U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Clinical Review Report: abobotulinumtoxinA (Dysport Therapeutic): (Ipsen Biopharmaceuticals Canada Inc.): Indication: For the symptomatic treatment of lower-limb spasticity in pediatric patients 2 years of age and older [Internet]. Ottawa (ON): Canadian Agency for Drugs and Technologies in Health; 2018 Aug.

Cover of Clinical Review Report: abobotulinumtoxinA (Dysport Therapeutic)

Clinical Review Report: abobotulinumtoxinA (Dysport Therapeutic): (Ipsen Biopharmaceuticals Canada Inc.): Indication: For the symptomatic treatment of lower-limb spasticity in pediatric patients 2 years of age and older [Internet].

Show details

Results

Findings From the Literature

A total of two studies were identified from the literature for inclusion in the systematic review (Figure 1). The included studies are summarized in Table 4 and described in the Included Studies Section. A list of excluded studies is presented in Appendix 3.

Figure 1. Flow Diagram for Inclusion and Exclusion of Studies.

Figure 1

Flow Diagram for Inclusion and Exclusion of Studies.

Table 4. Details of Included Studies.

Table 4

Details of Included Studies.

Included Studies

Description of Studies

Two trials, Study 141 and Study 701, met the inclusion criteria for this review. Study 141 was a pivotal trial.

Study 141 (N = 241) was a phase III, multi-centre, double-blind, prospective, randomized, placebo-controlled, single treatment cycle study that assessed the efficacy of aboBoNTA compared with placebo in children with dynamic equinus foot deformity associated with CP. Study 141 consisted of a screening period (day −7 to day 1), and patients received treatment on day 1 and were followed up for a minimum of 12 weeks and a maximum of 28 weeks (double-blind treatment period). All patients who had had at least 12 weeks of follow-up were considered to have completed the study. Patients were randomized into one of three treatment groups; aboBoNTA 10 U/kg, aboBoNTA 15 U/kg, or placebo, in a ratio of 1:1:1, and stratified according to age range (2 to 9 years and 10 to 17 years) and botulinum toxin (BoNT)–naive or non-naive status, as assessed at baseline. After randomization, aboBoNTA or placebo was administered by intramuscular injections into the gastrocnemius-soleus complex (GSC) of each affected lower limb. The dose of aboBoNTA administered was either 10 U/kg or 15 U/kg per affected GSC, so the total dose was 10 U/kg or 15 U/kg for unilateral injections and 20 U/kg or 30 U/kg for bilateral injections. Patients who required re-treatment at week 12, 16, 22, or 28 were offered entry into an open-label extension study (Study 147).

Study 701 (N = 52) was a phase III, multi-centre, double-blind, prospective, randomized placebo-controlled study that compared the efficacy and safety of a single administration of aboBoNTA or placebo in the treatment of pediatric dynamic equinus spasticity associated with CP. Following initial assessment of their LLS, eligible patients were randomized to receive a single treatment of either aboBoNTA (30 U/kg) or placebo. Study medication was distributed equally between both legs by injection into each of the gastrocnemius muscles of each limb. Each muscle was injected at two sites. The effects of the treatment were monitored over a minimum 16-week period. Post-treatment assessments were made at weeks 4, 8, and 16. If an investigator believed that a treatment effect was maintained at week 16, additional visits at weeks 24 and 36 were scheduled.

Populations

Inclusion and Exclusion Criteria

Study 141 included ambulatory male or female patients with CP between two and 17 years of age who had spastic lower limbs characterized by an equinus foot positioning during the stance phase of the gait and the ability to walk with or without walking aids. These patients were BoNT-naive or had received their last BoNT treatment of any type more than six months prior to study entry. Additionally, patients had a spasticity grade of between 2 and 4, inclusive, on the Tardieu Scale (TS), assessed at the ankle joint of the (most) affected lower limb to be injected with a spasticity angle of 10 degrees or more, and classified as Gross Motor Function Classification System (GMFCS) level I to III inclusive (where level I indicates that patients can walk without limitations; level II indicates that patients can walk with limitations; level III indicates that patients can walk using a hand-held mobility device; level IV indicates that patients have self-mobility with limitations and may use powered mobility; and level V refers to patients who are transported in a manual wheelchair). All patients had signed informed consent obtained from the child’s parent or guardian and signed consent from the child, when and where applicable. Patients were excluded if they had a current need for surgery or had previous surgery for spasticity of the GSC and/or hamstring muscles (and tendons) in the most affected leg to be injected. Furthermore, patients were excluded if there was evidence of non-ambulatory status, major limitation in the passive range of motion at the ankle, significant difference (> 2 cm) between the length of legs, serial casting in the past 12 weeks, previous injection of alcohol and/or phenol into the GSC and/or hamstrings in the most affected leg to be injected, severe athetoid or dystonic movements in the targeted lower limb(s), or known resistant or sensitivity to BoNT. Also excluded were patients who had received treatment with any drug that interferes either directly or indirectly with neuromuscular function or neuroblocking drugs used during surgery within the last 30 days prior to study treatment.

In Study 701, eligible patients aged between two and seven years were ambulatory, had a diagnosis of diplegic CP, had no evidence of fixed contracture (able to achieve 10 degrees passive ankle dorsiflexion in both legs), and had the potential to benefit from the injection of aboBoNTA to the gastrocnemius muscle. Patients were excluded if they had previously had surgery on the affected limbs or if there was a need for surgery within the next six months. Furthermore, patients were excluded if multi-level injections were required, if the patient had a significant foot deformity, if they had BoNT treatment within the previous nine months, had had previous phenol treatment for LLS, or had known hypersensitivity to BoNT. Also excluded were patients who had received an investigational new drug in the 30 days prior to entry, patients who were receiving aminoglycoside antibiotics or spectinomycin, patients with a generalized disorder of muscle activity (e.g., myasthenia gravis), and patients unwilling or unable to comply with the protocol. The parent/guardian of each patient gave written informed consent.

Baseline Characteristics

Patients enrolled in Study 141 were between two and 17 years of age (mean age 5.9 years). There were more male patients than female patients (60% versus 40%, respectively). The majority of patients were Caucasian (73.2%). More patients were treated in one leg than in both legs (59% versus 41%, respectively). Over half the patients (55.7%) were classified as GMFCS level I, and a third of the patients (33.2%) as level II. Approximately half of the patients (51.9%) were naive to treatment with any form of BoNT before entering the study. The three treatment groups were well balanced with respect to disease characteristics. All patients had a diagnosis of CP as defined by Rosenbaum. Approximately half the patients (49.8%) had spastic hemiparesis and 43.4% had spastic diparesis. Of the 102 patients with diparesis, 83 patients (81.4%) were injected in both legs versus 19 patients (18.7%) injected in one leg. The right leg was the most affected leg in just over half of all patients (54.9%). Epilepsy was reported for a higher proportion of patients in the aboBoNTA 10 U/kg and aboBoNTA 15 U/kg groups (10.1% and 12.7%, respectively) than in the placebo group (6.5%). The majority of patients (88.5%) were receiving some sort of non-drug therapy, casting/orthoses, and/or formal physiotherapy when they entered the study, and the number of patients receiving therapy was similar between–treatment groups (Table 5).

Table 5. Summary of Baseline Characteristics (ITT Population) for Study 141.

Table 5

Summary of Baseline Characteristics (ITT Population) for Study 141.

In Study 701, the aboBoNTA-treated patients were approximately a year older with consequently greater weight and height. Also, almost two-thirds of the aboBoNTA group was males, compared with half of the placebo group. All patients were Caucasian. The aboBoNTA and placebo groups were well balanced with regard to baseline maximum passive ankle dorsiflexion and Leeds Videographic Gait Assessment (VGA) at baseline. There was a substantial difference between groups in the Gross Motor Function Measure (GMFM) overall and goal-total scores, with aboBoNTA-treated patients being less functionally impaired than placebo-treated patients (Table 6).

Table 6. Summary of Baseline Characteristics for Study 701 (All-Patients-Treated Population).

Table 6

Summary of Baseline Characteristics for Study 701 (All-Patients-Treated Population).

Interventions

In Study 141, before administration, the powder was reconstituted at the investigational site with sterile, preservative-free saline (sodium chloride for injection 0.9%). Sterile saline was added to obtain a total volume to inject per patients of 2 mL per lower limb (i.e., 2 mL for unilateral and 4 mL for bilateral injections). Two aboBoNTA doses (10 U/kg or 15 U/kg per GSC injected into the affected leg (s)) were compared with placebo. The total dose was either 10 U/kg or 15 U/kg for unilateral injections, or 20 U/kg or 30 U/kg for bilateral injections. A total dose of either 10 U/kg or 15 U/kg of aboBoNTA was injected intramuscular into the gastrocnemius muscle and soleus muscle in four injection sites per affected lower limb. The 2 mL volume of injection per lower limb was split between gastrocnemius and soleus muscles according to a ratio of 3:2. The injection volume for each site is illustrated in Table 7 below. The maximum dose injected in patients was not to exceed 1,000 U or 30 U/kg, whichever was the lower value. The intramuscular injections were administered at the treatment visit into clinically indicated lower-limb muscles, using electrical stimulation or ultrasound (combined with complementary technique), in single dosing sessions. aboBoNTA was provided by the manufacturer as a white lyophilized powder in type I, 3 mL glass vials. Placebo was provided by the manufacturer in type I, 3.0 mL glass vials and was indistinguishable from aboBoNTA. The placebo contained only the excipients described for aboBoNTA. To maintain the blind, an independent reconstitutor prepared the study treatment in the syringes.

Table 7. Injection Volume in Gastrocnemius-Soleus Complex per Leg Without Hamstring Injections for Study 141.

Table 7

Injection Volume in Gastrocnemius-Soleus Complex per Leg Without Hamstring Injections for Study 141.

Concomitant use of anticholinergic drugs and concomitant treatment with dantrolene, tizanidine, or a gamma-aminobutyric acidergic (GABAergic) opioid or other anti-spasticity drug, including baclofen and benzodiazepines, were permitted during this study if the dosage had been stable for the four weeks prior to study treatment and was expected to remain at this stable dose throughout the study. Physiotherapy and the use of casts and orthoses were also permitted if they had been initiated prior to study entry (at least four weeks prior in the case of physiotherapy). In addition, both physiotherapy and the use of casts or orthoses had to continue at the same pre-study frequency and intensity until at least week 12. No new casts or orthoses were to be initiated until week 12, and no new physiotherapy was to be initiated less than four weeks prior to study entry or during the course of study up to the week 12 visit. The following was not permitting during the study:

  • the administration of BoNT into any site of the body other than the lower limb
  • use of any investigational new drug or device or off-label use of any drug
  • treatment with any drug that interfered either directly or indirectly with neuromuscular function (e.g., aminoglycoside antibiotics)
  • use of neuroblocking drugs, such as those used during surgery (e.g., curare).

In Study 701, patients were randomized to one of two treatment groups, receiving either aboBoNTA (30 U/kg) or placebo. Study medication was prepared to a final volume of 2 mL and was administered equally into two sites in the gastrocnemius muscle in both legs (0.5 mL per site). aboBoNTA was presented as a freeze-dried white pellet containing 500 units of Clostridium BoNTA–hemagglutinin complex together with 125 mcg of human albumin and 2.5 mg of lactose in a clear glass vial. Matching placebo supplies were presented in identical clear glass vials containing 125 mcg of human albumin and 2.5 mg of lactose. Blinding was achieved by supplying the study medication for each patient in identical patient packs. The use of BoNT during the study or during the nine months preceding the study was prohibited. Any oral anti-spasticity medication being taken prior to the study was to be continued at the same dose throughout the study period. Other concomitant medications were allowed at the discretion of the investigator. Regular physiotherapy and the use of walking aids and orthoses were also permitted to continue during the study. If orthoses were changed at entry, it was recommended that the baseline assessments be delayed until the patient had stabilized. Nine patients (35%) in the placebo treatment group and seven patients (27%) in the aboBoNTA treatment group were taking concomitant medications at entry. Antiepileptics and psychoanaleptics were the most frequently used concomitant medications.

Outcomes

In Study 141, the primary outcome was the change from baseline to week 4 in the Modified Ashworth Scale (MAS) score in the GSC at the ankle joint of the (most) affected lower limb. The first secondary outcome was Physician’s Global Assessment (PGA) at week 4. The second secondary outcome was goal attainment scaling (GAS) at week 4. Tertiary outcomes included:

  • mean change from baseline to week 12 (and at end of study [EOS] or early withdrawal [EW]) in the MAS score in the GSC at the ankle joint of the (most) affected lower limb
  • proportion of patients with at least one grade reduction in MAS score from baseline to week 4 (and to week 12 and EOS/EW) in the GSC at the ankle joint of the (most) affected lower limb
  • mean PGA score at week 12 (and EOS/EW)
  • mean GAS score at week 12 (and EOS/EW)

▬▬▬▬▬

▬▬▬▬▬

▬▬▬▬▬

▬▬▬▬▬

  • ▬▬▬▬▬

If relevant, the aforementioned tertiary efficacy end points were also assessed at week 16, week 22, and week 28.

In Study 141, effort was made in each centre to ensure that the same evaluating investigator assessed the same patients for the duration of the study. All investigators were trained in the use of the assessment scales prior to the start of the study in an attempt to minimize variability between centres. They were also given follow-up training during the study. The assessor who conducted the PGA of treatment response was different from the person who evaluated the MAS. When making their assessment, none of the assessors had knowledge of the scores obtained by the other assessor.

In Study 701, the primary efficacy variable was functional change, as assessed by the change from baseline in overall GMFM score without walking aids or orthoses at week 4 of the study. Secondary efficacy outcomes were: change in GMFM overall score at weeks 8 and 16 compared with baseline, change in GMFM total score at weeks 4, 8, and 16 compared with baseline, change in VGA scores at weeks 4 and 16 compared with baseline, change in Leeds Functional Mobility Questionnaire (FMQ) at weeks 4 and 16 compared with baseline, and subjective functional assessment of gait at weeks 4, 8, and 16.

Please refer to Appendix 5 for more information on the validity of the outcome measures described in this section.

Modified Ashworth Scale

The MAS is commonly used to measure increased muscle tone and spasticity due to different pathologies and neurologic conditions.31 The MAS was derived from the original Ashworth Scale to measure muscle resistance while moving the affected joint through its full range of movement in order to passively stretch the muscle.31 It provides a semiquantitative measure of this resistance to passive movement.32,33 The MAS is easy to use as it requires no additional equipment; hence, it is one of the most commonly used tools to measure spasticity and muscle rigidity in patients with CP34 or hypertonia.35 It is administered by a physician or therapist during the patient visit and comprises a six-point scale used to measure the degree of spasticity (intensity of muscle tone) as follows: 0 = no increase in muscle tone; 1 = slight increase in muscle tone, manifested by a catch and release or by minimal resistance at the end range of motion when the affected part(s) is moved in flexion or extension; 1+ = slight increase in muscle tone, manifested by a catch, followed by minimal resistance throughout the remainder (less than half) of the range of movement; 2 = more marked increase in muscle tone through most of the range of movement, but the affected part(s) is easily moved; 3 = considerable increase in muscle tone, passive movement is difficult; 4 = affected part(s) rigid in flexion or extension. 25,35,36 The MAS score is normally a categorical variable; however, for this review, it was treated as a continuous variable and, hence, needed to be transformed. The derived MAS scores that were used in this review were 0, 1, 2, 3, 4, and 5, which corresponded to the aforementioned original MAS scores of 0, 1, 1+, 2, 3, and 4 (as previously described), respectively.25 A higher MAS score indicates increased muscle tone, rigidity, or spasticity. There is no evidence of the validity of the MAS in children with spasticity and there is conflicting evidence on reliability. In Study 141, all investigators were trained in the use of the assessment scales prior to the start of the study in an attempt to minimize variability between centres. They were also given follow-up training during the study. One of the clinical experts consulted for this review indicated that a one-point difference in the MAS (in either direction) was clinically relevant; however, no peer-reviewed evidence was identified regarding a minimal clinically important difference (MCID) for the MAS in pediatric patients with LLS. The other clinical expert consulted for this review indicated that defining an MCID is challenging for the MAS, but considers that a clinically important change in a single patient must be at least a one-point change due to the nature of the MAS. However, based upon his clinical experience, a change between–treatment groups as low as 0.38 would be considered clinically significant when related to a group of patients receiving treatment.

Physician’s Global Assessment

In the pivotal study of this submission, the PGA of treatment response was conducted by the investigator by scoring responses to the question: “How would you rate the response to treatment in the patient’s lower limb(s) since the last injection?” on a nine-point categorical scale where −4 = markedly worse, −3 = much worse, −2 = worse, −1 = slightly worse, 0 = no change, +1 = slightly improved, +2 = improved, +3 = much improved, and +4 = markedly improved. Assessment of the PGA was undertaken independently by an investigator who was different from the one who assessed the MAS.22 No literature was identified regarding the psychometric properties (validity, reliability, or responsiveness) of the PGA for pediatric patients with LLS. In addition, no MCID for the PGA in this population has been identified.

Goal Attainment Scaling

GAS is a method of integrating the achievement of a number of individually set goals into a single goal attainment score.37 It has been applied in various areas of complex interventions, including spasticity management.38 Before the treatment, one or more individual goals are established by the patient (or their caregiver, if the patient is a child)39 and one or more researchers or practitioners (or others agreed upon by the practitioner). The clinician/researcher requires sufficient knowledge and experience when supporting patients to set realistic goals. In addition, they must be able to respect the patient’s ideology and what is important to them when setting goals (and thus able to avoid projecting their own goals and what they perceive to be important onto the patient) and they must have good negotiating skills in order to manage potentially unrealistic goals set by the patient.37 The number of goals can vary between patients in the same study and between patients in different studies. Numerical values ranging from −2 to +2 (a five-point scale) are used to describe the degree to which the goal(s) were or were not met.37 The expected target of achievement is set by the patient and treating team and given a value of 0. Outcomes that are less than expected are given values of −1 or −2 (the most unfavourable outcome) and outcomes that are better than expected are given values of +1 or +2 (the most favourable outcome). The originators of the GAS score transformed it to a standard variable (the T score), with scores ranging from 0 to 100, a mean of 50, and a standard deviation of 10. A change in the GAS T score of more than 10 appeared clinically important in adult patients with upper-limb spasticity (ULS) who had suffered diffuse brain injury or stroke or who had been diagnosed with multiple sclerosis and had been classified as responders (positive clinical outcome associated with BoNT treatment as identified by the treating physician) and nonresponders (negative or non-significant clinical outcome associated with BoNT treatment as identified by the treating physician).40 However, no validity or reliability studies have been conducted in children and, as a result, it is unclear if the psychometric properties observed in adults (particularly the responsiveness with GAS) apply to children. No MCID was identified for the GAS score in pediatric patients with LLS.

Tardieu Scale

The TS was developed by Tardieu et al. in 1954 to clinically measure spasticity by measuring the different angles of reaction when passing the muscle through stretches at different predefined velocities.41,42 This outcome measure was developed to more closely align with the 1979 Lance definition of spasticity, specifically, a “motor disorder characterized by a velocity-dependent increase in tonic stretch reflexes (muscle tone), with exaggerated tendon jerks, resulting from hyperexcitability of the stretch reflex, as one component of the upper motor neuron syndrome.”41 Spasticity is thus rated by examining the reaction difference of the muscle in question between the slowest and fastest stretch speed, both of which are performed by the same practitioner at the same time of day with the muscle being in the same resting position.41 The slow stretch assesses the passive range of motion and is slow enough to avoid producing a significant stretch reflex. The stretch at the fastest velocity is performed to maximize the involvement of the stretch reflex, thus producing a catch-and-release sensation (also termed clonus) that is dependent on the amount of spasticity present.41 Two parameters are used to measure the muscle spasticity, namely the spasticity angle X (which is the difference between slow-speed angle of arrest [V1] and the clonus or catch-and-release angle at the highest speed [V3]) and the spasticity grade Y (the grading of the intensity of the muscle reaction to the fastest stretch [V3] and is an ordinal variable). Larger spasticity angles correspond to more spasticity in the muscle. The spasticity is graded as follows: grade 0 = absence of spasticity as defined by a catch that is not followed by a release; grade 1 = passive movement is slowed down by mild resistance; grade 2 = passive movement (the catch and release) is transiently interrupted, grades 3 and 4 = severe spasticity; and non-ratable = a catch that is not followed by an obvious release occurring at inconsistent angles.41 Training has been shown to enhance the reliability of the TS, particularly in the angle of catch at fast speed (XV3), in all muscles except the knee flexors.41 In Study 141, all investigators were trained in the use of the assessment scales prior to the start of the study in an attempt to minimize variability between centres. They were also given follow-up training during the study. No MCID was identified in the literature with regard to pediatric patients with LLS. The TS at the ankle joint of the most affected lower limb was reported in Study 141.

Observational Gait Scale

The OGS is an objective outcome measure used to document gait changes (or impairments) of the upper motor syndrome in young children who have received injections of BoNT.22,43 It was derived from the Physician Rating Scale by expanding the scale from six to eight sections, including putting more emphasis on the knee-to-foot relationship during the standing phase. The gait parameter sections that make up the OGS include knee position in mid stance, initial foot contact, foot contact mid stance, timing of heel rise, hindfoot at mid stance, base of support, gait assistive devices, and change. The maximum score is 22 for each leg, which denotes a normal gait. In older children, the standard of assessing gait includes instrumented three-dimensional gait analysis; however, this is not always appropriate for children due their potential to be uncooperative and their small size.43 The child is recorded while walking and the investigator (e.g., someone with extensive knowledge of gait analysis) looks at the video in order to score each component.43 The OGS is a validated and reliable instrument to assess response to treatment for pediatric patients with spasticity. No MCID was identified in the literature regarding pediatric patients with LLS. The OGS in the most affected leg was reported in Study 141.

Pediatric Quality of Life Inventory Version 4.0 Generic Core Scales

The original Pediatric Quality of Life Inventory (PedsQL) was developed as a health-related quality-of-life (HRQoL) measure that addressed the paucity of appropriately validated and reliable instruments incorporating both the child and parental experience with chronic health conditions. The PedsQL uses a modular approach and incorporates generic and disease- and symptom-specific items that are appropriate for the assessment of pediatric chronic conditions.44 The PedsQL 4.0 Generic Core Scales comprise 23 items under the following modules: Physical Functioning (eight items), Emotional Functioning (five items), Social Functioning (five items), and School Functioning (five items).45 The Generic Core Scales comprise both a parent-proxy report and a child self-report that assess health perceptions. The child self-report format is specifically for three age groups: five to seven, eight to 12, and 13 to 18 years of age, while the corresponding parent-proxy reports are specifically for toddlers (ages two to four, for which there is no child self-assessment report), young children (ages five to seven), children (ages eight to 12), and adolescents (ages 13 to 18). The questions ask how much of a problem each item has been in the past month. A fivepoint Likert response scale is used across the child reports (from ages eight to 18) and the corresponding parent report and includes the following responses with corresponding scores: 0 = never a problem; 1 = almost never a problem; 2 = sometimes a problem; 3 = often a problem; and 4 = almost always a problem. In addition, a three-point scale is used for simplification and ease of use for children aged five to seven years (0 = not at all a problem; 2 = sometimes a problem; and 4 = a lot of a problem), with each of the response choices on the scale anchored to a happy, neutral, or sad face.45 The scores, which are reversed scored, are transformed linearly to a 0 to 100 scale, whereby 0 = 100, 1 = 75, 2 = 50, 3 = 25, and 4 = 0, with higher scores indicative of a higher HRQoL. The PedsQL Generic Core Scales have been validated and determined to be reliable and responsive in pediatric patients with chronic conditions. However, whether validity and responsiveness of the PedsQL holds true in pediatric patients with LLS is unknown, as the PedsQL has never been evaluated in this population and currently no known MCID exists for the PedsQL in pediatric patients with LLS.

Faces Pain Scale — Revised

The Faces Pain Scale (FPS) and Faces Pain Scale — Revised (FPS-R) were developed to measure pain in pediatric patients.46,47 Bieri et al.46 developed the FPS using a five-phase approach, with each phase helping lead to the development of the seven-faces (seven items) scale construct. The final phase examined the test–retest reliability and subsequently showed that a rank correlation coefficient of 0.79 was obtained when six-year-old children rated a painful experience over a two-week time period.46 Hicks et al.47 undertook the revising of the original FPS, as the seven-point version was not easily rescaled to either a 0 to 5 or 0 to 10 metric. Instead, they adapted the FPS to a six-face scale, with corresponding scoring of 0, 2, 4, 6, 8, and 10 (or 0, 1, 2, 3, 4, 5); a higher score indicates more pain.47 The clinical expert consulted for this review explained that pain is not normally associated with spasticity; therefore, the relevance of this outcome measure remains under question. No literature was identified regarding the psychometric properties (validity, reliability, or responsiveness) of the FPS for pediatric patients with LLS. In addition, no MCID for the FPS-R in this population has been identified.

Gross Motor Function Measure

The GMFM (and, subsequently, the 88-item GMFM [GMFM-88]48,49) is an outcome measure used to evaluate change in gross motor function over time in children with varying degrees of CP.50 The 85 items that made up the original GMFM (and the subsequent five additional items included in the GMFM-8848,49) were chosen because they were the items most likely to show change in patients with CP. Individuals items were combined into five separate areas of motor function to facilitate scoring. These dimensions include: A = lying and rolling; B = sitting; C = crawling and kneeling; D = standing; and E = walking, running, and jumping.48,50 Each individual item is scored on a four-point Likert scale (0 to 3), with assignments as follows: 0 = cannot do; 1 = initiates (< 10% of the task is completed); 2 = partially completes (10% to < 100% of the task); and 3 = task completion (100% of the task). Each dimension contributes equal weight; therefore, dimension scores are calculated using the following formula: child’s score ÷ maximum score × 100%. The total score is then obtained by adding up all of the dimension scores (per cent) and then dividing them by the total number of dimensions (five dimensions). To increase responsiveness, and if the therapist identifies specific goals, a goal-score total can also be calculated (using the same aforementioned algorithm for obtaining the total scores; however, this time, by dividing by the dimensions that were part of the goal setting).4850 It should be noted that the GMFM (and GMFM-88) only assesses how much of the task the child can perform (quantity) and does not measure how well the task is performed (quality).49 The GMFM is a validated instrument to assess response to treatment for pediatric patients with spasticity. No MCID was identified in the literature for the GMFM-88 with regard to pediatric patients with LLS. In Study 701, the total score was named the overall score.

Leeds Functional Mobility Questionnaire

In Study 701,51 the investigators used the Leeds FMQ, a 50-item questionnaire that was developed to identify and assess changes in the patient’s ability to manage everyday activities that are typically impaired in patients with LLS. It is administered as a structured interview with the patient’s parents and was administered at 0, 4, and 16 weeks post–aboBoNTA treatment. It is subdivided into three separate domains: sitting and standing, mobility, and other activities.51 There is no overall score for this rating instrument and each question is summarized and analyzed separately. Categorical data are generated from each question and assess the degree of difficulty when performing certain activities. A lower score indicates improved function. The Leeds FMQ was developed by the Regional Child Development Centre at St James’s University Hospital in Leeds, UK; however, it is still in the process of development.51 Hence, there has been no literature identified regarding its psychometric properties (validity, reliability, or responsiveness) for pediatric patients with LLS. In addition, no MCID for the Leeds FMQ in this population has been identified.

Leeds Videographic Gait Assessment

In Study 701,51 the investigators used the Leeds VGA to observe patient gait, viewed in both the sagittal and coronal planes. It was developed by the Leeds Regional Child Development Centre at St James’s University Hospital in Leeds, UK.51 In the study, patients walked along a walkway both with and without their normal splints and footwear at weeks 0, 4, and 16. The video clips were blinded and randomized to be reviewed by a panel of clinicians and physiotherapists who had experience in the management of children with walking difficulties associated with muscle spasticity. A standard score sheet was used to rate the following parameters, with each leg scored separately: initial foot contact, degree of knee flexion, presence/absence of rocker-bottom foot, hindfoot deformity (presence of valgus or varus), and walking aids used.51 No literature was identified regarding the psychometric properties (validity, reliability, or responsiveness) of the Leeds VGA for pediatric patients with LLS. In addition, no MCID for the Leeds VGA in this population has been identified.

Subjective Functional Assessment of Gait

This subjective functional assessment of gait was used by both the parent and investigator at each post-treatment visit to assess functional changes in response to treatment with aboBoNTA.51 Specifically, there is the parent’s and investigator’s opinion (scored separately) on the child’s functional changes, with the choices being presented as follows: good response; minimal response; no response; worse response; and not recorded.51 No literature was identified regarding any psychometric properties (validity, reliability, or responsiveness) of the subjective functional assessment of gait for pediatric patients with LLS. In addition, no MCID for the outcome measure in this population has been identified.

Harms

Adverse events (i.e., treatment-emergent adverse events [TEAEs], serious adverse events [SAEs], withdrawal due to adverse events [WDAEs], and notable adverse events [i.e., adverse events of special interest in this review]) were reported in both randomized controlled trials (RCTs).

In Study 141, a TEAE was defined as any adverse event that occurs during the treatment phase of the study if it: was not present prior to receiving the first intake of study medication; was present prior to receiving the first intake of study medication but the intensity increased during the treatment phase of the study; or it was present prior to receiving the first intake of study medication and the intensity was the same as it was prior to the first intake of study medication, however, during the active phase of the study the adverse event was related to the medication intake. An SAE was defined as any adverse event that is life-threatening or resulted in death, patient hospitalization, or prolongation of an existing hospitalization, a persistent or significant disability or incapacity, or a congenital anomaly or birth defect in the offspring of a patient who received the study treatment. An SAE was also defined as an important medical event that, based on appropriate medical judgment, may jeopardize the patient and may require medical and/or surgical intervention to prevent one of the outcomes listed previously.

In Study 701, an adverse event included any noxious, pathologic, or unintended change in anatomical, physiologic, or metabolic functions as indicated by physical signs, symptoms, and/or laboratory changes occurring in any phase of the clinical trial, whether associated with a drug or placebo and whether or not considered drug-related. An SAE was defined as any event that is fatal; life-threatening; permanently or temporarily disabling or incapacitating or results in hospitalization; or prolongs a hospital stay or is associated with congenital abnormality, cancer, or overdose (either accidental or intentional). In addition, any event the investigator regards as serious, or which would suggest any significant hazard, contraindication, side effect, or precaution that may be associated with the use of the drug, should be reported as a serious event.

Statistical Analysis

Study 141

In Study 141, the primary (MAS) and first secondary (PGA) efficacy end points were taken into account in the sample size calculation. The power used for the sample size calculations was equal to 85% for the primary efficacy end point and equal to 90% for the first secondary efficacy end point. The sample size needed per group was calculated for each end point separately, and then the larger one was retained. A total of 228 randomized patients (i.e., 76 randomized patients per treatment group) were necessary to demonstrate a statistically significant treatment effect on the primary efficacy end point with a type I error rate controlled at level 0.05 and a power of 85%, assuming: mean changes from baseline to week 4 in the MAS score of −1.3 and −0.9 in the aboBoNTA and placebo groups, respectively, a common standard deviation for the change from baseline to week 4 in the MAS score of 0.8, and a 3% dropout rate from baseline to week 4. A total of 165 randomized patients (i.e., 55 patients per treatment group) were necessary to demonstrate a statistically significant treatment effect on the mean PGA score with a two-sided comparison-wise type I error rate controlled at 0.05 and a power of 90%, assuming a between-group mean score difference of the PGA at week 4 of 0.7, a common standard deviation of the PGA score at week 4 of 1.1, and a 3% dropout rate at week 4. A targeted total sample size of 228 randomized patients (i.e., 76 randomized patients per treatment group) was considered sufficient to detect a treatment effect on both the primary and first secondary efficacy end points. Using a sample size of 228 as the larger of the two required figures meant the actual power for the PGA score comparison rose to 97%. The rationale for the above threshold for the MAS score was based on a previous clinical trial conducted in children with CP for lower extremity spasticity.52 That study assessed three different doses of onaBoNTA (low-dose group [1 U/kg], middle-dose group [3 U/kg], and high-dose group [5 U/kg]). The rationale for the aforementioned threshold or PGA was based on a previous clinical trial for ULS conducted in adult patients after a stroke.53

Two different statistical methodologies for the efficacy analyses were applied for the registrations in the US and non-US countries. Only the non-US approach and data are presented for the purposes of this review.

In non-US countries, superiority was based on the primary efficacy end point only. In order to control the family-wise type I error rate, the following two-step hierarchical testing procedure was applied for the testing of the superiority of each of the two aboBoNTA doses to placebo, where the superiority of aboBoNTA 15 U/kg to placebo for MAS at four weeks (primary efficacy outcome) was tested at a significance level of 0.05. If the P value associated with that testing was lower than 0.05, then the superiority of aboBoNTA 10 U/kg to placebo for MAS at four weeks (primary efficacy outcome) was tested at a significance level of 0.05. If the P value associated with that testing was lower than 0.05, it was then considered significant. In the event the hierarchical testing procedure was stopped at the end of step 1, the testing of the superiority of aboBoNTA 10 U/kg to placebo on the primary efficacy end point was performed to characterize the full clinical effect, but no formal statistical conclusion was drawn. Each of the two secondary efficacy end points was analyzed to compare each aboBoNTA dose with placebo at a 0.05 type I error rate.

Each tertiary efficacy end point was analyzed for exploratory purposes only to compare each aboBoNTA dose with placebo. No adjustment for multiplicity was completed for these analyses.

The primary efficacy analysis consisted of two contrast analyses within a single analysis of covariance (ANCOVA) model controlling for the baseline MAS score and the randomization stratification factors (age range and BoNT-naive or non-naive status as assessed at baseline) and the centre, all as fixed effects. The least squares (LS) means and the associated 95% CIs were calculated for the aboBoNTA and placebo groups, plus the differences in the LS means between these groups and the associated P values. The first secondary efficacy end point (mean PGA score at week 4) and the second secondary efficacy end point (mean GAS score at week 4) were analyzed using an analysis of variance (ANOVA) model, controlling for the randomization stratification factors (age range and BoNT treatment status) at baseline and at the centre, all as fixed effects. The LS means and the associated 95% CIs were calculated for the aboBoNTA and placebo groups, as were the differences in the LS means between these groups and the associated P values. For each of the tertiary end points, summary tables of raw values and change from baseline were provided at each visit. In ANOVA or analysis of covariance (ANCOVA) models, the LS means and the associated 95% CIs were calculated for the aboBoNTA and placebo groups, as were the differences in the LS means between these groups and the associated P values. The odds ratios and their 95% CIs were calculated from a logistic regression. To assess the impact of missing efficacy data at week 4, sensitivity analyses were performed with missing data imputed with baseline values (primary end point) or with the “markedly better” or “markedly worse” data (sensitivity analysis of the first secondary end point PGA data).

Subgroup analyses were performed on the primary and secondary efficacy end points in the intention-to-treat (ITT) population by BoNT status (naive or non-naive) at baseline. In the protocol for this review, a subgroup analysis by baseline severity of spasticity was identified; however, such analyses were not conducted.

Study 701

In Study 701, a sample size of 50 patients (25 patients per treatment group) was planned to provide 90% power to detect a clinically significant between-group difference of 10% in the overall GMFM score at the 0.05 significance level, allowing for a dropout rate of 5% to 10%. No rationale was provided on how the clinically significant between-group difference of 10% was selected. The primary efficacy end points in GMFM scores were analyzed using ANCOVA. For all other efficacy variables, analysis was performed using logistic regression. Centre, strata, and baseline scores were included in the model, as appropriate. No adjustments for multiplicity were performed. Missing data were imputed using the last observation carried forward (LOCF). No subgroup analysis was conducted in Study 701.

Analysis Populations

In Study 141, efficacy analyses were performed using the ITT population, which included all randomized patients who received at least one injection of study treatment and who had a MAS score in the GSC assessed both at baseline and at week 4. The per-protocol (PP) population was defined as all patients in the ITT population who did not have major protocol violations between baseline and week 4, inclusive. The safety population was defined as all randomized patients who received at least one injection of study treatment. The ITT population should be considered modified ITT given that appropriate ITT population would include all randomized patients regardless if they received treatment or had assessment after receiving treatments.

In Study 701, all safety and efficacy analyses were performed using the all-patients-treated (APT) population, which comprised all patients randomized to the study who received some study medication. The PP population comprised all patients in the APT population who did not have major protocol violations.

Patient Disposition

In Study 141, a total of 253 patients were screened, of whom 241 were enrolled into the study and were randomized. Of the randomized patients, a total of 239 patients received study treatment and 230 patients had at least 12 weeks of follow-up: 75 in the placebo group, 78 in the aboBoNTA 10 U/kg treatment group, and 77 in the aboBoNTA 15 U/kg treatment group (Table 8). Overall, 15 patients (6.2%) discontinued the study prematurely; 13 patients discontinued prior to or at week 12 (eight in the placebo group, two in the aboBoNTA 10 U/kg treatment group, and three in the aboBoNTA 15 U/kg treatment group); and two patients discontinued after week 12, both of whom were in the aboBoNTA 15 U/kg treatment group. The most common reason for discontinuation was consent withdrawal (seven patients). Two patients who were screening failures were randomized to the placebo group in error. The reasons given for these two patients withdrawing from the study were “does not meet entry criteria” and “other” (patient refused to enrol in the study). This was noted at the time and both patients were withdrawn from the study before receiving study treatment.

Table 8. Patient Disposition for Study 141.

Table 8

Patient Disposition for Study 141.

In Study 701, a total of 52 patients were randomized. There were no withdrawals and all patients completed the study up to week 16 (Table 9). A total of 16 patients were considered to have ongoing benefit and continued to week 24. Of these, 14 patients continued to week 36.

Table 9. Patient Disposition for Study 701.

Table 9

Patient Disposition for Study 701.

Exposure to Study Treatments

▬▬▬▬▬

In Study 701, with the exception of one placebo-treated patient and two aboBoNTA-treated patients who had only one leg treated, all other patients received the appropriate study medication specified by the randomization list.

Table 10. Exposure (Dose per Leg) for Study 141 — Safety Population.

Table 10

Exposure (Dose per Leg) for Study 141 — Safety Population.

Table 11. Number of Legs Treated (Dose per Leg) for Study 141 — Safety Population.

Table 11

Number of Legs Treated (Dose per Leg) for Study 141 — Safety Population.

Critical Appraisal

Internal Validity

The objectives of Study 141 were well defined. Randomization was stratified by patient age and previous exposure to BoNT treatment. Allocation concealment was sufficiently described. The randomization manager, who was a statistician independent from the study, prepared and kept the master randomization list for this study. The treatment arm allocations, as well as the treatment numbers supplied, were managed by an interactive voice response system (IVRS). The IVRS also managed all the logistical aspects for the study treatments. The sample size was determined based on the power (≥ 85%) to detect a difference of change from baseline (mean ± SD: 1.3 ± 0.8 and 0.9 ± 0.8 in the aboBoNTA and placebo groups, respectively) for MAS score at week 4 (P < 0.05) and based on the power (≥ 90%) to detect a between-group mean difference (mean ± SD: 0.7 ± 1.1) for the PGA score at week 4. The rationale for the aforementioned threshold for MAS was based on a previous clinical trial conducted in children with CP for lower extremity spasticity.52 That study assessed three different doses of onaBoNTA (1 U/kg, 3 U/kg, and 5 U/kg) and there was no placebo treatment group included, so it is not clear what the rationale was for using 0.9 for the change from baseline in the placebo group. The rationale for the aforementioned threshold for PGA was based on a previous clinical trial for ULS conducted in adult patients after a stroke, which is a different population than pediatric patients with LLS.53 Key patient baseline characteristics were balanced across treatment groups. The relevant concomitant medications (for spasticity) and the physiotherapy treatments were well described and balanced between the aboBoNTA and placebo groups. The outcome measurements (especially for the primary outcome and the secondary outcome) were well described. The overall dropout rate was low, but it was slightly higher in the placebo group (9.9%) than in the aboBoNTA treatment groups (2.5% in the aboBoNTA 10 U/kg treatment group and 6.3% in the aboBoNTA 15 U/kg treatment group). However, for the assessment of the primary outcome (MAS at 4 weeks), 99% of the patients randomized to the aboBoNTA treatment groups and 95% of those randomized to the placebo treatment group were included in this analysis. Hence the dropout rate does not seem to be a big limitation for the primary outcome.

In Study 141, effort was made in each centre to ensure the same evaluating investigator assessed the same patients for the duration of the study. All investigators were trained in the use of the assessment scales prior to the start of the study in an attempt to minimize variability between centres. They were also given follow-up training during the study. The assessor who conducted the PGA of treatment response was different from the person who evaluated the MAS. When making their assessment, none of the assessors had knowledge of the scores obtained by the other assessor. Providing training in the use of the assessment scales prior to the start of the study, and having the same evaluating investigator assessing the same patients for the duration of the study, would help in improving inter-rater reliability.

While, overall, Study 141 was generally well designed, as mentioned previously, some methodological limitations of the RCT need to be discussed in the interpretation of the results. Although the master randomization list was prepared independently and the allocation concealment was sufficient, how the randomization list was generated was not clearly described in the Clinical Study Report. While identical active and placebo vials were provided to maintain blinding for patients and investigators, there was a risk of unblinding in this trial as, overall, 48% of patients had previous exposure to BoNT treatment and were therefore likely to expect a reduction in symptoms after the injection. Placebo-treated patients would not experience this reduction in symptoms and, therefore, the patient and investigators might be able to identify treatment based on response, potentially impacting subjective outcomes, adverse effect reporting, and study dropout rates. Further, the criteria for re-treatment were not clearly defined. In addition, the analysis set for the primary analysis was identified as the ITT population; however, this is not a true ITT because the ITT population used in Study 141 was defined as all randomized patients who received at least one injection of study treatment and who had an MAS score in the GSC assessed both at baseline and at week 4, whereas the appropriate ITT population would include all randomized patients and, hence, the ITT population used in Study 141 should have been identified as a modified ITT population. Except for the primary outcome (MAS at week 4) which was analyzed based on a two-step statistical testing hierarchy to control type I error, the secondary outcomes (PGA and GAS at week 4), subgroup analyses, and all tertiary outcomes (such as MAS, PGA assessed at week 12, TS, OGS, FPS, and PedsQL) were analyzed for exploratory purpose only and no control for multiplicity of testing was employed, thus increasing the risk of type I error for all end points assessed, other than the primary end point; thus, all end points beyond the primary should be interpreted cautiously. In addition, subgroups were not analyzed appropriately, as no-test for interaction by previous BoNT experience was conducted to determine if there was an interaction between the treatment effect and previous exposure to BoNT. Also, the interpretation of data beyond week 12 in Study 141 should be interpreted with extreme caution due to the large number of dropouts. Furthermore, no MCIDs were established specific to a pediatric population with LLS, and the clinical significance of the benefit of aboBoNTA compared with placebo for all outcomes assessed was not clear from the literature.

The objectives of Study 701 were well defined. Eligible patients were randomly allocated to one of the two treatment groups following the randomization schedule, generated in blocks of four prior to the study. Each centre was supplied with blocks of four patient numbers, as required. Production of the randomization list and the blinding of study medication were performed by Penn Pharmaceuticals Ltd. Blinding was achieved by supplying the study medication for each patient in identical patient packs. Given the small number of patients included and that blocks of four were used, which potentially could make the allocation of participants predictable, selection bias might have been introduced. Also, balance may not have been achieved across the baseline variables, suggesting randomization was not successful, which may substantially bias the study results. In addition, it is not clearly described in the Clinical Study Report how the randomization list was generated. The sample size was determined based on the power (90%) to detect a clinically significant between-group difference of 10% in the overall GMFM score at the 0.05 significance level. However, no rationale was provided on how that clinically significant between-group difference of 10% was calculated. Study 701 did not show statistical significance between groups; this could be due to wrong assumptions made in the sample size calculation. The aboBoNTA and placebo groups were well balanced with regard to baseline maximum passive ankle dorsiflexion and Leeds VGA at baseline; however, there was a substantial difference between groups in the GMFM overall and goal-total scores with aboBoNTA-treated patients being less functionally impaired than placebo-treated patients. No patient discontinued the study. Also, the potential implications of conducting multiple statistical tests were not considered, and no adjustment was made for multiple testing despite secondary end points analyses, which would increase the risk of type I (false-positive) error; thus, all end points beyond the primary should be interpreted cautiously. Furthermore, no MCIDs were established specific to a pediatric population with LLS, and the clinical significance of the benefit of aboBoNTA compared with placebo for all outcomes assessed was not clear from the literature.

External Validity

There were no Canadian sites enrolled in both studies. According to the clinical expert consulted for this review, the population enrolled in both trials was generally representative of Canadian pediatric patients with LLS. The expert did note, however, that based on the baseline characteristics, the patient population appeared to be limited to ambulatory patients with mild to moderately severe spasticity. Only patients with LLS with a diagnosis of CP were included in both trials. No patients with LLS from other causes such as stroke, brain injury, or spinal cord injury were included in both trials; therefore, it is unclear whether the reported efficacy and safety of aboBoNTA could be generalized to pediatric patients who have LLS due to other causes or higher levels of functional impairment. (GMFCS levels IV and V). However, the clinical expert consulted for this review indicated that the underlying cause of the LLS would not impact the treatment strategy applied.

In Study 701, only the gastrocnemius was injected and not the GSC muscles, hence, the soleus was not injected as recommended in the Health Canada product monograph. The clinical expert consulted for this review indicated this might be an issue or a limitation and the response might differ from what would be seen when the muscles of the GSC are injected.

Efficacy

Only those efficacy outcomes identified in the review protocol are reported subsequently (Objective and Methods Section and Table 3). See Appendix 4 for detailed efficacy data.

Modified Ashworth Scale

MAS scores were assessed only in Study 141. The MAS was used to measure the intensity of muscle tone in the GSC at the ankle joint by measuring the resistance of the muscle to passive lengthening or stretching. The muscle tone of the affected leg was assessed in patients with unilateral lower-limb impairment, and of the most affected leg in patients with bilateral lower-limb impairment. The muscle tone was graded on a six-point scale, from 0 (no increase in tone) to 4 (affected parts rigid in flexion or extension).

In Study 141, the MAS scores at baseline (mean ± SD) were 3.1 ± 0.3, 3.1 ± 0.3, and 3.2 ± 0.4 in the aboBoNTA 10 U/kg/leg, aboBoNTA 15 U/kg/leg, and placebo groups, respectively. At week 4, the between-group mean difference in change from baseline was statistically significant (−0.49, 95% CI, −0.75 to −0.23, P = 0.0002) in the aboBoNTA 15 U/kg/leg group compared with the placebo group. Likewise, the between-group mean difference in change from baseline for the aboBoNTA 10 U/kg/leg group compared with placebo was statistically significant (−0.38, 95% CI, −0.64 to −0.13, P = 0.0029) (Table 12). MAS scores assessed at week 12 were analyzed as a tertiary outcome for exploratory purposes only. The improvement in MAS score observed for both aboBoNTA groups at week 4 appeared to be maintained at week 12 to a lesser extent. The MAS score results after week 4 and up to week 28 (for those who continued in the study) are presented in Table 18.

Table 12. Modified Ashworth Scale Score in the (Most) Affected Leg, Change From Baseline at Week 4 (Dose per Leg) for Study 141 — ITT Population.

Table 12

Modified Ashworth Scale Score in the (Most) Affected Leg, Change From Baseline at Week 4 (Dose per Leg) for Study 141 — ITT Population.

The sensitivity analysis of the primary efficacy end point, performed on all randomized patients who received study treatment by imputing missing data with baseline values, appeared to be in the same direction as the primary analysis, ▬▬▬▬▬

The subgroup analysis for the MAS assessed by previous exposure to BoNT treatment also showed an improvement in aboBoNTA 15 U/kg group compared with placebo, regardless of prior exposure to BoNT. An improvement was also seen in aboBoNTA 10 U/kg group compared with placebo regardless of prior exposure to BoNT (Table 19). No test for interaction by previous BoNT experience was conducted in order to determine if there was an interaction between the treatment effect and previous exposure to BoNT.

Responders Based on Modified Ashworth Scale

▬▬▬▬▬

Tardieu Scale Score

The TS score was analyzed in Study 141 for exploratory purposes only to compare each aboBoNTA dose with placebo. The spasticity angle (which is the difference between slow-speed angle of arrest and the clonus or catch-and-release angle at the highest speed, with larger spasticity angles corresponding to more spasticity in the muscle) and the spasticity grade Y (which is the grading of the intensity of the muscle reaction to the fastest stretch and is an ordinal variable ranging from 0 [defined as absence of spasticity as defined by a catch that is not followed by a release] to 4 [indicating severe spasticity]) are presented in Table 22.

▬▬▬▬▬

Goal Attainment Scaling

GAS at week 4 was analyzed as the second secondary outcome in Study 141. However, this outcome was not included in the statistical testing hierarchy and, hence, was not controlled for type I error and the level of significance may be inflated; therefore, statistical significance should be interpreted with caution for this outcome.

GAS was used to measure progress toward individual therapy goals. Between one and three individual goals (from a list of pre-selected goals) were defined for each patient by the physician, and the child’s parents (caregiver) where applicable, prior to treatment. The goal outcome scores were incorporated into a single aggregated overall GAS T score for each patient. If all goals were achieved as expected, the overall GAS score was 50.0. If the overall response was better than expected, the value was > 50.0 and, conversely, if the overall response was less than expected, the overall GAS score was < 50.0. Table 23 summarizes the goals selected at baseline for each treatment group. The five main goals chosen were “improved walking pattern,” “improved balance,” “decreased frequency of falling,” “decreased frequency of tripping,” and “improved endurance.” The most commonly chosen goal for each treatment group was “improved walking pattern.” ▬▬▬▬▬

Patients in both aboBoNTA treatment groups achieved a mean GAS score above 50.0, demonstrating that the overall response was better than expected. However, patients in the placebo group showed a less-than-expected response with a mean GAS score below 50.0. This result was statistically significant in both aboBoNTA treatment groups compared with placebo (Table 13). The outcome measure GAS at 4 weeks was not part of the hierarchical analysis plan and therefore was not adjusted for multiple comparisons; hence, the level of significance is inflated and results should be interpreted with caution. GAS scores assessed at week 12 were analyzed as a tertiary outcome for exploratory purposes only. The improvement in GAS score observed for both aboBoNTA groups at week 4 appeared to be maintained at week 12. The GAS score results after week 4 and up to week 28 are presented in Table 24.

Table 13. Goal Attainment Scaling Total Score at Week 4 (Dose per Leg) for Study 141 — ITT Population.

Table 13

Goal Attainment Scaling Total Score at Week 4 (Dose per Leg) for Study 141 — ITT Population.

The three most commonly chosen goals in the study were “improved walking pattern,” “improved balance,” and “decreased frequency of falling.” For “improved walking pattern” and “decreased frequency of falling,” there was a higher mean score in the aboBoNTA 10 U/kg treatment group and aboBoNTA 15 U/kg treatment group compared with the placebo group, and for the “improved balance” there was a similar mean score in all three treatment groups (Table 25).

The subgroup analysis for GAS assessed by previous exposure to BoNT treatment also showed an improvement in both aboBoNTA treatment groups compared with placebo, regardless of prior exposure to BoNT (Table 26). No test for interaction by previous BoNT experience was conducted to determine if there was an interaction between the treatment effect and previous exposure to BoNT.

Health-Related Quality of Life (Pediatric Quality of Life Inventory)

Pediatric Quality of Life Inventory Generic Core Scales

▬▬▬▬▬

▬▬▬▬▬

▬▬▬▬▬

▬▬▬▬▬

▬▬▬▬▬

Physician’s Global Assessment

The PGA of treatment response at week 4 was analyzed as the first secondary outcome in Study 141. The LS means of the PGA at week 4 were 1.54 (95% CI, 1.28 to 1.81) in the aboBoNTA 10 U/kg group, 1.50 (95% CI, 1.23 to 1.77) in the aboBoNTA 15 U/kg group, and 0.73 (95% CI, 0.46 to 0.99) in the placebo group, respectively (Table 14). The results of the PGA show that aboBoNTA (10 U/kg and 15 U/kg) were statistically significantly more effective than placebo (0.82 [95% CI, 0.50 to 1.14, P < 0.0001] and 0.77 [95% CI, 0.45 to 1.10, P < 0.0001], respectively). The PGA was not part of the hierarchical analysis plan and therefore was not adjusted for multiple comparisons, hence, the level of significance is inflated and results should be interpreted with caution. The PGA assessed at week 12 was analyzed as a tertiary outcome for exploratory purposes only. The improvement in PGA score observed for both aboBoNTA groups at week 4 was maintained at week 12 to a lesser extent. PGA findings assessed at weeks 12, 16, 20, and 24 are presented in Table 30.

Table 14. Physician’s Global Assessment of Treatment Response at Week 4 (Dose per Leg) for Study 141 — ITT Population.

Table 14

Physician’s Global Assessment of Treatment Response at Week 4 (Dose per Leg) for Study 141 — ITT Population.

The subgroup analysis for the PGA assessed by previous exposure to BoNT treatment also showed that, in both treatment groups, aboBoNTA was statistically significantly more effective than placebo, regardless of prior exposure to BoNT (Table 31). No test for interaction by previous BoNT experience was conducted to determine if there was an interaction between the treatment effect and previous exposure to BoNT.

Observational Gait Scale

▬▬▬▬▬

▬▬▬▬▬

Gross Motor Function Measure

In Study 701, the primary efficacy variable was functional change, as assessed by the change from baseline in overall GMFM score without walking aids or orthoses at week 4 of the study. There were no statistically significant between-group differences in the overall GMFM score at week 4 (Table 34).

In Study 701, change from baseline in overall GMFM score at weeks 8 and 16, and change from baseline in GMFM goal-total score at weeks 4, 8, and 16, were secondary efficacy variables. Table 35 summarizes the data for all patients treated and demonstrates there were no statistically significant between-group differences for both outcomes at all time points assessed.

Leeds Videographic Gait Assessment

In Study 701, change in VGA scores at weeks 4 and 16 compared with baseline was a secondary efficacy variable. The results for the APT population demonstrated statistically significant between-group differences at week 16, but not at week 4. Significantly more treated legs in the aboBoNTA group demonstrated a valgus or varus deformity of the hindfoot at week 16 (Table 36). Finally, fewer patients in the Dysport Therapeutic group used walking aids. In Study 701, all outcomes were not controlled for type I error and the level of significance may be inflated; therefore, any statistically significant results should be interpreted with caution.

Subjective Functional Assessment of Gait

In Study 701, subjective functional assessment at weeks 4, 8, and 16 was a secondary efficacy variable. Although at weeks 4 and 8 more patients in the aboBoNTA group demonstrated a good response (defined as an observable change that was also of functional benefit), no statistically significant between-group differences were reported at either time point (Table 37).

Leeds Functional Mobility Questionnaire

▬▬▬▬▬

Caregiver Burden Scale

Caregiver burden was an outcome identified as important to patients, according to the patient group input received for this review. This outcome was not assessed in either included trial.

Duration of Effect (Re-Treatment Intervals)

▬▬▬▬▬

▬▬▬▬▬

Harms

Only those harms identified in the review protocol are reported subsequently (see 2.2.1, Protocol).

Adverse Events

▬▬▬▬▬

▬▬▬▬▬

The incidence of TEAEs by dose per leg in Study 701 is presented in Table 17. At least one TEAE was reported in 39% and 50% of patients in the aboBoNTA and placebo groups, respectively. The most common TEAEs were rhinitis (15% in the aboBoNTA and placebo treatment groups), bronchitis (15% and 12% in the aboBoNTA and placebo groups, respectively), pharyngitis (12% in the aboBoNTA and placebo treatment groups), and viral infection (8% and 15% in the aboBoNTA and placebo groups, respectively).

Table 17. Harms for Study 701 — APT Population.

Table 17

Harms for Study 701 — APT Population.

Serious Adverse Events

In Study 141, one SAE was reported in the aboBoNTA 10 U/kg treatment group, no SAEs were reported in the aboBoNTA 15 U/kg treatment group, and five SAEs were reported in four patients in the placebo group (Table 15). In the aboBoNTA 10 U/kg treatment group, the SAE was adenoidal hypertrophy while, in the placebo group, the SAEs were gastroenteritis, pneumonia, rotavirus infection, head injury, and upper-limb fracture. Pneumonia and rotavirus infection SAEs occurred in one patient in the placebo group. In Study 701, one SAE was reported in the aboBoNTA group, where the patient suffered an episode of acute bronchitis. The event lasted for two days, during which he was hospitalized for monitoring. The patient recovered without sequelae. No SAEs were reported in the placebo group (Table 16).

Table 15. Harms (Dose per Leg) for Study 141 — Safety Population.

Table 15

Harms (Dose per Leg) for Study 141 — Safety Population.

Table 16. Harms by Total Dose Received for Study 141 — Safety Population.

Table 16

Harms by Total Dose Received for Study 141 — Safety Population.

Withdrawal Due to Adverse Events

In Study 141, one patient in the placebo group was withdrawn from the study because of an adverse event. This patient had Pelizaeus–Merzbacher disease. The investigator considered this event to be unrelated to study treatment. No other WDAE was reported in Study 141 (Table 15).

In Study 701, there were no WDAEs in either treatment arm.

Mortality

No death was reported in either study.

Notable Harms

Generalized weakness, dysphagia, respiratory failure, seizure, and incontinence were identified as the notable harms of interest based on the review protocol. In Study 141, the number of patients experiencing muscular weakness were 2 (2.5%), 0 (0%), and 1 (1.3%) in the aboBoNTA 10 U/kg, aboBoNTA 15 U/kg, and placebo groups, respectively, and the number of patients experiencing epilepsy was 2 (2.5%), 3 (3.5%), and 0 (0%) in the aboBoNTA 10 U/kg, aboBoNTA 15 U/kg, and placebo groups, respectively. All of the patients who reported epilepsy had a history of epilepsy. All five cases were in the aboBoNTA treatment groups and were assessed by the investigator as unrelated to study treatment. One patient in the aboBoNTA 15 U/kg group experienced dysphagia, while fecal incontinence and incontinence were each experienced by one patient in the aboBoNTA 10 U/kg group. There were no reports of patients experiencing respiratory failure (Table 15).

In Study 701, two patients in the aboBoNTA treatment group experienced urinary incontinence; no other notable harms of interest were reported (Table 16).

Copyright © 2018 Canadian Agency for Drugs and Technologies in Health.

The copyright and other intellectual property rights in this document are owned by CADTH and its licensors. These rights are protected by the Canadian Copyright Act and other national and international laws and agreements. Users are permitted to make copies of this document for non-commercial purposes only, provided it is not modified when reproduced and appropriate credit is given to CADTH and its licensors.

Except where otherwise noted, this work is distributed under the terms of a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International licence (CC BY-NC-ND), a copy of which is available at http://creativecommons.org/licenses/by-nc-nd/4.0/

Bookshelf ID: NBK540264

Views

  • PubReader
  • Print View
  • Cite this Page
  • PDF version of this title (962K)
  • Disable Glossary Links

Other titles in this collection

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...