Quest for the right Drug

|
עמוד הבית / פירדפסה / מידע מעלון לרופא

פירדפסה FIRDAPSE (AMIFAMPRIDINE)

תרופה במרשם תרופה בסל נרקוטיקה ציטוטוקסיקה

צורת מתן:

פומי : PER OS

צורת מינון:

טבליה : TABLETS

Pharmacological properties : תכונות פרמקולוגיות

Pharmacodynamic Properties

5.1     Pharmacodynamic properties

Pharmacotherapeutic group: other nervous system drugs, ATC code: N07XX05.

Mechanism of action
Amifampridine blocks voltage-dependent potassium channels, thereby prolonging pre-synaptic cell membrane depolarisation. Prolonging the action potential enhances the transport of calcium into the nerve ending. The resulting increase in intra-cellular calcium concentrations facilitates exocytosis of acetylcholine-containing vesicles, which in turn enhances neuromuscular transmission.

It improves muscle strength and resting compound muscle action potential (CMAP) amplitudes with an overall weighted mean difference of 1.69 mV (95% CI 0.60 to 2.77).


Pharmacodynamic effects
The pharmacodynamic profile of amifampridine has been studied for a range of doses. A prospective, placebo-controlled, randomised study in 26 patients with Lambert-Eaton myasthenic syndrome (LEMS) reported clinical efficacy for amifampridine at the standard recommended maximum dose of 60 mg/day (Sanders et al 2000). Two further studies in a total of 57 patients with LEMS have reported data from higher doses of amifampridine. McEvoy et al 1989 reported data from a short-term study in 12 patients with LEMS, which demonstrated that administration of amifampridine at doses up to 100 mg/day for a period of 3 days was effective in treating the autonomic and motor symptoms of LEMS. Sanders et al 1998 presented data on efficacy and safety of amifampridine treatment at doses up to 100 mg/day in 45 patients with LEMS who were treated for an average of 31 months. Therefore, in exceptional circumstances higher doses up to a maximum of 80 mg/day may be of benefit when given with the appropriate safety monitoring. It is recommended that dose titration from 60 mg/day to 80 mg/day is performed in 5 mg increments every 7 days. Upward dose titration should be discontinued if any adverse reaction or ECG abnormality is observed.

The effect of a single dose of 30 mg or 60 mg of amifampridine phosphate was used to evaluate the pharmacokinetic-QTc relationship of amifampridine concentration on cardiac repolarization exposure in healthy volunteers. This evaluation was conducted in a Phase 1, double-blind, randomized, crossover study to define the ECG effects of amifampridine phosphate at these doses compared to placebo and moxifloxacin (a positive control) in healthy men and women who are slow acetylators (n=52). There was no effect of amifampridine phosphate on heart rate, atrioventricular conduction or cardiac depolarization as measured by the heart rate, PR and QRS interval durations. No subjects developed new clinically relevant ECG morphological changes following administration of amifampridine phosphate. No effect was observed of amifampridine phosphate on cardiac repolarization as assessed using the QTc interval.

Clinical efficacy and safety

A double-blind, placebo-controlled, randomized withdrawal Study to evaluate the efficacy and safety of amifampridine phosphate in Patients with LEMS was conduct in adult patients 18 years of age or older (n=26). The patients were maintained on a stable dose and frequency of amifampridine phosphate for at least 7 days prior to randomization. In this four-day study, patients were randomized (1:1) to amifampridine phosphate (at patient’s optimal dose) or placebo on Day 0. Baseline assessments were obtained on day 0. The primary endpoints were change from baseline (CFB) in Patient Global Impression (SGI) and Quantitative Myasthenia Gravis (QMG) score at Day 4. A secondary efficacy endpoint was the change from baseline at Day 4 in CGI-I score, which was determined by treating physicians. Patients were allowed to use stable doses of peripherally acting cholinesterase inhibitors or corticosteroids. Patients with recent use of immunomodulatory therapies (e.g., azathioprine, mycophenolate, cyclosporine), rituximab, intravenous immunoglobulin G, and plasmapheresis were excluded from the study. Patients had a median age of 55.5 years (range: 31 to 75 years) and 62% were female and 38% were male.

Following the 4 day double-blind discontinuation period, patients treated with amifampridine phosphate maintained muscle strength compared to patients treated with placebo who showed worsening of muscle strength. Observed mean difference in QMG Total and SGI change from baseline score between treatments were -6.54 (95% CI: -9.78, -3.29; p=0.0004) and 2.95 (95% CI: 1.53, 4.38; p=0.0003) respectively, both statistically significant in favour of amifampridine phosphate. In addition, CGI-I scores at day 4 as determined by physicians showed significant improvement in patients remaining on amifampridine phosphate compared to placebo (p=0.0020).

Summary of Changes in Primary and Secondary Efficacy Endpoints from Baseline 
Assessment                      Amifampridine (n=13)                  Placebo (n=13) QMG Scoresa
LS Meand                                           0.00                              6.54 

LS Mean Diff (95% CI)                                       -6.54 (-9.78, -3.29) d p-value                                                           0.0004 SGI Scoresb
LS Meand                                          -0.64                              -3.59 LS Mean Diff (95% CI)                                        2.95 (1.53, 4.38) p-valued                                                          0.0003 CGI-I Scoresc
Mean (SD)                                       3.8 (0.80)                         5.5 (1.27) e p-value                                                           0.0020 a
Total QMG score range 0 – 39, 13 items, 0-3 points on each test. The more points = worse symptoms.
b
SGI is a 7-point scale which rates the global impression of the effects of study treatment (1=terrible to 7=delighted).
c      CGI-I is a 7-point scale based on changes in symptoms, behaviour, and functional abilities (1=very much improved to 7=very much worse).
d      CFB for QMG total score was modelled as the response, with fixed effects terms for treatment and QMG at Baseline.
e      p-value based on the Wilcoxon Rank Sum Test for treatment differences.


Pharmacokinetic Properties

5.2   Pharmacokinetic properties
Absorption

Orally administered amifampridine is rapidly absorbed in humans, reaching peak plasma concentrations by 0.6 to 1.3 hours (mean values).

In humans, the rate and extent of absorption of amifampridine is influenced by food (see Table 2).
There was a decrease in C max and AUC, and an increase in the time to reach maximum plasma concentrations when amifampridine phosphate was administered with food as compared to without food. A 2-fold increase in the time to reach C max (T max ) was observed in the presence of food.
Similarly C max and AUC 0-∞ were greater in the fasted state than in the fed state. Overall, food slowed and decreased the absorption of amifampridine with a lowering of exposure by C max on average by ~44% and lowered exposure by AUC ~20%. based on geometric mean ratios (fed-to-fasted).

Apparent plasma terminal elimination half-life differences were 3-4 fold between subjects in the food effect study. Bioavailability is approximately 93-100% based on recoveries of unmetabolised amifampridine and a major 3-N-acetylated amifampridine metabolite in urine.

Table 2: PK Parameters for Amifampridine in Fed and Fasted Subjects Following Administration of a Single Oral Dose of Amifampridine Phosphate

C max                     AUC 0-∞            T max                     t 1/2 Amifampridine
(ng/ml)                   (ng∙hr/ml)         (hr)                      (hr) 20 mg mean (S.D.), range        Mean (S.D.), range mean(S.D.), range         mean (S.D.), range 
Fasted (N=45)     59.1 (34.4), 16-137   117 (76.6), 22.1-271 0.637 (0.247), 0.25-1.5 2.5 (0.73), 1.23-4.31 

Fed* (N=46)       40.6 (31.3), 2.81-132 109 (76.4), 9.66-292 1.31 (0.88), 0.5-4.0    2.28 (0.704), 0.822-3.78 

* Eating a standardised high-fat meal
In a study of healthy volunteers, systemic exposure of amifampridine was notably influenced by the overall metabolic acetylation activity of NAT enzymes and NAT2 genotype. The NAT genes are highly polymorphic and result in phenotypes with variable acetylation activity rates ranging from slow to fast. In the healthy volunteer study, fast acetylators were defined by having a caffeine metabolite ratio >0.3 and slow acetylators with a caffeine metabolite ratio <0.2. There was significantly higher exposure to amifampridine in slow acetylators compared to fast acetylators. Statistically significant differences in amifampridine PK parameters C max , AUC 0-∞ , t 1/2 and apparent clearance was observed between fast and slow acetylators at all dose levels. In this study, slow acetylators experienced more adverse reactions than the fast acetylators. The safety profile in this study is consistent with adverse reactions observed with patients on amifampridine.

Table 3: Mean PK Parameters of Amifampridine in Healthy Subjects after Single Oral Doses (5-30mg) in Slow and Fast Acetylator Phenotypes

Amifampridine          5                          10                       20                       30 Dose (mg)
Subjects (N)      6         6               6             6          6            6           6              6 Acetylator       Fast     Slow             Fast         Slow        Fast        Slow         Fast          Slow Phenotype
Mean Amifampridine PK Parameters
AUC 0-t          2.89     30.1             9.55         66.3        24.7         142         43.5           230 (ng∙hr/ml )
AUC 0-∞          3.57     32.1             11.1         68.9        26.2         146         45.2           234 (ng∙hr/ml)
C max (ng/ml)    3.98     17.9             9.91         34.4        16.2        56.7         25.5          89.6 T max (hr)      0.750    0.830            0.805         1.14        1.04        1.07        0.810          1.29 t 1/2 (hr)      0.603     2.22             1.21         2.60        1.23        2.93         1.65          3.11 The mean caffeine acetylator ratio for these 12 subjects receiving four escalating doses were 0.408 and 0.172 for fast and slow acetylators types respectively.

Distribution

Distribution of amifampridine was studied in the rat. Following oral administration of radiolabeled [14C] amifampridine, radioactive material is rapidly absorbed from the gastrointestinal tract and widely distributed throughout the body. Concentrations in tissues are generally similar to or greater than concentrations in plasma, with the greatest concentration in organs of excretion (liver, kidney and the gastrointestinal tract) and some tissues of glandular function (lacrimal, salivary, mucous, pituitary and thyroid glands).

Biotransformation

In vitro and in vivo studies in humans indicate that amifampridine is metabolised to a single major 3- N-acetylated amifampridine metabolite.

Elimination

In humans, 93.2% to 100% of amifampridine is excreted into the urine within 24 hours after dosing as amifampridine (19%) and its 3-N-acetylated amifampridine metabolite (74.0% to 81.7%). The plasma elimination half-life is approximately 2.5 hours for the amifampridine and 4 hours for the 3-N- acetylated amifampridine metabolite.

The overall clearance of amifampridine is predominantly due to metabolism by N-acetylation and acetylator phenotype has a greater effect on an individual’s metabolism and elimination of amifampridine than does elimination by renal function (See Table 4).

Renal impairment

Exposure of amifampridine was generally higher in subjects with renal impairment than in subjects with normal renal function; however, NAT2 phenotype had a greater effect on an individual’s exposure to amifampridine than renal function status (See Table 4). Amifampridine exposure by AUC 0–∞ was up to 2-fold higher in slow acetylators and up to 3-fold higher in fast acetylators with 

severe renal impairment compared to subjects with normal renal function. Exposure by C max was marginally affected by renal impairment regardless of acetylation status.

In contrast, the 3-N-acetyl metabolite exposure levels were affected to a greater extent by renal impairment than those for amifampridine. The 3-N-acetyl metabolite exposure by AUC 0–∞ was up to 6.8-fold higher in slow acetylators and up to 4-fold higher in fast acetylators with severe renal impairment compared to subjects with normal renal function. Exposure by C max was only marginally affected by renal impairment regardless of acetylation status. Although the metabolite is inactive at potassium channels, potential off target effects due to accumulation are unknown.


Table 4: Mean PK Parameters of Amifampridine in Normal and Renal Impaired Subjects after Single Oral Dose Administration (10mg) in Slow and Fast Acetylator Phenotypes 
Renal Status              Normal                            Mild                              Moderate                    Severe 
Subjects (N)         4             4           4                     4                   4                 4         4               4 
NAT2 Phenotype            Fast       Slow           Fast                 Slow                Fast              Slow      Fast             Slow 
Mean Amifampridine PK Parameters

AUC 0-∞
(ng·h/ml)                10.7            59.1         16.1                81.3               14.3               126      32.8             119 
C max (ng/ml)            7.65            38.6         11.1                33.5               8.33               52.5     9.48             44.1 
T max (hr)               0.44            0.43         0.88                0.88               0.51               0.55     0.56             0.63  t 1/2 (hr)               1.63            2.71         1.86               2.95              1.72                 3.89     1.64             3.17 Mean 3-N-acetyl Amifampridine PK Parameters

AUC 0 -∞
(ng·h/ml)                 872                594           1264             1307             2724              1451     3525             4014 
C max (ng/ml)             170                115           208                 118           180                144     164              178 
T max (hr)                1.13           0.75            1.44              1.38             2.00               1.13    1.63       2.81  t   1/2   (hr)            4.32           4.08            5.35              7.71             13.61              6.99    18.22      15.7 


Hepatic impairment

There are no data on the pharmacokinetics of amifampridine in patients with hepatic impairment (see sections 4.2 and 4.4).

Paediatric population

There are no data on the pharmacokinetics of amifampridine in paediatric patients (see sections 4.2).
The effect of age on the pharmacokinetics of amifampridine has not been studied.

פרטי מסגרת הכללה בסל

התרופה תינתן לטיפול בתסמונת LEMS (Lambert Eaton myasthenic syndrome).

מסגרת הכללה בסל

התוויות הכלולות במסגרת הסל

התוויה תאריך הכללה תחום קליני Class Effect מצב מחלה
התרופה תינתן לטיפול בתסמונת LEMS (Lambert Eaton myasthenic syndrome).
שימוש לפי פנקס קופ''ח כללית 1994 לא צוין
תאריך הכללה מקורי בסל 23/01/2011
הגבלות תרופה מוגבלת לרישום ע'י רופא מומחה או הגבלה אחרת

בעל רישום

MEDISON PHARMA LTD

רישום

146 29 33301 01

מחיר

0 ₪

מידע נוסף

עלון מידע לרופא

08.03.22 - עלון לרופא

עלון מידע לצרכן

31.07.17 - עלון לצרכן 08.03.22 - עלון לצרכן עברית 26.05.20 - החמרה לעלון

לתרופה במאגר משרד הבריאות

פירדפסה

קישורים נוספים

RxList WebMD Drugs.com