Next Article in Journal
Ergonomic Challenges Inherent in Neonatal Resuscitation
Next Article in Special Issue
Differential Diagnosis of Pediatric Multiple Sclerosis
Previous Article in Journal
Digital Attention-Related Augmented-Reality Game: Significant Correlation between Student Game Performance and Validated Clinical Measures of Attention-Deficit/Hyperactivity Disorder (ADHD)
Previous Article in Special Issue
Clinical Approach to Pediatric Transverse Myelitis, Neuromyelitis Optica Spectrum Disorder and Acute Flaccid Myelitis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Long-Term Treatment Strategies of Pediatric Multiple Sclerosis, Including the use of Disease Modifying Therapies

The Mellen Center, Department of Neurology, Cleveland Clinic, Cleveland, OH 44195, USA
Children 2019, 6(6), 73; https://doi.org/10.3390/children6060073
Submission received: 22 March 2019 / Revised: 2 May 2019 / Accepted: 14 May 2019 / Published: 31 May 2019
(This article belongs to the Special Issue Multiple Sclerosis and Demyelinating Disorders in Children)

Abstract

:
Multiple sclerosis (MS) presenting in the pediatric years can lead to landmark disability levels younger in life than adult onset MS and so therefore early and effective treatment remains paramount for long-term outcomes. The goals of MS therapeutics in adults have widened to address multiple mechanisms: anti-inflammatory, neuroprotective, and myelin repair, yet the optimal paradigm for MS therapies in the pediatric population is not known. Pediatric onset MS add complexities due to the ongoing development of the central nervous system and the immune system. Clinical trials have led to an increasing number of pharmaceutical therapies for adult onset MS (AOMS), one POMS randomized controlled trial is completed and other trials are ongoing, yet due to the low prevalence of POMS, the dynamic landscape and risk management of the MS disease modifying therapies (DMT) it remains more difficult to complete trials in POMS. There is consensus that controlled clinical trials leading to appropriate and safe therapies for POMS are important for a multitude of reasons that include unique pediatric pharmacokinetics, short and long-term safety, developmental issues, clinical benefits, and regulatory approval. This review will focus on new treatment goals, paradigm, strategies, monitoring, compliance, and products in the long-term treatment of POMS. The discussion will focus on these new concepts and the published data related to DMT use in POMS. This review provides significant insight into new concepts of treatment goals and current approaches to enhance the lives of the POMS patients now and in the future.

1. Introduction

Multiple sclerosis (MS) presenting in the pediatric years can lead to landmark disability levels younger in life than adult onset Multiple sclerosis (AOMS) [1]. Early and effective treatment remains paramount to pediatric onset MS’s long-term outcomes. AOMS patients have increasing pharmacologic treatment options over the past 20 years and now controlled clinical trials in pediatric onset Multiple Sclerosis (POMS) have been legislatively mandated, leading to ongoing trials in POMS and one completed randomized double blind trial [2]. Current treatment of POMS most typically follows AOMS recommendations and treatment strategies, with the limitation of not addressing developmental stages of the patient and the developing immune system [3]. Treatment strategies and goals have been defined in AOMS and proposed in POMS [4]. Newer MS treatment strategies are gaining notoriety as a means of enhancing long term outcomes in MS. There is consensus that controlled clinical trials leading to appropriate and safe therapies for POMS are important for a multitude of reasons, including unique pediatric pharmacokinetics, short and long-term safety, developmental issues, clinical benefits, and regulatory approval [5]. This review will focus on new treatment goals, strategies, and products in the long-term treatment of POMS. The discussion will focus on these new concepts and the published data related to disease modifying therapy (DMT) use in POMS. This review provides significant insight into the new concepts of treatment goals and a current approach to enhance the lives of the POMS patients now and in the future.

2. Comprehensive Approach to POMS Care

Confirmation of the diagnosis of POMS is followed by a comprehensive approach to address the needs of the patient and family, including long-term use of DMT, social and school support, cognitive assessment, lifestyle assessment and modification as needed, symptom management, and mental health assessment and treatment. Typically, a team-based approach is needed to address these lifelong concerns of the POMS patient [6]. A POMS treatment team may include pediatricians, pediatric neurologists, neuroimmunologists, urologists, ophthalmologists, social workers, nurses, physical therapists, occupational therapists, neuropsychologists, health psychologists, cognitive therapists, nutritionists, and speech therapists. An MS Care Unit, a highly specialized treatment team, has been proposed due to the complexity of POMS DMT use and symptom management [7]. Treatment goals of POMS are similar to AOMS, yet there are additional POMS concerns. These unique concerns of the POMS patient are long-term DMT efficacy and safety, neurodevelopmental stage, pediatric pharmacokinetics, and pharmacodynamics. Cognitive dysfunction needs to be assessed and addressed early in the POMS population due to the threat of disabling cognitive outcomes [8]. Identification of the distinct injury and repair mechanisms unique to the younger patients may help to direct treatment paradigms and development of blood and imaging biomarkers, but for now the treatment recommendations vary widely.

3. Treatment Goals

Current MS treatment goals include modifying disease outcomes and maximizing patient safety. No evidence of disease activity (NEDA) is an accepted AOMS treatment goal referring to the absence of disease activity clinically and radiographically. NEDA has been redefined over the years to include more detailed disease outcomes. NEDA 3 has more detailed components: annualized relapse rate (ARR), number of new/newly enlarging T2 lesions (neT2), and no evidence of disease activity [9]. NEDA-3 was used a post hoc outcome in the adult RCTs of fingolimod treated versus placebo/Interferon β treated patients assessing outcomes of 3 RCT trials showing the strongest benefit in the youngest adult patients, ages 18–20, 21–30, or over 30 (all p < 0.05) and again proving NEDA as a realistic goal post for MS treatment [9]. NEDA 3 has further evolved to include cortical lesions (NEDA 3 + CL) [10]. Cognitive deficits are a common disabling symptom in POMS. AOMS cognitive deficits are associated with the presence of CLs. Inclusion of the cortical lesions in NEDA 3 + CL may be important in POMS where cortical lesions have been confirmed and may help to define POMS treatment goals [11].

4. Treatment Paradigms

The process of choosing a DMT should include a risk benefit discussion to clarify treatment goals of the provider, patient, and family. Clarifying individual’s priorities and trade- offs that may be acceptable to the patient and their family will focus the collective goals and risk tolerance. These priorities may be concordant or discordant between patient and provider and may depend on the patient and families’ understanding of the disease, neurocognitive status, and adherence to past medications [12,13]. Studies have shown the patient’s determinants of DMT choice include many variables: long term risk of disability, dosing, monitoring, cost, and safety of medication [14]. General practice guidelines and recommendations exist for DMT use in POMS, but the best treatment strategy is not known and is fluid, as more DMTs are available for use [15,16,17].
DMTs can be divided by their efficacy, mechanism of action, mode of administration, monitoring needs, safety, or by the length or frequency of the treatment [16]. DMT use should commence when the diagnosis is confirmed. Studies show frequent delays in treatment initiation in POMS. Mean DMT initiation delay of 20 months was seen in one POMS database review, which likely can lead to worsening disability due to ongoing inflammation and subsequent damage while awaiting treatment initiation [17,18]. Historically, the first line therapies for POMS have been injectable therapies including interferon β and glatiramer acetate supported by the AOMS and observational studies in POMS [19,20]. Long-term injectable DMT for relapsing forms of MS was first approved in the 1990s, see Table 1. From 2012–2019, nine new oral and infusion DMTs were approved for AOMS, enhancing the choices and pattern of use of DMTs for this population, see Table 2 and Table 3. The lack of randomized controlled trials (RCT) of DMT in POMS originally limited DMT use in POMS. One RCT has been completed in POMS, see Table 4 and many are ongoing. Currently there are 17 FDA approved DMTs, (see Table 5) for relapsing forms for AOMS and only one FDA approved DMT for POMS, and one FDA approved for adult onset primary progressive MS. EMA has approved Interferons and glatiramer acetate for patients 12 years of age and older. Canada has approved Fingolimod for POMS.
The pattern of use of DMT in POMS has changed with the availability of higher efficacy DMT. These previous “first line” therapies have frequent side effects, are more difficult, and have lower efficacy as compared to our newer DMT for AOMS [20]. The sequence of DMT varies according to provider’s experience and goals, patient’s preference and goals, payor, patient location, and even variability within one country can affect DMT choice [7]. The recent review of the United States’ Network of Pediatric MS Center’s database illustrated the change in pattern of DMT usage in POMS. The 2018 review included over 1000 POMS patients and the newer higher efficacy DMTs were used as first line in 42% of the POMS patients, the newer DMT included dimethyl fumarate (n = 102), natalizumab (n = 101), rituximab (n = 57), fingolimod (n = 37), daclizumab (n = 5), and teriflunomide (n = 3) showing the shift toward higher efficacy medications as first line for POMS in the US [17]. The DMTs are approved for use in AOMS at varying levels internationally (see Table 5 [29]). There are locations in the European Union and various payors in the US that dictate DMT algorithms. The high efficacy therapy protocol has been evaluated in uncontrolled smaller series in POMS, such as a Natalizumab series in 20 POMS patients showing the use of first line high efficacy DMT for POMS [30].
The trial of Fingolimod versus interferon β-1a in POMS is the first double blind, active comparator trial completed in POMS showing superior efficacy of fingolimod over interferon β-1a in reducing relapses in children and adolescents ages 12 to <18. The median duration of the trial regimen was 1.6 years. This trial confirmed POMS relapse rates were 2–3 times higher than AOMS [31]. The incidence of serious adverse events was higher in the fingolimod group as compared to the interferon β-1a group, serious infections were reported with fingolimod (see Table 6). Typical influenza-like symptoms were seen in the interferon group. There were 7 seizures reported in the trial, 6 with fingolimod, and 1 on interferon. Seizures have been reported more commonly in pediatric central nervous system demyelination syndromes, but this risk needs further monitoring. There were no reports of skin carcinoma that were seen in the AOMS trials with Fingolimod. Rebound disease, MS activity after cessation of fingolimod, has been reported in AOMS, yet not in POMS to date [32]. The discontinuation rates were lower for fingolimod as compared to interferon β-1a, half of the interferon β-1a. Discontinuation was commonly related to disease activity. The discontinuation rates related to adverse events were overall low: (Fingolimod 4.7 vs. IFN 2.8%) [2]. The fingolimod versus interferon β-1a trial is the first completed RCT in POMS and provides needed data addressing unique features of treatment issues in POMS.
Infusion therapies for MS have a longer lasting immune effect compared to oral DMT medications with variable windows of immune changes, natalizumab’s effects may last 4–16 weeks [26]. Alemtuzumab and Natalizumab have shown similar effects on relapse rates, and yet disability progression was less in Natalizumab as compared to Alemtuzumab, no controlled data in POMS [33]. Alemtuzumab has had effective pediatric use in heart and kidney transplants with known risks including immunosuppression and autoimmunity [34,35], and is currently being investigated as escalation therapy for POMS (ClinicalTrials.gov Identifier: NCT03368664). Rituximab has been assessed in a class 1 phase II study in AOMS showing significant improvement in relapse rate and MRI lesion load [36]. A reduction in relapse rate was shown in adolescents with POMS administered Rituximab [37]. The risk of severe infections including PML justifies randomized evaluation of these infusion therapies in POMS to clarify the long-term risk of these products in this unique population.
The dosing of certain pharmaceutical products in pediatric patients must take into account the unique absorption, distribution, metabolism, and elimination (ADME) of this age group [38]. The level of hepatic metabolizing enzymes may be significantly different in various age ranges and can differ between sexes as well. The renal glomerular filtration rate reaches adult values at 1 year of age and is therefore less of a concern [38]. The pharmacokinetics of a particular agent and potential differences according to the age of the child or adolescent must be taken into account due to variability. Pharmacodynamic studies in POMS are recommended as efficacy may be influenced by the differences in efficacy and tolerability according to the level of the developing immune system [38].

5. Monitoring Treatment in POMS

Monitoring of MS disease activity and DMT safety will depend on the DMT choice, the individual patient’s disease activity, and comorbidities (see Table 1, Table 2 and Table 3 for DMT monitoring needs). Due to risk of disability from POMS and concern over short-term and long-term safety of DMT POMS patients need regular clinical monitoring [1]. See Table 1, Table 2 and Table 3 for safety monitoring depending on DMT choice. Medication switches due to poor tolerance or noncompliance in POMS has been reported at a 16% rate in the first year of treatment [17]. An International Pediatric MS Study Group consensus statement defines inadequate treatment response in POMS: if the patient has been fully compliant on treatment for 6 months and demonstrates (1) no reduction in relapse rate or new T2 or contrast enhancing lesions (as compared to pretreatment); or (2) 2 or more confirmed relapses (clinical or MRI) within a 12-month period [38].

6. Escalation of Therapy

The optimal sequence of DMT is not known; there is lack of clarity on treatment sequencing in AOMS and POMS. There are uncontrolled studies of DMT used in the context of POMS showing similar efficacy as in the AOMS population. The terminology of first line versus second line is being replaced with the concepts of escalation and induction, as well as individualized therapy [4]. A 2011 open label multicenter series of POMS patients showed 55.8% of the patients with disease stability who therefore did not need to escalate therapy, while the rest of the patients needed 2–4 further DMT in 2 years due to refractory disease. The rate of change was similar if the patients started with Interferon or glatiramer [17]. The pattern of “first line” DMT choice is changing over time due to observational and controlled studies suggesting higher efficacy of newer DMT as compared with lower efficacy early approved DMT in AOMS and POMS, see primary and secondary outcomes of RCT in various DMT, see Table 7. The unique features of POMS have not been studied in all DMT [19]. The concept of utilizing high efficacy treatment in POMS from the disease onset has been proposed, the long term outcomes of which are not yet known [39]; the concern for long term disability in this population validates this proposal [40].
Escalation therapy refers to starting with low efficacy product and increasing the efficacy of the DMT depending on the NEDA status and tolerability. Comparison of the DMT AOMS studies has inherent limitations; multiple comparisons have been reported including a value based comparison from the Institute for Clinical and Economic review, ICER, looking at the strength of the evidence, patient goals, and costs for each FDA approved DMT products [47]. A standardized ICER evaluation of all MS DMT concluded the newest FDA AOMS approved product, ocrelizumab dominating the other DMTs when cost was compared with supportive care for the AOMS patient, showing it as the best value in the US market or as cost effective as a first line treatment for RRMS [27]. There is no similar ICER evaluation for DMT use in POMS. In the AOMS DMT ICER analysis Alemtuzumab dominated as the second line DMT over the other defined second line products: natalizumab, fingolimod and ocrelizumab for RRMS, providing more quality due to high efficacy and lower cost, while the other DMTs were similar in terms of cost and health outcomes. Maintaining up-to-date education on the changing landscape of DMT and risk tolerance also likely plays a role in the rate and timing of escalation therapy in various providers [28]. There has been global concern over the cost and access to DMTs, especially financial barriers to the patients and their families.

7. Immune Reconstitution

Immune reconstitution (IR) therapy, a “resetting” of the immune system, intermittent or noncontiguous therapy, applies to various medications and procedures. There have been suggestions that these noncontiguous AOMS therapies may indeed be cost effective, as well as improve quality of life [48]. Autologous stem cell transplantation for severe cases of POMS has been an early example of this type of IR therapy. Concerns over IR treatment related mortality in early studies predominated, yet there was evidence of long-lasting (3 years), change in the T cell repertoire as well as clinical stability in AOMS patients following autologous stem cell transplantation [49,50]. A recent AOMS randomized trial of myeloablative stem cell therapies (SCT) in relapsing AOMS showed delayed time to disease progression when compared to other DMT [51]. Mesenchymal stem cells (MSC) are present in various tissues including bone marrow, adipose and umbilical cord blood, and their immune altering capabilities have been exploited in various disease states. MSC are reported to have minimal known risk, potential immune alteration, and CNS regeneration, yet the lack of controlled studies in POMS precludes their current use in this population [52]. Pediatric neurologic conditions treated with MSC include autism, cerebral palsy, and spinal muscular atrophy (SMA). MSC use in pediatric SMA was noted to be low risk yet not long-lasting, as there was recurrence of symptoms in SMA. The recurrence of symptoms in pediatric SMA may mean that MSC may indeed be a “maintenance” therapy after all [53].

8. Clinical Trials in POMS

Due to concern over the lack of RCT data addressing the unique features of POMS, there is now a demand from the Federal (FDA) and European (EMA) regulatory agencies for a Pediatric Investigation plan for approval of a new biologic agent [38,54] The IPMSSG addressed the challenges of completing a RCT in the POMS population with the need for controlled trials to determine the safety and efficacy of the DMT in POMS in the IPMSSG consensus statement of 2012 [38]. The unique ethical issues of this population have been clarified: immunological immaturity, risk of community acquired infections, neurodevelopmental factors as well as short and long-term toxicities [55]. There are ongoing POMS clinical trials with dimethyl fumarate, teriflunomide and the infusion alemtuzumab looking at efficacy and safety of these products in POMS. DMT pattern of use in AOMS in regards to escalation versus early high efficacy DMT is being examined in two ongoing AOMS trials: (DELIVER-MS (NCT03535298) and TREAT-MS (NCT03500328) [56] yet the optimal approach for POMS is not known. The long-term safety of DMT use in POMS does not extend beyond the data of the fingolimod RCT in POMS.

9. Compliance and Adherence

Compliance and adherence of DMT are important issues in POMS as it involves both the patient and level of caregiver support. The POMS patients’ compliance and adherence need to be assessed regularly as a rate of 1–47% nonadherence has been seen, can lead to poor outcomes, and may be difficult to dramatically improve with behavioral interviewing or electronic monitoring [57,58]. The patient’s perception and preferences of the DMT’s risk, side effects, and dosing likely affect compliance as well. Compliance can have a long-term effect on disability outcomes as seen in a series of MS patients on injectable medication with the highest compliance reporting lowest disability progression [15,59]. Compliance with DMT for 6 months is assumed in the IPMSSG statement on DMT efficacy [38].

10. Biomarkers in MS

Biomarker is an objective measure of a biologic process or the pharmacologic response to a therapeutic intervention. Biomarkers may play a role in the DMT choice, DMT risk management, and predicting treatment response. Biomarkers would therefore help to effectively and efficiently tailor the care POMS patient and guide the caregiver and family. There are proposed biomarkers for the diagnosis of MS, the DMT choice, and DMT maintenance of the various disease modifying products, yet the majority of the data is in the AOMS (see Table 8 below). Neutralizing antibodies to the interferons have been known to lessen efficacy and are used to guide therapy with the current recommendation to stop interferons due to high titers of neutralizing antibody [60]. John Cunningham visual index, or JC index is used to gauge the safety of using Natalizumab due to risk of Progressive multifocal encephalopathy or PML. JC status, prior use of immunosuppressive products, and length of use of Natalizumab increase the risk of PML. If Biomarkers range from blood to CSF tests and include neutralizing antibodies leading to less efficacy of the DMT to serum or CSF evidence of neurotrauma, as that seen with Neurofilament light chain (NFL), a marker of white matter axonal injury. Glial fibrillary acidic proteins at elevated levels can also be seen in AOMS and in the future may be used as a biomarker of disease activity. POMS study showed high levels of NFL in the CSF of pediatric cases with acquired CNS demyelination [61]. A low cost, simple test, such as a blood test, to predict and/or monitor the treatment response which could enhance the long-term outcomes of the patient due to enhanced active monitoring of the MS activity would be welcome as it could minimize patient discomfort due to tests, as well as minimize their cost.
There are possible biomarkers to guide therapy choice and to determine disease activity; neurofilament light chain (NFL) is one such biomarker. NFL and glial fibrillary acidic protein (GFAP) concentrations in the serum and CSF have correlated with overall disease activity, reflecting a recent relapse, disease progression, or MRI activity with a new or active lesion and may reflect axonal injury [62,63]. A blood test as a means of MS disease assessment would be a novel and effective way of managing a chronic disease that now relies on expensive MRIs for assessment of subclinical disease activity.

11. Neuroregeneration in POMS

Demyelination, axon loss, and marginal tissue repair lead to eventual disability in MS. Acute axonal damage is more extensive in POMS as compared with AOMS [64]. New treatment targets include remyelination, neuroprotection, and/or neuroregeneration in MS. Pediatric MS can lead to disability and may take more years as compared with AMS, but it does cause early neurocognitive deficits and eventual physical disability as well. Improved neural repair has been postulated in POMS, but the mechanism is unknown. Remyelination is impaired in MS and intensified due to disease duration and advanced age. Remyelination has been tested in AOMS and a successful phase II remyelination trial with clemastine has been reported to possibly improve myelin and therefore nerve function in the optic nerve [65]. In AOMS, blood tests have revealed a product, Coco, a bone morphogenetic proteins antagonist. Related to remyelination failure, alternation of this antagonist may induce further repair [66]. There are vaccines in an early phase of trial aiming to use immune tolerance for neuroprotection. More research is needed to repair and prevent disability in POMS.

12. Conclusions

POMS patients have a growing armamentarium of therapeutic products to enhance their long-term outcome and improve their quality of life. POMS has unique features when compared with AOMS: more inflammatory disease, more frequent relapses, neurocognitive disability levels, rates of cerebral atrophy, quantity of axonal damage, and MRI lesion accrual [4]; recommended POMS therapies should address these unique features. Due to the complexity of POMS, it is best to seek out an MS Care unit with a team-based approach and knowledge of risk management with the multiple DMTs. Shared decision making, a discussion of the goals of the DMT and the goals of the patient and family are paramount for achieving long-term success with the treatment course. The treatment goals should be expressed early and defined regularly to enhance compliance. Risk of the DMTs can be severe, and priorities need to be defined; the DMTs with black box warnings need to be reviewed in detail with the patient and their family. Higher efficacy DMTs are now used more often in POMS, and as such, less disability and relapses are being reported. Complementary and lifestyle “treatments” and/or modification of higher risk environmental factors may also enhance long-term outcomes in POMS and can be considered in collaboration with needed pharmaceutical products [61]. There is one completed randomized controlled clinical trial in POMS confirming the efficacy and safety of fingolimod in this population. Biomarkers before and during treatments can help predict and monitor treatment response. There are ongoing clinical trials in POMS to add to the efficacy and safety data of more DMTs for this unique population. Newer treatment goals of nerve repair and/or remyelination have not been systematically studied in POMS, but are goals to work toward in order to lessen and/or repair disability. For now, identifying the patient and family goals, defining MS Care Unit’s treatment goals, optimal DMT management, and a healthy lifestyle can serve the patient and family in the long run.

Funding

This research received no external funding.

Conflicts of Interest

Mary Rensel serves as a consultant or speaker for Biogen, Teva, Genzyme and Novartis. She receives educational grant funding from Genzyme and research grant funding from NMSS and MedImmune.

References

  1. Renoux, C.; Vukusic, S.; Mikaeloff, Y.; Edan, G.; Clanet, M.; Dubois, B.; Debouverie, M.; Brochet, B.; Lebrun-Frenay, C.; Pelletier, J.; et al. Natural History of Multiple Sclerosis with Childhood Onset. N. Engl. J. Med. 2007, 356, 2603–2613. [Google Scholar] [CrossRef] [PubMed]
  2. Chitnis, T.; Arnold, D.L.; Banwell, B.; Brück, W.; Ghezzi, A.; Giovannoni, G.; Greenberg, B.; Krupp, L.; Rostásy, K.; Tardieu, M.; et al. Trial of Fingolimod versus Interferon Beta-1a in Pediatric Multiple Sclerosis. N. Engl. J. Med. 2018, 379, 1017–1027. [Google Scholar] [CrossRef] [PubMed]
  3. Schwarz, A.; Balint, B.; Korporal-Kuhnke, M.; Jarius, S.; von Engelhardt, K.; Fürwentsches, A.; Bussmann, C.; Ebinger, F.; Wildemann, B.; Haas, J. B-cell populations discriminate between pediatric- and adult-onset multiple sclerosis. Neurol. Neuroimmunol. Neuroinflamm. 2017, 4, e309. [Google Scholar] [CrossRef] [PubMed]
  4. Chitnis, T.; Ghezzi, A.; Bajer-Kornek, B.; Boyko, A.; Giovannoni, G.; Pohl, D. Pediatric multiple sclerosis: Escalation and emerging treatments. Neurology 2016, 87, S103–S109. [Google Scholar] [CrossRef] [PubMed]
  5. Chitnis, T.; Tardieu, M.; Amato, M.P.; Banwell, B.; Bar-Or, A.; Ghezzi, A.; Kornberg, A.; Krupp, L.B.; Pohl, D.; Rostasy, K.; et al. International Pediatric MS Study Group Clinical Trials Summit: Meeting report. Neurology 2013, 80, 1161–1168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Cappa, R.; Theroux, L.; Brenton, J.N. Pediatric Multiple Sclerosis: Genes, Environment, and a Comprehensive Therapeutic Approach. Pediatr. Neurol. 2017, 75, 17–28. [Google Scholar] [CrossRef] [PubMed]
  7. Soelberg Sorensen, P.; Giovannoni, G.; Montalban, X.; Thalheim, C.; Zaratin, P.; Comi, G. The Multiple Sclerosis Care Unit. Mult. Scler. J. 2019, 25, 627–636. [Google Scholar] [CrossRef]
  8. Julian, L.; Serafin, D.; Charvet, L.; Ackerson, J.; Benedict, R.; Braaten, E.; Brown, T.; O’Donnell, E.; Parrish, J.; Preston, T.; et al. Cognitive Impairment Occurs in Children and Adolescents With Multiple Sclerosis: Results From a United States Network. J. Child Neurol. 2013, 28, 102–107. [Google Scholar] [CrossRef]
  9. Gärtner, J.; Chitnis, T.; Ghezzi, A.; Pohl, D.; Brück, W.; Häring, D.A.; Karlsson, G.; Putzki, N. Relapse Rate and MRI Activity in Young Adult Patients with Multiple Sclerosis: A Post Hoc Analysis of Phase 3 Fingolimod Trials. Mult. Scler. J. Exp. Translat. Clin. 2018, 4, 205521731877861. [Google Scholar] [CrossRef]
  10. Puthenparampil, M.; Cazzola, C.; Zywicki, S.; Federle, L.; Stropparo, E.; Anglani, M.; Rinaldi, F.; Perini, P.; Gallo, P. NEDA-3 status including cortical lesions in the comparative evaluation of natalizumab versus fingolimod efficacy in multiple sclerosis. Ther. Adv. Neurol. Disord. 2018, 11, 175628641880571. [Google Scholar] [CrossRef]
  11. Maranzano, J.; Till, C.; Assemlal, H.-E.; Fonov, V.; Brown, R.; Araujo, D.; O’Mahony, J.; Yeh, E.A.; Bar-Or, A.; Marrie, R.A.; et al. Detection and clinical correlation of leukocortical lesions in pediatric-onset multiple sclerosis on multi-contrast MRI. Mult. Scler. 2018, 25. [Google Scholar] [CrossRef] [PubMed]
  12. Brett Hauber, A.; Fairchild, A.O.; Reed Johnson, F. Quantifying Benefit–Risk Preferences for Medical Interventions: An Overview of a Growing Empirical Literature. Appl. Health Econ. Health Policy 2013, 11, 319–329. [Google Scholar] [CrossRef] [PubMed]
  13. Bruce, J.M.; Bruce, A.S.; Lynch, S.; Thelen, J.; Lim, S.-L.; Smith, J.; Catley, D.; Reed, D.D.; Jarmolowicz, D.P. Probability discounting of treatment decisions in multiple sclerosis: Associations with disease knowledge, neuropsychiatric status, and adherence. Psychopharmacology 2018, 235, 3303–3313. [Google Scholar] [CrossRef] [PubMed]
  14. Poulos, C.; Kinter, E.; van Beek, J.; Christensen, K.; Posner, J. Preferences of Patients with Multiple Sclerosis for Attributes of Injectable Multiple Sclerosis Treatments in the United Kingdom and France. Int. J. Technol. Assess. Health Care 2018, 34, 425–433. [Google Scholar] [CrossRef] [PubMed]
  15. Rae-Grant, A.; Day, G.S.; Marrie, R.A.; Rabinstein, A.; Cree, B.A.C.; Gronseth, G.S.; Haboubi, M.; Halper, J.; Hosey, J.P.; Jones, D.E.; et al. Practice guideline recommendations summary: Disease-modifying therapies for adults with multiple sclerosis: Report of the Guideline Development, Dissemination, and Implementation Subcommittee of the American Academy of Neurology. Neurology 2018, 90, 777–788. [Google Scholar] [CrossRef] [PubMed]
  16. Karussis, D.; Petrou, P. Immune reconstitution therapy (IRT) in multiple sclerosis: The rationale. Immunol. Res. 2018, 66, 642–648. [Google Scholar] [CrossRef] [PubMed]
  17. Yeh, E.A.; Waubant, E.; Krupp, L.B.; Ness, J.; Chitnis, T.; Kuntz, N.; Ramanathan, M.; Belman, A.; Chabas, D.; Gorman, M.P.; et al. Multiple Sclerosis Therapies in Pediatric Patients with Refractory Multiple Sclerosis. Arch. Neurol. 2011, 68, 437. [Google Scholar] [CrossRef] [PubMed]
  18. Comi, G.; Radaelli, M.; Sørensen, P.S. Evolving concepts in the treatment of relapsing multiple sclerosis. Lancet 2017, 389, 1347–1356. [Google Scholar] [CrossRef]
  19. Krysko, K.M.; Graves, J.; Rensel, M.; Weinstock-Guttman, B.; Aaen, G.; Benson, L.; Chitnis, T.; Gorman, M.; Goyal, M.; Krupp, L.; et al. Use of newer disease-modifying therapies in pediatric multiple sclerosis in the US. Neurology 2018, 91, e1778–e1787. [Google Scholar] [CrossRef]
  20. Waldman, A.T.; Gorman, M.P.; Rensel, M.R.; Austin, T.E.; Hertz, D.P.; Kuntz, N.L. Network of Pediatric Multiple Sclerosis Centers of Excellence of National Multiple Sclerosis Society Management of pediatric central nervous system demyelinating disorders: Consensus of United States neurologists. J. Child Neurol. 2011, 26, 675–682. [Google Scholar] [CrossRef]
  21. Ghezzi, A.; on behalf of the Immunomodulatory Treatment of Early onset MS (ITEMS) Group. Immunomodulatory treatment of early onset multiple sclerosis: Results of an Italian Co-operative Study. Neurol. Sci. 2005, 26, s183–s186. [Google Scholar] [CrossRef] [PubMed]
  22. Higurashi, N.; Hamano, S.; Eto, Y. Secondary progressive multiple sclerosis in childhood--interferon beta 1b treatment. No To Hattatsu 2006, 38, 209–213. [Google Scholar] [PubMed]
  23. Ghezzi, A.; Amato, M.P.; Capobianco, M.; Gallo, P.; Marrosu, G.; Martinelli, V.; Milani, N.; Milanese, C.; Moiola, L.; Patti, F.; et al. Disease-modifying drugs in childhood-juvenile multiple sclerosis: Results of an Italian co-operative study. Mult. Scler. J. 2005, 11, 420–424. [Google Scholar] [CrossRef] [PubMed]
  24. Gorman, M.P. Daclizumab Use in Patients with Pediatric Multiple Sclerosis. Arch. Neurol. 2012, 69, 78. [Google Scholar] [CrossRef] [PubMed]
  25. Alroughani, R.; Das, R.; Penner, N.; Pultz, J.; Taylor, C.; Eraly, S. Safety and Efficacy of Delayed-Release Dimethyl Fumarate in Pediatric Patients with Relapsing Multiple Sclerosis (FOCUS). Pediatr. Neurol. 2018, 83, 19–24. [Google Scholar] [CrossRef] [PubMed]
  26. Plavina, T.; Muralidharan, K.K.; Kuesters, G.; Mikol, D.; Evans, K.; Subramanyam, M.; Nestorov, I.; Chen, Y.; Dong, Q.; Ho, P.-R.; et al. Reversibility of the effects of natalizumab on peripheral immune cell dynamics in MS patients. Neurology 2017, 89, 1584–1593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Zimmermann, M.; Brouwer, E.; Tice, J.A.; Seidner, M.; Loos, A.M.; Liu, S.; Chapman, R.H.; Kumar, V.; Carlson, J.J. Disease-Modifying Therapies for Relapsing–Remitting and Primary Progressive Multiple Sclerosis: A Cost-Utility Analysis. CNS Drugs 2018, 32, 1145–1157. [Google Scholar] [CrossRef]
  28. Saposnik, G.; Montalban, X.; Selchen, D.; Terzaghi, M.A.; Bakdache, F.; Montoya, A.; Fruns, M.; Caceres, F.; Oh, J. Therapeutic Inertia in Multiple Sclerosis Care: A Study of Canadian Neurologists. Front. Neurol. 2018, 9. [Google Scholar] [CrossRef]
  29. Tillery, E.E.; Clements, J.N.; Howard, Z. What’s new in multiple sclerosis? Ment. Health Clin. 2017, 7, 213–220. [Google Scholar] [CrossRef]
  30. Kornek, B.; Aboul-Enein, F.; Rostasy, K.; Milos, R.-I.; Steiner, I.; Penzien, J.; Hellwig, K.; Pitarokoili, K.; Storm van’s Gravesande, K.; Karenfort, M.; et al. Natalizumab Therapy for Highly Active Pediatric Multiple Sclerosis. JAMA Neurol. 2013, 70, 469. [Google Scholar] [CrossRef]
  31. Gorman, M.P.; Healy, B.C.; Polgar-Turcsanyi, M.; Chitnis, T. Increased relapse rate in pediatric-onset compared with adult-onset multiple sclerosis. Arch. Neurol. 2009, 66, 54–59. [Google Scholar] [CrossRef] [PubMed]
  32. Hatcher, S.E.; Waubant, E.; Nourbakhsh, B.; Crabtree-Hartman, E.; Graves, J.S. Rebound Syndrome in Patients With Multiple Sclerosis After Cessation of Fingolimod Treatment. JAMA Neurol. 2016, 73, 790. [Google Scholar] [CrossRef] [PubMed]
  33. Kalincik, T.; Brown, J.W.L.; Robertson, N.; Willis, M.; Scolding, N.; Rice, C.M.; Wilkins, A.; Pearson, O.; Ziemssen, T.; Hutchinson, M.; et al. Treatment effectiveness of alemtuzumab compared with natalizumab, fingolimod, and interferon beta in relapsing-remitting multiple sclerosis: A cohort study. Lancet Neurol. 2017, 16, 271–281. [Google Scholar] [CrossRef]
  34. Das, B.; Dimas, V.; Guleserian, K.; Lacelle, C.; Anton, K.; Moore, L.; Morrow, R. Alemtuzumab (Campath-1H) therapy for refractory rejections in pediatric heart transplant recipients. Pediatr. Transplant. 2017, 21, e12844. [Google Scholar] [CrossRef] [PubMed]
  35. Knechtle, S.J.; Fernandez, L.A.; Pirsch, J.D.; Becker, B.N.; Chin, L.T.; Becker, Y.T.; Odorico, J.S.; D’Alessandro, A.M.; Sollinger, H.W. Campath-1H in renal transplantation: The University of Wisconsin experience. Surgery 2004, 136, 754–760. [Google Scholar] [CrossRef] [PubMed]
  36. Hauser, S.L.; Waubant, E.; Arnold, D.L.; Vollmer, T.; Antel, J.; Fox, R.J.; Bar-Or, A.; Panzara, M.; Sarkar, N.; Agarwal, S.; et al. B-Cell Depletion with Rituximab in Relapsing–Remitting Multiple Sclerosis. N. Engl. J. Med. 2008, 358, 676–688. [Google Scholar] [CrossRef]
  37. Tzaribachev, N.; Koetter, I.; Kuemmerle-Deschner, J.B.; Schedel, J. Rituximab for the treatment of refractory pediatric autoimmune diseases: A case series. Cases J. 2009, 2, 6609. [Google Scholar] [CrossRef]
  38. Chitnis, T.; Tenembaum, S.; Banwell, B.; Krupp, L.; Pohl, D.; Rostasy, K.; Yeh, E.A.; Bykova, O.; Wassmer, E.; Tardieu, M.; et al. Consensus statement: Evaluation of new and existing therapeutics for pediatric multiple sclerosis. Mult. Scler. J. 2012, 18, 116–127. [Google Scholar] [CrossRef]
  39. McGinley, M.; Rossman, I.T. Bringing the HEET: The Argument for High-Efficacy Early Treatment for Pediatric-Onset Multiple Sclerosis. Neurotherapeutics 2017, 14, 985–998. [Google Scholar] [CrossRef]
  40. Waubant, E.; Chabas, D.; Okuda, D.T.; Glenn, O.; Mowry, E.; Henry, R.G.; Strober, J.B.; Soares, B.; Wintermark, M.; Pelletier, D. Difference in Disease Burden and Activity in Pediatric Patients on Brain Magnetic Resonance Imaging at Time of Multiple Sclerosis Onset vs Adults. Arch. Neurol. 2009, 66. [Google Scholar] [CrossRef]
  41. Gold, R.; Kappos, L.; Arnold, D.L.; Bar-Or, A.; Giovannoni, G.; Selmaj, K.; Tornatore, C.; Sweetser, M.T.; Yang, M.; Sheikh, S.I.; et al. Placebo-Controlled Phase 3 Study of Oral BG-12 for Relapsing Multiple Sclerosis. N. Engl. J. Med. 2012, 367, 1098–1107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Cohen, J.A.; Barkhof, F.; Comi, G.; Hartung, H.-P.; Khatri, B.O.; Montalban, X.; Pelletier, J.; Capra, R.; Gallo, P.; Izquierdo, G.; et al. Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. N. Engl. J. Med. 2010, 362, 402–415. [Google Scholar] [CrossRef] [PubMed]
  43. Hauser, S.L.; Bar-Or, A.; Comi, G.; Giovannoni, G.; Hartung, H.-P.; Hemmer, B.; Lublin, F.; Montalban, X.; Rammohan, K.W.; Selmaj, K.; et al. Ocrelizumab versus Interferon Beta-1a in Relapsing Multiple Sclerosis. N. Engl. J. Med. 2017, 376, 221–234. [Google Scholar] [CrossRef] [PubMed]
  44. Syed, Y.Y. Ocrelizumab: A Review in Multiple Sclerosis. CNS Drugs 2018, 32, 883–890. [Google Scholar] [CrossRef] [PubMed]
  45. O’Connor, P.; Wolinsky, J.S.; Confavreux, C.; Comi, G.; Kappos, L.; Olsson, T.P.; Benzerdjeb, H.; Truffinet, P.; Wang, L.; Miller, A.; et al. Randomized Trial of Oral Teriflunomide for Relapsing Multiple Sclerosis. N. Engl. J. Med. 2011, 365, 1293–1303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Kretzschmar, B.; Pellkofer, H.; Weber, M.S. The Use of Oral Disease-Modifying Therapies in Multiple Sclerosis. Curr. Neurol. Neurosci. Rep. 2016, 16. [Google Scholar] [CrossRef] [PubMed]
  47. Guarnera, C.; Bramanti, P.; Mazzon, E. Comparison of efficacy and safety of oral agents for the treatment of relapsing–remitting multiple sclerosis. Drug Des. Dev. Ther. 2017, 11, 2193–2207. [Google Scholar] [CrossRef]
  48. Walter, E.; Berger, T.; Bajer-Kornek, B.; Deisenhammer, F. Cost-utility analysis of alemtuzumab in comparison with interferon beta, fingolimod, and natalizumab treatment for relapsing-remitting multiple sclerosis in Austria. J. Med. Econ. 2018, 1–12. [Google Scholar] [CrossRef]
  49. Tyndall, A. Application of autologous stem cell transplantation in various adult and pediatric rheumatic diseases. Pediatr. Res. 2012, 71, 433–438. [Google Scholar] [CrossRef] [Green Version]
  50. Muraro, P.A.; Douek, D.C.; Packer, A.; Chung, K.; Guenaga, F.J.; Cassiani-Ingoni, R.; Campbell, C.; Memon, S.; Nagle, J.W.; Hakim, F.T.; et al. Thymic output generates a new and diverse TCR repertoire after autologous stem cell transplantation in multiple sclerosis patients. J. Exp. Med. 2005, 201, 805–816. [Google Scholar] [CrossRef]
  51. Burt, R.K.; Balabanov, R.; Burman, J.; Sharrack, B.; Snowden, J.A.; Oliveira, M.C.; Fagius, J.; Rose, J.; Nelson, F.; Barreira, A.A.; et al. Effect of Nonmyeloablative Hematopoietic Stem Cell Transplantation vs Continued Disease-Modifying Therapy on Disease Progression in Patients With Relapsing-Remitting Multiple Sclerosis: A Randomized Clinical Trial. JAMA 2019, 321, 165. [Google Scholar] [CrossRef] [PubMed]
  52. Drela, K.; Siedlecka, P.; Sarnowska, A.; Domanska-Janik, K. Human mesenchymal stem cells in the treatment of neurological diseases. Acta Neurobiol. Exp. 2013, 73, 38–56. [Google Scholar]
  53. Feng, M.; Lu, A.; Gao, H.; Qian, C.; Zhang, J.; Lin, T.; Zhao, Y. Safety of Allogeneic Umbilical Cord Blood Stem Cells Therapy in Patients with Severe Cerebral Palsy: A Retrospective Study. Stem Cells Int. 2015, 2015, 1–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Bavdekar, S. Pediatric clinical trials. Persp. Clin. Res. 2013, 4, 89. [Google Scholar] [CrossRef] [PubMed]
  55. Tenembaum, S.N. Ethical challenges in paediatric clinical trials in multiple sclerosis. Ther. Adv. Neurol. Disord. 2012, 5, 139–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Mahajan, K.R.; Nakamura, K.; Ontaneda, D. Treatment decisions in MS: Shifting the goal posts or changing how we see them? Mult. Scler. J. 2018, 24, 1523–1525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Thannhauser, J.E.; Mah, J.K.; Metz, L.M. Adherence of Adolescents to Multiple Sclerosis Disease-Modifying Therapy. Pediatr. Neurol. 2009, 41, 119–123. [Google Scholar] [CrossRef] [PubMed]
  58. Yeh, E.A.; Grover, S.A.; Powell, V.E.; Alper, G.; Banwell, B.L.; Edwards, K.; Gorman, M.; Graves, J.; Lotze, T.E.; Mah, J.K.; et al. Impact of an electronic monitoring device and behavioral feedback on adherence to multiple sclerosis therapies in youth: Results of a randomized trial. Qual. Life Res. 2017, 26, 2333–2349. [Google Scholar] [CrossRef] [PubMed]
  59. Fox, R.J.; Salter, A.R.; Tyry, T.; Sun, J.; You, X.; Laforet, G.; Campagnolo, D. Treatment Discontinuation and Disease Progression with Injectable Disease-Modifying Therapies: Findings from the North American Research Committee on Multiple Sclerosis Database. Int. J. MS Care 2013, 15, 194–201. [Google Scholar] [CrossRef]
  60. Polman, C.H.; Bertolotto, A.; Deisenhammer, F.; Giovannoni, G.; Hartung, H.-P.; Hemmer, B.; Killestein, J.; McFarland, H.F.; Oger, J.; Pachner, A.R.; et al. Recommendations for clinical use of data on neutralising antibodies to interferon-beta therapy in multiple sclerosis. Lancet Neurol. 2010, 9, 740–750. [Google Scholar] [CrossRef]
  61. Boesen, M.S.; Jensen, P.E.H.; Magyari, M.; Born, A.P.; Uldall, P.V.; Blinkenberg, M.; Sellebjerg, F. Increased cerebrospinal fluid chitinase 3-like 1 and neurofilament light chain in pediatric acquired demyelinating syndromes. Mult. Scler. Relat. Disord. 2018, 24, 175–183. [Google Scholar] [CrossRef] [PubMed]
  62. Novakova, L.; Zetterberg, H.; Sundström, P.; Axelsson, M.; Khademi, M.; Gunnarsson, M.; Malmeström, C.; Svenningsson, A.; Olsson, T.; Piehl, F.; et al. Monitoring disease activity in multiple sclerosis using serum neurofilament light protein. Neurology 2017, 89, 2230–2237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Högel, H.; Rissanen, E.; Barro, C.; Matilainen, M.; Nylund, M.; Kuhle, J.; Airas, L. Serum glial fibrillary acidic protein correlates with multiple sclerosis disease severity. Mult. Scler. J. 2018. [Google Scholar] [CrossRef] [PubMed]
  64. Pfeifenbring, S.; Bunyan, R.F.; Metz, I.; Röver, C.; Huppke, P.; Gärtner, J.; Lucchinetti, C.F.; Brück, W. Extensive acute axonal damage in pediatric multiple sclerosis lesions: Axonal Damage in Pediatric MS. Ann. Neurol. 2015, 77, 655–667. [Google Scholar] [CrossRef] [PubMed]
  65. Green, A.J.; Gelfand, J.M.; Cree, B.A.; Bevan, C.; Boscardin, W.J.; Mei, F.; Inman, J.; Arnow, S.; Devereux, M.; Abounasr, A.; et al. Clemastine fumarate as a remyelinating therapy for multiple sclerosis (ReBUILD): A randomised, controlled, double-blind, crossover trial. Lancet 2017, 390, 2481–2489. [Google Scholar] [CrossRef]
  66. Mausner-Fainberg, K.; Penn, M.; Golan, M.; Benhamou, M.; Gertel, S.; Karni, A. Reduced levels of Coco in sera of multiple sclerosis patients: A potential role in neuro-regeneration failure. J. Neuroimmunol. 2019, 15, 36–40. [Google Scholar] [CrossRef] [PubMed]
Table 1. Summary of injectable disease modifying therapy for pediatric-onset multiple sclerosis (POMS).
Table 1. Summary of injectable disease modifying therapy for pediatric-onset multiple sclerosis (POMS).
Interferon Β 1a
Mechanism of ActionShifts cytokine balance to a more anti-inflammatory profile; reduces trafficking of inflammatory cells across the blood brain barrier
DosingDose titration to start 22 or 44 µg sq three times weekly; 30 µg im weekly; or 125 µg sq every other week
Side effectinjection-site erythema, influenza-like illness, pyrexia, headache, myalgia, chills, injection-site pain, injection-site pruritus, arthralgia and asthenia
MonitoringCBC and LFT at baseline and q 6 months
Pregnancy test prior to use, it is not recommended to get pregnant or nurse while on this product
Adverse eventsdepression
elevated transaminases and thyroid abnormalities
Neutralizing antibodies to interferons
POMS publications[21]
POMS RCTOngoing, ClinicalTrials.gov Identifier: NCT03870763
Interferon β-1b
Mechanism of ActionShifts cytokine balance to a more anti-inflammatory profile; reduces trafficking of inflammatory cells across the blood brain barrier
Dosing0.25mg sq every other day
Side effectsSee Interferon β-1a
Monitoring
Adverse events
POMS publications[22]
POMS RCTnone
Glatiramer Acetate
Mechanism of actionModulates function of antigen-presenting cells; induces differentiation of CD4 + T cells into Th2 cells
Dosing 20 mg sq every day or 40 mg sq three times a week
Side effectsinjection-site reactions, chest pain, rash, dyspnea, and vasodilation
MonitoringNo blood work required
Adverse eventsLipoatrophy, skin necrosis
POMS publications[23]
POMS RCTnone
Daclizumab
Mechanism of actionHumanized monoclonal antibody to alpha subunit of IL-2 receptor, reducing IL–2-mediated activation of lymphocytes
Dosing150 mg sq once monthly
Side effectsinfluenza, bronchitis, eczema, lymphadenopathy, nasopharyngitis, upper respiratory tract infection, rash, and dermatitis. Rash (7% to 11%), depression (7% to 10%), upper respiratory tract infection (9% to 17%), pharyngitis (25%), and elevated ALT levels (5% to 6%) http://www.accessdata.fda.gov/drugsatfda_docs/label/2016/761029s000lbl.pdf
Monitoring Assess for Hepatitis B and C, Daclizumab is contraindicated in Hepatitis B and C
Assess for LFT at baseline, preexisting hepatic disease or impairment (ALT or aspartate transaminase ≥2 times the upper limit) or history of autoimmune hepatitis is contraindicated
Pregnancy test prior to use, it is not recommended to get pregnant or nurse while on this product
Adverse eventsAutoimmune Encephalitis (removed from US market 2018)
Colitis
Elevated Liver enzymes
POMS publication[24]
POMS RCTnone
Abbreviations: sq = subcutaneous, im = intramuscular, CBC = complete blood count, LFT = liver function test.
Table 2. Summary of oral disease modifying therapy for POMS.
Table 2. Summary of oral disease modifying therapy for POMS.
Fingolimod
Mechanism of ActionSphingosine 1 phosphate receptor modulator
Dosing0.25 or 0.5 mg by mouth once a day
Side effectsHeadache
Back pain
Diarrhea cough
AELow ALC
QTc prolongation
Respiratory Infection
High ALT
Rebound disease activity
Rare PML
Bradycardia
First- and second-degree block
Rebound disease sinusitis back pain first-dose monitoring requirement, contraindications in heart disease, risk of macular edema skin cancers
Monitoringcontraindications in heart disease
VZV serum titer, pre FDO
OCT pre and 3 months post FDO use with caution with other medications that prolong QTc interval
Requires FDO < 6 h of observation with first dose with hourly vitals
EKG before and after FDO
CBC with diff baseline, 3 months then every 6 months
JC index
Avoid live vaccines while on Fingolimod and for 2 months after it is stopped.
Pregnancy test prior to use, it is not recommended to get pregnant or nurse while on this product
POMS publications[9]
POMS RCTCompleted [2]
FDA approved in POMSApproved in POMS 2018
Dimethyl Fumarate
Mechanism of ActionIncrease Nuclear factor erythroid 2 (NF-E2)-related factor 2
Dosing120 mg oral twice a day × 7 days then 240 mg oral twice a day
Side effectsFlushing, diarrhea, nausea, vomiting, and abdominal pain
Adverse eventsLow absolute lymphocyte count
Rare PML case
MonitoringBaseline CBC with diff and LFT then q 12 months
Pregnancy test prior to use, it is not recommended to get pregnant or nurse while on this product
POMS publications[25]
POMS RCTYes, ongoing BG00012, ClinicalTrials.gov Identifier: NCT02283853
ClinicalTrials.gov Identifier: NCT03870763
FDA approved for POMSNo
Teriflunomide
Mechanism of ActionInhibits pyrimidine biosynthesis
Dosing7 mg or 14 mg oral once a day
Side effectsDiarrhea
Nausea
Headache
Adverse eventsDecreased hair density
Elevated Alanine aminotransferase
Increased blood pressure
Paresthesia in the upper extremities
Major birth defects and embyrolethality in animal studies relatively long half-life of approximately 19 days, elimination can take >8 months
MonitoringCBC with diff, LFT baseline, LFT q month × 6, Blood pressure monitoring
Effective birth control compliance in male and female patients
Contraindicated in severe hepatic impairment and those of child bearing potential not using effective birth control (http://products.sanofi.us/aubagio/aubagio.pdf)
Pregnancy test prior to use, it is not recommended to get pregnant or nurse while on this product
Rapid elimination due to long half lifeIf drug-induced liver injury or pregnancy is suspected, discontinue AUBAGIO and start an accelerated elimination procedure with cholestyramine or activated charcoal
POMS PublicationsNone
POMS RCTYes, ongoing ClinicalTrials.gov Identifier: NCT02201108
FDA approved for POMSNone
Abbreviations: ALC = absolute lymphocyte count, FDO = First dose observation, LFT = liver function tests, VZV = Varicella Zoster virus, PML = progressive multifocal leukoencephalopathy.
Table 3. Summary of infusion disease modifying therapy for POMS.
Table 3. Summary of infusion disease modifying therapy for POMS.
Natalizumab
Mechanism of ActionInhibits the transmigration of immune cells across the blood-brain barrier thus inhibiting inflammation in the central nervous system.
Dosing300 mg Intravenous every 4 weeks or 3–5 mg/kg IV every 4 weeks
Side effectsInfusion reactions
Headaches
Fatigue
Adverse events PML
Infections
Neutralizing antibodies
JC antibodies
Rebound disease
Encephalitis
Meningitis
Acute retinal necrosis due to herpes infection
Monitoring Risk evaluation mitigation strategy due to risk of PML
Monitor for the risk of PML while on Natalizumab and at least 6 months after stopping Natalizumab
Monitor patients for signs or symptoms of PML, if any concerning signs, hold treatment and evaluate the patient as needed, clinically, MRI and possibly cerebrospinal fluid for JC virus DNA
John Cunningham virus index q 3 months
CBC and LFT q 6 months
Natalizumab antibodies
Monitor 1 h after infusion
Pregnancy test prior to use, it is not recommended to get pregnant or nurse while on this product
PMLProgressive Multifocal Leukoencephalopathy risk factors:
  • Prior use of immunosuppressants
  • Presence of anti-JCV antibodies
  • Length of time on Natalizumab
POMS publications[26,27,28]
POMS RCTnone
FDA approval in POMSnone
Alemtuzumab
Mechanism of actionmonoclonal antibody against CD52 and leads to depletion of mature lymphocytes
Dosing12 mg IV daily × 5 year 1 then 12 mg IV daily × 3 in year 2 and then as needed depending on disease activity
Side effectsInfusion reactions rash, headache, vomiting, nausea, diarrhea, flushing, abdominal pain, pyrexia, nasopharyngitis, urinary tract infection, fatigue, insomnia, upper respiratory tract infection, herpes viral infection, urticaria, pruritus, thyroid gland disorders, fungal infections, arthralgia, extremity pain, back pain, sinusitis, oropharyngeal pain, paresthesia, and dizziness
Adverse eventsAutoimmune conditions
Infections
Possible cancer
Premedication needed for infusion
Antiviral needed for risk of viral infection until CD4 > 200 cells/u/L
MonitoringVaricella Zoster virus titers before infusion, vaccinate as needed
All necessary immunizations, including varicella zoster vaccine, should be completed at a minimum of 6 weeks prior to administration of alemtuzumab
risk evaluation mitigation strategy program due to risk of serious adverse reactions
Continuous observation × 2 h after infusion
Labs and urine month × 5 years after last infusion
CD4
Risk management strategy
Pregnancy test prior to use, it is not recommended to get pregnant or nurse while on this product
POMS Publicationsnone
POMS RCTYes, ongoing (ClinicalTrials.gov Identifier: NCT03368664)
FDA approval in POMSnone
Ocrelizumab
Mechanism of ActionHumanized form of the CD20 monoclonal antibody
Dosing 300 mg IV repeated in 2 weeks then 600 mg IV every 6 months.
Pretreatment with IV steroids and Benadryl
Side effectsInfusion reaction
Adverse eventsUpper respiratory tract infections (40% to 49%), lower respiratory tract infections (8% to 10%), infusion reactions (34% to 40%), and infection of the skin and/or subcutaneous tissue (14%) https://www.gene.com/download/pdf/ocrevus_prescribing.pdf [Reflist]
MonitoringRisk evaluation mitigation strategy due to risk of autoimmune disorder risk and cancer risks
Screen for Tuberculosis and Hepatitis B prior to use
Vaccinate at least 6 weeks prior to therapy
Monitor 1 h after infusion
Infection
Possible breast cancer
Pregnancy test prior to use, it is not recommended to get pregnant or nurse while on this product
POMS PublicationsNone
POMS RCTNone
FDA approval in POMSNone
Abbreviations: IV = intravenous, RCT = randomized controlled trials, CBC = complete blood count, LFT = liver function tests, PML = progressive multifocal leukoencephalopathy.
Table 4. Randomized controlled trial of fingolimod versus interferon in POMS.
Table 4. Randomized controlled trial of fingolimod versus interferon in POMS.
POMS outcomes Age 10–17 [2]Interferon β-1aFingolimod
n108107
Dosing interferon β-1a at a dose of 30 μg per week0.5 mg per day (0.25 mg per day for patients with a body weight of ≤40 kg
Adverse events95.3% serious AE n = 7 (infection, supraventricular tachycardia)88%serious AE n = 18, 16.8% (infection, leukopenia)
Convulsions n = 1, 0.9%Convulsions n = 6, (5.6%)
AE > 10%AE > 10%
Nervous system disorder 42%Leukopenia
Eye disorder
Psychiatric 10.3Gastrointestinal disorders 34%
Headache 29%Infection and infestation 59%
Influenza 11%
Respiratory tract upper 15.9%
Nervous system disorder 43%
Headache 31.8%
Psychiatric 18.7%
Skin disorders 16.8%Skin disorders 18.7%
Completed trial 87.4%88 (81.5%)100 [93.5%]
Primary End Point, Adjusted Annualized Relapse Rate 0.670.12, 82% decrease compared with IFN B-1a, P < 0.0001
MRI New T2 lesion, 53% absolute difference
Secondary End Point Annualized Rate of New or Newly Enlarging T2 Lesions9.274.39 relative difference, 53%; P < 0.001
Reasons for Permanent Discontinuation of MedicationN = 11, lack of treatment effect Elevated liver aminotransferase levels, macular edema, cardiac arrhythmias or electrocardiographic (ECG) abnormalities, and pregnancy
Table 5. Global disease modifying therapy dates of approval.
Table 5. Global disease modifying therapy dates of approval.
DMTFDAEMACanadaAustralia
Interferons AOMS1995, 2002, 2009, 2012, 2013, 2014 (pegylated)199519951996
1997
1998
2008
2014
Interferons POMS Age 12–adult
Glatiramer Acetate AOMS1996, 2014 200219971997
2015
Glatiramer Acetate POMS Age 12–adult
Fingolimod AOMS2010201020112011
Fingolimod POMS2018
Age 10–17
2018
Age 10–17
Siponimod AOMS2019
Siponimod POMSnone
Daclizumab AOMS2016, removed from market 2018 2016xxxx
Teriflunomide AOMS2012201320132016
Teriflunomide POMSnone
Dimethyl fumarate AOMS2013201420132017
Dimethyl fumarate POMSnone
Cladribine AOMS2019201720172010, removed from market 2011
Reapproved 2019
Cladribine POMSnone
Natalizumab AOMS2004, returned to market in 20062006, 2011 SC20062006
Natalizumab POMSnone
Alemtuzumab AOMS2014201320132015
reserved for those with inadequate
responses to 2 or more MS medications
Alemtuzumab POMSnone
Ocrelizumab AOMS2017201820172017
RRMSRRMS
PPMSPPMS
Ocrelizumab POMSnone
Abbreviations: SC—second line, RRMS—relapsing remitting MS, PPMS—primary progressive MS, AOMS—adult onset Multiple Sclerosis, POMS—pediatric onset multiple sclerosis.
Table 6. FDA Black Box warning and contraindications.
Table 6. FDA Black Box warning and contraindications.
NatalizumabProgressive Multifocal Encephalopathy (PML)
Contraindicated if history of PML.
TeriflunomideContraindicated in patients with pre-existing acute or chronic liver disease
contraindicated in women of childbearing potential
Hepatotoxicity and risk of teratogenicity
AlemtuzumabRisks of serious and sometimes fatal autoimmune conditions, infusion reactions, and various malignancies.
It is contraindicated in human immunodeficiency virus due to CD4 + lymphocyte count reductions
DaclizumabDaclizumab is contraindicated in Hepatitis B and C or liver impairment
Removed from US market due to Autoimmune encephalitis
Risk of severe liver injury and immune-mediated disorders, such as skin reactions, lymphadenopathy, and noninfectious colitis
FingolimodFingolimod is contraindicated: if there is a cardiac event or vascular event like a stroke or TIA, irregular heart rate in the past 6 months and or medication that slows the QTc
SiponimodPatients with a CYP2C9*3/*3 genotype (4)
In the last 6 months, experienced myocardial infarction, unstable angina, stroke, TIA, decompensated heart failure requiring hospitalization, or Class III/IV heart failure (4)
Presence of Mobitz type II second-degree, third-degree AV block, or sick sinus syndrome, unless patient has a functioning pacemaker (4)
Table 7. Primary outcome in Randomized Controlled Trials in AOMS.
Table 7. Primary outcome in Randomized Controlled Trials in AOMS.
Reduction in ARRRelative Risk Reduction to PlaceboReduction in Sustained DisabilityReduction in MRI New T2 LesionsReduction in New Gad Enhancing Lesions
Alemtuzumab Not tested vs placebo
Dimethyl fumarate [41] vs. placebo53,48%44%34%, 38%85%90%
Fingolimod [42]See Table 6 for POMS data23%
Natalizumab68% 42%83%
Daclizumab (removed from the market willingly by Biogen)45% compared with IFN β 1a 45–54% 54% new or new enlarging T2
Ocrelizumab [43,44]44, 46% compared with IFN β 1a46–47%Slow disability in PPMS
Teriflunomide [45] 31%29%69% against placeboFree from gad lesions 89% compared with placebo
Cladribine [46]66% Slowed 3-month disability 33% 86%
Table 8. Potential serum biomarkers in MS.
Table 8. Potential serum biomarkers in MS.
ProductPotential Biomarker
InterferonNeutralizing antibody
NatalizumabNatalizumab antibody
JC index
L Selectin
Neurofilament light chain
FingolimodVZV status
Alemtuzumabinterleukin (IL)-21
CD4
CD19
DMFALC
AllNeurofilament light chain
Chitinase3-like 1
Glial fibrillary acidic proteins

Share and Cite

MDPI and ACS Style

Rensel, M. Long-Term Treatment Strategies of Pediatric Multiple Sclerosis, Including the use of Disease Modifying Therapies. Children 2019, 6, 73. https://doi.org/10.3390/children6060073

AMA Style

Rensel M. Long-Term Treatment Strategies of Pediatric Multiple Sclerosis, Including the use of Disease Modifying Therapies. Children. 2019; 6(6):73. https://doi.org/10.3390/children6060073

Chicago/Turabian Style

Rensel, Mary. 2019. "Long-Term Treatment Strategies of Pediatric Multiple Sclerosis, Including the use of Disease Modifying Therapies" Children 6, no. 6: 73. https://doi.org/10.3390/children6060073

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop