Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Anthony T. Reder is active.

Publication


Featured researches published by Anthony T. Reder.


Lancet Neurology | 2014

Safety and efficacy of fingolimod in patients with relapsing-remitting multiple sclerosis (FREEDOMS II): a double-blind, randomised, placebo-controlled, phase 3 trial

Peter A. Calabresi; Ernst Wilhelm Radue; Douglas S. Goodin; K Rammohan; Anthony T. Reder; Timothy Vollmer; Mark A. Agius; Ludwig Kappos; Tracy Stites; Bingbing Li; Linda Cappiello; Philipp von Rosenstiel; Fred D. Lublin

BACKGROUND Fingolimod has shown reductions in clinical and MRI disease activity in patients with relapsing-remitting multiple sclerosis. We further assessed the efficacy and safety of fingolimod in such patients. METHODS We did this placebo-controlled, double-blind phase 3 study predominantly in the USA (101 of 117 centres). Using a computer-generated sequence, we randomly allocated eligible patients-those aged 18-55 years with relapsing-remitting multiple sclerosis-to receive fingolimod 0·5 mg, fingolimod 1·25 mg, or placebo orally once daily (1:1:1; stratified by study centre). On Nov 12, 2009, all patients assigned to fingolimod 1·25 mg were switched to the 0·5 mg dose in a blinded manner after a review of data from other phase 3 trials and recommendation from the data and safety monitoring board, but were analysed as being in the 1·25 mg group in the primary outcome analysis. Our primary endpoint was annualised relapse rate at month 24, analysed by intention to treat. Secondary endpoints included percentage brain volume change (PBVC) from baseline and time-to-disability-progression confirmed at 3 months. This trial is registered with ClinicalTrilals.gov, number NCT00355134. FINDINGS Between June 30, 2006, and March 4, 2009, we enrolled and randomly allocated 1083 patients: 370 to fingolimod 1·25 mg, 358 to fingolimod 0·5 mg, and 355 to placebo. Mean annualised relapse rate was 0·40 (95% CI 0·34-0·48) in patients given placebo and 0·21 (0·17-0·25) in patients given fingolimod 0·5 mg: rate ratio 0·52 (95% CI 0·40-0·66; p<0·0001), corresponding to a reduction of 48% with fingolimod 0·5 mg versus placebo. Mean PBVC was -0·86 (SD 1·22) for fingolimod 0·5 mg versus -1·28 (1·50) for placebo (treatment difference -0·41, 95% CI -0·62 to -0·20; p=0·0002). We recorded no statistically significant between-group difference in confirmed disability progression (hazard rate 0·83 with fingolimod 0·5 mg vs placebo; 95% CI 0·61-1·12; p=0·227). Fingolimod 0·5 mg caused more of the following adverse events versus placebo: lymphopenia (27 [8%] patients vs 0 patients), increased alanine aminotransferase (29 [8%] vs six [2%]), herpes zoster infection (nine [3%] vs three [1%]), hypertension (32 [9%] vs 11 [3%]), first-dose bradycardia (five [1%] vs one [<0·5%]), and first-degree atrioventricular block (17 [5%] vs seven [2%]). 53 (15%) of 358 patients given fingolimod 0·5 mg and 45 (13%) of 355 patients given placebo had serious adverse events over 24 months, which included basal-cell carcinoma (ten [3%] patients vs two [1%] patients), macular oedema (three [1%] vs two [1%]), infections (11 [3%] vs four [1%]), and neoplasms (13 [4%] vs eight [2%]). INTERPRETATION Our findings expand knowledge of the safety profile of fingolimod and strengthen evidence for its beneficial effects on relapse rates in patients with relapsing-remitting multiple sclerosis. We saw no effect of fingolimod on disability progression. Our findings substantiate the beneficial profile of fingolimod as a disease-modifying agent in the management of patients with relapsing-remitting multiple sclerosis. FUNDING Novartis Pharma AG.


Neurology | 1996

Validation of the Functional Assessment of Multiple Sclerosis quality of life instrument

David Cella; K. Dineen; Barry G. W. Arnason; Anthony T. Reder; Kimberly Webster; G. Karabatsos; C. Chang; Stephen Lloyd; F. Mo; J. Stewart; Dusan Stefoski

Based on scientific literature and interviews with clinicians and patients, we developed a quality of life instrument for use with people with MS called the Functional Assessment of Multiple Sclerosis (FAMS).The initial item pool consisted of 88 questions: 28 from the general version of the Functional Assessment of Cancer Therapy quality of life instrument, plus 60 generated by patients, providers, and literature review. The validation samples comprised a mail survey cohort (N = 377) and a clinical cohort (N = 56). Both cohorts provided evidence for internal consistency of the derived subscales, test-retest reliability, content validity, concurrent validity, and construct validity. Principal components and Rasch measurement model analyses were applied sequentially to survey sample data, reducing test length to 44 questions, divided into six subscales: mobility, symptoms, emotional well-being (depression), general contentment, thinking/fatigue, and family/social well-being. Fifteen initially rejected questions were added back as miscellaneous (unscored) questions for their potential clinical and empirical value. The mobility subscale was strongly predictive of the Kurtzke Extended Disability Status Scale and the Scripps Neurologic Rating Scales. The other five subscales were not, indicating they measure aspects of patient quality of life not captured by the neurologic exam. The final 59-item English language instrument (FAMS version 2) is available for inclusion in clinical trials and clinical practice. NEUROLOGY 1996;47: 129-139


Journal of Neuroimmunology | 1991

Cytokine levels in the cerebrospinal fluid and serum of patients with multiple sclerosis

Davide Maimone; Susan Gregory; Barry G. W. Arnason; Anthony T. Reder

Interleukin (IL) 1 beta, tumor necrosis factor alpha (TNF alpha), and IL-6 are cytokines which mediate cellular responses during immune activation and inflammation. In multiple sclerosis (MS) they might be responsible for T-cell activation (IL-1 beta), for demyelination (TNF alpha), and for immunoglobulin (Ig) synthesis (IL-6) within the central nervous system. We studied IL-1 beta, TNF alpha, and IL-6 levels in the cerebrospinal fluid (CSF) of 34 patients with MS, 43 patients with non-inflammatory neurological diseases (NIND), and 19 patients with inflammatory neurological diseases (IND). IL-6 was found in the CSF of 29% of MS, 7% of NIND, and 47% of IND patients. TNF alpha was detected in the CSF of 23% of MS, 7% of NIND, and 29% of IND. CSF IL-6 and TNF alpha levels were significantly higher in MS and IND than in NIND. IL-1 beta was rarely detected in the CSF of any group. At least one cytokine was detected in 52% of MS CSF, 11% of NIND CSF, and 64% of IND CSF. In MS patients, no relationship was observed between the incidence or the amount of intrathecal IgG synthesis or oligoclonal bands and the presence of any cytokine. We also evaluated cytokine levels in paired sera from 11 MS and 13 NIND patients. Low levels of IL-6 were detected in most sera from MS and NIND patients. TNF alpha was detected in only two MS sera, and IL-1 beta was undetectable in any sample. Our results indicate that increased CSF levels of the cytokines IL-6 and TNF alpha occur frequently in MS and IND, but there is no obvious relationship to intrathecal Ig synthesis.


Multiple Sclerosis Journal | 2012

Recommendations for a Brief International Cognitive Assessment for Multiple Sclerosis (BICAMS)

Dawn Langdon; Maria Pia Amato; J. B S Boringa; Bruno Brochet; Foley F; S. Fredrikson; Hämäläinen P; H.-P. Hartung; Krupp L; Iris-Katharina Penner; Anthony T. Reder; Ralph H. B. Benedict

Background: Cognitive impairment in MS impacts negatively on many patients at all disease stages and in all subtypes. Full clinical cognitive assessment is expensive, requiring expert staff and special equipment. Test versions and normative data are not available for all languages and cultures. Objective: To recommend a brief cognitive assessment for multiple sclerosis (MS) that is optimized for small centers, with one or few staff members, who may not have neuropsychological training and constructed to maximize international use. Methods: An expert committee of twelve members representing the main cultural groups that have so far contributed considerable data about MS cognitive dysfunction was convened. Following exhaustive literature review, peer-reviewed articles were selected to cover a broad spectrum of cultures and scales that targeted cognitive domains vulnerable to MS. Each was rated by two committee members and candidates scales were rated on psychometric qualities (reliability, validity, and sensitivity), international application, ease of administration, feasibility in the specified context, and acceptability to patients. Results: The committee recommended the Symbol Digit Modalities Test, if only 5 minutes was available, with the addition of the California Verbal Learning Test – Second Edition and the Brief Visuospatial Memory Test – Revised learning trials if a further 10 minutes could be allocated for testing. Conclusions: A brief cognitive assessment for MS has been recommended. A validation protocol has been prepared for language groups and validation studies have commenced.


Annals of Neurology | 2011

Genome-wide meta-analysis identifies novel multiple sclerosis susceptibility loci

Nikolaos A. Patsopoulos; Federica Esposito; Joachim Reischl; Stephan Lehr; David Bauer; Jürgen Heubach; Rupert Sandbrink; Christoph Pohl; Gilles Edan; Ludwig Kappos; David Miller; Javier Montalbán; Chris H. Polman; Mark Freedman; Hans-Peter Hartung; Barry G. W. Arnason; Giancarlo Comi; Stuart D. Cook; Massimo Filippi; Douglas S. Goodin; Paul O'Connor; George C. Ebers; Dawn Langdon; Anthony T. Reder; Anthony Traboulsee; Frauke Zipp; Sebastian Schimrigk; Jan Hillert; Melanie Bahlo; David R. Booth

To perform a 1‐stage meta‐analysis of genome‐wide association studies (GWAS) of multiple sclerosis (MS) susceptibility and to explore functional consequences of new susceptibility loci.


Neurology | 2008

Mechanisms of action of disease-modifying agents and brain volume changes in multiple sclerosis

Robert Zivadinov; Anthony T. Reder; Massimo Filippi; Alireza Minagar; Olaf Stüve; Hans Lassmann; Michael K. Racke; Michael G. Dwyer; Elliot M. Frohman; Omar Khan

Disease-modifying agents (DMAs), including interferon beta (IFNβ) and glatiramer acetate (GA), are the mainstays of long-term treatment of multiple sclerosis (MS). Other potent anti-inflammatory agents like natalizumab and different types of chemotherapeutics are increasingly being used for treatment of MS, particularly in patients with breakthrough disease activity. Brain volume (BV) loss occurs early in the disease process, accelerates over time, and may be only partially affected by DMA therapy. Low-dose, low frequency IFNβ administered once weekly and GA appear to partially reduce BV decline over the second and third years of treatment. High dose, high frequency IFNβ demonstrated no clear effect on BV loss during this time period. Current evidence suggests that changes in BV after immunoablation may not be due entirely to the resolution of edema but may be related to potential chemotoxicity of high dose cyclophosphamide. Natalizumab reduces the development of BV decline in the second and third years of treatment. IV immunoglobulin showed a positive effect on decelerating BV reduction in relapsing and advanced stages of MS. These differences between DMAs may be explained by the extent of their therapeutic effects on inflammation and on the balance between inhibition or promotion of remyelination and neuronal repair in the CNS. We described the mechanisms of action by which DMAs induce accelerated, non–tissue-related BV loss (pseudoatrophy) in the short term but, in the long run, may still potentially lead to permanent BV decline. The effects of corticosteroid therapy on changes in BV in patients with MS help clarify the mechanisms through which potent anti-inflammatory treatments may prevent, stabilize, or induce BV loss.


Journal of Clinical Investigation | 1997

INCREASED CD80+ B CELLS IN ACTIVE MULTIPLE SCLEROSIS AND REVERSAL BY INTERFERON BETA -1B THERAPY

Kursad Genc; D L Dona; Anthony T. Reder

Costimulatory molecules help determine T cell responses. CD80 (B7-1) and CD86 (B7-2), costimulatory proteins on antigen-presenting cells, bind to CD28 on T cells. When costimulation is coupled with a signal through the T cell receptor (TCR), T cell proliferation and cytokine secretion are induced. However, TCR signaling without CD80/CD86CD28 costimulation causes anergy. During multiple sclerosis (MS) exacerbations, circulating immune cells are activated, Th1 cytokine levels in the blood are elevated, and blood-derived immune cells destroy brain oligodendroglia. In the experimental autoimmune encephalomyelitis model of MS, CD80 on antigen-presenting cells induces Th1 cell responses; CD86 enhances generation of Th2 cells. Variation in CD80 and CD86 expression is likely to influence immune regulation in MS. We demonstrate that the number of circulating CD80(+) lymphocytes is increased significantly during MS exacerbations, but is normal in stable MS. These CD80(+) lymphocytes are predominantly B cells, based on two-color flow cytometry. The number of CD71(+) and HLA-DR+ lymphocytes and monocytes is also increased in active MS. Therapy with IFN beta-1b markedly reduces the number of circulating CD80(+) B cells and increases CD86(+) monocyte number. HLA-DR+, CD71(+), and CD25(+) mononuclear cell numbers are also reduced by therapy. The number of CD80(+) cells may be a useful surrogate marker during IFN-beta therapy, and reduction of CD80-mediated costimulation may be one therapeutic mechanism by which IFN-beta acts in MS.


Neurology | 2007

Neutralizing antibodies to interferon beta: Assessment of their clinical and radiographic impact: An evidence report: Report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology

Douglas S. Goodin; Elliot M. Frohman; B. Hurwitz; Paul O'Connor; J. J. Oger; Anthony T. Reder; J. C. Stevens

The clinical and radiologic impact of developing neutralizing antibodies (NAbs) to interferon beta (IFN ) while on this therapy for multiple sclerosis (MS) is assessed. On the basis of Class II and III evidence, it is concluded that treatment of patients with MS with IFN (Avonex, Betaseron, or Rebif) is associated with the production of NAbs (Level A). NAbs in the serum are probably associated with a reduction in the radiographic and clinical effectiveness of IFN treatment (Level B). In addition, the rate of NAb production is probably less with IFN -1a treatment than with IFN -1b treatment, although the magnitude and persistence of this difference is difficult to determine (Level B). Finally, it is probable that there is a difference in seroprevalence due to variability in the dose of IFN injected or in the frequency or route of its administration (Level B). Regardless of the explanation, it seems clear that IFN -1a (as it is currently formulated for IM injection) is less immunogenic than the current IFN preparations (either IFN -1a or IFN -1b) given multiple times per week subcutaneously (Level A). However, because NAbs disappear in some patients even with continued IFN treatment (especially in patients with low titers), the persistence of this difference is difficult to determine (Level B). Although the finding of sustained high-titer NAbs ( 100 to 200 NU/mL) is associated with a reduction in the therapeutic effects of IFN on radiographic and clinical measures of MS disease activity, there is insufficient information on the utilization of NAb testing to provide specific recommendations regarding when to test, which test to use, how many tests are necessary, or which cutoff titer to apply (Level U). NEUROLOGY 2007;68:977–984 The development of neutralizing antibodies (NAbs) to proteins administered therapeutically is often associated with a reduction in the biologic actions that these proteins exert. It is therefore surprising that the clinical and radiographic impact of NAbs to interferon beta (IFN ) in the treatment of multiple sclerosis (MS) is controversial. This assessment evaluates the clinical and radiographic impact of NAbs in this setting and considers some of the difficulties in this research area that may explain the ongoing controversy. In this regard, it is useful for readers to appreciate the complexity of this particular biologic system.1-9 Thus, a brief overview of IFN biology is provided in the supplementary material to this asFrom the University of California, San Francisco (D.S.G.); University of Texas Southwestern (E.M.F.), Dallas; Duke University Medical College (B.H.), Durham, NC; St. Michaels Hospital (P.W.O.), Toronto, Ontario, Canada; University of BC (J.J.O.), Vancouver, British Columbia, Canada; The University of Chicago (A.T.R.), Oak Park, IL; and Lutheran Medical Office (J.C.S.), Fort Wayne, IN. Disclosure: The authors report no conflicts of interest. Received August 10, 2006. Accepted in final form December 7, 2006. Approved by the Therapeutics and Technology Subcommittee on July 28, 2006; by the Practice Committee on November 11, 2006; and by the AAN Board of Directors on January 4, 2007. Address correspondence and reprint requests to the American Academy of Neurology, 1080 Montreal Avenue, St. Paul, MN 55116; e-mail: [email protected] Additional material related to this article can be found on the Neurology Web site. Go to www.neurology.org and scroll down the Table of Contents for the March 27 issue to find the title link for this article. Copyright


Lancet Neurology | 2010

Recommendations for clinical use of data on neutralising antibodies to interferon-beta therapy in multiple sclerosis

Chris H. Polman; Antonio Bertolotto; Florian Deisenhammer; Gavin Giovannoni; Hans-Peter Hartung; Bernhard Hemmer; Joep Killestein; Henry F. McFarland; Joel Oger; Andrew R. Pachner; John Petkau; Anthony T. Reder; Stephen C. Reingold; Huub Schellekens; Per Soelberg Sørensen

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.


Neurology | 2012

Survival in MS A randomized cohort study 21 years after the start of the pivotal IFNβ-1b trial

Douglas S. Goodin; Anthony T. Reder; George C. Ebers; Gary Cutter; Marcelo Kremenchutzky; Joel Oger; Dawn Langdon; Mark Rametta; Karola Beckmann; T.M. DeSimone; Volker Knappertz

Objective: To examine the effects of interferon beta (IFNβ)-1b on all-cause mortality over 21 years in the cohort of 372 patients who participated in the pivotal randomized clinical trial (RCT), retaining (in the analysis) the original randomized treatment-assignments. Methods: For this randomized long-term cohort study, the primary outcome, defined before data collection, was the comparison of all-cause mortality between the IFNβ-1b 250 μg and placebo groups from the time of randomization through the entire 21-year follow-up interval (intention-to-treat, log-rank test for Kaplan-Meier survival curves). All other survival outcomes were secondary. Results: After a median of 21.1 years from RCT enrollment, 98.4%(366 of 372) of patients were identified, and, of these, 81 deaths were recorded (22.1% [81 of 366]). Patients originally randomly assigned to IFNβ-1b 250 μg showed a significant reduction in all-cause mortality over the 21-year period compared with placebo (p = 0.0173), with a hazard ratio of 0.532 (95% confidence interval 0.314–0.902). The hazard rate of death at long-term follow-up by Kaplan-Meier estimates was reduced by 46.8% among IFNβ-1b 250 μg–treated patients (46.0% among IFNβ-1b 50 μg–treated patients) compared with placebo. Baseline variables did not influence the observed treatment effect. Conclusions: There was a significant survival advantage in this cohort of patients receiving early IFNβ-1b treatment at either dose compared with placebo. Near-complete ascertainment, together with confirmatory findings from both active treatment groups, strengthens the evidence for an IFNβ-1b benefit on all-cause mortality. Classification of Evidence: This study provides Class III evidence that early treatment with IFNβ-1b is associated with prolonged survival in initially treatment-naive patients with relapsing-remitting multiple sclerosis.

Collaboration


Dive into the Anthony T. Reder's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xuan Feng

University of Chicago

View shared research outputs
Top Co-Authors

Avatar

Jack P. Antel

Montreal Neurological Institute and Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jack P. Antel

Montreal Neurological Institute and Hospital

View shared research outputs
Top Co-Authors

Avatar

Karola Beckmann

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Martin T. Lowy

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Anthony Traboulsee

University of British Columbia

View shared research outputs
Researchain Logo
Decentralizing Knowledge