Abstract
Multiple sclerosis (MS) in children and adolescents accounts for 3–10% of the whole MS population, and is characterized by a relapsing course in almost all cases. The frequency of relapses is higher than in adult onset MS, at least in the first years of evolution. The objective of treatment is to speed the recovery after a relapse, to prevent the occurrence of relapses, and to prevent disease progression and neurodegeneration. The use of drugs for MS in children and adolescents has not been studied in clinical trials, so their use is mainly based on results from trials in adults and from observational studies. There is a consensus to treat acute relapses with intravenous high-dose corticosteroids. The possibility of preventing relapses and disease progression is based on the use of immunomodulatory agents. Interferon-beta (IFNB) and glatiramer acetate (GA) have been demonstrated to be safe and well tolerated in pediatric MS patients, and also to reduce relapse rate and disease progression. Cyclophosphamide and natalizumab could be offered as second-line treatment in patients with a poor response to IFNB or GA. New oral and injectable drugs will be available in the near future: if safe and well tolerated in the long-term follow up of adults with MS, they could be tested in the pediatric MS population.
Keywords: adolescence, childhood, cyclophosphamide, glatiramer acetate, interferon beta, intravenous immunoglobulin, intravenous methylprednisolone, mitoxantrone, multiple sclerosis, natalizumab
Introduction
The onset of multiple sclerosis (MS) typically occurs in adults at about 30 years of age; however the onset before 18 years of age is being increasingly recognized worldwide, accounting for 3–10% of all individuals with MS [Banwell et al. 2007]. The clinical features of pediatric MS (Ped-MS) have been delineated in several retrospective, prospective and longitudinal studies. Overall, they are not different from those of the adult form, but some findings seem to be peculiar to Ped-MS, being more frequent in comparison with adult MS (A-MS):
the onset with cerebellar and brainstem dysfunction [Banwell et al. 2007; Ghezzi et al. 2002];
the polysymptomatic presentation, with fever, headache, lethargy, meningism, seizures (acute disseminated encephalomyelitis-like onset), especially in very young patients [Banwell et al. 2007];
the evolution with a high number of relapses, specially in the first years of the disease, and an annualized relapse rate of 1–1.9, which is higher than in A-MS [Gorman et al. 2009; Renoux et al. 2007; Ghezzi et al. 2002; Gusev et al. 2002];
the evolution, with a predominance of the relapsing–remitting (RR) course, in more than 90% of cases, and the very low frequency of the primary progressive course [Banwell et al. 2007];
the progression, with a longer interval but a lower age to reach mild and severe disability.
In fact, the majority of Ped-MS patients develop mild and severe disability, as well as the shift to the secondary progressive phase of MS, after a longer disease interval but at a younger age, compared with A-MS [Renoux et al. 2007; Boiko et al. 2002]. So, at a given age, patients with onset in childhood are more disabled than those with a later onset.
The frequency of relapses (or the interattack interval) in the first few years after disease onset is a negative prognostic factor as it correlates with an increased disease severity and with an earlier entry into the secondary progressive phase of MS [Banwell et al. 2007].
Recent studies have demonstrated that approximately one third of children and adolescents with MS develop cognitive dysfunction early: processing speed, attention, language and working memory are involved, with a negative impact on academic functioning and on social relationships [Amato et al. 2008; Banwell and Anderson, 2005; MacAllister et al. 2005].
The high number of relapses in the first years of the disease, and the high frequency of patients with the RR course suggest that the inflammatory process is more pronounced in children with MS compared with adults. This pattern is also suggested by the frequent pleocytosis in the cerebrospinal fluid (CSF) [Pohl et al. 2004], and by the aspect of magnetic resonance imaging (MRI) lesions [Waubant et al. 2009; Chabas et al. 2008]. It seems reasonable to expect that drugs targeting the inflammatory process could have a positive beneficial effect in patients with Ped-MS. As in A-MS, the objective of the treatment is to prevent the occurrence of brain damage, and to limit the damage once it has occurred. In other words:
to speed the recovery and prevent disability after a relapse;
to prevent the occurrence of relapses;
to prevent disease progression, disability and cognitive impairment.
As diagnosis of MS has a major impact on patients and their families, psychological and social support should be offered to reduce the emotional impact of MS, to face the physical and psychological limitations in family, school and social activities, and to strengthen the compliance with medication. In addition to pharmacological treatment, the care of children and adolescents with MS requires a multidisciplinary team, with occupational and physiotherapists, psychologists and social workers, in addition to pediatric or adult neurologists [Banwell et al. 2007].
The effects of drugs in Ped-MS have not been formally evaluated in clinical trials, so their use is based on results in A-MS, experts’ opinion, case reports, and data from observational studies. There are no approved guidelines for the treatment of Ped-MS, but recently a proposal has been published by a group of adult and child neurologists, with the objective of defining a shared approach for the treatment of children and adolescents with MS [Ghezzi et al. 2010].
In the present review, the most relevant studies related to the treatment of Ped-MS are presented, with a hint to possible future therapeutic strategies.
Management of acute MS relapses
Corticosteroids remain a mainstay of treatment for relapses in MS of adults, with class I and class II evidence of speeding functional recovery after acute attacks [Goodin et al. 2002]. Based on the evidence from A-MS studies, there is a general consensus to treat acute relapses of MS in children and adolescents with intravenous methylprednisolone 10–30 mg/kg for 3–5 days [Ghezzi et al. 2010; Banwell, 2005; Ghezzi 2005], although the effectiveness of corticosteroids has not been clearly demonstrated by specific studies. In adults with MS, oral corticosteroids [Morrow et al. 2004; Barnes et al. 1997] have a similar effectiveness and the benefit of a more practical route of administration, but this schedule of administration has not been tested in pediatric patients.
The treatment of acute relapses with corticosteroids hastens the recovery but does not modify the long-term evolution of the disease [Compston and Coles, 2002; Optic Neuritis Study Group, 1997]. Only a single class II study [Zivadinov et al. 2001] has shown that regular pulsed corticosteroids are useful in the long-term management: in a cohort of 88 adult MS randomly assigned to pulses of methylprednisolone (MP) (5 g over 5 consecutive days every 4 months for 3 years, subsequently every 6 months for 2 years) or MP at the same dose schedule only for relapses, the relapse rate was equivalent but disability was less in subjects treated with pulsed MP.
In patients with a severe attack and/or an incomplete recovery after high-dose corticosteroids, a second pulse of corticosteroids can be offered [Banwell, 2005]. Alternatively, intravenous immunoglobulins can be administered, usually 2 g/kg over 2–5 days. This treatment does not have proven efficacy in treating acute relapses, and only has beneficial effects in preventing some relapses in adults with RRMS, demonstrated by studies with methodological limitations and small size [Elovaara et al. 2008; Banwell, 2005; Goodin et al. 2002].
For patients suffering from severe steroid-resistant relapses, plasma exchange can be offered as a therapeutic option, based on results of randomized studies in adults and sporadic use in single MS pediatric cases [Lehmann et al. 2006; Weinshenker et al. 1999; Takahashi et al. 1997].
Treatment to prevent relapses and disease progression
First-line disease-modifying treatment of Ped-MS
Three beta-interferons (Avonex® 30 µg given intramuscularly [i.m.] once a week, Rebif® 22 µg or 44 µg given subcutaneously [s.c.] three times a week, Betaferon® 250 µg given s.c. every other day) and glatiramer acetate (GA; Copaxone®, 20 mg given s.c. every day) are licensed for the treatment of RRMS, and one beta-interferon (Betaferon® 250 µg given s.c. every other day) for the treatment of secondary progressive MS.
There is a large body of evidence, based on results of randomized, double-blind, placebo-controlled trials with class I evidence, that these immunomodulatory agents (IAs):
reduce relapse rate and, to a lesser degree, the progression of disability in adults with RRMS [PRISM Study Group, 1998; Jacobs et al. 1996; Johnson et al. 1995; The IFNB Multiple Sclerosis Study Group and the UBC MS/MRI Analysis Group, 1995; The IFNB Multiple Sclerosis Study Group, 1993];
delay the time to a second MS attack [Comi et al. 2009, 2001; Kappos et al. 2006; Jacobs et al. 2000] and the risk of confirmed disability [Kappos et al. 2007b], if given in subjects at their first demyelinating episode;
have a profound effect on MRI lesions and activity [Soriani et al. 2009]; in patients treated with interferon-beta (IFNB)-1a 22 µg weekly, this drug reduced cerebral atrophy, compared with placebo [Filippi et al. 2004].
The effect of IAs to reduce relapse rate and disease progression has been confirmed in postmarketing studies, with a clear demonstration that IAs are also more effective in everyday clinical practice if started earlier [Trojano et al. 2009, 2007].
There is also some evidence that IFNB improves cognitive functioning [Patti et al. 2010; Kappos et al. 2009; Fisher et al. 2000].
These positive clinical effects of IAs in adults, and their overall positive safety profile, have encouraged the use of these drugs in Ped-MS patients. At present, there have been no formal clinical trials of IAs in the Ped-MS population; the effect of these medications has been evaluated in some preliminary studies on a small number of cases [Mikaeloff et al. 2001; Waubant et al. 2001], mainly addressed to evaluate their safety, and subsequently in studies including a larger number of cases [Ghezzi et al. 2009a, 2005; Banwell et al. 2006; Tenembaum and Segura, 2006; Pohl et al. 2005; Kornek et al. 2003], also providing data on the clinical effects.
In two studies, the data of patients treated with IAs were compared with those of an untreated group. In a study of 16 patients randomly assigned to IFNB-1a 30 µg i.m. weekly or placebo, the active treatment reduced relapse rate, disability progression and MRI measures [Pakdaman et al. 2006]. In a cohort of 197 Ped-MS patients, 24 patients treated with IFNB showed a significant reduction in relapse rate compared with subjects who did not receive any therapy [Mikaeloff et al. 2008].
The main demographic and clinical findings of the cohorts of Ped-MS patients treated with IAs are summarized in Table 1. In comparison with the pretreatment phase, a reduction of relapse rate is reported in almost all studies, as well as a stabilization of disability, assessed by means of Expanded Disability Status Scale (EDSS) score, confirming the positive effect on disease evolution observed in A-MS patients.
Table 1.
n | Age at MS onset (years) | Treatment duration (months) | Side effects | Clinical results | |
---|---|---|---|---|---|
AVONEX | |||||
Waubant et al. [2001] | 9 | 11 | 17 | • Flu-like symptoms (44%) • Injection site reaction (11%) | No impact on relapse rate |
Pakdaman et al. [2006] | 16 (8 treated) | 48 | • No significant side effects • No treatment discontinuations | In treated group: • significantly fewer relapses and disability progression. • fewer new MRI lesions | |
Ghezzi et al. [2009a] | 77 | 11.4 | 53.6 | • Flu-like syndrome (24.7%) • Headache (19.5%) • Myalgia (9,1%) • Fatigue (6.5%) • Skin injection reaction (6.5%) • Psychological disturbances (2.6%) • Increased liver enzymes (5.2%) • Thyroid dysfunction (10.4%) • Lymphopenia (1.3%) | • Decreased relapse rate (from 2.5 to 0.4) • Final EDSS unchanged • 26% of patients lost to follow up or stopped the therapy (after 3.9 years) • 30% of patients shifted to other drugs (mean follow up duration 5.3 yeas) |
REBIF | |||||
Tenembaum and Segura [2006] | 24 | 9.3 | 44.4 | • 2 serious adverse events (chronic arthritis 1, attempted suicide 1) • Flu-like symptoms (58%) • Myalgia/arthralgia (17%) • Injection site reaction (75%) • Abnormal liver enzymes (33%) | • Significant reduction of relapse rate • Decreased EDSS in subjects ≤ 10 years old |
Pohl et al. [2005] | 51 | 13.4 | 21.6 | • Injection site reaction (71%) • Flu-like symptoms (65%) • Gastrointestinal symptoms (10%) • Abnormal liver enzymes (35%) • Abnormal blood counts (39%) • Discontinued 9/51 | • Decreased relapse rate (from 1.9 to 0.8) • EDSS score stable in 48/51 |
Ghezzi et al. [2009a] | 39 | 12.6 | 59.9 | • Injection skin reaction (18%) • Flu-like syndrome (7.7%) • Headache (7.7%) • Myalgia (5.1%) • Fatigue (2.8%) • Nausea (2.8%) • Lymphopenia (7.7%) | • Decreased relapse rate (from 3.2 to 0.9) • Final EDSS unchanged • 64% of patients shifted to other treatments (mean follow up duration 3.6 yeas) |
BETAFERON | |||||
Banwell et al. [2006] | 43 | 10.9 | 29.2 | • No serious or unexpected events • Flu-like symptoms (35%) • Injection site reaction (20.9%) • Abnormal liver enzymes (21%) • Discontinued 25/431 | • Reduction of relapse rate of 50% |
COPAXONE | |||||
Kornek et al. [2003] | 7 | 13.7 | 24 | • No laboratory abnormalities • Transient systemic reaction (14%) | • 28% relapse-free • No change in mean EDSS |
Ghezzi et al. [2009a] | 14 | 13.1 | 74.6 | • Occasionally chest pain • No hematological abnormality | • Decreased relapse rate (from 2.9 to 0.2) • Final EDSS slightly improved |
AVONEX/REBIF/BETAFERON | |||||
Mikaeloff et al. [2001] | 13 2 1 | 13.1 | 12 | • Flu-like symptoms (69%) • Injection site reaction (19%) • Transient abnormal liver enzymes (6%) | • Treatment failure in 25% • EDSS stable at the end of follow up |
Mikaeloff et al. [2008] | 12 9 3 | Mean 11.0 | 17.1 | NA | • Significant delay to the subsequent attack • Less frequent (but not significant) occurrence of severe disability |
EDSS, Expanded Disability Status Scale.
The frequency of adverse events reported in these studies is similar to those observed in a meta-analysis of A-MS patients [Filippini et al. 2003] (Table 2). The wide range of frequency of adverse events, summarized in Table 2, is likely to be due to the use of different methods of evaluation and IFNB types.
Table 2.
Adverse events | Frequency in Ped-MS (range) | Mean frequency in adult MS [Filippini et al. 2003] |
---|---|---|
Flu-like symptoms | 8–71% | 47% |
Injection skin reaction | 7–75% | 61% |
Headache | 8–28% | 50% |
Myalgia | 5–17% | 25% |
Fatigue | 3–6% | 18% |
Nausea | 3–10% | 32% |
Increased liver enzyme | 6–33% | – |
Thyroid dysfunction | 8% | – |
Data in children are less consistent: Tenembaum and Segura [2006] treated eight children under 10 years of age with Rebif, and observed a statistically significant reduction of relapse rate. Ghezzi et al. [2009a] treated 23 patients under 12 years of age with IAs, finding a reduction of relapse rate and a stabilization of EDSS at the end of follow up. IAs were well tolerated, with a frequency of adverse events similar to those observed in the whole cohort of adolescents.
The available literature on the treatment with Copaxone is limited to a small case series [Ghezzi et al. 2009a; Kornek et al. 2003]. A reduction of relapses has been shown, together with a stabilization of disability. No major clinical adverse events and laboratory abnormalities were reported during the treatment with GA. Dyspnea, chest pain, transient systemic reaction (flushing, dizziness) and fatigue have been observed in 7–14% of pediatric patients treated with GA. Hematological and hepatic abnormalities are not reported in patients treated with GA.
The overall positive results of IFNB and GA in Ped-MS has led the European Regulatory Agency to modify the label of drugs including the following statement: “limited published data suggest that safety profile in adolescents from 12 to 16 years of age receiving IFNB or GA is similar to that seen in adults (… )”, whereas there is not enough information on the use of IFNB or GA in children below 12 years of age.
Clinical results have also been confirmed after a long-term follow up of more than 4 years (Table 3) [Ghezzi et al. 2009a].
Table 3.
Avonex n = 77 | Rebif/Betaferon n = 39 | Copaxone n = 14 | |
---|---|---|---|
Mean age of MS onset | 11.4 ± 3.1 | 12.6 ± 2.6 | 13.1 ± 1.5 |
Treatment duration | 53. ± 27.0 | 59.9 ± 39.5 | 74.6 ± 35.5 |
Annualized pretreatment relapse rate | 2.5 ± 1.9 | 3.2 ± 2.5 | 2.9 ± 1.8 |
Relapse rate during the treatment | 0.4 ± 0.6 | 0.9 ± 1.1 | 0.2 ± 0.3 |
EDSS pretreatment | 1.3 ± 1.0 | 1.7 ± 0.8 | 1.1 ± 0.4 |
Final EDSS | 1.5 ± 1.2 | 2.0 ± 1.4 | 1.0 ± 0.9 |
EDSS, Expanded Disability Status Scale.
Based on the evidence that:
IAs are safe and well tolerated in children and adolescents with MS;
IAs have confirmed an effect in reducing relapse rate and disease progression in this population;
IAs are more advantageous in A-MS if administered in early phases of the disease;
a panel of experts [Ghezzi et al. 2010] has recommended to start early the therapy with IAs in children and adolescents with relapsing MS, to prevent the occurrence of relapses, to prevent the accumulation of disability and to reduce brain damage. Subjects at the first demyelinating episode must be monitored clinically and with MRI study; a second MRI is strongly recommended 3–6 months later. Treatment should be offered if a new clinical episode occurs or a second MRI shows disease activity (new T2 or gadolinium-enhanced [Gd+] lesions). There is no agreement on the use of IAs in Ped-MS patients after the first demyelinating episode, but it could be considered in some selected cases with an aggressive onset (many/large MRI lesions, Gd+ lesions).
In almost all patients, full doses of IAs are administered. Most authors suggest initiation of IFNB at one quarter to one half of the recommended adult dose, reaching the full dose within 1–3 months if it is well tolerated. Hematological tests (blood cell count, liver enzymes, bilirubin, thyroid function) should be performed at months 0, 1, 3, and every 3 months in the first year, and then successively every 3–6 months [Ghezzi et al. 2010]. In a cohort of patients treated before 12 years of age, 16 of them were treated with Avonex, 5 with Rebif, 2 with GA, with the same dose used in adolescents and adults. Adverse events and tolerability were similar to those observed in the whole cohort of pediatric patients [Ghezzi et al. 2009a].
Failure of first-line treatment
Relapses and disease progression can occur in spite of treatment with IAs. As no medication prevents the risk of a new attack by 100%, in clinical practice it is not simple to decide whether to continue the treatment with the same drug in a patient who continues to have relapses. Clinical findings (number or relapses, frequency of relapses during the treatment compared to the pretreatment phase, increase of EDSS score), and MRI data (new T2 lesions, Gd+ lesions) have been included as criteria to define treatment failure, but at present no consensus has been reached on the use of clinical or MRI markers [Portaccio et al. 2006; Rio et al. 2006; Freedman et al. 2004].
A panel of experts [Ghezzi et al. 2010] has suggested that patients may be deemed as failing to show an adequate response to a given medication if they meet one or more of the following criteria:
relapse frequency, evaluated for 12 months of sustained, compliant therapy, is maintained at, or exceeds, the pretreatment relapse rate (assuming an annualized relapse rate of >1);
severity of relapses on treatment is such that rescue therapy (intravenous immunoglobulins, plasma exchange) is consistently required for the management of acute relapses;
physical disability accrual exceeds at least 2 points on the EDSS, sustained for more than 3 months.
The presence of two or more new T2 lesions or one Gd+ lesion on MRI scans obtained at least 1 year after sustained therapy with IFNB is indicative of a likelihood of further relapses in A-MS [Rio et al.2009, 2008; Tomassini et al. 2006; Rudick et al. 2004], but this finding was not included as proof of treatment failure in Ped-MS as data are not available regarding the natural history of lesion accrual in treated and untreated patients.
For patients treated with IFNB, there is evidence that the presence and persistence of neutralizing antibodies (NABs) to IFNB is related to a lack of biological activity of IFNB [Farrell et al. 2008; Malucchi et al. 2008; Sørensen et al. 2006, 2005]. Patients failing IFNB or demonstrating IFNB NABs can be offered therapy with GA [Capobianco et al. 2008]; an increase of IFNB dosage can also be considered for patients without NABs. Patients failing GA therapy can be considered for treatment with IFNB.
In a study of 130 Ped-MS patients, approximately 30% of cases altered treatment between IFNB and GA: the change of treatment regimen appeared to be advantageous, as the relapse rate continued to be lower with respect to the pretreatment value [Ghezzi et al. 2009a].
For patients with a very active form of MS, treatment with second-line drugs should be considered.
Second-line treatments in Ped-MS
Natalizumab (NA), mitoxantrone and cyclophosphamide (Cy) can be offered as options for adults with very active/rapidly progressing RRMS and poor response despite treatment with IFNB or GA.
NA has shown a strong effect on disease activity in a double-blind, placebo-controlled trial [Polman et al. 2006] reducing the relapse rate by 68% and slowing disease progression by 54% after 2 years of treatment. In a post-hoc study, disease activity was suppressed completely in 29.5% of cases and they remained free of clinical and radiological activity (defined by the criteria of new/enlarged lesions, or Gd+ lesions) after 2 years of treatment [Havrdova et al. 2009].
NA is administered i.v. every 28 days and is well tolerated; however, the occurrence of JC-virus-mediated progressive multifocal leukoencephalopathy (PML), with a risk of about 1:1000, the possible increased risk of melanoma, the potential hepatotoxicity must be taken into account [Kappos et al. 2007a; Yousry et al. 2006]. Many postmarketing studies are ongoing in adults to establish these risks in clinical practice.
NA was demonstrated to be effective in two single cases with very active MS and failure to first line treatments [Appleton and Boggild, 2009; Boriell et al. 2009], and in three MS patients with many relapses and MRI activity who were treated with NA 3-5 mg/kg every 28 days, obtaining a suppression of disease activity [Huppke et al. 2008]. NA was safe and well tolerated in a cohort of 38 subjects with juvenile Crohn’s disease, treated with this drug [Hyams et al. 2007].
Preliminary data of a cohort of 17 cases with active MS (3.1 relapses in the previous year) and MRI activity (2.3 Gd+ lesions at baseline), a mean age of 14.2 years, and a mean of 10 NA infusions have been presented recently [Ghezzi et al. 2009b]. Following NA treatment, the relapse rate decreased from 3.1 to 0.1, EDSS from 2.7 to 2 at the last observation, and no subject developed Gd+ lesions during the treatment. NA was well tolerated and the dose was the same as used in adults.
Data are not available on long-term follow up of NA, and further studies are necessary to better define the role of NA as a second-line treatment of Ped-MS, although these preliminary data show that NA is very effective and well tolerated in Ped-MS.
Mitoxantrone is approved for the treatment of active MS in adults, and it could also be offered as a second-line therapy in selected Ped-MS patients with active MS, but the increased risk of severe adverse events such as leukemia, cardiomyopathy, infections and infertility, in addition to side effects such as nausea, vomiting and hair loss, strongly limit the use of this drug. In particular, some recent studies have shown a high risk of leukemia, 1:333 in a study including 5472 cases [Ellis and Boggild, 2009] and 1:140 in an Italian multicenter survey of 2854 cases [Martinelli et al. 2009]. There are no data on the use of mitoxantrone in Ped-MS.
Cy is not licensed for MS treatment, but it has a powerful effect of reducing disease activity in A-MS. In a recent paper Cy has been used in a group of 17 Ped-MS patients with a mean age of 15 years and evidence of disease activity, as they received a mean of 2.5 steroid courses in the previous year, in spite of other first- (14 cases) and second-line (9 cases) treatments [Makhani et al. 2009]. Three different treatment regimens were used: induction therapy alone, induction therapy with pulse maintenance therapy, and pulse maintenance therapy alone. The dose ranged from 500 to 1000 mg/m2. EDSS score was reduced in most cases after 1 year. Three children remained relapse free, 12 reduced their annualized relapse rate from 3.8 to 1.6. Adverse events were frequent (nausea and vomiting in 15 patients, lymphopenia in 16, anemia in 10, alopecia in 10, menstrual disorders in 5 out of 10 girls; in rare cases thrombocytopenia, hematuria, infections, fatigue, urticaria, myalgias); they were severe in a few patients (amenorrhea in 3, sterility in 1, osteoporosis in 2, bladder cancer in 1). For these reasons the authors concluded that Cy should be considered a second-line treatment for carefully selected and monitored children with aggressive MS refractory to first-line therapies.
To conclude, limited published data seem to suggest that NA and Cy are effective in Ped-MS with active course. Further studies are necessary to define the dose, the treatment duration, and the occurrence of short- and long-term adverse effects.
New treatments
Many new oral drugs are under evaluation in phase II or phase III trials [Gasperini and Ruggieri, 2009]:
Cladribine is a new oral immunosuppressant with a strong effect in reducing clinical relapses, disease progression, and MRI activity. A phase III trial has randomized 1326 patients to receive two different cumulative doses of cladribine or placebo in a 1:1:1 ratio [Giovannoni et al. 2010]; the active treatment significantly reduced the annualized relapse rate (0.14 and 0.15, respectively, versus 0.33), the risk of disease progression, and brain lesion count on MRI. A phase III trial evaluating the effect of cladribine to delay the conversion to MS after a first demyelinating episode is ongoing.
Fingolimod is another powerful oral immunomodulating agent; in a phase III trial including 1292 patients, oral fingolimod at a dose of either 1.25 or 0.5 mg significantly reduced annualized relapse rate compared with IFNB-1a 30 µg weekly (0.20 and 0.16, respectively, versus 0.33) [Cohen et al. 2010]. In another study [Kappos et al. 2010] the same doses of fingolimod were compared with placebo after 24 months of treatment. Both doses of fingolimod reduced relapse rate (0.16 and 0.18, respectively, versus 0.40), the probability of disease progression (16.6% and 17.7%, respectively, versus 24.1%), and MRI measures.
Teriflunomide has been demonstrated to reduce MRI activity in a phase II study, together with some positive clinical outcome measures [O’Connor et al. 2006]; a phase III trial is ongoing.
Fumarate reduced several measures of MRI activity and reduced relapse rate by 32% in a phase II study [Kappos et al. 2008]; a phase III study is ongoing.
Laquinimod has shown an effect on MRI and clinical measures given at the dose of 0.6 mg in a phase II study enrolling 306 patients [Comi et al. 2008]; a phase III trial is ongoing.
Monoclonal antibodies targeting cells involved in the immunopathological process of MS (CD20+ B cells, Rituximab; CD25+ B cells, Daclizumab; CD52 on lymphocytes and monocytes, Alentuzumab) are promising drugs for the treatment of MS. Rituximab was effective in a phase I study including RRMS [Bar-Or et al. 2008], and partially effective in primary progressive MS patients with evidence of MRI activity [Hawker et al. 2009]. It was effective in an adolescent with a severe evolution of MS [Karenfort et al. 2009]. Daclizumab reduced disease activity in RRMS patients who experienced persistent MS disease activity with IFNB therapy in a phase II study [Bielekova et al. 2009].
Alentuzumab strongly reduced the rate of sustained accumulation of disability and the annualized relapse rate in comparison with Rebif in a phase II trial [The CAMMS223 Trial Investigators, 2008]. A phase III trial is ongoing. The major risk of monoclonal antibodies is related to the development of PML [Carson et al. 2009].
Conclusions
At present, no formal clinical trials of IAs have been performed in Ped-MS patients, and the use of medications is mainly based on results on clinical trials in adults. IAs such as IFNB and GA appear to be safe and well tolerated in children and adolescents with MS.
According to the suggestions of a panel of experts, IAs should be given to children and adolescents with active RRMS to reduce the risk of relapses and the risk of disease progression. A careful clinical follow up of patients must be planned to prevent and minimize adverse effects, and to adapt the treatment according to the clinical evolution of MS.
In cases that have a poor response to a drug, clinicians can consider the option to increase the dose (if possible) or to switch subjects to other first-line or second-line treatments. Cy and NA can be offered to patients with an active evolution and a poor response to first-line treatments. Further studies are needed to better evaluate their safety.
In the future many oral or injectable agents will become available for the treatment of adults with MS. Most of them seem to have a stronger clinical effect compared with currently available drugs, others will probably have an equal clinical effect but with the advantage of an oral administration. The extension of a new drug to Ped-MS needs careful demonstration of its safety in adults and careful evaluation of potentially negative effects in children given its mechanism of action. In particular, the risk of infections should be carefully considered, owing to the greater exposure to infections in young subjects, of cancer, and the potential effects on reproductive function and physical development, with careful surveillance of short- and long-term safety and tolerability. Further studies will define their possible use in Ped-MS. It will become possible to select the most useful and best-tolerated drug for each patient.
Conflict of interest statement
Dr. Angelo Ghezzi received honoraria for speaking from Bayer Schering, Biogen-Dompé, Merck-Serono, and Novartis, for consultancy from Actelion, Merck-Serono, and Novartis, received support for participation to National and International Congresses from Bayer Schering, Biogen-Dompé, Merck-Serono Novartis, Sanofi-Aventis.
References
- Amato M., Goretti B., Ghezzi A., Lori S., Zipoli V., Portaccio E., et al. (2008) Cognitive and psychosocial features of childhood and juvenile MS. Neurology 70: 1891–1897 [DOI] [PubMed] [Google Scholar]
- Appleton R.E., Boggild M. (2009) Natalizumab in paediatric multiple sclerosis and service implication. Dev Med Child Neurol 51: 758–759 [DOI] [PubMed] [Google Scholar]
- Banwell B. (2005) Treatment of children and adolescents with multiple sclerosis. Expert Rev Neurotherapeutics 5: 391–401 [DOI] [PubMed] [Google Scholar]
- Banwell B.L., Anderson P.E. (2005) The cognitive burden of multiple sclerosis in children. Neurology 64: 891–894 [DOI] [PubMed] [Google Scholar]
- Banwell B., Ghezzi A., Bar-Or A., Mikaeloff Y., Tardieu M. (2007) Multiple sclerosis in children: clinical diagnosis, therapeutic strategies, and future directions. Lancet Neurol 6: 887–902 [DOI] [PubMed] [Google Scholar]
- Banwell B., Reder A.T., Krupp L., Tenembaum S., Eraksoy M., Alexey B., et al. (2006) Safety and tolerability of interferon beta-1b in pediatric multiple sclerosis. Neurology 66: 472–476 [DOI] [PubMed] [Google Scholar]
- Barnes D., Hughes R.A., Morris R.W., Wade-Jones O., Brown P., Britton T., et al. (1997) Randomised trial of oral and intravenous methylprednisolone in acute relapses of multiple sclerosis. Lancet 349: 902–906 [DOI] [PubMed] [Google Scholar]
- Bar-Or A., Calabresi P.A., Arnold D., Markowitz C., Shafer S., Kasper L.H., et al. (2008) Rituximab in relapsing–remitting multiple sclerosis: a 72-week, open-label, phase I trial. Ann Neurol 63: 395–400 [DOI] [PubMed] [Google Scholar]
- Bielekova B., Howard T., Packer A.N., Richert N., Blevins G., Ohayon J., et al. (2009) Effect of anti-CD25 antibody daclizumab in the inhibition of inflammation and stabilization of disease progression in multiple sclerosis. Arch Neurol 66: 483–489 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boiko A., Vorobeychik G., Paty D., Devonshire V., Sadovnick D., the UBC MS Clinic Neurologists (2002) Early onset multiple sclerosis: a longitudinal study. Neurology 59: 1006–1010 [DOI] [PubMed] [Google Scholar]
- Boriell G., Prosperino L., Lucchetti A., Pozzilli C. (2009) Natalizumab treatment in pediatric multiple sclerosis: a case report. Eur J Paediatr Neurol 13: 67–71 [DOI] [PubMed] [Google Scholar]
- Capobianco M., Rizzo A., Malucchi S., Sperli F., Di Sapio A., Oggero A., et al. (2008) Glatiramer acetate is a treatment option in neutralising antibodies to interferon-beta-positive patients. Neurol Sci 29: S227–S229 [DOI] [PubMed] [Google Scholar]
- Carson K.R., Focosi D., Major E.O., Petrini M., Richey E.A., West D.P., et al. (2009) Monoclonal antibody-associated progressive multifocal leucoencephalopathy in patients treated with rituximab, natalizumab, and efalizumab: a Review from the Research on Adverse Drug Events and Reports (RADAR) Project. Lancet Oncol 10: 816–824 [DOI] [PubMed] [Google Scholar]
- Chabas D., Castello-Trivino T., Mowry E.M., Strober J.B., Glenn O.A., Waubant E. (2008) Vanishing MS T2-bright lesions before puberty: a distinct MRI phenotype. Neurology 71: 1090–1093 [DOI] [PubMed] [Google Scholar]
- Cohen J., Barkhof F., Comi G., Hartung H.P., Khatri B.O., Montalban X., et al. (2010) Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. N Engl J Med 362: 402–415 [DOI] [PubMed] [Google Scholar]
- Comi G., Filippi M., Barkhof F., Durelli L., Edan G., Fernandez O., et al. (2001) Effect of early interferon treatment on conversion to definite multiple sclerosis: a randomised study. Lancet 357: 1576–1582 [DOI] [PubMed] [Google Scholar]
- Comi G., Martinelli V., Rodegher M., Moiola L., Bajenaru O., Carra A., et al. (2009) Effect of glatiramer acetate on conversion to clinically definite multiple sclerosis in patients with clinically isolated syndrome (PreCISe study): a randomised, double-blind, placebo-controlled trial. Lancet Neurol 374: 1503–1511 [DOI] [PubMed] [Google Scholar]
- Comi G., Pulizzi A., Rovaris M., Abramsky O., Arbizu T., Boiko A., et al. (2008) Effect of laquinimod on MRI-monitored disease activity in patients with relapsing–remitting multiple sclerosis: a multicentre randomised double-blind placebo-controlled phase IIb study. Lancet 371: 2085–2092 [DOI] [PubMed] [Google Scholar]
- Compston A., Coles A. (2002) Multiple sclerosis. Lancet 359: 1221–1231 [DOI] [PubMed] [Google Scholar]
- Ellis R., Boggild M. (2009) Therapy related acute leukaemia with mitoxantrone: what is the risk and can we minimise it? Mult Scler 15: 505–508 [DOI] [PubMed] [Google Scholar]
- Elovaara I., Apostolski S., van Doorn P., Apostolski S., van Dororn P., Gilhus N.E., et al. (2008) EFNS guidelines for the use of intravenous immunoglobulin in treatment of neurological disorders. Eur J Neurol 15: 893–908 [DOI] [PubMed] [Google Scholar]
- Farrell R., Kapoor R., Leary S., Rudge P., Thompson A., Miller D. (2008) Neutralizing anti-interferon beta antibodies are associated with reduced side effects and delayed impact on efficacy of interferon-beta. Mult Scler 14: 212–218 [DOI] [PubMed] [Google Scholar]
- Filippi M., Rovaris M., Inglese M., Barkhof F., De Stefano N., Smith S., et al. (2004) Interferon beta-1a for brain tissue loss in patients at presentation with syndromes suggestive of multiple sclerosis: a randomised, double-blind, placebo-controlled trial. Lancet 364: 1489–1496 [DOI] [PubMed] [Google Scholar]
- Filippini G., Munari L., Incorvaia B., Ebers G.C., Polman C., D'Amico R., et al. (2003) Interferons in relapsing remitting multiple sclerosis: a systematic review. Lancet 361: 545–552 [DOI] [PubMed] [Google Scholar]
- Fisher J.S., Priore R.L., Jacobs L.D., Cookfair D.L., Rudick R.A., Herndon R.M., et al. (2000) Neuropsychological effects of interferon-beta-1a in relapsing multiple sclerosis. Multiple Sclerosis Collaborative Research Group. Ann Neurol 48: 885–892 [PubMed] [Google Scholar]
- Freedman M.S., Patry D.G., Grand’Maison F., Myles M.L., Paty D.W., Selchen D.H., et al. (2004) Treatment optimization in multiple sclerosis. Can J Neurol Sci 31: 157–168 [DOI] [PubMed] [Google Scholar]
- Gasperini C., Ruggieri S. (2009) New oral drugs for multiple sclerosis. Neurol Sci Suppl 2): S179–S183 [DOI] [PubMed] [Google Scholar]
- Ghezzi A. (2005) Childhood–juvenile multiple sclerosis: clinical characteristics and treatment. Expert Rev Neurotherapeutics 5: 403–411 [DOI] [PubMed] [Google Scholar]
- Ghezzi A., Amato M.P., Annovazzi P., Capobianco M., Gallo P., La Mantia L., et al. (2009a) Long-term results of immunomodulatory treatment in children and adolescents with multiple sclerosis: the Italian experience. Neurol Sci 30: 193–199 [DOI] [PubMed] [Google Scholar]
- Ghezzi A., Amato M.P., Capobianco M., Gallo P., Marrosu G., Martinelli V., et al. (2005) Disease modifying drugs in childhood-juvenile multiple sclerosis: results of the ITEMS (immunomodulatory treatment of early-onset MS) study group. Mult Scler 11: 420–424 [DOI] [PubMed] [Google Scholar]
- Ghezzi, A., Banwell, B., Boyko, A., Amato MP, Anlar B., Blinkenberg M. et al (2010) The management of MS in children: A European view, Mult. Scler. in press. [DOI] [PubMed]
- Ghezzi A., Pozzilli C., Brescia-Morra V., Bortolon F., Capra R., Grimaldi L., et al. (2009b) Safety and effectiveness of natalizumab in paediatric multiple sclerosis: results of 17 patients. Mult Scler 15(Suppl 2): 774–774 [Google Scholar]
- Ghezzi A., Pozzilli C., Liguori M., Marrosu M.G., Dilani N., Milanese C., et al. (2002) Prospective study of multiple sclerosis with early onset. Mult Scler 8: 115–118 [DOI] [PubMed] [Google Scholar]
- Giovannoni G., Comi G., Cook S., Rammohan K., Rieckmann P., Soelberg Sørensen P., et al. (2010) A placebo-controlled trial of oral cladribine for relapsing multiple sclerosis. N Engl J Med 362: 416–426 [DOI] [PubMed] [Google Scholar]
- Goodin D.S., Frohman E.M., Garmany G.P., Halper J., Likosky W.H., Lublin F.D., et al. (2002) Disease modifying therapies in multiple sclerosis: Subcommittee of the American Academy of Neurology and the MS Council for Clinical Practice Guidelines. Neurology 58: 169–178 [DOI] [PubMed] [Google Scholar]
- Gorman M.P., Healy B.C., Polgar-Turcsanyi M., Chitnis T. (2009) Increased relapse rate in pediatric-onset compared with adult-onset multiple sclerosis. Arch Neurol 66: 54–59 [DOI] [PubMed] [Google Scholar]
- Gusev E., Boiko A., Bikova O., Maslova O., Guseva M., Boiko S., et al. (2002) The natural history of early onset multiple sclerosis: comparison of data from Moscow and Vancouver. Clin Neurol Neurosurg 104: 203–207 [DOI] [PubMed] [Google Scholar]
- Havrdova E., Galetta S., Hutchinson M., Stefoski D., Bates D., Polman C.H., et al. (2009) Effect of natalizumab on clinical and radiological disease activity in multiple sclerosis: a retrospective analysis of the Natalizumab Safety and Efficacy in Relapsing–Remitting Multiple Sclerosis (AFFIRM) study. Lancet Neurol 8: 254–260 [DOI] [PubMed] [Google Scholar]
- Hawker K., O'Connor P., Freedman M.S., Calabresi P.A., Antel J., Simon J., et al. (2009) Rituximab in patients with primary progressive multiple sclerosis: results of a randomized double-blind placebo-controlled multicenter trial. Ann Neurol 66: 460–471 [DOI] [PubMed] [Google Scholar]
- Huppke P., Stark W., Zurcher C., Huppke B., Brück W., Gärtner J. (2008) Natalizumab use in pediatric multiple sclerosis. Arch Neurol 65: 1655–1658 [DOI] [PubMed] [Google Scholar]
- Hyams J.S., Wilson D.C., Thomas A., et al. (2007) Natalizumab therapy for moderate to severe Crohn disease in adolescents. J Pediatr Gastroenterol Nutr 44: 185–191 [DOI] [PubMed] [Google Scholar]
- Jacobs L.D., Beck R.W., Simon J.H., Kinkel R.P., Brownscheidle C.M., Murray T.J., et al. (2000) Intramuscular interferon beta-1a therapy initiated during a first demyelinating event in multiple sclerosis. CHAMPS Study Group. N Engl J Med 343: 898–904 [DOI] [PubMed] [Google Scholar]
- Jacobs L.D., Cookfair D.L., Rudick R.A., Herndon R.M., Richert J.R., Salazar A.M., et al. (1996) Intramuscular interferon beta 1-a for disease progression in exacerbating-remitting multiple sclerosis. Ann Neurol 39: 285–294 [DOI] [PubMed] [Google Scholar]
- Johnson K.P., Brooks B.R., Cohen J.A., Herndon R.M., Richert J.R., Salazar A.M., et al. (1995) Copolymer 1 reduces relapse rate and improves disability in relapsing–remitting multiple sclerosis: results of a phase III multicenter, double- blind, placebo-controlled trial. Neurology 45: 1268–1276 [DOI] [PubMed] [Google Scholar]
- Kappos L., Bates D., Hartung H.P., Havrdova E., Miller D., Polman C.H., et al. (2007a) Natalizumab treatment for multiple sclerosis: recommendations for patient selection and monitoring. Lancet Neurol 6: 431–441 [DOI] [PubMed] [Google Scholar]
- Kappos L., Freedman M.S., Polman C., Edan G., Hartung H.P., Miller D.H., et al. (2007b) Effect of early versus delayed interferon beta-1b treatment on disability after a first clinical event suggestive of multiple sclerosis: a 3-year follow-up analysis. Lancet Neurol 370: 389–397 [DOI] [PubMed] [Google Scholar]
- Kappos L., Freedman M., Polman C.H., Edan G., Hartung H.P., Miller D.H., et al. (2009) Long-term effect of early treatment with interferon beta-1b after a first clinical event suggestive of multiple sclerosis: 5-year active treatment extension of the phase 3 BENEFIT trial. Lancet Neurol 11: 987–997 [DOI] [PubMed] [Google Scholar]
- Kappos L., Gold R., Miller D.H., Macmanus D.G., Havrdova E., Limmroth V., et al. (2008) Efficacy and safety of oral fumarate in patients with relapsing-remitting multiple sclerosis: a multicentre randomised double-blind placebo-controlled phase Iib study. Lancet 372: 1463–1472 [DOI] [PubMed] [Google Scholar]
- Kappos L., Polman C., Freedman M.S., Edan G., Hartung H.P., Miller D.H., et al. (2006) Treatment with interferon beta-1b delays conversion to clinically definite and McDonald MS in patients with clinically isolated syndromes. Neurology 67: 1242–1249 [DOI] [PubMed] [Google Scholar]
- Kappos L., Radue E.W., O’Connor P., Polman C., Hohlfeld R., Calabresi P., et al. (2010) A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med 362: 387–401 [DOI] [PubMed] [Google Scholar]
- Karenfort M., Kieseier B.C., Tibussek D., Assmann B., Schaper J., Mayatepek E. (2009) Rituximab as a highly effective treatment in a female adolescent with severe multiple sclerosis. Dev Med Child Neurol 51: 159–161 [DOI] [PubMed] [Google Scholar]
- Kornek B., Bernert G., Balassy C., Geldner J., Prayer D., Feucht M. (2003) Glatiramer acetate treatment in patients with childhood and juvenile onset multiple sclerosis. Neuropediatrics 34: 120–126 [DOI] [PubMed] [Google Scholar]
- Lehmann H.C., Hartung H.P., Hetzel G.R., Stüve O., Kieseier B.C. (2006) Plasma exchange in neuroimmunological disorders: Part 1: Rationale and treatment of inflammatory central nervous system disorders. Arch Neurol 63: 930–935 [DOI] [PubMed] [Google Scholar]
- MacAllister W.S., Belman A.L., Milazzo M., Weisbrot D.M., Christodoulou C., Scherl W.F., et al. (2005) Cognitive functioning in children and adolescents with multiple sclerosis. Neurology 64: 1422–1425 [DOI] [PubMed] [Google Scholar]
- Makhani N., Gorman M.P., Branson H.M., Stazzone L., Banwell B.L., Chitnis T. (2009) Cyclophosphamide therapy in pediatric multiple sclerosis. Neurology 72: 2076–2082 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malucchi S., Gilli F., Caldano M., Marnetto F., Valentino P., Granieri L., et al. (2008) Predictive markers for response to interferon therapy in patients with multiple sclerosis. Neurology 70: 1119–1127 [DOI] [PubMed] [Google Scholar]
- Martinelli V., Capra R., Cocco E. (2009) High incidence of acute leucemia in multiple sclerosis patients treated with mitoxantrone: a retrospective multicentre Italian study. Mult Scler (Suppl 2): S167–S170 [Google Scholar]
- Mikaeloff Y., Caridde G., Tardieu M., Suissa S. (2008) Effectiveness of early beta interferon on the first attack after confirmed multiple sclerosis: a comparative study. Eur J Paediatr Neurol 12: 205–209 [DOI] [PubMed] [Google Scholar]
- Mikaeloff Y., Moreau T., Debouverie M., Pelletier J., Lebrun C., Gout O., et al. (2001) Interferon-beta treatment in patients with childhood-onset multiple sclerosis. J Pediatr 139: 443–446 [DOI] [PubMed] [Google Scholar]
- Morrow S.A., Stoian C.A., Dmitrovic J., Chan S.C., Metz L.M. (2004) The bioavailability of IV methylprednisolone and oral prednisone in multiple sclerosis. Neurology 63: 1079–1080 [DOI] [PubMed] [Google Scholar]
- O’Connor P.W., Freedman M.S., Bar-Or A., Rice G.P., Confavreux C., Paty D.W., et al. (2006) A phase II study of the safety and efficacy of teriflunomide in multiple sclerosis with relapses. Neurology 66: 894–900 [DOI] [PubMed] [Google Scholar]
- Optic Neuritis Study Group (1997) The 5-year risk of MS after optic neuritis: experience of the optic neuritis treatment trial. Neurology 49: 1404–1413 [DOI] [PubMed] [Google Scholar]
- Pakdaman H., Fallah A., Sahraian M.A., Pakdaman R., Meysamie A. (2006) Treatment of early onset multiple sclerosis with suboptimal dose of interferon beta-1a. Neuropediatrics 37: 257–260 [DOI] [PubMed] [Google Scholar]
- Patti F., Amato M.P., Bastianello S., Caniatti L., Di Monte E., Ferrazza P., et al. (2010) Effects of immunomodulatory treatment with subcutaneous interferon beta-1a on cognitive decline in mildly disabled patients with relapsing-remitting multiple sclerosis. Mult Scler 16: 68–77 [DOI] [PubMed] [Google Scholar]
- Pohl D., Rostasy K., Gartner J., Hanefeld F. (2005) Treatment of early onset multiple sclerosis with subcutaneous interferon beta-1a. Neurology 65: 888–890 [DOI] [PubMed] [Google Scholar]
- Pohl D., Rostasy K., Reiber H., Hanefeld F. (2004) CSF characteristics in early-onset multiple sclerosis. Neurology 63: 1966–1967 [DOI] [PubMed] [Google Scholar]
- Polman C.H., O’Connor P.W., Havrdova E., Hutchinson M., Kappos L., Miller D.H., et al. (2006) A randomized, placebo-controlled trial of Natalizumab for relapsing multiple sclerosis. N Engl J Med 354: 899–910 [DOI] [PubMed] [Google Scholar]
- Portaccio E., Zipoli V., Siracusa G., Sorbi S., Amato M.P. (2006) Response to interferon-beta therapy in relapsing–remitting multiple sclerosis: a comparison of different clinical criteria. Mult Scler 12: 281–286 [DOI] [PubMed] [Google Scholar]
- PRISM Study Group (1998) Randomized double-blind placebo controlled study of interferon beta 1-a in relapsing-remitting multiple sclerosis. Lancet 352: 1498–1504 [PubMed] [Google Scholar]
- Renoux C., Vukusic S., Mikaeloff Y., Mikaeloff Y., Edan G., Clanet M., et al. (2007) Natural history of multiple sclerosis with childhood onset. N Engl J Med 365: 2603–2613 [DOI] [PubMed] [Google Scholar]
- Rio J., Castello J., Rovira A., Tintoré M., Sastre-Garriga J., Horga A., et al. (2009) Measures in the first year of therapy predict the response to interferon beta in MS. Mult Scler 15: 848–853 [DOI] [PubMed] [Google Scholar]
- Rio J., Nos C., Tintorè M., Téllez N., Galán I., Pelayo R., et al. (2006) Defining the response to interferon-beta in relapsing-remitting multiple sclerosis patients. Ann Neurol 59: 344–352 [DOI] [PubMed] [Google Scholar]
- Rio J., Rovira A., Tintoré M., Huerga E., Nos C., Tellez N., et al. (2008) Relationship between MRI lesion activity and response to IFNβ in relapsing remitting multiple sclerosis patients. Mult Scler 14: 479–484 [DOI] [PubMed] [Google Scholar]
- Rudick R., Lee J., Simon J., Ransohoff R.M., Fisher E. (2004) Defining interferonβ response status in multiple sclerosis. Ann Neurol 56: 548–555 [DOI] [PubMed] [Google Scholar]
- Sørensen P.S., Deisenhammer F., Duda P., Hohlfeld R., Myhr K.M., Palace J., et al. (2005) Guidelines on use of anti-IFN-beta antibody measurements in multiple sclerosis: report of an EFNS Task Force on IFN-beta antibodies in multiple sclerosis. Eur J Neurol 12: 817–827 [DOI] [PubMed] [Google Scholar]
- Sørensen P.S., Tscherning T., Mathiesen H.K., Langkilde A.R., Ross C., Ravnborg M., et al. (2006) Neutralizing antibodies hamper IFNbeta bioactivity and treatment effect on MRI in patients with MS. Neurology 67: 1681–1683 [DOI] [PubMed] [Google Scholar]
- Soriani M.P., Bonzano L., Roccatagliata L., Cutter G.R., Mancardi G.L., Bruzzi P., et al. (2009) Magnetic resonance imaging as a potential surrogate for relapses in multiple sclerosis. A meta-analytic approach. Ann Neurol 65: 268–275 [DOI] [PubMed] [Google Scholar]
- Takahashi I., Sawaishi Y., Takeda O., Enoki M., Takada G. (1997) Childhood multiple sclerosis treated with plasmapheresis. Pediatr Neurol 17: 83–87 [DOI] [PubMed] [Google Scholar]
- Tenembaum S.N., Segura M.J. (2006) Interferon beta-1a treatment in childhood and juvenile-onset multiple sclerosis. Neurology 67: 511–513 [DOI] [PubMed] [Google Scholar]
- The CAMMS223 Trial Investigators (2008) Alentuzumab vs. interferon beta-1a in early multiple sclerosis. N Engl J Med 359: 1786–1801 [DOI] [PubMed] [Google Scholar]
- The IFNB Multiple Sclerosis Study Group (1993) Interferon beta-1b is effective in relapsing–remitting multiple sclerosis: I clinical results of a multicenter, randomised, double-blind, placebo-controlled trial. Neurology 43: 655–661 [DOI] [PubMed] [Google Scholar]
- The IFNB Multiple Sclerosis Study Group and the UBC MS/MRI Analysis Group (1995) Interferon beta-1b in the treatment of MS: final outcome of the randomised controlled trial. Neurology 53: 1277–1285 [PubMed] [Google Scholar]
- Tomassini V., Paolillo A., Russo P., Giugni E., Prosperini L., Gasperini C., et al. (2006) Predictors of long-term clinical response to interferon β therapy in relapsing multiple sclerosis. J Neurol 253: 287–293 [DOI] [PubMed] [Google Scholar]
- Trojano M., Pellegrini F., Paolicelli D., Fuiani A., Zipoli V., Zimatore G.B., et al. (2007) New natural history of interferon-beta treated relapsing multiple sclerosis. Ann Neurol 61: 300–306 [DOI] [PubMed] [Google Scholar]
- Trojano M., Pellegrini F., Fuiani A., Paolicelli D., Zimatore G.B., Tortorella C., et al. (2009) Real-life impact of early interferon beta therapy in relapsing multiple sclerosis. Ann Neurol 66: 513–520 [DOI] [PubMed] [Google Scholar]
- Waubant E., Chabas D., Okuda D., Glenn O., Mowry E., Henry R.G., et al. (2009) Difference in disease burden and activity in pediatric patients on brain magnetic resonance imaging at time of multiple sclerosis onset vs adults. Arch Neurol 66: 967–971 [DOI] [PubMed] [Google Scholar]
- Waubant E., Hietpas J., Stewart T., Dyme Z., Herbert J., Lacy J., et al. (2001) Interferon beta-1a in children with multiple sclerosis is well tolerated. Neuropediatrics 32: 211–213 [DOI] [PubMed] [Google Scholar]
- Weinshenker B.G., O'Brien P.C., Petterson T.M., Noseworthy J.H., Lucchinetti C.F., Dodick D.W., et al. (1999) A randomized trial of plasma exchange in acute central nervous system inflammatory demyelinating disease. Ann Neurol 46: 878–886 [DOI] [PubMed] [Google Scholar]
- Yousry T.A., Major E.O., Ryschkewitsch C., Fahle G., Fischer S., Hou J., et al. (2006) Evaluation of patients treated with natalizumab for progressive multifocal leukoencephalopathy. N Engl J Med 354: 924–933 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zivadinov R., Rudick R.A., De Masi, Nasuelli D., Ukmar M., Pozzi-Mucelli R.S., et al. (2001) Effects of IV methylprednisolone on brain atrophy in relapsing–remitting MS. Neurology 57: 1239–1247 [DOI] [PubMed] [Google Scholar]