Skip to main content
Neurotherapeutics logoLink to Neurotherapeutics
. 2012 Nov 27;10(1):106–110. doi: 10.1007/s13311-012-0156-3

Enhancing Neural Transmission in Multiple Sclerosis (4-Aminopyridine Therapy)

Andrew D Goodman 1,, Robert Thompson Stone 1,3
PMCID: PMC3557354  PMID: 23184313

Abstract

Enhancing neural transmission by improving axonal conduction and synaptic neurotransmitter release is a novel strategy to improve symptoms in multiple sclerosis. Dalfampridine (4-aminopyridine extended-release) is a first-in-class medication that targets the damaged nervous system through blockage of voltage-gated potassium channels. Through a series of clinical trials, dalfampridine (dosed at 10 mg twice daily) has been found to improve walking speed by approximately 25 % on average in one third of individuals with multiple sclerosis regardless of disease stage. Furthermore, it significantly improves patients' perception of their ambulatory disability and may improve lower extremity strength. Given the mechanism of action, the most serious adverse effect is its pro-convulsant property, which occurs more frequently at high serum concentrations. The most common adverse events include increased falls, urinary tract infections, dizziness, insomnia, and headaches. Despite these potential side-effects, the vast majority of individuals who derive benefit continue on the treatment. The exact mechanism of action is uncertain, as is the reason for response variability. The medication serves as proof-of-concept that targeting axonal transmission can improve disability in multiple sclerosis.

Electronic supplementary material

The online version of this article (doi:10.1007/s13311-012-0156-3) contains supplementary material, which is available to authorized users.

Keywords: Multiple sclerosis, Gait disorder, Potassium channel, Neural transmission, Dalfampridine

Introduction

Multiple sclerosis (MS) is the most common disease of the central nervous system in young adults leading to sustained disability. A wide variety of deficits contribute to disability, including visual disturbances, weakness, ataxia, and gait dysfunction, fatigue, bladder and bowel difficulties, and cognitive dysfunction. The disease involves immune-mediated destruction of the central nervous system myelin, as well as a variable amount of direct axonal injury. Although the underlying mechanism of disability progression remains uncertain, inflammatory demyelination is likely a significant contributor, as it leads to slowed conduction velocity along affected axons. Current disease-modifying therapies target the heightened immune response, but do not directly target the damaged nervous system.

Dalfampridine (4-aminopyridine extended-release; termed fampridine sustained-release in pre-2010 literature and outside the United States) is a novel therapy that directly targets demyelinated axons presumably by improving conduction and facilitating synaptic transmission. 4-aminopyridine (4-AP) is a broad-spectrum antagonist of voltage-gated potassium (Kv) channels [1, 2]. The compound has been previously studied for spinal cord injury, neuromuscular junction disorders, and Alzheimer’s disease [3, 4]. A related compound 3,4 diaminopyridine has also been studied in MS, as well as neuromuscular disease, most notably Lambert-Eaton syndrome [5].

Primarily based on 2 phase 3 randomized, placebo-controlled clinical trials, dalfampridine has been shown to improve walking speed on the timed 25-foot walk (T25FW) test by approximately 25 % on average in one third of subjects [6, 7] Gait dysfunction is 1 of the leading causes of physical disability among patients with MS. Recently diagnosed patients with relapsing-remitting MS can have reduced walking speed and stride length, as well as balance impairment, suggesting that the process leading to gait dysfunction frequently begins early in the disease [8]. Overall, approximately 75 % of MS patients struggle with mobility problems leading to loss of independence, physical appearance of disability, and unemployment [9]. As such, gait dysfunction contributes significantly to reduced quality of life [10, 11]. Thus, improving mobility through enhanced neural transmission, in addition to targeting inflammatory demyelination, can lead to enhancement of quality of life for MS patients.

Mechanism of Action

Demyelination of central axons leads to exposure of the paranodal and juxtaparanodal regions (areas immediately adjacent to the nodes of Ranvier). These axonal regions are rich in Kv channels. It is theorized that exposure of these channels in demyelinated axons contributes significantly to dysfunction of action potential propagation as it causes the cell to move closer to voltage equilibrium for potassium [1]. In addition, exposure of slow Kv channels impairs axonal generation of repetitive impulses by interfering with hyperpolarization [12]. 4-AP also increases pre-synaptic transmission by increasing end-terminal influx of calcium [13]. This may be due to the indirect effect of Kv blockage on calcium influx or direct effects of 4-AP on voltage-gated calcium channels, thereby increasing cytoplasmic free calcium [1416].

Remyelination of damaged axons depends on proliferation of oligodendrocytes. The oligodendrocytes originate from oligodentrocyte precursor cells (OPC), and thus OPC proliferation is likely to be important in recovery from relapses. There is evidence that certain Kv channel subtypes play a role in OPC proliferation, and thus blockage could hypothetically help facilitate remyelination [17, 18]. 4-AP could also have an immunomodulatory role as immune cells (e.g., T and B cells, microglia, and macrophages) also express Kv channels. Furthermore, the number of Kv channels is significantly upregulated when these cells are activated and seem to play a role in activation and proliferation [14, 19, 20].

Pharmacology and Pharmacokinetics

The Kv channel has 4 alpha subunits, each containing 6 membrane-spanning, alpha-helical regions. The subunits form pores that are either homotetramers or heterotetramers [1]. 4-AP is a broad-spectrum Kv channel-blocking agent that has the greatest affinity for slowly inactivating channels. It is part of a family of monoamino and diamino pyridine derivatives, where there is an amino substitution in the fourth position of the pyridine ring [1, 12]. 4-AP is a small, lipid soluble, and readily crosses the blood brain barrier. 4-AP binds to the Kv channel from the cytoplasmic side at the opening of the pore, thus occluding it [1].

Early studies of 4-AP showed an increase in action potential duration and amplitude when measured on experimentally demyelinated mammalian peripheral nerve fibers [21, 22]. Small human clinical trials using intravenous and orally administered immediate-release formulations of 4-AP were subsequently completed in MS and spinal cord injury [2328]. Pharmacologic trials have shown that immediate-release 4-AP has a mean time to maximum concentration of 1 h and a mean half-life of 3.5 h. The maximum observed plasma concentration for the 10 mg and 25 mg doses were 46.4 and >100 ng/mL, respectively. The peak serum concentration was clearly dose related and followed a linear distribution [2]. The medication is primarily excreted renally with a total urinary excretion of 99 % after 24 h. It has negligible protein binding and limited conversion to metabolites [29, 30].

Immediate-release formulations of 4-AP were found to have a narrow therapeutic window, with higher concentrations acting as a proconvulsant [31]. This finding, along with the need for frequent dosing throughout the day, motivated creation of an extended-release formulation (initially entitled sustained-release). This extended-release formulation has a mean time to maximum concentration of 3.2 to 5 h and a mean half-life of 5.2 to 6.4 h. The maximum observed plasma concentration for the 10 mg dose was 22 ng/mL (50 % lower than the immediate release formulation) [29, 30]. Thus, the sustained-release formulation had lower peak concentrations, and could be dosed less frequently. It is important to note, that in the setting of moderate-to-severe renal impairment (i.e., creatinine clearance, <40 mL/min), peak plasma concentration was 1.5 to 2 times higher with time to maximum concentration of 6 to 8 h and a half-life of 8 to 14 h [29].

Clinical Trials

For the last 30 years, small safety and efficacy trials of 4-AP were completed in MS patients. Initial studies showed improvement in visual performance, motor function, coordination, gait, and oculomotor function. No serious adverse effects were identified in these early studies [32, 33]. In 1994, Bever et al. [34] published a randomized, placebo-controlled trial testing the immediate-release formulation of 4-AP (fampridine). The trial documented an improvement in visual contrast-sensitivity and strength, but did not find a significant difference in Expanded Disability Status Scale scores. Serum concentrations >60 ng/mL were associated with a higher incidence of adverse events. One subject had a generalized tonic-clonic seizure at a concentration of 104 ng/mL, and another had an acute confusional episode at 114 ng/mL.

Schwid et al. [35] published a small, double blind, placebo-controlled crossover study of dalfampridine (4-AP sustained release) in 1997 that helped identify walking speed as an outcome of interest. They used a dose of 17.5 mg twice daily for 1 week. The study found improvement in timed gait and nonsignificant improvement in timed stair climbing, grip strength, or Expanded Disability Status Scale. Importantly, no serious adverse events were documented. In 2007 and 2008, Goodman et al. [36, 37] published 2 randomized, double-blind, placebo-controlled dose trials of dalfampridine; the first trial assessed the safety and efficacy of dose escalation, and the second trial compared parallel groups of 3 doses. The latter trial found an increase in baseline lower extremity strength in the 3-drug treatment groups, but found that serious adverse events were more common in the higher-dosed groups. In the initial study, 2 subjects had convulsions at 30 to 35 mg twice daily dosing. In the latter study, 2 subjects experienced seizures, and 1 had encephalopathy in the 20 mg group.

These preliminary studies informed the design of the first phase 3, randomized, controlled, double-blind clinical trial [6]. This study evaluated dalfampridine (10 mg twice daily dosing) for 14 weeks versus a placebo in 301 subjects. The mean peak dalfampridine concentration was 27.6 ng/mL. In the dalfampridine group, there were 35 % of subjects versus 8 % in the placebo group who were deemed “timed walk responders.” Responders were defined as patients who had a faster walking speed on the T25FW for at least 3 of 4 visits during the double-blind treatment period compared with their maximum speed for any of the 5 off-drug visits. Mean improvement of walking speed in responders was 25 % versus 5 % in the placebo group. This improvement was maintained during the 14-week treatment period. This study also used the 12-item MS walking scale, which evaluates a subject’s perception of their ambulatory disability (with a lower score correlating with a lower perceived disability). The change in score had significantly improved in the responder group (-6.84 vs 0.05 in the nonresponders). This was an important observation as it supported the conclusion that the improvement in walking speed to be not only statistically, but also clinically, relevant [6]. A recent report evaluated what change in the T25FW would statistically be considered “minimally important,” and found the estimate to be around 20 % [38]. The trial also assessed clinical global impression scores as well as lower extremity strength, which were significantly improved in responders [6].

Goodman et al. [7] published the results of the second phase 3 trial in 2010. The study evaluated dalfampridine 10 mg twice daily dosing for 9 weeks versus placebo in 239 subjects. The mean peak dalfampridine concentration was 28.5-30.2 ng/mL. 43 % of subjects in the dalfampridine group were found to be treatment responders (vs 9 % in the placebo group). Improvement in walking speed in responders was 25 % in the treatment group versus 8 % in the placebo group. Again, the responder group showed significant improvement in the 12-item MS walking scale score (-6.04 vs 0.85), and improvement in a measure of lower extremity strength [7]. Table 1 summarizes the efficacy data from the 2 phase 3 trials.

Table 1.

Summary of Efficacy Measures in 2 Pivotal, Phase III Clinical Trials

Efficacy measure Goodman, et al. (2009) [6] Goodman, et al. (2010) [7]
T25FW responders
 (vs placebo) 35 % (8 %) 42.9 % (9.3 %)
 Average change in walking speed in responders (vs placebo) 25.2 % (4.7 %) 24.7 % (7.7 %)
 Average change in MSWS-12 score (vs placebo) -6.84 (0.05) -6.04 (0.85)
 Average change in LEMMT score (vs placebo) 0.18 (0.04) 0.145 (0.042)

LEMMT = Lower Extremity Manual Muscle Test [6, 7]; MSWS-12 = 12-Item Multiple Sclerosis Walking Scale; T25FW test = timed 25-foot walk test

All differences shown were significant with p value < 0.05

Safety

The most severe adverse events identified by trials have been seizures and acute encephalopathy. These seem to occur with high serum concentrations of the medication (>100 ng/mL), and were more common with the immediate-release formulation [4]. The theorized mechanism for its proconvulsant effect is increased neuronal excitability from potassium channel blockade. When estimating the increased seizure risk associated with the medication, 1 must account for the overall increased incidence of seizures in the MS population (2 % vs 1 % in the general population) [39]. In an initial study by Bever et al. [34] on immediate-release 4-AP, 1 patient had a generalized-tonic clonic seizure with a serum concentration of 104 ng/mL [4]. In the dalfampridine dose ranging trials by Goodman [36], 2 patients had seizures in each study on doses ranging from 20 to 35 mg [36, 37]. In the phase 3 pivotal trials, only 1 seizure occurred in the initial study, but it was associated with sepsis secondary to community-acquired pneumonia [6]. There have been reports of toxicity related to pharmacies independently compounding an immediate-release formulation of the medication. A concern is that these preparations may not always meet uniformity requirements [31]. Overall, it seems that when the extended-release preparation is used at the appropriate dosage (10 mg twice daily), the incidence of seizures is no higher than that of the MS population as a whole. However, given the risk for seizures at higher serum concentrations, the medication should be avoided in those with a history of seizures or renal dysfunction.

Other serious adverse events occurring on treatment (although not clearly related to the medication) have been severe anxiety, sepsis (secondary to community-acquired pneumonia), cellulitis, pyelonephritis, patellar fracture, coronary artery disease, and cholelithiasis. The most common adverse effects are summarized in Table 2, and these include increased frequency of falls, urinary tract infections, insomnia, dizziness, and headache. Less frequently encountered adverse effects have included fatigue, nausea, upper respiratory infections, asthenia, back pain, balance dysfunction, arthralgia, and paresthesias [6, 7]. There is no indication that the medication increases the incidence of MS relapses. There is no indication of cardiotoxicity at standard doses, although there are reports of cardiac arrhythmias in patients taking unregulated formulations [31]. Data from open-label extension studies show no new long-term safety concerns and 73 % continued usage as of 2008 [40].

Table 2.

Most Frequent Adverse Events Recorded in 2 Pivotal, Phase III Clinical Trials*

Most Frequent AEs Goodman, et al. (2009) [6] Goodman, et al. (2010) [7]
Urinary tract infection 14 % 17.5 %
Fall 16 % 11.7 %
Insomnia 8 % 10 %
Headache 6 % 9.2 %
Dizziness 8 % 8.3 %
Nausea 6 % 8.3 %
Asthenia 6 % 8.3 %
Upper respiratory infection 6 % 5.8 %
Back pain 6 % 5.8 %
Balance disorder 6 % 5.8 %

*[6, 7]

As previously described, the medication is excreted via the kidneys, and thus patients with moderate-to-severe renal insufficiency have 1.5 to 2 times higher peak serum concentrations [29]. Caution should be used in patients with mild renal impairment (creatinine clearance, 51-80 mL/min), but no specific dose adjustment is recommended. The medication should be avoided in patients with moderate-to-severe renal impairment (creatinine clearance, <51 mL/min).

Conclusion and Future Directions

Dalfampridine is a unique treatment that is believed to work by improving axonal conduction and facilitating synaptic transmission. It significantly increases walking speed in approximately one third of individuals, which can help improve independence, employment opportunity, and ultimately quality of life. Seizure is the adverse event of most concern, but when used at standard dosing, the incidence is not likely higher than that of the general MS population.

It is unclear why some patients (30-40 %) are treatment responders and others are not. It has been proposed that lesion location may influence response to treatment. Responders may have demyelinated axons more relevant to gait, and thus they are more likely to have benefit from treatment. However, the subjects included in each pivotal trial had T25FW for >8 seconds, and so it is likely that the majority of patients had demyelinating lesions relevant to ambulation. The discrepancy could involve individual polymorphisms of voltage-gated potassium channels rendering specific channels more or less susceptible to the medication. Future studies to evaluate the cause of the discrepancy may allow identification of potential responders.

Dalfampridine serves as proof-of-concept that targeting the damaged nervous system can improve symptoms. Alternate mechanisms for enhancing axonal conduction should be sought. If treatments are developed with such varied mechanisms, significant symptomatic improvement could potentially be achieved through combination therapies.

Electronic supplementary material

ESM 1 (510.7KB, pdf)

(PDF 510 kb)

Acknowledgments

Required Author Forms

Disclosure forms provided by the authors are available with the online version of this article.

References

  • 1.Judge SI. Potassium channel blockers in multiple sclerosis: neuronal Kv channels and effects of symptomatic treatment. Pharmacol Ther 2006;111:224–259 [DOI] [PubMed]
  • 2.Hayes KC, Katz MA, Devane JG, et al. Pharmacokinetics of an immediate-release oral formulation of fampridine (4-aminopyridine) in normal subjects and patients with spinal cord injury. J Clin Pharmacol. 2003;43:379–385. doi: 10.1177/0091270003251388. [DOI] [PubMed] [Google Scholar]
  • 3.Murray NM, Newsom-Davis J. Treatment with oral 4-aminopyridine in disorders of neuromuscular transmission. Neurology. 1981;31:265–271. doi: 10.1212/WNL.31.3.265. [DOI] [PubMed] [Google Scholar]
  • 4.Wesseling H, Agoston S, VanDam GB, et al. Effects of 4-aminopyridine in elderly patients with Alzheimer’s disease. N Engl J Med. 1984;210:988–989. doi: 10.1056/NEJM198404123101514. [DOI] [PubMed] [Google Scholar]
  • 5.Bever CT, Anderson PA, Leslie J, et al. Treatment with oral 3,4 diaminopyridine improves leg strength in multiple sclerosis patients. Neurology. 1996;47:1457–1462. doi: 10.1212/WNL.47.6.1457. [DOI] [PubMed] [Google Scholar]
  • 6.Goodman AD, Brown TR, Krupp LB, et al. Sustained-release oral fampridine in multiple sclerosis: a randomized, double-blind, controlled trial. Lancet. 2009;373:732–738. doi: 10.1016/S0140-6736(09)60442-6. [DOI] [PubMed] [Google Scholar]
  • 7.Goodman AD, Brown TR, Edwards KR, et al. A phase 3 trial of extended release oral dalfampridine in multiple sclerosis. Ann Neurol. 2010;68:494–502. doi: 10.1002/ana.22240. [DOI] [PubMed] [Google Scholar]
  • 8.Martin CL, Phillips BA, Kilpatrick TJ, et al. Gait and balance impairment in early multiple sclerosis in the absence of clinical disability. Mult Scler. 2006;12:620–628. doi: 10.1177/1352458506070658. [DOI] [PubMed] [Google Scholar]
  • 9.Hobart JC, Riazi A, Lamping DI, et al. Measuring the impact of MS on walking ability: the 12-Item MS Walking Scale (MSWS-12) Neurology. 2003;60:31–36. doi: 10.1212/WNL.60.1.31. [DOI] [PubMed] [Google Scholar]
  • 10.Sutliff MH. Contribution of impaired mobility to patient burden in multiple sclerosis. Curr Med Res Opin. 2010;26:109–119. doi: 10.1185/03007990903433528. [DOI] [PubMed] [Google Scholar]
  • 11.Zwibel HL. Contribution of impaired mobility and general symptoms to the burden of multiple sclerosis. Adv Ther. 2009;26:1043–1057. doi: 10.1007/s12325-009-0082-x. [DOI] [PubMed] [Google Scholar]
  • 12.Korenke AR, Rivey MP, Allington DR. Sustained-release fampridine for symptomatic treatment of multiple sclerosis. Ann Pharmacother. 2008;42:1458–1465. doi: 10.1345/aph.1L028. [DOI] [PubMed] [Google Scholar]
  • 13.Smith KJ, Felts PA, John GR. Effects of 4-aminopyridine on demyelinated axons, synapses and muscle tension. Brain. 2000;123:171–184. doi: 10.1093/brain/123.1.171. [DOI] [PubMed] [Google Scholar]
  • 14.Espejo C, Montalban X. Dalfampridine in multiple sclerosis: from symptomatic treatment to immunomodulation. Clin Immunol. 2012;142:84–92. doi: 10.1016/j.clim.2011.06.004. [DOI] [PubMed] [Google Scholar]
  • 15.Tibbs GR, Barrie AP, Van Mieghem FJ, Mcmahon HT, Nicholls DG. Repetitive action potentials in isolated nerve terminals in the presence of 4-aminopyridine: effects on cytosolic free Ca2+ and glutamate release. J Neurochem. 1989;53:1693–1699. doi: 10.1111/j.1471-4159.1989.tb09232.x. [DOI] [PubMed] [Google Scholar]
  • 16.Wu ZZ, Li DP, Chen SR, Pan HL. Aminopyridines potentiate synaptic and neuromuscular transmission by targeting the voltage-activated calcium channel beta subunit. J Biol Chem. 2009;284:36453–36461. doi: 10.1074/jbc.M109.075523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Chittajallu R, Chen Y, Wang X, et al. Regulation of Kv1 subunit expression in oligodendrocyte progenitor cells and their role in G1/S phase progression of the cell cycle. Proc Natl Acad Sci U S A. 2002;99:2350–2355. doi: 10.1073/pnas.042698399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Vautier F, Belachew S, Chittajallu R, Gallo V. Shaker-type potassium channel subunits differentially control oligodendrocyte progenitor proliferation. Glia. 2004;48:337–345. doi: 10.1002/glia.20088. [DOI] [PubMed] [Google Scholar]
  • 19.Decoursey TE, Chandy KG, Gupta S, Cahalan MD. Mitogen induction of ion channels in murine T lymphocytes. J Gen Physiol. 1987;89:405–420. doi: 10.1085/jgp.89.3.405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Chandy KG, Decoursey TE, Chahalan MD, Mclaughlin C, Gulta S. Voltage-gated potassium channels are required for human T lymphocyte activation. J Exp Med. 1984;160:369–385. doi: 10.1084/jem.160.2.369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Sherratt RM, Bostock H, Sears TA. Effects of 4-aminopyridine on normal and demyelinated mammalian nerve fibres. Nature. 1980;283:570–572. doi: 10.1038/283570a0. [DOI] [PubMed] [Google Scholar]
  • 22.Bowe CM, Kocsis JD, Targ EF, Waxman SG. Physiological effects of 4-aminopyridine on demyelinated mammalian motor and sensory fibers. Ann Neurol. 1987;22:264–268. doi: 10.1002/ana.410220212. [DOI] [PubMed] [Google Scholar]
  • 23.Stefoski D, Davis FA, Fitzsimmons WE, Luskin SS, Rush J, Parkhurst GW. 4-Aminopyridine in multiple sclerosis: prolonged administration. Neurology. 1991;41:1344–1348. doi: 10.1212/WNL.41.9.1344. [DOI] [PubMed] [Google Scholar]
  • 24.Van Diemen HA, Polman CH, van Dongen TM, et al. The effect of 4-aminopyridine on clinical signs in multiple sclerosis: a randomized, placebo-controlled, double-blind, cross-over study. Ann Neurol. 1992;32:123–130. doi: 10.1002/ana.410320203. [DOI] [PubMed] [Google Scholar]
  • 25.Polman CH, Bertelsmann FW, van Loenen AC, Koetsier JC. 4-aminopyridine in the treatment of patients with multiple sclerosis. Long term efficacy and safety. Arch Neurol. 1994;51:292–296. doi: 10.1001/archneur.1994.00540150090022. [DOI] [PubMed] [Google Scholar]
  • 26.Hayes KC, Blight AR, Potter PJ, et al. Preclinical trial of 4-aminopyridine in patients with chronic spinal cord injury. Paraplegia. 1993;38:7–15. doi: 10.1038/sc.1993.40. [DOI] [PubMed] [Google Scholar]
  • 27.Donovan WH, Halter JA, Graves DE, et al. Intravenous infusion of 4-AP in chronic spinal cord injured subjects. Spinal Cord 2000;38728-732. [DOI] [PubMed]
  • 28.Hansebout RR, Blight AR, Fawcett S, Reddy K. 4-Aminopyridine in chronic spinal cord injury: A controlled, double-blind, crossover study in eight patients. J Neurotrauma. 1993;10:1–18. doi: 10.1089/neu.1993.10.1. [DOI] [PubMed] [Google Scholar]
  • 29.Smith W, Swan S, Marbury T, Henney H. III Single-dose pharmacokinetics of sustained-release fampridine (fampridine-sr) in healthy volunteers and adults with renal impairment. J Clin Pharmacol. 2010;50:151–159. doi: 10.1177/0091270009344857. [DOI] [PubMed] [Google Scholar]
  • 30.Bever CT, Jr, Young D, Tierney D, et al. The pharmacokinetics and tolerability of a slow-release formulation of 4-aminopyridine in multiple sclerosis patients (abstract) Neurology. 1995;45(suppl 4):A351. [Google Scholar]
  • 31.Cornblath DR, Bienen EJ, Blight A. The safety profile of dalfampridine extended release in multiple sclerosis clinical trials. Clin Ther. 2012;34:1056–1069. doi: 10.1016/j.clinthera.2012.03.007. [DOI] [PubMed] [Google Scholar]
  • 32.Jones RE, Heron JR, Foster DH. Effects of 4-aminopyridine in patients with multiple sclerosis. J Neurol Sci. 1983;60:353–362. doi: 10.1016/0022-510X(83)90145-4. [DOI] [PubMed] [Google Scholar]
  • 33.Davis FA, Stefoski D, Rush J. Orally administered 4-aminopyridine improves clinical signs in multiple sclerosis. Ann Neurol. 1990;27:186–192. doi: 10.1002/ana.410270215. [DOI] [PubMed] [Google Scholar]
  • 34.Bever CT, Young D, Anderson A, et al. The effects of 4-aminopyridine in multiple sclerosis patients: results of a randomized, placebo-controlled, double-blind, concentration-controlled, crossover trial. Neurology. 1994;44:1054. doi: 10.1212/WNL.44.6.1054. [DOI] [PubMed] [Google Scholar]
  • 35.Schwid SR, Petrie MD, McDermott MP, et al. Quantitative assessment of sustained-release 4-aminopyridine for symptomatic treatment of multiple sclerosis. Neurology. 1997;48:817–821. doi: 10.1212/WNL.48.4.817. [DOI] [PubMed] [Google Scholar]
  • 36.Goodman AD, Cohen JA, Cross A, et al. Fampridine-SR in multiple sclerosis: a randomized, double-blind, placebo-controlled, dose-ranging study. Mult Scl. 2007;13:357–368. doi: 10.1177/1352458506069538. [DOI] [PubMed] [Google Scholar]
  • 37.Goodman AD, Brown TR, Cohen JA, et al. Dose comparison trial of sustained-release fampridine in multiple sclerosis. Neurology. 2008;71:1134–1141. doi: 10.1212/01.wnl.0000326213.89576.0e. [DOI] [PubMed] [Google Scholar]
  • 38.Coleman CI, Sobieraj DM, Marinucci LN. Minimally important clinical difference of the Times 25-Foot Walk Test: results from a randomized controlled trial in patients with multiple sclerosis. Curr Med Res Opin. 2012;28:49–56. doi: 10.1185/03007995.2011.639752. [DOI] [PubMed] [Google Scholar]
  • 39.Sokic DV, Stojsavljevic N, Drulovic J, et al. Seizures in multiple sclerosis. Epilepsia. 2001;42:72–79. doi: 10.1046/j.1528-1157.2001.48699.x. [DOI] [PubMed] [Google Scholar]
  • 40.Goodman AD. Interim analysis of open-label extension studies of dalfampridine extended release tablets in patients with multiple sclerosis. Neurology. 2010;74(suppl 2):A101. [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

ESM 1 (510.7KB, pdf)

(PDF 510 kb)


Articles from Neurotherapeutics are provided here courtesy of Elsevier

RESOURCES