Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Aug 15.
Published in final edited form as: J Neurol Sci. 2011 Jun 11;307(1-2):41–45. doi: 10.1016/j.jns.2011.05.024

Quantitative differences in the immunomodulatory effects of Rebif and Avonex in IFN-β 1a treated multiple sclerosis patients

George P Christophi 2,5, Jennifer A Christophi 4, Ross C Gruber 3, Cornelia Mihai 1, Luis J Mejico 1, Paul T Massa 1,2, Burk Jubelt 1,2,*
PMCID: PMC3395433  NIHMSID: NIHMS305631  PMID: 21658727

Abstract

Interferon-β (IFN-β) is a current effective treatment for multiple sclerosis (MS) and exerts its therapeutic effects by down-modulating the systemic immune response and cytokine signaling. In clinical practice there are several formulations of interferon including a low dose of IFN-β 1a formulation of 30μg IM once weekly (Avonex) and a high dose formulation of 44 μg SC three times weekly (Rebif). Recent studies suggest that Rebif is more efficacious compared to Avonex in preventing relapses and decreasing MRI activity in relapsing remitting MS (RRMS) patients. This study examines whether there are quantitative gene expression changes in interferon-treated RRMS patients that can explain the difference in efficacy and side effects between Rebif and Avonex. Herein, RRMS patients were treated for three months with IFN-β 1a and the levels of plasma cytokines and gene expression in peripheral blood mononuclear cells were examined. Thirty-two normal subjects were compared to thirty-two RRMS patients, of which ten were treated with Rebif and ten with Avonex. Rebif and Avonex both significantly and equally suppressed plasma TNF-α and IL-6 levels. Rebif suppressed IL-13 significantly more than Avonex. Rebif also significantly suppressed the levels of the chemokines CCL17 and RANTES, the protease ADAM8, and COX-2 at a higher degree compared to Avonex. The STAT1-inducible genes IP-10 and caspase 1 were significantly increased with Rebif compared to Avonex. In conclusion, the higher dosed, more frequently administered IFN-β 1a Rebif when compared to IFN β-1a Avonex has more potent immunomodulatory effects. These quantitative results might relate to efficacy and side-effect profile of the two IFN-β 1a formulations and provide prospective practical clinical tools to monitor treatment and adjust dosage.

Keywords: Multiple sclerosis, CNS Demyelination, Autoimmunity interferon, dosage, efficacy, cytokines, inflammatory gene expression, Signaling, Signal Transducers and Activators of Transcription STAT, nuclear factor kappa-B (NF-κB), peripheral immune cells, EAE/TMEV

INTRODUCTION

Multiple Sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS) that remains a major cause of disability (1). However, disease-modifying agents that decrease exacerbation rates and slow progression of disability are now available for use in treatment (26). One of these agents is interferon-β 1a (IFN-β 1a), which reduces the frequency of clinical exacerbations, reduces magnetic resonance imaging (MRI) disease activity, and slows disease progression (79).

IFN-β is a pleiotropic cytokine with diverse mechanisms of action including antiviral, immune-stimulating and immunosuppressive actions (10, 11). IFN β primarily signals by phosphorylating the tyrosine residues of the Janus kinase (JAK) and signal transducers and activators of transcription (STAT) proteins like STAT1 and STAT3, which leads to transcription factor translocation into the nucleus promoting gene expression (12). Multiple mechanisms in which IFN-β modulates the immune response have been reported, including down regulation of class II major histocompatiability complex (MHC) molecules and limiting the migration of immune cells across the blood brain barrier (BBB) (1316). Furthermore, IFN-β acts to decrease the elevated blood levels of T cell-derived cytokines IFN-γ, IL-4/IL13 and TNF-α seen in MS (1720), which otherwise act to increase the proinflammatory transcription factors STAT 1, STAT 6 and NF-κB in MS patients (12, 14, 2124).

IFN-β 1a treatment of patients with RRMS in vivo and also in vitro leukocyte cultures can reduce the activation of the transcription factors NF-κB and STAT6 and decrease downstream inflammatory gene expression regulated by those transcription factors (25). However, IFN-β 1a (Rebif) treatment signals directly to activate the transcription factor STAT1 and increase the expression genes containing STAT1 responsive sites in their promoter. Additionally, IFN-β induces the activation of intracellular regulatory proteins. The inhibitory effects of IFN-β 1a treatment on cytokine signaling are at least partly mediated by the induced expression of the tyrosine phosphatase SHP-1 that acts as a broad negative regulator of STAT-1, STAT6, and NF-κB signaling and results in decreased inflammatory gene expression (25).

IFN-β 1a is approved to treat MS patients with two different dosages and delivery methods. The Interferon-β 1a Avonex is delivered at 30 μg intramuscularly (im) once a week (7), while Interferon-β 1a Rebif is delivered at 44 μg subcutaneously (sc) three times a week (3). Clinically comparative studies have shown that the high-dosed, more frequently administered Rebif is significantly more effective than the low-dosed, less frequently administered Avonex (9, 26). Specifically, after 24 weeks of treatment 75 % of Rebif patients did not have a relapse versus 63% of patients taking Avonex, with an odds ratio of 1.9 (1.3–2.6) and 48% of Rebif treated patients had no new MRI activity versus 33% of patients treated with Avonex (p=0.0001). This superior efficacy of Rebif over Avonex could still be demonstrated at an additional medium time on study of up to 64 weeks (27). In addition, disease activity in the low dosed IFN-β 1a Avonex group could be partially suppressed by switching subjects to the high dosed IFN-β 1a Rebif (28). On the other hand, Rebif is associated with increased side effects compared to Avonex such as injection site irritation and mild flu-like symptoms (26, 29). Importantly, some studies demonstrate that different formulations and dosages of IFN-β treatment elicit different gene and protein expression profiles that might relate to treatment efficacy (3032).

The present study investigates whether the pronounced clinical efficacy of high-dosed IFN-β 1a Rebif compared to the low-dosed IFN-β 1a Avonex is reflected in plasma cytokine and inflammatory gene expression levels in PBMCs that have been previously implicated in the pathogenesis of MS and are regulated by interferon treatment. Furthermore, we evaluated whether we could detect deferential and quantitative gene expression changes that could provide a reliable marker for IFN-β 1a response or dosage adjustment. Here we demonstrate that three-month treatment with Rebif had more potent immunomodulatory activity compared to Avonex. Rebif was more effective compared to Avonex at reducing expression of several NF-κB-responsive inflammatory genes including the chemokines CCL17 and RANTES, the protease ADAM8, and COX2, which are implicated in MS pathogenesis. Furthermore, Rebif was significantly more effective compared to Avonex at inducing the STAT1-inducible chemokine IP-10 and caspase 1. The differential modulation of these genes could at least partially contribute to the pronounced clinical efficacy of the high-dosed IFN-β 1a in RRMS and also provide novel molecular tools to monitor therapeutic effects and adjust dosage.

METHODS

Patient selection

Patients were clinically diagnosed as having definite MS (33) of the relapsing-remitting (RR) type (34). All patients selected had not received any disease modifying treatment, IFN-β, glatiramir acetate, steroids, or other immunosuppressive agents, at least three months prior to donating blood. RRMS patients gave blood before and after a three-month treatment with recombinant IFN-β 1a (Rebif or Avonex). Table I provides additional information on the patients and normal subjects used in this study. The average age of the patients was 40 years old, 70% female patients, an average age at onset of 35 years, and an average EDSS score just before treatment of 2.5. There was no significant difference among the various groups and patient characteristics were similar to previous relevant studies (9). The Institutional Review Board of SUNY Upstate University approved all studies and both patients and normal controls granted informed consent before providing blood.

Table I.

Biometric data of MS patients and normal subjects used in the study.

Patient Category Number of Subjects Age (Years) Gender Age at Onset Disease Duration EDSS Score
Normal Subjects 32 41±12 22F, 10M --- --- ---
RR MS 32 41±9 29F, 13M 36±9 5.9± 5 2.5±1.4
REBIF Tx 10 38±9 7F, 3M 34±8 4.0±2 2.4±1.7
AVONEX Tx 10 43±8 7F, 3M 35±9 6.8±4 2.5±0.9

RR MS are untreated Relapsing Remitting multiple sclerosis patients. The data are shown in mean value ± SD. For the gender, F stands for females and M stands for males. The age at onset of clinically diagnosed MS and disease duration are shown in years. The clinical symptoms were measured on Kurtzuke’s Expanded Disability Status Scale (EDSS) at the time the initial blood was drawn. Patients with RR MS gave blood before and after a three-month treatment with recombinant interferon β-1a (Rebif or Avonex) and EDSS scores shown here were calculated just before treatment with interferon β-1a.

PBMC isolation

Patients and normal subjects donated 60 ml of blood collected in heparinized tubes. Blood was diluted 1:1 with HBSS and overlaid onto lymphocyte separation medium (Cellgro, Herndon, VA). After centrifugation, the plasma was collected and used to quantify cytokine levels, while the 10 ml of the interface containing the PBMCs were collected and washed twice with HBSS (12). Freshly isolated cells were suspended in STAT- 60 (Tel-Test, Friendswood, TX) for RNA extraction. Plasma was used for cytokine analysis using ELISA.

Real-Time RT-PCR

Total RNA was isolated using RNA STAT-60. RNA was quantified spectrophotometrically and 0.5 μg of total RNA was converted into cDNA. Briefly, total RNA and random primers (Invitrogen, Carlsbad, CA) were incubated at 72 degrees for 10 minutes and Superscript II RT enzyme (Invitrogen, Carlsbad, CA) was used for reverse transcription (35). cDNA was used for quantitative real time PCR using SYBR Green kit (Abgene, Epson, UK). Serial dilutions of cDNA containing a known copy number of each gene were used in each quantitative PCR run to generate a standard curve relating copy number with threshold amplification cycle (36). A blank/negative control (cDNA reaction without RNA) was run with each RT-PCR assay and the samples always had lower threshold amplification cycle than the negative control. Gene expression levels were calculated during the logarithmic amplification phase by determining the initial mRNA copy number using the standard curve (37). Amplification of each gene specific fragment was confirmed by examination of melting peaks, by agarose gel electrophoresis, and DNA sequencing. The primers used in the study were previously documented (12, 25).

Cytokine ELISA

The levels of the cytokines IFN-γ, IL-4, IL-13, TNF-α and IL-6 were measured using R&D Systems DuoSet ELISA kits (R&D Systems) following the manufacturer’s protocol.

Statistical Analysis

Data are presented in means with standard error values. The p-values were generated using the unpaired Student’s t-test value of less than 0.05, which was chosen to indicate statistical significance between two sample means.

RESULTS

Normal subjects and RRMS patients who were treated with either Rebif or Avonex donated blood before and three months after treatment. First, we quantified the levels of the plasma cytokines by ELISA in normal subjects, untreated RRMS, Rebif-treated RRMS, and Avonex-treated RRMS patients (Table II). The levels of IFN-γ, a cytokine that induces STAT1 activation, were significantly higher in the plasma of untreated MS patients compared to normal subjects, and IFNβ-1a treatment failed to significantly alter IFN-γ levels. The levels of IL-4 and IL-13, cytokines that signal through STAT6, were significantly higher in the plasma of untreated MS patients as compared to normal subjects and treatment with IFN-β 1a had no effect on IL-4 levels. However, treatment with Rebif caused a significant two-fold decrease in IL-13 levels, while Avonex did not, and importantly there was a significant difference between Rebif-treated and Avonex-treated MS patients. Furthermore, the levels of the NF-κB-inducible cytokines TNF-α and IL-6 were significantly higher in untreated MS patients as compared to normal controls. Both Rebif and Avonex significantly reduced IL-6 and TNF-α levels, but there was not a significant difference between the two treatments.

Table II.

Inflammatory profile of normal subjects, untreated RR MS patients, and in vivo three-month treated RR MS patients with IFN β-1a Rebif or Avonex.

Gene Normal Subjects Untreated MS Patients Rebif MS Patients Avonex MS Patients
Cytokine Levels in Plasma (pg/mL) IFN-γ 10±4 569±207 467±262 512±311
IL-4 12±7 201±81 207±102 143±78
IL-13 67±26 523±125 212±82*$ 381±113
TNF-α 33±8 390±156 243±92* 267±125*
IL-6 18±5 152±28 97±25* 83±42 *
mRNA levels (relative to normal subjects IP-10 1±0.2 3.2±1.3 12.4±3.3*$ 6.3±2.4*
Caspase 1 1±0.3 2.5±0.6 6.3±2.0* 4.2±2.6
RANTES 1±0.1 12.3±2.7 4.4±1.9*$ 8.7±2.3*
MMP9 1±0.2 4.2±0.9 2.2±0.7* 3.3±1.0
VCAM1 1±0.4 6.4±2.1 2.5±1.2* 4.1±1.7
COX-2 1±0.1 7.2±1.8 3.8±1.3*$ 5.1±0.6*
CCL17 1±0.2 4.1±0.6 1.6±0.5*$ 3.0±0.9
ADAM8 1±0.4 4.9±1.0 2.3±1.2*$ 3.8±0.7*
Arginase I 1±0.2 3.7±0.6 2.1±0.9* 2.4±0.8*
CXCR3 1±0.2 1.7±0.5 1.3±0.2 1.9±0.6
CCR4 1±0.3 1.5±0.4 1.2±0.7 1.3±0.4
SHP-1 1±0.1 0.5±0.1 2.2±0.4* 1.7±0.4*
Actin 1±0.1 1.0±0.2 1.1±0.3 1.2±0.3
GAPDH 1±0.1 1.1±0.2 0.9±0.2 1.1±0.3

Several inflammatory indicators were quantified in the plasma or in freshly isolated PBMCs of normal subjects (n=32), untreated RR MS patients (n=32), and Rebif-treated RR MS patients (n=10) or Avonex (n=10). The levels of the cytokines IFN-γ, TNF-α, IL-6, IL-4, and IL-13 were quantified in the plasma by ELISA and are recorded in pg cytokine per mL plasma. The mRNA levels of chemokine IP-10/CXCL10, Caspase 1, the T-cell chemokine RANTES, the metalloprotease MMP9, the vascular adhesion molecule VCAM 1, the cyclooxygenase 2 (COX-2), the chemokine CCL17/TARC, the protease ADAM8, the enzyme arginase I, the chemokine receptors CXCR3 and CCR4, the tyrosine protein phosphatase SHP-1, and the housekeeping control genes beta-actin and GAPDH were quantified by real-time RT-PCR in freshly isolated PBMCs of normal subjects, MS patients, and IFN-β treated MS patients. The mRNA levels were normalized to the levels of normal subjects that are shown as 1. The values indicate statistical means ± standard error values,

indicates significance of P<0.05 between normal subjects and untreated MS patients and

*

indicates significance of P<0.05 between untreated and IFN-β treated MS patients, and

$

indicates significance between Rebif treated and Avonex treated patients.

Next, it was important to quantify the levels of several cytokine-inducible genes that where shown to be modulated in MS by interferon treatment (25, 38, 39). Several genes were quantified by real time RT-PCR in freshly isolated PBMC of normal subjects, RRMS patients and in three-month treated Rebif or Avonex RRMS patients (table II). First, we quantified the expression levels of the STAT1-inducibe genes the chemokine IP-10 (interferon gamma-induced protein)/CXCL10 and caspase 1, which were elevated in PBMC of MS patients as compared to normal subjects, in accordance with previous reports (4042). Treatment with either IFN-β 1a Rebif or Avonex significantly increased the expression of IP-10 and caspase 1, which is not surprising since IFN-β signals primarily to activate STAT1. Importantly, Rebif significantly induced higher levels of IP-10 mRNA compared to Avonex treatment.

Furthermore, we examined the expression of NF-κB responsive genes: the chemokine RANTES (Regulated upon Activation, Normal T-cell Expressed)/CCL5, and Secreted, the metalloprotease MMP9, cyclooxygenase-2 (COX-2), and vascular cell adhesion protein 1 (VCAM1) (Table II). All these genes have been previously implicated in the pathogenesis of MS and have been shown to be elevated in leukocytes and in the CNS of MS patients (12, 4346). As expected, RANTES, MMP9, COX-2, and VCAM1 were significantly elevated in leukocytes of untreated MS patients as compared to normal subjects. The levels of RANTES and COX2 were suppressed following treatment with either Rebif or Avonex compared to untreated patients. Importantly, Rebif treatment reduced RANTES and COX2 to a greater degree compared to Avonex treatment. MMP9 and VCAM1 mRNA levels were significantly reduced by Rebif treatment but not by Avonex treatment compared to untreated patients.

We also quantified the expression of the STAT6/NF-κB responsive genes, the chemokine TARC (Thymus and Activation Regulated Chemokine)/CCL17 and the MBP-cleaving protease ADAM8, and the STAT6 responsive gene arginase I (12, 4750). The expression levels of these genes were significantly higher in untreated MS patients as compared to normal subjects (Table II). Rebif-treated MS patients had significantly lower levels of CCL17, ADAM8 and Arginase I as compared to untreated MS patients. Avonex-treated MS patients had significantly reduced ADAM8 and Arginase 1 levels as compared to normal subjects. Importantly, the reductions seen in CCL17 and ADAM8 were significantly greater in Rebif-treated MS patients as compared to Avonex-treated MS patients.

The phosphatase SHP-1 is deficient in untreated MS patients (12) and Rebif and Avonex treatment resulted in a significant upregulation of SHP-1 (Table II). Although no significant differences were observed between the two different treatments, there was a trend showing Rebif treatment inducing higher levels of SHP-1 compared to Avonex treatment. In addition, we did not observe any differences in the expression levels of the chemokine receptors CXCR3 or CCR4, which are not directly regulated by STAT1, STAT6 or NF-κB. Finally, the levels of the housekeeping control genes beta-actin and GAPDH were similar in all groups serving as an important internal control.

DISCUSSION

The high-dosed, high-frequency IFN-β 1a Rebif is more effective than low-dosed, low-frequency IFN-β 1a Avonex in reducing relapses and MRI activity in patients with RRMS (9, 26). This study aimed to establish an immunological/molecular basis for this difference in efficacy and document collectively possible biomarkers that can potentially explain the difference in efficacy and can be monitor treatment and adjust dosage.

Multiple sclerosis is an immune-mediated disease and leukocytes in the blood and in CNS lesions have increased activation of the transcription factors STAT1, STAT6, and NF-κB that might be the result of increased plasma levels of the corresponding cytokines IFN-γ, IL-4/IL13, and TNF-α (12, 14, 2024, 5157). In turn, these elevated transcription factors drive the expression of several inflammatory genes that might contribute to the enhanced demyelinating activity of CNS-infiltrating leukocytes. Here, we demonstrate that three-month treatment of RRMS patients with Rebif compared to Avonex results in differential and quantitative changes in cytokines and their downstream inflammatory gene expression.

First, we examined and quantified the levels of the plasma cytokines that were shown to be upregulated in MS and correlated with the acute phase of MS (1720). IFN-β 1a treatment significantly decreased TNF-α and IL-6 levels but there were no significant differences between Rebif and Avonex treatment. The levels of the cytokines IFN-γ and IL-4 were not significantly affected following treatment with either agent. However, Rebif treatment caused a significant reduction in plasma IL-13 compared to Avonex treatment. Importantly, IL-13 is highly elevated in lymphocytes of MS patients during relapse and return near baseline during remission (54). Interestingly, mast cells are a main source for IL-13 and are believed to play a major role in lesion formation in MS and EAE (58, 59).

In addition to cytokine levels, IFN-β modulates the activation of the transcription factors STAT1, STAT6, and NF-κB and their downstream inflammatory gene expression (25). IFN-β signals to primarily activate STAT1, which is widely considered an inflammatory transcription factor inducing several genes that mediate CNS demyelination (25). This seems paradoxical in the light that IFN-β has therapeutic effects in MS; however the activation of STAT1 might be related to the incomplete action of IFN-β and its side-effect profile. Importantly, unlike IFN-γ, IFN-β also activates the transcription factor STAT3 (10, 11). In turn, STAT3 signaling could mediate many of the anti-inflammatory effects of IFN-β treatment in MS though mediating and augmenting the anti-inflammatory IL-10/STAT3 signaling pathway, inducing expression of intracellular molecules like SHP-1 that can inhibit cytokine signaling, and mediate NF-κB and STAT6 inhibition (6063). Furthermore, augmented NF-κB activation through either increased TNF-α, IL-1b, IL-17 levels or aberrant signaling is considered to be central in MS pathogenesis and an important target of IFN-β therapy (12, 64, 65). Therefore, the fact that Rebif has more potent inhibitory effect on NF-κB-inducible genes might contribute to enhanced clinical benefit through decreased immune cell infiltration and activation, dismissed expression of neurotoxic molecules, and reduced oligodendrocyte injury.

We examined the expression of STAT1 responsive genes, the chemokine IP-10 that chemo-attracts primarily T-cells into CNS, and caspase 1 that is involved in apoptosis and proteolytic processing of IL-1β (40, 66). Expression levels of IP-10 and caspase 1 were elevated in leukocytes of RRMS patients compared to normal subjects. IP-10 levels were significantly further increased following three-month Rebif treatment as compared to Avonex treatment. These data suggest that the higher frequency and high-dosed of Rebif translates into more potent long-term immunomodulatory effects compared to low-dosed Avonex. Interestingly, the increased levels of the chemokine IP-10 following IFN-β treatment closely correlated with the flu-like symptoms occurring in treated MS patients (40) and several studies report that that Rebif treatment is associated with slightly more frequent flu-like symptoms compared to Avonex (26, 29).

Both Rebif and Avonex treatment had a suppressive effect on the mRNA levels of several inflammatory genes regulated by the transcription factors NF-κB and STAT6 in leukocytes from RRMS patients. However Rebif treatment resulted in a significantly greater reduction in the expression of RANTES, COX2, CCL17 and ADAM8 compared to Avonex. In addition to serving as biomarkers to disease activity, these genes play a major role in MS pathogenesis. For example chemokines play an important role in attracting immune cells into the CNS, enzymes like COX-2 process neurotoxic molecules, and proteases like ADAM 8 can directly degrade myelin (46, 49, 67). The genes that showed differential regulation among Rebif and Avonex are induced through the NF-κB activation, suggesting that Rebif treatment more effectively inhibits the NF-κB activation. These data in all suggest that Rebif treatment is associated with an augmented immunosuppressive response compared to Avonex, which is likely to translate into clinical benefits and at least partly explain the increased efficacy seen in treating MS patients (9).

This study demonstrated that indeed there are underling gene expression changes that parallel the difference in clinical efficacy between the two interferon-β 1a formulations, Rebif and Avonex. At the same time, the study has certain limitations including the small sample size of patients, the fact that gene expression was examined only at a single three-month time point after treatment, and the lack of direct correlation of gene expression levels to clinical outcomes of the patients used in this study. These limitations might partially explain why not all the results were homogeneous and why significant differences were not reached with all the genes examined. Nonetheless, this study clearly demonstrates that Rebif and Avonex treatment result in differential gene expression changes that correlate to previously documented clinical responses. More studies are needed to firmly establish a direct correlation between clinical outcomes and gene expression profiles following interferon-β 1a treatment. Therefore, it is important that this study can provide the groundwork and contribute in developing molecular tools that can objectively and promptly monitor the therapeutic effects of interferon β-1a treatment and possibly help adjust dosages in clinical practice.

Acknowledgments

We thank Carol Ozark for collection of normal subject blood samples. We would also like to thank Mike Panos and Christophoros P Christophi with their help in RNA isolation. We would like to thank Dr. Isobel Scarisbrick at Mayo Clinic for providing cDNA plamids used in performing real time RT-PCR. This work was supported in part by research grants from Serono, Inc. to Burk Jubelt, the National Multiple Sclerosis Society (RG2569C5) to Paul T. Massa, and NIH grant (NS041593) to Paul T. Massa.

Footnotes

None of the authors has any potential financial conflict of interest related to this manuscript and the results of this report have been generated, analyzed, and interpreted independently of any sponsor participation or influence.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Noseworthy JH, Lucchinetti C, Rodriguez M, Weinshenker BG. Multiple sclerosis. N Engl J Med. 2000;343(13):938–52. doi: 10.1056/NEJM200009283431307. [DOI] [PubMed] [Google Scholar]
  • 2.Coles AJ, Compston DA, Selmaj KW, Lake SL, Moran S, Margolin DH, et al. Alemtuzumab vs. interferon beta-1a in early multiple sclerosis. N Engl J Med. 2008;359(17):1786–801. doi: 10.1056/NEJMoa0802670. [DOI] [PubMed] [Google Scholar]
  • 3.PRISMS. Randomised double-blind placebo-controlled study of interferon beta-1a in relapsing/remitting multiple sclerosis. PRISMS (Prevention of Relapses and Disability by Interferon beta-1a Subcutaneously in Multiple Sclerosis) Study Group. Lancet. 1998;352(9139):1498–504. [PubMed] [Google Scholar]
  • 4.Johnson KP, Brooks BR, Cohen JA, Ford CC, Goldstein J, Lisak RP, et al. Copolymer 1 reduces relapse rate and improves disability in relapsing-remitting multiple sclerosis: results of a phase III multicenter, double-blind placebo-controlled trial. The Copolymer 1 Multiple Sclerosis Study Group. Neurology. 1995;45(7):1268–76. doi: 10.1212/wnl.45.7.1268. [DOI] [PubMed] [Google Scholar]
  • 5.Kappos L, Radue EW, O’Connor P, Polman C, Hohlfeld R, Calabresi P, et al. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med. 2010;362(5):387–401. doi: 10.1056/NEJMoa0909494. [DOI] [PubMed] [Google Scholar]
  • 6.Antel JP, Miron VE. Central nervous system effects of current and emerging multiple sclerosis-directed immuno-therapies. Clin Neurol Neurosurg. 2008;110(9):951–7. doi: 10.1016/j.clineuro.2008.03.021. [DOI] [PubMed] [Google Scholar]
  • 7.Jacobs LD, Cookfair DL, Rudick RA, Herndon RM, Richert JR, Salazar AM, et al. Intramuscular interferon beta-1a for disease progression in relapsing multiple sclerosis. The Multiple Sclerosis Collaborative Research Group (MSCRG) Ann Neurol. 1996;39(3):285–94. doi: 10.1002/ana.410390304. [DOI] [PubMed] [Google Scholar]
  • 8.PRISMS-4. PRISMS-4: Long-term efficacy of interferon-beta-1a in relapsing MS. Neurology. 2001;56(12):1628–36. doi: 10.1212/wnl.56.12.1628. [DOI] [PubMed] [Google Scholar]
  • 9.Panitch H, Goodin DS, Francis G, Chang P, Coyle PK, O’Connor P, et al. Randomized, comparative study of interferon beta-1a treatment regimens in MS: The EVIDENCE Trial. Neurology. 2002;59(10):1496–506. doi: 10.1212/01.wnl.0000034080.43681.da. [DOI] [PubMed] [Google Scholar]
  • 10.Platanias LC. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol. 2005;5(5):375–86. doi: 10.1038/nri1604. [DOI] [PubMed] [Google Scholar]
  • 11.van Boxel-Dezaire AH, Rani MR, Stark GR. Complex modulation of cell type-specific signaling in response to type I interferons. Immunity. 2006;25(3):361–72. doi: 10.1016/j.immuni.2006.08.014. [DOI] [PubMed] [Google Scholar]
  • 12.Christophi GP, Hudson CA, Gruber RC, Christophi CP, Mihai C, Mejico LJ, et al. SHP-1 deficiency and increased inflammatory gene expression in PBMCs of multiple sclerosis patients. Lab Invest. 2008;88(3):243–55. doi: 10.1038/labinvest.3700720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Prat A, Biernacki K, Antel JP. Th1 and Th2 lymphocyte migration across the human BBB is specifically regulated by interferon beta and copolymer-1. J Autoimmun. 2005;24(2):119–24. doi: 10.1016/j.jaut.2005.01.004. [DOI] [PubMed] [Google Scholar]
  • 14.Gobin SJ, Montagne L, Van Zutphen M, Van Der Valk P, Van Den Elsen PJ, De Groot CJ. Upregulation of transcription factors controlling MHC expression in multiple sclerosis lesions. Glia. 2001;36(1):68–77. doi: 10.1002/glia.1096. [DOI] [PubMed] [Google Scholar]
  • 15.Javed A, Reder AT. Therapeutic role of beta-interferons in multiple sclerosis. Pharmacol Ther. 2006;110(1):35–56. doi: 10.1016/j.pharmthera.2005.08.011. [DOI] [PubMed] [Google Scholar]
  • 16.Axtell RC, Steinman L. Type 1 interferons cool the inflamed brain. Immunity. 2008;28(5):600–2. doi: 10.1016/j.immuni.2008.04.006. [DOI] [PubMed] [Google Scholar]
  • 17.Link H. The cytokine storm in multiple sclerosis. Mult Scler. 1998;4(1):12–5. doi: 10.1177/135245859800400104. [DOI] [PubMed] [Google Scholar]
  • 18.Ozenci V, Kouwenhoven M, Huang YM, Kivisakk P, Link H. Multiple sclerosis is associated with an imbalance between tumour necrosis factor-alpha (TNF-alpha)- and IL-10-secreting blood cells that is corrected by interferon-beta (IFN-beta) treatment. Clin Exp Immunol. 2000;120(1):147–53. doi: 10.1046/j.1365-2249.2000.01175.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Franciotta D, Zardini E, Bergamaschi R, Andreoni L, Cosi V. Interferon gamma and interleukin 4 producing T cells in peripheral blood of multiple sclerosis patients undergoing immunomodulatory treatment. J Neurol Neurosurg Psychiatry. 2003;74(1):123–6. doi: 10.1136/jnnp.74.1.123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hohnoki K, Inoue A, Koh CS. Elevated serum levels of IFN-gamma, IL-4 and TNF-alpha/unelevated serum levels of IL-10 in patients with demyelinating diseases during the acute stage. J Neuroimmunol. 1998;87(1–2):27–32. doi: 10.1016/s0165-5728(98)00053-8. [DOI] [PubMed] [Google Scholar]
  • 21.Frisullo G, Angelucci F, Caggiula M, Nociti V, Iorio R, Patanella AK, et al. pSTAT1, pSTAT3, and T-bet expression in peripheral blood mononuclear cells from relapsing-remitting multiple sclerosis patients correlates with disease activity. J Neurosci Res. 2006;84(5):1027–36. doi: 10.1002/jnr.20995. [DOI] [PubMed] [Google Scholar]
  • 22.Zeis T, Graumann U, Reynolds R, Schaeren-Wiemers N. Normal-appearing white matter in multiple sclerosis is in a subtle balance between inflammation and neuroprotection. Brain. 2008;131(Pt 1):288–303. doi: 10.1093/brain/awm291. [DOI] [PubMed] [Google Scholar]
  • 23.Cannella B, Raine CS. Multiple sclerosis: cytokine receptors on oligodendrocytes predict innate regulation. Ann Neurol. 2004;55(1):46–57. doi: 10.1002/ana.10764. [DOI] [PubMed] [Google Scholar]
  • 24.Eggert M, Goertsches R, Seeck U, Dilk S, Neeck G, Zettl UK. Changes in the activation level of NF-kappa B in lymphocytes of MS patients during glucocorticoid pulse therapy. J Neurol Sci. 2008;264(1–2):145–50. doi: 10.1016/j.jns.2007.08.026. [DOI] [PubMed] [Google Scholar]
  • 25.Christophi G, Panos M, Hudson C, Mihai C, Jubelt B, Massa P. Interferon-b Treatment in Multiple Sclerosis Attenuates Inflammatory Gene Expression Through Inducible Activity of the Phosphatase SHP-1. Clinical Immunology. 2009;133(1):27–44. doi: 10.1016/j.clim.2009.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Oliver BJ, Kohli E, Kasper LH. Interferon therapy in relapsing-remitting multiple sclerosis: A systematic review and meta-analysis of the comparative trials. J Neurol Sci. 2011;302(1–2):96–105. doi: 10.1016/j.jns.2010.11.003. [DOI] [PubMed] [Google Scholar]
  • 27.Panitch H, Goodin D, Francis G, Chang P, Coyle P, O’Connor P, et al. Benefits of high-dose, high-frequency interferon beta-1a in relapsing-remitting multiple sclerosis are sustained to 16 months: final comparative results of the EVIDENCE trial. J Neurol Sci. 2005;239(1):67–74. doi: 10.1016/j.jns.2005.08.003. [DOI] [PubMed] [Google Scholar]
  • 28.Schwid SR, Thorpe J, Sharief M, Sandberg-Wollheim M, Rammohan K, Wendt J, et al. Enhanced benefit of increasing interferon beta-1a dose and frequency in relapsing multiple sclerosis: the EVIDENCE Study. Arch Neurol. 2005;62(5):785–92. doi: 10.1001/archneur.62.5.785. [DOI] [PubMed] [Google Scholar]
  • 29.Schwid SR, Panitch HS. Full results of the Evidence of Interferon Dose-Response-European North American Comparative Efficacy (EVIDENCE) study: a multicenter, randomized, assessor-blinded comparison of low-dose weekly versus high-dose, high-frequency interferon beta-1a for relapsing multiple sclerosis. Clin Ther. 2007;29(9):2031–48. doi: 10.1016/j.clinthera.2007.09.025. [DOI] [PubMed] [Google Scholar]
  • 30.Kappos L, Achtnichts L, Dahlke F, Kuhle J, Naegelin Y, Sandbrink R, et al. Genomics and proteomics: role in the management of multiple sclerosis. J Neurol. 2005;252(Suppl 3):iii21–iii27. doi: 10.1007/s00415-005-2013-3. [DOI] [PubMed] [Google Scholar]
  • 31.Bertolotto A, Sala A, Malucchi S, Marnetto F, Caldano M, Di Sapio A, et al. Biological activity of interferon betas in patients with multiple sclerosis is affected by treatment regimen and neutralising antibodies. J Neurol Neurosurg Psychiatry. 2004;75(9):1294–9. doi: 10.1136/jnnp.2004.037259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hong J, Zang YC, Hutton G, Rivera VM, Zhang JZ. Gene expression profiling of relevant biomarkers for treatment evaluation in multiple sclerosis. J Neuroimmunol. 2004;152(1–2):126–39. doi: 10.1016/j.jneuroim.2004.03.004. [DOI] [PubMed] [Google Scholar]
  • 33.McDonald WI, Compston A, Edan G, Goodkin D, Hartung HP, Lublin FD, et al. Recommended diagnostic criteria for multiple sclerosis: guidelines from the International Panel on the diagnosis of multiple sclerosis. Ann Neurol. 2001;50(1):121–7. doi: 10.1002/ana.1032. [DOI] [PubMed] [Google Scholar]
  • 34.Lublin FD, Reingold SC. Defining the clinical course of multiple sclerosis: results of an international survey. National Multiple Sclerosis Society (USA) Advisory Committee on Clinical Trials of New Agents in Multiple Sclerosis. Neurology. 1996;46(4):907–11. doi: 10.1212/wnl.46.4.907. [DOI] [PubMed] [Google Scholar]
  • 35.Christophi GP, Hudson CA, Panos M, Gruber RC, Massa PT. Modulation of macrophage infiltration and inflammatory activity by the phosphatase SHP-1 in virus-induced demyelinating disease. J Virol. 2009;83(2):522–39. doi: 10.1128/JVI.01210-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Christophi GP, Isackson PJ, Blaber S, Blaber M, Rodriguez M, Scarisbrick IA. Distinct promoters regulate tissue-specific and differential expression of kallikrein 6 in CNS demyelinating disease. J Neurochem. 2004;91(6):1439–49. doi: 10.1111/j.1471-4159.2004.02826.x. [DOI] [PubMed] [Google Scholar]
  • 37.Christophi GP, Panos M, Hudson CA, Christophi RL, Mersich AT, Blystone SD, et al. Macrophages of multiple sclerosis patients display deficient SHP-1 expression and enhanced inflammatory phenotype. Lab Invest. 2009 doi: 10.1038/labinvest.2009.32. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Goertsches R, Serrano-Fernandez P, Moller S, Koczan D, Zettl UK. Multiple sclerosis therapy monitoring based on gene expression. Curr Pharm Des. 2006;12(29):3761–79. doi: 10.2174/138161206778559786. [DOI] [PubMed] [Google Scholar]
  • 39.O’Doherty C, Villoslada P, Vandenbroeck K. Pharmacogenomics of Type I interferon therapy: a survey of response-modifying genes. Cytokine Growth Factor Rev. 2007;18(3–4):211–22. doi: 10.1016/j.cytogfr.2007.04.012. [DOI] [PubMed] [Google Scholar]
  • 40.Buttmann M, Merzyn C, Hofstetter HH, Rieckmann P. TRAIL, CXCL10 and CCL2 plasma levels during long-term Interferon-beta treatment of patients with multiple sclerosis correlate with flu-like adverse effects but do not predict therapeutic response. J Neuroimmunol. 2007;190(1–2):170–6. doi: 10.1016/j.jneuroim.2007.08.009. [DOI] [PubMed] [Google Scholar]
  • 41.Huang WX, Huang P, Hillert J. Increased expression of caspase-1 and interleukin-18 in peripheral blood mononuclear cells in patients with multiple sclerosis. Mult Scler. 2004;10(5):482–7. doi: 10.1191/1352458504ms1071oa. [DOI] [PubMed] [Google Scholar]
  • 42.Salmaggi A, Gelati M, Dufour A, Corsini E, Pagano S, Baccalini R, et al. Expression and modulation of IFN-gamma-inducible chemokines (IP-10, Mig, and I-TAC) in human brain endothelium and astrocytes: possible relevance for the immune invasion of the central nervous system and the pathogenesis of multiple sclerosis. J Interferon Cytokine Res. 2002;22(6):631–40. doi: 10.1089/10799900260100114. [DOI] [PubMed] [Google Scholar]
  • 43.Rice GP, Hartung HP, Calabresi PA. Anti-alpha4 integrin therapy for multiple sclerosis: mechanisms and rationale. Neurology. 2005;64(8):1336–42. doi: 10.1212/01.WNL.0000158329.30470.D0. [DOI] [PubMed] [Google Scholar]
  • 44.Moriuchi H, Moriuchi M, Fauci AS. Nuclear factor-kappa B potently up-regulates the promoter activity of RANTES, a chemokine that blocks HIV infection. J Immunol. 1997;158(7):3483–91. [PubMed] [Google Scholar]
  • 45.Teleshova N, Pashenkov M, Huang YM, Soderstrom M, Kivisakk P, Kostulas V, et al. Multiple sclerosis and optic neuritis: CCR5 and CXCR3 expressing T cells are augmented in blood and cerebrospinal fluid. J Neurol. 2002;249(6):723–9. doi: 10.1007/s00415-002-0699-z. [DOI] [PubMed] [Google Scholar]
  • 46.Carlson NG, Rojas MA, Redd JW, Tang P, Wood B, Hill KE, et al. Cyclooxygenase-2 expression in oligodendrocytes increases sensitivity to excitotoxic death. J Neuroinflammation. 2010;7:25. doi: 10.1186/1742-2094-7-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Richens J, Fairclough L, Ghaemmaghami AM, Mahdavi J, Shakib F, Sewell HF. The detection of ADAM8 protein on cells of the human immune system and the demonstration of its expression on peripheral blood B cells, dendritic cells and monocyte subsets. Immunobiology. 2007;212(1):29–38. doi: 10.1016/j.imbio.2006.06.012. [DOI] [PubMed] [Google Scholar]
  • 48.Xu L, Hilliard B, Carmody RJ, Tsabary G, Shin H, Christianson DW, et al. Arginase and autoimmune inflammation in the central nervous system. Immunology. 2003;110(1):141–8. doi: 10.1046/j.1365-2567.2003.01713.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Amour A, Knight CG, English WR, Webster A, Slocombe PM, Knauper V, et al. The enzymatic activity of ADAM8 and ADAM9 is not regulated by TIMPs. FEBS Lett. 2002;524(1–3):154–8. doi: 10.1016/s0014-5793(02)03047-8. [DOI] [PubMed] [Google Scholar]
  • 50.Narikawa K, Misu T, Fujihara K, Nakashima I, Sato S, Itoyama Y. CSF chemokine levels in relapsing neuromyelitis optica and multiple sclerosis. J Neuroimmunol. 2004;149(1–2):182–6. doi: 10.1016/j.jneuroim.2003.12.010. [DOI] [PubMed] [Google Scholar]
  • 51.Link J, Soderstrom M, Olsson T, Hojeberg B, Ljungdahl A, Link H. Increased transforming growth factor-beta, interleukin-4, and interferon-gamma in multiple sclerosis. Ann Neurol. 1994;36(3):379–86. doi: 10.1002/ana.410360309. [DOI] [PubMed] [Google Scholar]
  • 52.Hermans G, Stinissen P, Hauben L, Van den Berg-Loonen E, Raus J, Zhang J. Cytokine profile of myelin basic protein-reactive T cells in multiple sclerosis and healthy individuals. Ann Neurol. 1997;42(1):18–27. doi: 10.1002/ana.410420106. [DOI] [PubMed] [Google Scholar]
  • 53.Rohowsky-Kochan C, Molinaro D, Cook SD. Cytokine secretion profile of myelin basic protein-specific T cells in multiple sclerosis. Mult Scler. 2000;6(2):69–77. doi: 10.1177/135245850000600203. [DOI] [PubMed] [Google Scholar]
  • 54.Ochi H, Osoegawa M, Wu XM, Minohara M, Horiuchi I, Murai H, et al. Increased IL-13 but not IL-5 production by CD4-positive T cells and CD8-positive T cells in multiple sclerosis during relapse phase. J Neurol Sci. 2002;201(1–2):45–51. doi: 10.1016/s0022-510x(02)00189-2. [DOI] [PubMed] [Google Scholar]
  • 55.Ishizu T, Osoegawa M, Mei FJ, Kikuchi H, Tanaka M, Takakura Y, et al. Intrathecal activation of the IL-17/IL-8 axis in opticospinal multiple sclerosis. Brain. 2005;128(Pt 5):988–1002. doi: 10.1093/brain/awh453. [DOI] [PubMed] [Google Scholar]
  • 56.Killestein J, Den Drijver BF, Van der Graaff WL, Uitdehaag BM, Polman CH, Van Lier RA. Intracellular cytokine profile in T-cell subsets of multiple sclerosis patients: different features in primary progressive disease. Mult Scler. 2001;7(3):145–50. doi: 10.1177/135245850100700302. [DOI] [PubMed] [Google Scholar]
  • 57.Feng X, Petraglia AL, Chen M, Byskosh PV, Boos MD, Reder AT. Low expression of interferon-stimulated genes in active multiple sclerosis is linked to subnormal phosphorylation of STAT1. J Neuroimmunol. 2002;129(1–2):205–15. doi: 10.1016/s0165-5728(02)00182-0. [DOI] [PubMed] [Google Scholar]
  • 58.Theoharides TC, Kempuraj D, Kourelis T, Manola A. Human mast cells stimulate activated T cells: implications for multiple sclerosis. Ann N Y Acad Sci. 2008;1144:74–82. doi: 10.1196/annals.1418.029. [DOI] [PubMed] [Google Scholar]
  • 59.Sayed BA, Walker ME, Brown MA. Cutting edge: Mast cells regulate disease severity in a relapsing-remitting model of multiple sclerosis. J Immunol. 2010;186(6):3294–8. doi: 10.4049/jimmunol.1003574. [DOI] [PubMed] [Google Scholar]
  • 60.Christophi GP, Hudson CA, Gruber R, Christophi CP, Massa PT. Promoter-specific induction of the phosphatase SHP-1 by viral infection and cytokines in CNS glia. J Neurochem. 2008 doi: 10.1111/j.1471-4159.2008.05337.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Guarda G, Braun M, Staehli F, Tardivel A, Mattmann C, Forster I, et al. Type I interferon inhibits interleukin-1 production and inflammasome activation. Immunity. 2011;34(2):213–23. doi: 10.1016/j.immuni.2011.02.006. [DOI] [PubMed] [Google Scholar]
  • 62.Murray PJ. Understanding and exploiting the endogenous interleukin-10/STAT3-mediated anti-inflammatory response. Curr Opin Pharmacol. 2006;6(4):379–86. doi: 10.1016/j.coph.2006.01.010. [DOI] [PubMed] [Google Scholar]
  • 63.Riley JK, Takeda K, Akira S, Schreiber RD. Interleukin-10 receptor signaling through the JAK-STAT pathway. Requirement for two distinct receptor-derived signals for anti-inflammatory action. J Biol Chem. 1999;274(23):16513–21. doi: 10.1074/jbc.274.23.16513. [DOI] [PubMed] [Google Scholar]
  • 64.Yan J, Greer JM. NF-kappa B, a potential therapeutic target for the treatment of multiple sclerosis. CNS Neurol Disord Drug Targets. 2008;7(6):536–57. doi: 10.2174/187152708787122941. [DOI] [PubMed] [Google Scholar]
  • 65.Ramgolam VS, Sha Y, Jin J, Zhang X, Markovic-Plese S. IFN-beta inhibits human Th17 cell differentiation. J Immunol. 2009;183(8):5418–27. doi: 10.4049/jimmunol.0803227. [DOI] [PubMed] [Google Scholar]
  • 66.Wandinger KP, Sturzebecher CS, Bielekova B, Detore G, Rosenwald A, Staudt LM, et al. Complex immunomodulatory effects of interferon-beta in multiple sclerosis include the upregulation of T helper 1-associated marker genes. Ann Neurol. 2001;50(3):349–57. doi: 10.1002/ana.1096. [DOI] [PubMed] [Google Scholar]
  • 67.Huang D, Han Y, Rani MR, Glabinski A, Trebst C, Sorensen T, et al. Chemokines and chemokine receptors in inflammation of the nervous system: manifold roles and exquisite regulation. Immunol Rev. 2000;177:52–67. doi: 10.1034/j.1600-065x.2000.17709.x. [DOI] [PubMed] [Google Scholar]

RESOURCES