capture-ELISA (cELISA) to measure binding antibodies (BAbs). For MxA an upper normal threshold of 87 (RE) was considered, 20 TRU/mL was the threshold for NAbs, and 1 U for BAbs positivity. Thirty-seven out of 167 patients (22%) were MxA-negative; of these, 22 were both BAbs and NAbs+, whereas 12 were BAbs+ but Nabs-, and three were both BAbs and NAbs-. The following conclusions were drawn from the study: (1) MxA mRNA should be measured after 1 year of IFNβ therapy; (2) after 1 year of IFNβ treatment, absence of IFNβ bioactivity was detected in 22% of the patients; (3) different biological phenomena and reduced compliance explain this absence; (4) identification of the reason for absence of IFN bioactivity improves patients' management.
Collapse
|
20
|
Hecker M, Goertsches RH, Fatum C, Koczan D, Thiesen HJ, Guthke R, Zettl UK. Network analysis of transcriptional regulation in response to intramuscular interferon-β-1a multiple sclerosis treatment. THE PHARMACOGENOMICS JOURNAL 2010; 12:134-46. [PMID: 20956993 DOI: 10.1038/tpj.2010.77] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Interferon-β (IFN-β) is one of the major drugs for multiple sclerosis (MS) treatment. The purpose of this study was to characterize the transcriptional effects induced by intramuscular IFN-β-1a therapy in patients with relapsing-remitting form of MS. By using Affymetrix DNA microarrays, we obtained genome-wide expression profiles of peripheral blood mononuclear cells of 24 MS patients within the first 4 weeks of IFN-β administration. We identified 121 genes that were significantly up- or downregulated compared with baseline, with stronger changed expression at 1 week after start of therapy. Eleven transcription factor-binding sites (TFBS) are overrepresented in the regulatory regions of these genes, including those of IFN regulatory factors and NF-κB. We then applied TFBS-integrating least angle regression, a novel integrative algorithm for deriving gene regulatory networks from gene expression data and TFBS information, to reconstruct the underlying network of molecular interactions. An NF-κB-centered sub-network of genes was highly expressed in patients with IFN-β-related side effects. Expression alterations were confirmed by real-time PCR and literature mining was applied to evaluate network inference accuracy.
Collapse
Affiliation(s)
- M Hecker
- Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Jena, Germany.
| | | | | | | | | | | | | |
Collapse
|
21
|
Polman CH, Bertolotto A, Deisenhammer F, Giovannoni G, Hartung HP, Hemmer B, Killestein J, McFarland HF, Oger J, Pachner AR, Petkau J, Reder AT, Reingold SC, Schellekens H, Sørensen PS. Recommendations for clinical use of data on neutralising antibodies to interferon-beta therapy in multiple sclerosis. Lancet Neurol 2010; 9:740-50. [PMID: 20610349 DOI: 10.1016/s1474-4422(10)70103-4] [Citation(s) in RCA: 164] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The identification of factors that can affect the efficacy of immunomodulatory drugs in relapsing-remitting multiple sclerosis (MS) is important. For the available interferon-beta products, neutralising antibodies (NAb) have been shown to affect treatment efficacy. In June, 2009, a panel of experts in MS and NAbs to interferon-beta therapy convened in Amsterdam, Netherlands, under the auspices of the Neutralizing Antibodies on Interferon beta in Multiple Sclerosis consortium, a European-based project of the 6th Framework Programme of the European Commission, to review and discuss data on NAbs and their practical consequences for the treatment of patients with MS on interferon beta. The panel believed that information about NAbs and other markers of biological activity of interferons (ie, myxovirus resistance protein A [MxA]) can be integrated with clinical and imaging indicators to guide individual treatment decisions. In cases of sustained high-titre NAb positivity and/or lack of MxA bioactivity, a switch to a non-interferon-beta therapy should be considered. In patients who are doing poorly clinically, therapy should be switched irrespective of NAb or MxA bioactivity.
Collapse
Affiliation(s)
- Chris H Polman
- Department of Neurology, MS Center Amsterdam, Free University Medical Center, Amsterdam, Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Rani MRS, Xu Y, Lee JC, Shrock J, Josyula A, Schlaak J, Chakraborthy S, Ja N, Ransohoff RM, Rudick RA. Heterogeneous, longitudinally stable molecular signatures in response to interferon-beta. Ann N Y Acad Sci 2010; 1182:58-68. [PMID: 20074275 DOI: 10.1111/j.1749-6632.2009.05068.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Interferons (IFNs) are widely used in therapy for viral, neoplastic, and inflammatory disorders, but clinical response varies among patients. The biological basis for variable clinical response is not known. We determined the primary molecular response to IFN-beta (IFN-beta) injections in 35 treatment-naïve multiple sclerosis (MS) patients using a customized cDNA macroarray with 186 interferon-stimulated genes (ISGs). Our results revealed striking interindividual heterogeneity, both in the magnitude as well as the nature of the primary molecular response to IFN-beta injections. Despite marked between-subject variability in the molecular response, responses within individual subjects were stable over a 6-month interval. Our data suggest that clinical response to IFN-beta therapy for MS differs among patients because of qualitative rather than quantitative variability in the primary molecular response to the drug.
Collapse
Affiliation(s)
- M R Sandhya Rani
- Neuroinflammation Research Center, Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Harris VK, Sadiq SA. Disease biomarkers in multiple sclerosis: potential for use in therapeutic decision making. Mol Diagn Ther 2010; 13:225-44. [PMID: 19712003 DOI: 10.1007/bf03256329] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disorder of the brain and spinal cord that predominantly affects white matter. MS has a variable clinical presentation and has no 'diagnostic' laboratory test; this often results in delays to definite diagnosis. In confronting the disease, early diagnosis and appropriate, timely therapeutic intervention are critical factors in ensuring favorable long-term outcomes. The availability of reliable biomarkers could radically alter our management of MS at critical phases of the disease spectrum. Identification of markers that could predict the development of MS in high-risk populations would allow for intervention strategies that may prevent evolution to definite disease. Work with anti-myelin antibodies and the ongoing analysis of microarray gene expression have thus far not yielded biomarkers that predict future disease development. Similarly, extensive studies with serum and cerebrospinal fluid (CSF) have not yielded a disease-specific and sensitive diagnostic biomarker for MS. Establishment of disease diagnosis always leads to questions about long-term prognosis because in an individual patient the natural history of the disease is clinically unpredictable. Biomarkers that correlate with myelin loss, spinal cord disease, grey matter and subcortical demyelination need to be developed in order to accurately predict the disease course. The bulk of effort in biomarker development in MS has been concentrated in the area of monitoring disease activity. At present, a disease 'activation' panel of CSF biomarkers would include the following: interleukin-6 or its soluble receptor, nitric oxide and nitric oxide synthase, osteopontin, and fetuin-A. Although disease activity in MS is predominantly inflammatory, disease progression is likely to be the result of neurodegeneration. Therefore, the roles of proteins indicative of neuronal, axonal, and glial loss such as neurofilaments, tau, 14-3-3 proteins, and N-acetylaspartate are all under investigation, as are proteins affecting remyelination and regeneration, such as Nogo-A. With the increasing awareness of cognition dysfunction in MS, molecules such as apolipoprotein and proteins in the amyloid precursor protein pathway implicated in dementia are also being examined. Serum biomarkers that help monitor therapeutic efficacy such as the titer of antibody to beta-interferon, a first-line medication in MS, are established in clinical practice. Ongoing work with biomarkers that reflect drug bioavailability and factors that distinguish between medication responders and nonresponders are also under investigation. The discovery of new biomarkers relies on applying advances in proteomics along with microarray gene and antigen analysis and will hopefully result in the establishment of specific biomarkers for MS.
Collapse
Affiliation(s)
- Violaine K Harris
- Multiple Sclerosis Research Center of New York, New York, New York 10019, USA
| | | |
Collapse
|
24
|
A cytokine immunosensor for Multiple Sclerosis detection based upon label-free electrochemical impedance spectroscopy using electroplated printed circuit board electrodes. Biosens Bioelectron 2009; 25:506-9. [DOI: 10.1016/j.bios.2009.07.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2009] [Revised: 07/02/2009] [Accepted: 07/21/2009] [Indexed: 11/18/2022]
|
25
|
Deisenhammer F. Neutralizing antibodies to interferon-beta and other immunological treatments for multiple sclerosis: prevalence and impact on outcomes. CNS Drugs 2009; 23:379-96. [PMID: 19453200 DOI: 10.2165/00023210-200923050-00003] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Biopharmaceuticals can induce antibodies, which interact with and neutralize the therapeutic effect of such drugs and are therefore termed neutralizing antibodies (NAbs). In the treatment of multiple sclerosis, NAbs against interferon (IFN)-beta and natalizumab have been recognized. The prevalence of NAbs against different IFNbeta preparations varies widely, mainly depending on the product but also on other factors such as amino acid sequence variations, glycosylation, formulation, route and frequency of application, dose, duration of treatment and patient characteristics (human leukocyte antigen [HLA] status). IFNbeta-1a given intramuscularly induces significantly less NAbs than any other IFNbeta formulation. The longitudinal development of NAbs also differs between IFNbeta preparations, with higher reversion rates in IFNbeta-1b-treated compared with IFNbeta-1a-treated patients. The negative effect of NAbs on various outcome measures is very consistent across many studies, specifically when observation periods are longer than 2 years. NAbs against natalizumab occur less frequently (6%) and, like NAbs against IFNbeta, they are associated with a loss of clinical and radiological efficacy of the drug.
Collapse
Affiliation(s)
- Florian Deisenhammer
- Department of Neurology, Innsbruck Medical University, Anichstrasse 35, Innsbruck 6020, Austria.
| |
Collapse
|
26
|
Sellebjerg F, Krakauer M, Hesse D, Ryder LP, Alsing I, Jensen PEH, Koch-Henriksen N, Svejgaard A, Soelberg Sørensen P. Identification of new sensitive biomarkers for thein vivoresponse to interferon-β treatment in multiple sclerosis using DNA-array evaluation. Eur J Neurol 2009; 16:1291-8. [DOI: 10.1111/j.1468-1331.2009.02716.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
27
|
Hesse D, Frederiksen JL, Koch-Henriksen N, Schreiber K, Stenager E, Heltberg A, Ravnborg M, Bendtzen K, Sellebjerg F, Sorensen PS. Methylprednisolone does not restore biological response in multiple sclerosis patients with neutralizing antibodies against interferon-β. Eur J Neurol 2009; 16:43-7. [DOI: 10.1111/j.1468-1331.2008.02336.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
28
|
Weinstock-Guttman B, Ramanathan M, Zivadinov R. Interferon-beta treatment for relapsing multiple sclerosis. Expert Opin Biol Ther 2008; 8:1435-47. [PMID: 18694361 DOI: 10.1517/14712598.8.9.1435] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Recombinant forms of IFN-beta were the first therapeutic intervention found to be effective at interfering with the course of multiple sclerosis (MS), a chronic and debilitating disease affecting the CNS in young adults. OBJECTIVE/METHODS To examine the application of IFN-beta to MS treatment by a review of relevant literature. RESULTS The different IFN-beta products available are similar in their clinical effects. However, the response to IFN-beta therapy is only partial and the most efficient individual-specific dose, route and frequency of administration are not elucidated fully. The mechanism of action of IFN-beta in MS is also not understood fully but its immunomodulatory effects are probably more important than its anti-proliferative and antiviral activities. CONCLUSIONS Although new therapeutic approaches are being sought to better treat MS, IFN-beta remains one of the most recognized and approved worldwide therapeutic options for this disease.
Collapse
Affiliation(s)
- Bianca Weinstock-Guttman
- The Jacobs Neurological Institute, Department of Neurology, University at Buffalo, State University of New York, Buffalo, NY 14203, USA.
| | | | | |
Collapse
|
29
|
Rinta S, Kuusisto H, Raunio M, Paalavuo R, Levula M, Lehtimäki T, Elovaara I. Apoptosis-related molecules in blood in multiple sclerosis. J Neuroimmunol 2008; 205:135-41. [PMID: 18963025 DOI: 10.1016/j.jneuroim.2008.09.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 09/02/2008] [Accepted: 09/05/2008] [Indexed: 12/23/2022]
Abstract
A failure in apoptosis of lymphocytes may lead to harmful immunoreactivity in MS. We analyzed apoptosis-related molecules including TRAIL, sFas, sFasL and MIF in the blood of 117 MS patients and controls to answer whether these molecules may be used in the evaluation of disease activity and immunomodulatory effect of IFN-beta. Increased levels of sTRAIL, sFasL and MIF were found in sera of untreated patients with MS relapse indicating their association with MS disease activity. IFN-beta treated patients in remission had increased TRAIL mRNA, sTRAIL, sFaL and MIF that most likely reflect bioactivity of IFN-beta.
Collapse
Affiliation(s)
- Sanna Rinta
- Neuroimmunology Unit, Department of Neurology, Finn-Medi 3, Biokatu 10, 33014 University of Tampere, Finland
| | | | | | | | | | | | | |
Collapse
|
30
|
Weinstock-Guttman B, Bhasi K, Badgett D, Tamaño-Blanco M, Minhas M, Feichter J, Patrick K, Munschauer F, Bakshi R, Ramanathan M. Genomic effects of once-weekly, intramuscular interferon-beta1a treatment after the first dose and on chronic dosing: Relationships to 5-year clinical outcomes in multiple sclerosis patients. J Neuroimmunol 2008; 205:113-25. [PMID: 18950872 DOI: 10.1016/j.jneuroim.2008.09.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Revised: 08/25/2008] [Accepted: 09/02/2008] [Indexed: 01/10/2023]
Abstract
PURPOSE To characterize gene expression in multiple sclerosis (MS) patients after the first dose and chronic dosing of 30 microg, once weekly, intramuscular interferon-beta1a (IFN-beta) and to delineate the pharmacogenomic differences between Good Responders and Partial Responders to IFN-beta therapy. METHODS The treatment responses after the first IFN-beta dose and chronic IFN-beta dosing were assessed in 22 relapsing MS patients (17 females, 5 males; average age: 41.5+/-SD 10.4 years). Gene expression profiles in peripheral blood mononuclear cells were obtained prior to treatment and at 1, 2, 4, 8, 24, 48, 120, 168 h after the first IFN-beta dose and at 1, 6 and 12 months after chronic dosing with once-weekly 30 microg IFN-beta-1a intramuscularly. Repeated measures statistics with false discovery rate control were used. The functional characteristics, biological pathways and transcription factor sites were analyzed. RESULTS Of the 1000 genes modulated following the first dose and upon chronic dosing of IFN-beta in MS patients, approximately 35% were up-regulated and 65% were down- regulated; the percentage of modulated genes in common was approximately 50%. The expression of the pharmacodynamic mRNA markers of IFN-beta effect showed differences in time profiles for the Good Responder and Partial Responders to IFN-beta therapy and the Jak-STAT, TNFRSF10B, IL6, TGFbeta, retinoic acid and CDC42 pathways were differentially modulated. The patients with side effects to therapy showed differences in the TGFbeta1, IFNG/STAT3 and TNF pathways. CONCLUSIONS Gene expression is a valuable tool for understanding the molecular mechanisms of IFN-beta action in MS patients.
Collapse
|
31
|
Translation towards personalized medicine in Multiple Sclerosis. J Neurol Sci 2008; 274:68-75. [PMID: 18789804 DOI: 10.1016/j.jns.2008.07.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Revised: 07/19/2008] [Accepted: 07/24/2008] [Indexed: 12/24/2022]
Abstract
In recent years the realization that the concept 'one drug fits all' - does not work, created the need to shift gears from 'treating the disease' to 'treating the patient', and implementation of 'Personalized Medicine' where treatment is tailored to the individual. In chronic and progressive diseases, such as Multiple Sclerosis (MS), the need for tailored therapeutics is especially imperative, as the consequences of an ineffective medication might be irreversible dysfunction. In recent years accumulating evidence indicates that MS is not a single disease and that patients with different disease subtypes respond differently to a medication. Environment and genetics are among the factors that determine disease subtype and activity, and the patient's response to medication. Additional factors include demographic characteristics such as gender and age, as well as chrono-biological indicators. During the last few years, advances and availability of new technologies have brought genome-wide gene expression profiling studies to many medical fields, including MS. Genomic technologies have also stimulated pharmacogenetics studies, that aim to identify genetic factors that affect response to treatment. However, pharmacogenetics information is still immature to allow its translation to clinical practice in MS. Notably, one of the major limitations in obtaining reproducible data across MS pharmacogenetics studies has been the lack of a consensus as to the appropriate method for determining clinical response. In light of the rapid advances in technology and progress in applying individualized treatment strategies in other diseases, 'Personalized Medicine' for MS seems feasible within the coming years.
Collapse
|
32
|
Millonig A, Dressel A, Bahner D, Bitsch A, Bogumil T, Elitok E, Kitze B, Tumani H, Weber F, Gneiss C, Deisenhammer F. MxA protein an interferon beta biomarker in primary progressive multiple sclerosis patients. Eur J Neurol 2008; 15:822-6. [DOI: 10.1111/j.1468-1331.2008.02190.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
33
|
Hilpert J, Beekman JM, Schwenke S, Kowal K, Bauer D, Lampe J, Sandbrink R, Heubach JF, Stürzebecher S, Reischl J. Biological response genes after single dose administration of interferon beta-1b to healthy male volunteers. J Neuroimmunol 2008; 199:115-25. [PMID: 18565596 DOI: 10.1016/j.jneuroim.2008.04.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 04/21/2008] [Accepted: 04/25/2008] [Indexed: 11/30/2022]
Abstract
Treatment with interferon beta-1b (IFNB-1b) is clinically effective in multiple sclerosis patients. However, the mechanism of action is only partially understood, and validated biological response markers are lacking. We assessed IFNB-1b-induced transcriptional changes by microarray technology. Healthy male volunteers received 250 mug IFNB-1b or placebo in a double-blind, randomized controlled trial (n=5 per group). Most transcripts demonstrated peak levels after 6-12 h and returned to baseline after 48 h. We identified 227 differentially regulated genes including novel and previously described markers. This panel may become a valuable tool for development of new IFNB-1b formulations and assessment of clinical drug effects.
Collapse
Affiliation(s)
- Jan Hilpert
- Clinical Pharmacology, Bayer Schering Pharma AG, Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Although the occurrence of neutralizing antibodies (NAbs) to interferon (IFN)-beta has been acknowledged since the pivotal trials of IFN-beta in multiple sclerosis (MS), the effect of these antibodies has for several reasons been debated. The main reason for the controversies has been insufficient knowledge of the fact that clinically relevant NAbs do not appear until 12-18 months after initiation of IFN-beta therapy which make studies of 2 years or less unsuited to assess the clinical relevance of NAbs. Further, changes in NAb affinity occur and contribute to increase NAb effects by time. The present paper reviews our current knowledge of NAbs and stresses the importance of using measurements of NAbs routinely. It is concluded that NAb titres are important for the biological response to IFN-beta. Patients with low or intermediate titres may have preserved a full or partial biological response and might still benefit from IFN-beta therapy. However, persistent high titres of NAbs indicate an abrogation of the biological response and, hence, absence of therapeutic efficacy, and this observation should lead to a change of therapy. The application of the existing information about NAbs in clinical practice would lead to improved efficacy of IFN-beta treatment for the benefit of patients with MS.
Collapse
Affiliation(s)
- D Hesse
- Department of Neurology, Danish Multiple Sclerosis Research Center, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | | |
Collapse
|
35
|
Weinstock-Guttman B, Tamaño-Blanco M, Bhasi K, Zivadinov R, Ramanathan M. Pharmacogenetics of MXA SNPs in interferon-beta treated multiple sclerosis patients. J Neuroimmunol 2006; 182:236-9. [PMID: 17126411 DOI: 10.1016/j.jneuroim.2006.10.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Revised: 10/12/2006] [Accepted: 10/13/2006] [Indexed: 12/22/2022]
Abstract
The myxovirus resistance A (MXA) mRNA has been extensively investigated for assessing the biologic responses of multiple sclerosis (MS) patients to interferon-beta (IFN-beta) therapy. The objective of this study was to evaluate the associations between two MXA promoter region single nucleotide polymorphisms (rs2071430 and rs17000900) and the gene expression responses, clinical and MRI phenotypes in IFN-beta treated MS patients. The rs2071430 and rs17000900 SNPs, which are located in or near an interferon-stimulated response element (ISRE), were genotyped in 179 relapsing MS patients. Quantitative MRI measurements were available for 101 patients on IFN-beta monotherapy. Gene expression was assessed in 22 anti-interferon-beta neutralizing antibody negative patients. No significant association was found between the MXA genotype at these two SNPs and clinical, MRI and MXA gene expression in MS patients treated with IFN-beta therapy.
Collapse
|