Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Aging Cell ; 22(2): e13765, 2023 02.
Article in English | MEDLINE | ID: mdl-36633253

ABSTRACT

Genome-wide association studies (GWAS) have validated a strong association of atherosclerosis with the CDKN2A/B locus, a locus harboring three tumor suppressor genes: p14ARF , p15INK4b , and p16INK4a . Post-GWAS functional analysis reveals that CUX is a transcriptional activator of p16INK4a via its specific binding to a functional SNP (fSNP) rs1537371 on the atherosclerosis-associated CDKN2A/B locus, regulating endothelial senescence. In this work, we characterize SATB2, another transcription factor that specifically binds to rs1537371. We demonstrate that even though both CUX1 and SATB2 are the homeodomain transcription factors, unlike CUX1, SATB2 is a transcriptional suppressor of p16INK4a and overexpression of SATB2 competes with CUX1 for its binding to rs1537371, which inhibits p16INK4a and p16INK4a -dependent cellular senescence in human endothelial cells (ECs). Surprisingly, we discovered that SATB2 expression is transcriptionally repressed by CUX1. Therefore, upregulation of CUX1 inhibits SATB2 expression, which enhances the binding of CUX1 to rs1537371 and subsequently fine-tunes p16INK4a expression. Remarkably, we also demonstrate that IL-1ß, a senescence-associated secretory phenotype (SASP) gene itself and a biomarker for atherosclerosis, induces cellular senescence also by upregulating CUX1 and/or downregulating SATB2 in human ECs. A model is proposed to reconcile our findings showing how both primary and secondary senescence are activated via the atherosclerosis-associated p16INK4a expression.


Subject(s)
Atherosclerosis , Matrix Attachment Region Binding Proteins , Humans , Atherosclerosis/genetics , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Endothelial Cells/metabolism , Genome-Wide Association Study , Homeodomain Proteins/genetics , Matrix Attachment Region Binding Proteins/genetics , Phenotype , Repressor Proteins/genetics , Transcription Factors/genetics , Interferon beta-1b/pharmacology
2.
Inflammopharmacology ; 30(2): 667-681, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35249177

ABSTRACT

3-Nitropropionic acid (3-NP) model serves as a beneficial tool to evaluate the effect of novel treatments for Huntington's disease (HD). The aim of the present study was to demonstrate the neuroprotective effect of recombinant human erythropoietin (rhEPO) and interferon-beta-1b (IFN-ß-1b) in 3-NP-induced neurotoxicity in rats. Rats were injected with 3-NP (10 mg/kg/day, i.p) for 2 weeks and were divided into five subgroups; the first served as the HD group, the second received rhEPO (5000 IU/kg/every other day, i.p.) for 2 weeks, the third received rhEPO starting from the 5th day of 3-NP injection, the fourth received IFN-ß-1b (300,000 units, every day other day, s.c) for 2 weeks, and the last received IFN-ß-1b starting from the 5th day of 3-NP injection. All treatments significantly improved motor and behavior performance of rats. Moreover, all treatments markedly restored mitochondrial function as well as brain-derived neurotrophic factor level, and reduced oxidative stress biomarkers, pro-inflammatory mediators, nuclear factor kappa B expression, caspase-3, and Bax/Bcl2 ratio in the striatum. In conclusion, the present study demonstrates the neuroprotective potential of rhEPO or IFN-ß-1b on 3-NP-induced neurotoxicity in rats. Furthermore, our study suggests that activation of JAK2/STAT3 or JAK1/STAT3 may contribute to the neuroprotective activity of rhEPO or IFN-ß-1b, respectively. We also found that early treatment with rhEPO did not confer any benefits compared with late rhEPO treatment, while early IFN-ß-1b showed a marked significant benefit compared with late IFN-ß-1b.


Subject(s)
Erythropoietin , Neuroprotective Agents , Animals , Erythropoietin/pharmacology , Humans , Interferon beta-1b/pharmacology , Neuroprotective Agents/pharmacology , Nitro Compounds , Propionates , Rats , Rats, Wistar , Signal Transduction
3.
Fundam Clin Pharmacol ; 34(5): 530-547, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32603486

ABSTRACT

Patients with COVID-19 are sometimes already being treated for one or more other chronic conditions, especially if they are elderly. Introducing a treatment against COVID-19, either on an outpatient basis or during hospitalization for more severe cases, raises the question of potential drug-drug interactions. Here, we analyzed the potential or proven risk of the co-administration of drugs used for the most common chronic diseases and those currently offered as treatment or undergoing therapeutic trials for COVID-19. Practical recommendations are offered, where possible.


Subject(s)
Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Prescription Drugs/pharmacology , Analgesics/pharmacology , Anti-Asthmatic Agents/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Anticoagulants/pharmacology , Antineoplastic Agents/pharmacology , Antitubercular Agents/pharmacology , Antiviral Agents/pharmacology , Betacoronavirus , COVID-19 , Cardiovascular Agents/pharmacology , Drug Interactions , Humans , Hydroxychloroquine/pharmacology , Hypoglycemic Agents/pharmacology , Hypolipidemic Agents/pharmacology , Interferon beta-1b/pharmacology , Pandemics , Prescription Drugs/pharmacokinetics , Psychotropic Drugs/pharmacology , Receptors, Interleukin/antagonists & inhibitors , Risk Assessment , SARS-CoV-2 , Thyroid Hormones/pharmacology , COVID-19 Drug Treatment
4.
J Immunol ; 203(8): 2222-2238, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31527198

ABSTRACT

Microglia being the resident macrophage of brain provides neuroprotection following diverse microbial infections. Japanese encephalitis virus (JEV) invades the CNS, resulting in neuroinflammation, which turns the neuroprotective role of microglia detrimental as characterized by increased microglial activation and neuronal death. Several host factors, including microRNAs, play vital roles in regulating virus-induced inflammation. In the current study, we demonstrate that the expression of miR-301a is increased in JEV-infected microglial cells and human brain. Overexpression of miR-301a augments the JEV-induced inflammatory response, whereas inhibition of miR-301a completely reverses the effects. Mechanistically, NF-κB-repressing factor (NKRF) functioning as inhibitor of NF-κB activation is identified as a potential target of miR-301a in JEV infection. Consequently, miR-301a-mediated inhibition of NKRF enhances nuclear translocation of NF-κB, which, in turn, resulted in amplified inflammatory response. Conversely, NKRF overexpression in miR-301a-inhibited condition restores nuclear accumulation of NF-κB to a basal level. We also observed that JEV infection induces classical activation (M1) of microglia that drives the production of proinflammatory cytokines while suppressing alternative activation (M2) that could serve to dampen the inflammatory response. Furthermore, in vivo neutralization of miR-301a in mouse brain restores NKRF expression, thereby reducing inflammatory response, microglial activation, and neuronal apoptosis. Thus, our study suggests that the JEV-induced expression of miR-301a positively regulates inflammatory response by suppressing NKRF production, which might be targeted to manage viral-induced neuroinflammation.


Subject(s)
Antiviral Agents/pharmacology , Encephalitis Virus, Japanese/drug effects , Encephalitis Virus, Japanese/immunology , Encephalitis, Japanese/drug therapy , Encephalitis, Japanese/immunology , Interferon beta-1b/pharmacology , MicroRNAs/metabolism , Repressor Proteins/antagonists & inhibitors , Animals , Cells, Cultured , Encephalitis Virus, Japanese/metabolism , Encephalitis, Japanese/metabolism , Female , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Male , Mice , Mice, Inbred BALB C , MicroRNAs/genetics , Repressor Proteins/metabolism
5.
J Neuroimmunol ; 337: 577062, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31521828

ABSTRACT

Interleukins (ILs)-22, 32α and 34 were monitored in the sera of relapsing-remitting multiple sclerosis (RRMS) patients at different time intervals with or without interferon ß-1b, interferon ß-1a and fingolimod treatments. The results showed that sera of untreated RRMS patients were statistically higher in concentration of IL-22 (P < .001), but not IL-32α and IL-34, than those of healthy individuals. Interestingly, interferon ß-1b, interferon ß-1a and fingolimod treatments led to a significant decrease of serum concentrations of ILs-22 and 32α, but not 34, at 6 and 12 months of treatment, compared to their initial concentrations before initiating therapy. The correlation analysis revealed that the changes of serum IL-22 (r = 0.814) and, to a lesser extent, IL-32α (r = 0.381) concentrations were positively correlated with those of expanded disability status score. In conclusion, serum IL-22 concentration may be a potential marker for MS disease severity and efficacy of treatment.


Subject(s)
Fingolimod Hydrochloride/therapeutic use , Interferon beta-1a/therapeutic use , Interferon beta-1b/therapeutic use , Interleukins/blood , Multiple Sclerosis, Relapsing-Remitting/blood , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Adolescent , Adult , Biomarkers/blood , Female , Fingolimod Hydrochloride/pharmacology , Humans , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Interferon beta-1a/pharmacology , Interferon beta-1b/pharmacology , Interleukins/antagonists & inhibitors , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Random Allocation , Young Adult , Interleukin-22
6.
J Med Internet Res ; 21(7): e14373, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31359863

ABSTRACT

BACKGROUND: Accurate measurement of medication adherence using classical observational studies typically depends on patient self-reporting and is often costly and slow. In contrast, digital observational studies that collect data directly from the patient may pose minimal burden to patients while facilitating accurate, timely, and cost-efficient collection of real-world data. In Germany, ~80% of patients with multiple sclerosis (MS) treated with interferon beta 1b (Betaferon) use an electronic autoinjector (BETACONNECT), which automatically records every injection. Patients may also choose to use a medical app (myBETAapp) to document injection data and their well-being (using a "wellness tracker" feature). OBJECTIVE: The goal of this pilot study was to establish a digital study process that allows the collection of medication usage data and to assess medication usage among patients with MS treated with interferon beta-1b who use myBETAapp. METHODS: The PROmyBETAapp digital observational study was a mixed prospective and retrospective, noninterventional, cohort study conducted among users of myBETAapp in Germany (as of December 2017: registered accounts N=1334; actively used accounts N=522). Between September and December 2017, users received two invitations on their app asking them to participate. Interested patients were provided detailed information and completed an electronic consent process. Data from consenting patients' devices were collected retrospectively starting from the first day of usage if historical data were available in the database and collected prospectively following consent attainment. In total, 6 months of data on medication usage behavior were collected along with 3 months of wellness tracker data. Descriptive statistics were used to analyze persistence, compliance, and adherence to therapy. RESULTS: Of the 1334 registered accounts, 96 patients (7.2%) provided informed consent to participate in the study. Of these, one patient withdrew consent later. For another patient, injection data could not be recorded during the study period. Follow-up of the remaining 94 patients ended in May 2018. The mean age of participants was 46.6 years, and 50 (53%) were female. Over the 6-month study period, persistence with myBETAapp usage was 96% (90/94), mean compliance was 94% of injections completed, and adherence (persistence and ≥80% compliance) was 89% (84/94). There was no apparent difference between male and female participants and no trend across age groups. The wellness tracker was used by 21% of participants (20/94), with a mean of 3.1 entries per user. CONCLUSIONS: This study provides important information on medication usage among patients with MS treated with interferon beta-1b and on consenting behavior of patients in digital studies. In future studies, this approach may allow patients' feedback to be rapidly implemented in existing digital solutions. TRIAL REGISTRATION: ClinicalTrials.gov NCT03134573; https://clinicaltrials.gov/ct2/show/NCT03134573.


Subject(s)
Interferon beta-1b/therapeutic use , Medication Adherence/statistics & numerical data , Mobile Applications/standards , Multiple Sclerosis/drug therapy , Cohort Studies , Female , Humans , Interferon beta-1b/pharmacology , Male , Middle Aged , Multiple Sclerosis/pathology , Pilot Projects , Prospective Studies , Retrospective Studies
7.
AAPS J ; 21(2): 26, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30737590

ABSTRACT

Recombinant human interferon-ß (rhIFN-ß) therapy is the first-line treatment in relapsing-remitting forms of multiple sclerosis (MS). The mechanism of action underlying its therapeutic activity is only partially understood as IFN-ßs induce the expression of over 1000 genes modifying multiple immune pathways. Currently, assessment of potency for IFN-ß products is based on their antiviral effect, which is not linked to its therapeutic effect. Here, we explore the use of a multiplexed gene expression system to more broadly characterize IFN-ß bioactivity. We find that MM6 cells stimulated with US-licensed rhIFN-ßs induce a dose-dependent and reproducible pattern of gene expression. This pattern of gene expression was used to compare the bioactivity profile of biosimilar candidates with the corresponding US-licensed rhIFN-ß products, Rebif and Betaseron. While the biosimilar candidate for Rebif matched the pattern of gene expression, there were differences in the expression of a subset of interferon-inducible genes including CXCL-10, CXCL-11, and GBP1 induced by the biosimilar candidate for Betaseron. Assessment of product impurities in both products suggested that the difference was rooted in the presence of innate immune response modulating impurities (IIRMIs) in the licensed product. These studies indicate that determining the expression levels for an array of reporter genes that monitor different pathways can be informative as part of the demonstration of biosimilarity or comparability for complex immunomodulatory products such as IFN-ß, but the sensitivity of each gene to potential impurities in the product should be examined to fully understand the results.


Subject(s)
Adjuvants, Immunologic/pharmacology , Biosimilar Pharmaceuticals/pharmacology , Drug Contamination , Gene Expression/drug effects , Immunity, Innate/drug effects , Cell Line, Tumor , Gene Expression Profiling/methods , Humans , Interferon beta-1a/pharmacology , Interferon beta-1b/pharmacology , Recombinant Proteins/pharmacology
8.
Mult Scler ; 25(6): 837-847, 2019 05.
Article in English | MEDLINE | ID: mdl-29761737

ABSTRACT

BACKGROUND: Long-term follow-up from the randomized trial of interferon beta-1b (IFNB-1b) permitted the assessment of different definitions of no evidence of disease activity (NEDA) for predicting long-term outcome in multiple sclerosis (MS). OBJECTIVE: To examine the predictive validity of different NEDA definitions. METHODS: Predictive validity for negative disability outcomes (NDOs) at 16 years and survival at 21 years post-randomization were assessed. NEDA in the first 2 years was defined as follows: clinical NEDA: no relapses or Expanded Disability Status Scale (EDSS) progression from baseline to Year 2; NEDA-3a: no relapses, no confirmed ⩾1-point EDSS progression, and no new T2-active lesions; NEDA-3b: no relapses, no EDSS progression, and no increase in T2 burden of disease (T2-BOD); and NEDA-4: no relapses, no EDSS progression, and no increase in T2-BOD or atrophy. NDOs were defined as death, need for wheelchair, EDSS ⩾6, or progressive MS. RESULTS: A total of 245 and 371 patients were evaluated at 16 and 21 years, respectively. Clinical NEDA predicted NDOs ( p = 0.0029), as did baseline EDSS ( p < 0.0001), baseline T2-BOD ( p < 0.0001), and change in T2-BOD ( p = 0.0033). IFNB-1b treatment ( p = 0.0251), relapse rate in the 2 years before study start ( p = 0.0260), T2-BOD at baseline ( p = 0.0014), and change in T2-BOD ( p = 0.0129) predicted survival at 21 years. CONCLUSION: Clinical NEDA predicted long-term disability outcome. By contrast, definitions of NEDA that included on-therapy changes in magnetic resonance imaging variables did not increase the predictive validity.


Subject(s)
Adjuvants, Immunologic/pharmacology , Disease Progression , Interferon beta-1b/pharmacology , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Severity of Illness Index , Adult , Female , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/physiopathology , Prognosis , Randomized Controlled Trials as Topic , Reproducibility of Results
9.
Eur J Neurol ; 25(3): 527-534, 2018 03.
Article in English | MEDLINE | ID: mdl-29205701

ABSTRACT

BACKGROUND AND PURPOSE: We have previously shown that patients with multiple sclerosis receiving immunomodulatory treatment have reduced seroprotection rates after influenza immunization. The aim of this study was to further investigate the influence of immunomodulatory therapies on the antibody response and seroprotection rates in patients immunized with seasonal influenza vaccine in 2012/2013 compared with healthy controls. METHODS: Ninety patients receiving fingolimod, glatiramer acetate, interferon beta-1a/1b, natalizumab or no therapy were compared with 62 healthy controls. All subjects received the inactivated split virus vaccine in 2012 and serum samples were collected pre-vaccination and 3, 6 and 12 months post-vaccination. The vaccine responses were evaluated by the hemagglutination inhibition assay and adjusted for age and gender. RESULTS: No significant differences in rates of protection against H1N1 for interferon beta-1a/1b and glatiramer acetate were observed as compared with controls at 3, 6 and 12 months. Fingolimod provided reduced protection at all time points post-vaccination, whereas natalizumab displayed reduced protection at 3 and 6 months. Patients without immunomodulation did not display protection rates that were significantly different from the controls at 3 and 12 months. CONCLUSION: These findings suggest that patients with multiple sclerosis receiving fingolimod or natalizumab should be considered for a second dose of the vaccine in cases of insufficient protection. Our results further indicate that new immunomodulatory treatment regimens should be systematically evaluated for their influence on influenza-specific vaccine responses.


Subject(s)
Antibodies, Viral/blood , Fingolimod Hydrochloride/pharmacology , Glatiramer Acetate/pharmacology , Immunogenicity, Vaccine/immunology , Immunologic Factors/pharmacology , Influenza Vaccines/immunology , Interferon beta-1b/pharmacology , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Natalizumab/pharmacology , Adult , Female , Humans , Male , Middle Aged , Seasons
10.
J Neurol Sci ; 379: 298-303, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28716266

ABSTRACT

Multiple sclerosis (MS) is a chronic demyelinating central nervous system (CNS) disease that involve oligodendrocyte loss and failure to remyelinate damaged brain areas causing a progressive neurological disability. Studies in MS mouse model suggest that cannabinoids ameliorate symptoms as spasticity, tremor and pain reducing inflammation via cannabinoid-mediated system. The aim of our study is to investigate the changes in cannabinoid type 1 (CNR1) and 2 (CNR2) receptors mRNA expression levels and promoter methylation in peripheral blood mononuclear cells (PBMCs) of MS secondary progressive (MSS-SP) patients treated with Sativex®. Our cohort included MSS-SP patients, that at the time of Sativex® treatment, are treated (n=7), not treated (n=11) or that had terminated interferon-ß-1b (IFN-ß-1b) therapy (n=12). By Methylation Sensitive High Resolution Melting (MS-HRM), we characterized the methylation profile of CNR1 and CNR2 promoter region, while the relative mRNA transcript levels of these two genes were evaluated in the same samples by Quantitative Real-Time PCR (qRT-PCR) analysis. We did not find different pattern of cytosine-phosphate-guanine (CpG) methylation in the CNR1/CNR2 promoter region of all MSS-SP patients treated with Sativex®. In addition, CNR1 and CNR2 expression did not significantly differ in MSS-SP patients not treated with IFN-ß-1b vs. them that have suspended, while in MSS-SP patients treated with IFN-ß-1b during Sativex® therapy we found a specific decrease of the CNR2 expression levels. These results suggest that the different expression of cannabinoid receptors by Sativex® treatment in leukocytes might be regulated through a molecular mechanism that involve interferon modulation.


Subject(s)
Cannabidiol/pharmacology , Dronabinol/pharmacology , Leukocytes, Mononuclear/metabolism , Methylation/drug effects , Multiple Sclerosis, Chronic Progressive/genetics , Promoter Regions, Genetic/genetics , Receptor, Cannabinoid, CB1/genetics , Receptor, Cannabinoid, CB2/genetics , Drug Combinations , Female , Humans , Interferon beta-1b/pharmacology , Interferon beta-1b/therapeutic use , Male , Middle Aged
11.
Neurol Sci ; 38(2): 337-342, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27885448

ABSTRACT

Multiple sclerosis (MS) can impair cognitive functions even in the early stages. The Brief International Cognitive Assessment for Multiple Sclerosis (BICAMS) battery is very short and highly sensitive and can be used to evaluate cognitive status in the disease. Several clinical trials have shown beneficial effects of disease-modifying drugs (DMDs) on long-term cognitive measures which may even reduce cognitive deficits in MS patients. Relapsing remitting MS patients using DMDs were enrolled in the study and monitored for 12 months. BICAMS and the Expanded Disability Status Scale were applied to the study group. We evaluated and monitored 161 newly diagnosed cases of definite MS by the end of the trial. 110 patients (68.2%) were female. One hundred and two healthy subjects (female to male ratio 68:34) were enrolled into the study. MS patients were categorized into three DMT groups: IFNB1-a SC, IFNB1-b, and GA. Mean scores of all three cognitive tests (SDMT, BVMT-R, and CVLT-II) were significantly higher in the control group than in the MS patients. The number of cognitively impaired patients decreased from 31.7 to 21.7% on the basis of CVLT (p = 0.024), and 42 (26.1%) to 30 (18.6%) on the basis of BVMT-R at month 12. A significant difference was determined in terms of cognitive status between MS patients using both IFNB and GA and the healthy control group. Ours is the first study to compare IFNB and GA in terms of evaluating cognitive involvement and to use the BICAMS battery in monitoring treatment.


Subject(s)
Cognitive Dysfunction/drug therapy , Immunologic Factors/pharmacology , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Outcome Assessment, Health Care , Severity of Illness Index , Adult , Cognitive Dysfunction/etiology , Female , Glatiramer Acetate/administration & dosage , Glatiramer Acetate/pharmacology , Humans , Immunologic Factors/administration & dosage , Interferon beta-1a/administration & dosage , Interferon beta-1a/pharmacology , Interferon beta-1b/administration & dosage , Interferon beta-1b/pharmacology , Male , Multiple Sclerosis, Relapsing-Remitting/complications , Single-Blind Method , Young Adult
12.
Neurología (Barc., Ed. impr.) ; 31(4): 247-254, mayo 2016. ilus, tab, graf
Article in Spanish | IBECS | ID: ibc-151304

ABSTRACT

INTRODUCCIÓN: El estudio BENEFIT ha mostrado los beneficios del uso precoz del interferón beta 1 b (IFNβ-1b). El objetivo del trabajo fue estimar la eficiencia del tratamiento precoz vs. diferido del IFNβ-1b en pacientes con un síndrome desmielinizante aislado (SDA) indicativo de esclerosis múltiple (EM) en España. MÉTODOS: Se desarrolló un modelo de Markov desde la perspectiva social, con un horizonte temporal de 2 años hasta toda la vida. Una cohorte de 1.000 pacientes con SDA y estados de salud definidos por la Expanded Disability Syndrome Scale (EDSS) fue tratada o no con IFNβ-1b al inicio. Los datos del BENEFIT se usaron para la progresión en la EDSS y las transiciones a EM. Los costes se estimaron de la literatura. Las utilidades derivaron del EQ-5D y publicaciones y la mortalidad de tablas de mortalidad y de la EDSS. Costes (€ de 2013) y resultados se descontaron al 3% anual. Se realizó un análisis de sensibilidad probabilístico. RESULTADOS: En el caso base, tanto la razón de coste utilidad incremental (RCUI) como la razón de coste efectividad incremental (RCEI) del IFNβ-1b vs. no tratamiento fueron dominantes (más eficaz y menos costoso) bajo la perspectiva social. Bajo la perspectiva del SNS, la RCUI fue de 40.702 €/AVAC y la RCEI de 13 €/recaída evitada. CONCLUSIÓN: El tratamiento precoz con IFNβ-1b después de un SDA frente al tratamiento diferido es eficiente desde la perspectiva social, pero puede no ser eficiente desde la perspectiva del SNS al no tener en cuenta los costes no sanitarios


INTRODUCTION: The BENEFIT study has demonstrated the benefits of early treatment with interferon beta 1 b (IFNβ-1b). The objective of this study was to estimate the efficiency of early vs delayed IFNβ-1b treatment in patients with clinically isolated syndrome (CIS) suggestive of multiple sclerosis (MS) in Spain. METHODS: A Markov model reflecting the social perspective was developed with time horizons ranging from 2 years to lifetime. A cohort of 1000 patients with CIS, whose health status had been measured on the Expanded Disability Symptom Scale (EDSS), included patients who received early IFNβ-1b treatment and those who did not. Data from the BENEFIT study were used to model EDSS progression and transitions to MS. Costs were estimated from published literature. Patient utilities were derived from EQ-5D data and published data. Mortality was estimated using life tables and EDSS data. Costs (€ at 2013 rates) and outcomes were discounted at 3% per annum. A probabilistic sensitivity analysis was performed. RESULTS: In the base case, both the incremental cost utility ratio (ICUR) and the incremental cost effectiveness ratio (ICER) of IFNβ-1b versus no treatment were dominant (more effective and less costly) from a social perspective. From the perspective of the Spanish Health System, the ICUR was € 40,702/QALY and the ICER was € 13/relapse avoided. CONCLUSION: Early treatment with IFNβ-1b after a CIS versus delayed treatment is efficient from a social perspective, but it may not be efficient from the perspective of the NHS which does not take non health-related costs into account


Subject(s)
Humans , Male , Female , Adult , Interferon beta-1b/administration & dosage , Interferon beta-1b/pharmacology , Interferon beta-1b/therapeutic use , Cost-Benefit Analysis/economics , Cost-Benefit Analysis , Cost Efficiency Analysis , Multiple Sclerosis/complications , Multiple Sclerosis/prevention & control , Multiple Sclerosis/therapy , Evaluation of the Efficacy-Effectiveness of Interventions , 50303 , Cohort Studies , Spain/epidemiology
13.
J Interferon Cytokine Res ; 36(3): 192-203, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26824268

ABSTRACT

Human serum albumin (HSA)-free formulation of Escherichia coli-derived human interferon beta (IFNß-1b) with a high percentage of monomeric protein and low immunogenicity is developed and characterized in the current study. UV spectroscopy, fluorescence spectroscopy, dynamic light scattering, sodium dodecyl sulfate polyacrylamide gel electrophoresis, Western blotting, Micro-Flow Imaging, resonant mass measurement, size exclusion, and reversed-phase high performance liquid chromatographies were applied to assess the effect of excipients on the stability of IFNß-1b to establish a HSA-free formulation. The antiviral activity of IFNß-1b was evaluated using human lung carcinoma cell line. Immune tolerant mice to hIFNß were used to assess the immunogenicity of the HSA-free formulated IFNß-1b in comparison to Betaferon drug product and Avonex drug substance as standards through IgG titering of plasma. HSA-free formulated IFNß-1b, including 200 mM L-arginine, 200 mM trehalose, and 0.1% n-dodecyl ß-D-maltoside in 10 mM sodium acetate buffer, pH 7.4, showed the highest biological activity. The stability of IFNß-1b in the HSA-free formulation was monitored for 3 weeks at 4°C and 37°C with relative humidity of 10% and 75%, respectively. Protein aggregation and immunogenicity in transgenic mice were decreased in the HSA-free formulated IFNß-1b compared to Betaferon. The stability, biological activity, and immunogenicity of the HSA-free formulation and Betaferon were evaluated. Incubation of formulations at 4°C and 37°C for 3 weeks showed that the HSA-free formulated IFNß-1b was more stable and less immunogenic in transgenic FVB/N mice. Low immunogenicity and the absence of HSA, which reduces the potential risk of viral infection (eg, HIV and HCV), are promising for clinical studies.


Subject(s)
Antibodies, Viral/blood , Antiviral Agents/pharmacology , Encephalomyocarditis virus/drug effects , Interferon beta-1b/pharmacology , A549 Cells , Animals , Antiviral Agents/immunology , Antiviral Agents/isolation & purification , Cloning, Molecular , Encephalomyocarditis virus/growth & development , Enzyme-Linked Immunosorbent Assay , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Immune Tolerance , Interferon beta-1b/biosynthesis , Interferon beta-1b/isolation & purification , Mice , Mice, Transgenic , Protein Stability , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology
14.
Folia Morphol (Warsz) ; 75(2): 257-263, 2016.
Article in English | MEDLINE | ID: mdl-26711647

ABSTRACT

BACKGROUND: Interferon beta (IFNb) was the first proven drug for the treatment of multiple sclerosis (MS). The diagnosis of MS frequently occurs in women at childbearing age (especially in twenties and thirties). Therefore, the pregnancy process is major concern for many women with MS. Data on women exposed to IFNb during pregnancy are limited. The aim of our study was to investigate the teratogenic potential of IFNb on embryonic development via embryo culture technique. Recently, this technique has been often used for determining teratogenic effect of pharmacologic drugs and potential teratogens on embryonic development. MATERIALS AND METHODS: In this study, IFNb was applied to the culture medium and after 48 h of culture effects of IFNbs (1000 IU/IFNb-1a and 1000 IU/IFNb-1b) on embryonic development were morphologically investigated. RESULTS: According to morphologic scoring system, total morphologic score, somite number and protein contents were similar between control group and two experimental groups (p > 0.05). On the other hand, yolk sac diameter, crown- -rump length and head length were significantly decreased in two experimental groups compared with control group (p < 0.05). CONCLUSIONS: Consequently, IFNb-1a and IFNb-1b, applied to the culture medium, have no macroscopic teratogenic effect on embryonic development. However, in respect of morphometric measurements, IFNb-1a and IFNb-1b have caused growth retardation in embryo. This research related to interferon was the first study using vitro embryo culture technique; thus, in our point of view, future studies which will be performed by using different doses of IFN will contribute to the literature.


Subject(s)
Interferon beta-1a/pharmacology , Interferon beta-1b/pharmacology , Female , Humans , Multiple Sclerosis
15.
J Neurol ; 262(11): 2466-71, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26239222

ABSTRACT

Multiple sclerosis (MS) is a chronic demyelinating neurodegenerative disease of the CNS that requires long-term treatment. The identification of patient characteristics that can help predict disease outcomes could improve care for patients with MS. The objective of this study is to identify predictors of disease activity in patients from the BEYOND trial. This regression analysis of patients with relapsing-remitting MS from BEYOND examined the predictive value of patient characteristics at baseline and after 1 year of treatment with interferon beta-1b 250 µg every other day for clinical and MRI outcomes after year 1 of the study. 857 and 765 patients were included in the analyses of clinical and MRI outcomes, respectively. In multivariate analyses of age, a higher number of relapses in the past 2 years, ≥3 new MRI lesions in the first year, and, especially, a higher number of relapses in year 1 predicted the future occurrence of relapses. By contrast, age, MRI activity, and the presence of neutralizing antibodies in the first year were principally predictive of future MRI activity. In patients with continued clinical disease activity or substantial MRI activity on therapy, an alternative therapeutic approach should be strongly considered.


Subject(s)
Adjuvants, Immunologic/pharmacology , Interferon beta-1b/pharmacology , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Outcome Assessment, Health Care , Adjuvants, Immunologic/administration & dosage , Adult , Age Factors , Female , Follow-Up Studies , Humans , Interferon beta-1b/administration & dosage , Magnetic Resonance Imaging , Male , Middle Aged , Prognosis , Recurrence
16.
Clin Exp Immunol ; 182(2): 162-72, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26212126

ABSTRACT

Multiple sclerosis (MS) is an inflammatory, demyelinating and neurodegenerative disease triggered by infiltration of activated T cells into the central nervous system. Interferon (IFN)-ß is an established, safe and effective treatment for patients with relapsing-remitting MS (RRMS). The cytokine can inhibit leucocyte infiltration into the central nervous system; however, little is known about the precise molecular mechanisms. Previously, in vitro application of IFN-ß1b was shown to reduce CXCL12/CXCR4-mediated monocyte migration. Here, we analysed the effects of IFN-ß1b on CXCR4-dependent T cell function. In vitro exposure to IFN-ß1b (1000 U/ml) for 20 h reduced CXCR4-dependent chemotaxis of primary human T cells from healthy individuals and patients with RRMS. Investigating the IFN-ß1b/CXCR4 signalling pathways, we found no difference in phosphorylation of ZAP70, ERK1/2 and AKT despite an early induction of the negative regulator of G-protein signalling, RGS1 by IFN-ß1b. However, CXCR4 surface expression was reduced. Quantitative real time-PCR revealed a similar reduction in CXCR4-mRNA, and the requirement of several hours' exposure to IFN-ß1b supports a transcriptional regulation. Interestingly, T cells from MS patients showed a lower CXCR4 expression than T cells from healthy controls, which was not reduced further in patients under IFN-ß1b therapy. Furthermore, we observed no change in CXCL12-dependent chemotaxis in RRMS patients. Our results demonstrate clearly that IFN-ß1b can impair the functional response to CXCR4 by down-regulating its expression, but also points to the complex in vivo effects of IFN-ß1b therapy.


Subject(s)
Chemotaxis/drug effects , Interferon beta-1b/pharmacology , Receptors, CXCR4/metabolism , T-Lymphocytes/drug effects , Adult , Blotting, Western , Cell Movement/drug effects , Cells, Cultured , Female , Gene Expression/drug effects , Gene Expression/immunology , Humans , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/genetics , Multiple Sclerosis, Relapsing-Remitting/immunology , Receptors, CXCR4/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Young Adult
17.
Psychiatry Clin Neurosci ; 69(10): 649-57, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25907350

ABSTRACT

AIMS: The aim of this study was to evaluate the effect of various disease-modifying therapies (DMT) on quality of life in multiple sclerosis (MS). METHODS: This was a three-arm parallel study with balanced randomization in which 90 newly diagnosed, definite MS subjects referred to Ghaem Medical Center, Mashhad, Iran were enrolled between 2006 and 2009. Patients were randomly allocated into three DMT groups: Avonex, Rebif and Betaferon. Health-related quality of life was assessed in MS patients at baseline and 12 months after treatment with DMT using the MS Quality of Life-54 questionnaire. RESULTS: Both mental and physical health scores improved within all three treatment groups after 12 months of treatment; however, this increase was only significant in the mental health composite in the Betaferon group (P = 0.024). Betaferon had the highest mental health score change (14.04) while this change was 7.26 for Avonex (P = 0.031) and 5.08 for Rebif (P = 0.017). A physical health composite score comparison among the three treatment groups revealed no significant results. CONCLUSIONS: With a positive impact of DMT on mental and physical dimensions of QOL in MS patients, initiation of treatment soon after diagnosis is recommended. In MS patients with more mental issues and fewer physical disabilities, Betaferon might be considered as a better choice of treatment.


Subject(s)
Adjuvants, Immunologic/pharmacology , Interferon beta-1a/pharmacology , Interferon beta-1b/pharmacology , Multiple Sclerosis/drug therapy , Outcome Assessment, Health Care , Quality of Life , Adjuvants, Immunologic/administration & dosage , Adult , Female , Humans , Interferon beta-1a/administration & dosage , Interferon beta-1b/administration & dosage , Male , Single-Blind Method
18.
Clin Ter ; 166(1): e62-6, 2015.
Article in English | MEDLINE | ID: mdl-25756269

ABSTRACT

Chemokine (C-X-C motif) receptor 3 (CXCR3) and its ligands (MIG, IP-10) play an important role in multiple sclerosis (MS). The CXCR3 receptor is expressed on the majority of T cells in the cerebrospinal fluid (CSF) of patients with MS, suggesting that the CXCR3 receptor may mediate the trafficking of T cells into the central nervous system. IP-10, and MIG were found to be elevated in the CSF of patients with MS during relapse. These chemokines were also detected in actively demyelinating lesions, and upregulation of CXCR3 expression on peripheral blood CD4+ lymphocytes was associated with MS relapses.Treatment with Interferon (IFN)-ß-1a or IFN-ß-1b was associated with increased IP-10. Natalizumab that exerts impressive therapeutic effects in patients with MS induces a marked decline of Th1 chemokines (MIG, IP-10, I-TAC) in CSF.


Subject(s)
Cytokines/metabolism , Multiple Sclerosis/physiopathology , Receptors, CXCR3/biosynthesis , Adult , Cerebrospinal Fluid/metabolism , Chemokine CXCL10/biosynthesis , Chemokine CXCL9/biosynthesis , Humans , Interferon beta-1a/pharmacology , Interferon beta-1b/pharmacology , Ligands , Male , T-Lymphocytes/metabolism
19.
Hum Mol Genet ; 24(11): 3192-205, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25721402

ABSTRACT

We analysed gene expression microarray data from whole blood samples from 228 multiple sclerosis (MS) patients either untreated or treated with one of three alternative commonly used interferon beta (IFNß) disease modifying drugs: Avonex (×1 weekly), Betaseron (every second day) or Rebif (×3 weekly). Patient injections were not timed to coordinate sample collections, thus providing a global transcriptomic profile for each population of patients studied. Three hundred and fifty one genes were significantly differentially expressed by at least one of the IFNß drugs. Despite the different drug sources with distinct injection and dosage protocols, a striking similarity was found in the identity and functional classes of the differentially expressed genes induced. Using the 25 most-upregulated genes, we defined a robust IFNß gene expression signature that quantifies the IFN activation state per blood sample collected irrespective of the type of IFNß therapy. This 25-gene signature also defined basal IFN activation states among untreated MS patients, which differed among individuals but remained relatively constant per patient with time. The maximum drug-induced IFN-activation state was similar for all three drugs despite a 1.7-2.0-fold diminished average effect for Avonex. This and a more erratic effect of Avonex per patient across longitudinal measurements is likely a result of its reduced injection frequency. In summary, we have defined a robust blood-derived type I IFN gene signature from MS patients. This signature could potentially serve to generically quantify the systemic Type I IFN activation status for any other clinical manifestation, inclusive of other autoimmune diseases.


Subject(s)
Adjuvants, Immunologic/pharmacology , Interferon beta-1a/pharmacology , Interferon beta-1b/pharmacology , Multiple Sclerosis/blood , Transcriptome/drug effects , Adjuvants, Immunologic/therapeutic use , Humans , Interferon beta-1a/therapeutic use , Interferon beta-1b/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...