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: Voretigene Neparvovec (Luxturna): (Novartis Pharmaceuticals Canada Inc.): Indication: Vision loss, inherited retinal dystrophy [Internet]. Ottawa (ON): Canadian Agency for Drugs and Technologies in Health; 2021 Jan.

Cover of Clinical Review Report: Voretigene Neparvovec (Luxturna)

Clinical Review Report: Voretigene Neparvovec (Luxturna): (Novartis Pharmaceuticals Canada Inc.): Indication: Vision loss, inherited retinal dystrophy [Internet].

Show details

Executive Summary

An overview of the submission details for the drug under review is provided in Table 1.

Table 1. Submitted for Review.

Table 1

Submitted for Review.

Introduction

Inherited retinal dystrophy (IRD) consists of a broad group of genetic retinal disorders that are associated with progressive visual dysfunction. These conditions are caused by mutations in more than 260 different genes, including the retinal pigment epithelium 65 kDa protein (RPE65) gene.1 For patients with IRD caused by biallelic mutations in the RPE65 gene, common clinical diagnoses include Leber congenital amaurosis (LCA) and retinitis pigmentosa (RP).2 As mutations in a gene are more directly linked to the underlying molecular pathogenesis, it is now considered more appropriate to categorize this group of disorders by the individual disease-causing gene (genotype).3 Symptoms of LCA can become evident from as young as 2 to 3 months of age.2,4 There is progressive, profound reduction of visual acuity (VA), concentric reduction of visual fields (VFs), night blindness (nyctalopia), and nystagmus.2,46 Patients with a clinical diagnosis of RP tend to have a more variable onset and slower progression of disease than those with LCA. Symptoms of RP usually begin around 10 years of age and these patients tend to have better preservation of visual function compared with those with a clinical diagnosis of LCA.7,8 Visual impairment in individuals with RPE65-mediated IRD can present at a range of ages, from infancy to adolescence, with the unavoidable progression to complete blindness occurring as early as the preschool years or as late as the third decade of life.4,9 Due to the complexity of signs and symptoms, diagnosis based solely on clinical exam and electroretinogram is insufficient. A comprehensive assessment of medical and ocular history, clinical eye exams, optical coherence tomography (OCT) imaging, assessment of VFs, and an analysis of family history with genetic testing are essential to accurately diagnosing and classifying IRD.10,11 Genetic testing in IRD plays an important role in improving the accuracy of diagnosis and prognosis and is critical for guiding treatment decisions and identifying appropriate patients for clinical trials.10 The exact prevalence and incidence of RPE65-mediated IRD is uncertain. CADTH reviewers estimated that the number of individuals with RPE65-mediated IRD in Canada ranges from 129 to 378, as noted in the CADTH Pharmacoeconomic Review Report. The sponsor estimated that the number of individuals with RPE65-mediated IRD in Canada ranges from 42 to 277.12

In Canada, there are currently no pharmacological treatment options for patients with IRDs. The clinical experts consulted by CADTH for this review indicated that the standard of care for patients with IRD is supportive in nature and focuses on monitoring, psychological support, mobility training, and visual rehabilitation to maintain the patients’ ability to perform activities of daily living (ADL) and improve health-related quality of life (HRQoL).

The indication for voretigene neparvovec is for the treatment of adult and pediatric patients with vision loss due to IRD caused by confirmed biallelic RPE65 mutations and who have sufficient viable retinal cells. Disease-causing biallelic RPE65 mutations should be confirmed by an accredited laboratory using validated assay methods.13 Voretigene neparvovec is a 5 × 1012 vector genomes (vg) per millilitre concentrate for solution for subretinal injection. The dose of voretigene neparvovec for each eye is 1.5 × 1011 vg.

The objective of this report was to perform a systematic review of the beneficial and harmful effects of voretigene neparvovec administered as a single dose of 1.5 × 1011 vg in each eye via subretinal injection for the treatment of adult and pediatric patients with vision loss due to IRD caused by confirmed biallelic RPE65 mutations and who have sufficient viable retinal cells.

Stakeholder Engagement

The information in this section is a summary of input provided by the patient groups who responded to CADTH’s call for patient input and from clinical expert(s) consulted by CADTH for the purpose of this review.

Patient Input

One patient group submission was received for this review, which was authored jointly by Canada’s largest blindness organizations: Fighting Blindness Canada (FBC), the Canadian Council of the Blind (CCB), the CNIB Foundation, and Vision Loss Rehabilitation Canada (VLRC).

The submission is mainly oriented around a survey that was prepared in consultation with an independent researcher; the analysis was done by FBC and the submission was developed collaboratively by all of the submitting organizations.

Approximately 71% of the survey group respondents reported having RP, 4% reported having LCA, and the remaining reported a broader spectrum of IRDs. Seven individuals had the RPE65 mutation treated by voretigene neparvovec, which in each case was confirmed by genetic testing. Even though VA was maintained for some participants, 67% of the overall group were diagnosed as legally blind, over 70% had moderate-to-severe low vision, and 12% had near or total blindness.

Among the survey participants, 54% responded that IRD has negatively affected their employment or school status, with 22% rating the impact of their disease on their ability to perform job or school responsibilities as very severe. Survey participants reported struggling through school, requiring assistive technologies, or having to quit schooling as a result of the daily challenges being too difficult to overcome. Maintaining a stable career was flagged as being equally challenging.

Patients indicated that their eyesight interferes to some degree with most daily activities, including mobility and getting around, hobbies and leisure, socializing and interacting with others, looking after their appearance, reading a book or newspaper, and using a phone or iPad. Patients worry about their condition getting worse; struggle with challenges presented by daily activities, including parenting; experience long wait times for appointments; feel anxiety and uncertainty about the future and the impact of their diseases on their families; and, in some cases, are impacted by a lack of meaningful work, education, or social life.

Patients expressed a desire for a cure for the condition entirely, improved night vision and mobility at night, or facilitation of regular day-to-day activities such as social interactions, maintaining personal relationships, work, and study. Even if the treatments were to only enhance vision and mobility at night, most survey participants reported this would improve their overall quality of life. Many respondents also indicated that a stabilization of vision would be valuable, something that would at least halt progression if retinal damage cannot be reversed.

Clinician Input

The information in this section is based on input received from a panel of 6 clinical specialists consulted by CADTH for the purpose of this review.

Currently, in the absence of treatment for inherited retinal disease, the ultimate objective is to help each patient achieve their self-described life goals despite the limitation in their visual functioning. The most important outcome for patients is to maintain or improve their mobility, independence, and ability to achieve their life goals. The ideal treatment would be one that delays or stops disease progression and maintains the vision that patients had before they initiated therapy.

The clinical experts indicated there is currently no pharmacologic treatment available for the treatment of patients with vision loss due to IRD caused by confirmed biallelic RPE65 mutations. The current treatment is supportive care where treatment of patients with progressive and inevitable vision loss revolves mainly around counselling the patient. Given there is currently no pharmacologic treatment available, clinical experts anticipated that voretigene neparvovec would cause a shift in the current treatment paradigm.

Patients best suited for treatment with voretigene neparvovec are those who are at least 4 years of age with sufficient viable retinal cells. Patients who are younger than 4 years of age may benefit from treatment with voretigene neparvovec; however, the surgery and administration of treatment in that age group is difficult and the assessment of treatment response is influenced by the inability to measure and monitor visual function, as the testing is not designed for young children under 4 years of age.

The most vulnerable patients with RPE65 mutations appear to be those 10 years to 20 years of age, as patients in that age group lose considerable VF and start to lose their cone and rod functions. Therefore, it would be better to intervene before patients reach that age group, but age on its own should not be the only criteria for treating patients.

The clinical experts indicated that age on its own should not be the criterion for treating patients but rather the presence of sufficient viable retinal cells. The panellists also indicated that patients who might benefit from treatment should not be distinguished as having LCA or RP, as patients diagnosed with either LCA or RP caused by RPE65 mutations are both suited for treatment with voretigene neparvovec.

The clinical experts indicated that measuring viable retinal cells is not a straightforward procedure. The panellists also indicated that there is no benchmark or threshold of viable retinal cells that can be used to objectively define the sufficient viable retinal cells criterion. The clinical experts also indicated that a numerical cut-off could not be applied universally throughout all OCT technologies and generations. In clinical practice, the presence of sufficient viable retinal cells would be determined by the treating physician using OCT examinations to measure the area of remaining viable photoreceptors, which would be supplemented by VA and visual function tests.

The clinical experts indicated that it is expected that treatment response will wane over time; however, it is uncertain how long the treatment effect will last and when the treatment effect will start waning.

The clinical experts indicated that a medical geneticist would be required to complete the genetic testing, and that the genetic diagnosis, patient selection, and pre- and post-surgical evaluations (safety and efficacy) should be confirmed by an inherited retinal disease specialist, and that treatment with voretigene neparvovec should be initiated and administered by a retinal surgeon experienced in performing sub-macular injection and managing its complications.

Clinical Evidence

Pivotal Studies and Protocol Selected Studies

Description of Studies

Study 301 (N = 31) was a phase III, open-label, randomized controlled trial designed to evaluate the efficacy and safety of sequential subretinal injection of voretigene neparvovec into each eye in patients diagnosed with LCA due to RPE65 mutations. Randomization, which followed screening and confirmation of study eligibility, occurred in a 2:1 ratio of intervention (voretigene neparvovec) to control and used a block design stratified by age (≥ 10 years versus < 10 years) and mobility testing passing level (pass at ≥ 125 lux versus < 125 lux), as determined at screening. A total of 31 patients in 2 study sites in the US (the study enrolled international patients, including 1 patient from Canada) were randomized to either the treatment group (n = 21) or the control group (n = 10). Patients randomized to the voretigene neparvovec group received a dose of 1.5 × 1011 vg of voretigene neparvovec in each eye; the non-simultaneous subretinal injections were to occur within an 18-day period (12 days ± 6 days). Patients randomized to the control group did not receive voretigene neparvovec, sham injection, or corticosteroids for a period of at least 1 year from baseline evaluations. Following repeated retinal and visual function analysis, including mobility testing at 1 month, 3 months, 6 months, and 1 year, patients in the control group were crossed over to receive non-simultaneous injections of 1.5 × 1011 vg of voretigene neparvovec in each eye (within 18 days) after 1 year of randomization, provided they still met all study eligibility criteria. The primary end point was change in bilateral multi-luminance mobility testing (MLMT) performance at year 1 relative to baseline. Secondary end points were change in full-field sensitivity threshold (FST) averaged over both eyes at year 1 relative to baseline, change in assigned first eye MLMT performance at year 1 relative to baseline, and change in VA averaged over both eyes at year 1 relative to baseline.

In Study 301, there were more females (n = 18, 58%) than males (n = 13, 42%) and patients were primarily White (n = 21, 68%). At the time of randomization, the mean patient age was 15.1 (standard deviation [SD] = 10.9) years, with a range of 4 years to 44 years. Patients randomized to the voretigene neparvovec group were slightly younger (mean = 14.7 years; SD = 11.8 years) than those randomized to the control group (mean = 15.9 years; SD = 9.5 years). More patients with better baseline MLMT performance (< 125 lux) were enrolled in the voretigene neparvovec group (57%) than in the control group (40%).

Efficacy Results

Treatment with voretigene neparvovec resulted in statistically significant improvements in navigational ability in low-to-moderate light conditions (measured by MLMT score). The mean bilateral MLMT change score after 1 year was 1.8 (SD = 1.1) for the voretigene neparvovec group and 0.2 (SD = 1.0) for the control group. The difference in change from baseline in bilateral MLMT between the voretigene neparvovec and control treatment groups at 1 year was 1.6 (95% confidence interval [CI], 0.72 to 2.41; P = 0.001) which was statistically significant in favour of voretigene neparvovec; however, the difference between the treatment groups did not exceed 2 points. Eleven patients (52%) in the voretigene neparvovec group had an MLMT score change of 2 or more (the difference considered meaningful by the FDA and the European Medicines Agency [EMA]). In contrast, only 1 patient (10%) in the control group had a score change of 2 and none of the patients in the control group had a score change greater than 2. Although 62% of patients in the voretigene neparvovec group achieved a score of 6 on the MLMT (the maximum possible score) following administration of voretigene neparvovec, compared with no patients in the control group achieving a score of 6 on the MLMT, the observed mean increase in MLMT score of 1.8 observed in the voretigene neparvovec group could be an underestimate of the within-groups magnitude of the change due to the potential ceiling effect. Improvements in the MLMT score observed at 1 year seemed to be maintained until the 4-year follow-up.

Patients treated with voretigene neparvovec experienced a mean improvement in FST greater than 2 log units, whereas mean FST did not change in the control group; the mean change from baseline to year 1 was −2.08 (standard error [SE] = 0.29) log10(candela second per square metre [cd.s/m2]) for the voretigene neparvovec group and 0.04 (SE = 0.44) log10(cd.s/m2) for the control group. There were statistically significant improvements in full-field light sensitivity with voretigene neparvovec (mean difference versus control = −2.11 log units; 95% CI, −3.19 to −1.04; P < 0.001) at 1 year. This between-group difference exceeded the suggested clinical significance threshold of 10 decibels (dB) or 1 log unit for clinical significance.14 The improvements were sustained for 4 years after the second-eye injection, where for all patients who received treatment with voretigene neparvovec, the mean change from injection baseline at 4 years after the second-eye injection was −2.00 (SE = 1.35) log10(cd.s/m2). The clinical experts explained that the changes seen would be clinically meaningful in terms of improving visual function.

A longitudinal repeated measures analysis of VA using the Holladay scale averaged over both eyes showed a mean change from baseline to 1 year of −0.16 (SE = 0.07) logarithm of the minimum angle of resolution (LogMAR) for the voretigene neparvovec group and 0.01 (SE = 0.10) LogMAR for the control group, resulting in a mean treatment-effect difference of −0.16 LogMAR (95% CI, −0.41 to 0.08; P = 0.17), an 8-letter improvement. This difference was neither statistically significant nor clinically meaningful. Using the Lange scale for off-chart VA results, the mean between-group treatment difference was −0.16 LogMAR (95% CI, −0.31 to −0.01; nominal P = 0.035) corresponding to a 7.5-letter improvement on the eye chart for people who had voretigene neparvovec. All changes were smaller than the accepted clinically meaningful change (≥ 0.30 LogMAR). By year 3, little further change was seen in VA for either arm after treatment. The clinical experts consulted by CADTH for this review explained that even a small change would be important for patients. The clinical experts also noted that even if there were no improvement, preventing vision deterioration would be important for the patient’s quality of life.

For the patient-completed Visual Function Questionnaire (VFQ), the mean change from baseline to year 1 was 2.6 (SD = 1.8) for the voretigene neparvovec group and 0.1 (SD = 1.4) for the control group, for a mean between-group treatment difference of 2.4 (95% CI, 1.0 to 3.8; nominal P = 0.001). For the parent-completed surveys, the mean change from baseline to year 1 was 3.9 (SD = 1.9) for the voretigene neparvovec group and −0.2 (SD = 1.3) for the control group, for a mean between-group treatment difference of 4.0 (95% CI, 2.1 to 6.0; nominal P = 0.002). Although the VFQ assessed the patients’ ability to perform ADL for those patients who received voretigene neparvovec, the questionnaire did not contain any items to specifically assess HRQoL for patients. In addition, the VFQ used in Study 301 was not assessed psychometrically and, given the modifications made to the original National Eye Institute VFQ, the minimal important differences (MIDs) identified in the literature for that measure were not considered directly generalizable to the version used in Study 301.

Long-term data for patients in the voretigene neparvovec group suggest durable improvements in visual performance across multiple end points for at least 4 years following voretigene neparvovec administration. Similarly, after crossing over to voretigene neparvovec, patients in the control/voretigene neparvovec group exhibited visual performance improvements comparable to those observed in the voretigene neparvovec group, effects that were maintained through at least 3 years following bilateral voretigene neparvovec administration. Study 301 aims to follow patients for up to 15 years after treatment. The current duration of follow-up is limited to 5 years and longer-term efficacy and safety data for voretigene neparvovec are awaited. The duration of treatment effect is unclear. Also, given the small sample size (30 patients), there is uncertainty around the generalizability of the results observed for the long-term treatment effect. The clinical experts indicated that voretigene neparvovec would likely provide long-term benefits, although this was associated with substantial uncertainty.

Subgroup analyses of interest to this review were based on age, clinical phenotype, and number of viable retinal cells. No preplanned subgroup analyses were conducted in Study 301. The sponsor conducted a post hoc subgroup analysis by age (younger than 18 years at first injection versus 18 years or older at first injection); however, this analysis should be considered hypothesis-generating. Also, it is not clear if this subgroup analysis was conducted at the request of a regulator, and no rationale was provided for the age cut-off. The clinical experts consulted by CADTH indicated that greater clinical benefit from treatment is expected earlier in the condition when there are more viable retinal cells for the gene replacement therapy to restore. While patient age can be used as a proxy to estimate how advanced the condition is, due to the heterogeneous nature of RPE65 mutation–associated retinal dystrophy and the differences in the age of onset and disease progression, retinal cell viability should be assessed on an individual patient basis, regardless of patient’s age (as long as the patient is at least 4 years of age), to determine if the specific individual is likely to respond to voretigene neparvovec.

Harms Results

All patients in Study 301 experienced at least 1 treatment-emergent adverse event (TEAE). Most adverse events were mild in severity and no patient had any adverse events that led to study discontinuation or death.

The most frequently reported TEAEs in the voretigene neparvovec group were leukocytosis (in 45% of patients); vomiting (in 40% of patients); nasopharyngitis, headache, and pyrexia (in 35% of patients for each); oropharyngeal pain, cough, and nausea (in 30% of patients for each); increased intraocular pressure (in 20% of patients); and cataract and hematuria in 15% of patients for each.

Overall, 13 (65%) patients in the voretigene neparvovec group had at least 1 TEAE considered to be related to the study drug administration procedure. The TEAEs most often considered to be probably related to the administration procedure were cataract and increased intraocular pressure (n = 3 [15%] patients for each).

During the control period, 2 (10%) patients in the voretigene neparvovec group experienced 3 serious adverse events (SAEs) at time points distant from vector administration. One patient experienced a possible seizure requiring hospitalization and 1 patient experienced an adverse reaction to medications administered during oral surgery, which required hospitalization.

At the time of the data cut-off for the Clinical Study Report (CSR), which provided updated results of safety data through July 2, 2018, including follow-up for up to 5 years after the second injection for some patients, 6 SAEs occurred in 5 patients, including convulsion (1 event), adverse drug reactions (2 events) and retinal disorder (1 event of foveal thinning and loss of vision), retinal detachment (1 event), pneumonia (1 event), and menorrhagia (1 event).

One ocular SAE occurred in Study 301, where a patient with pre-existing atrophy of the retina who received voretigene neparvovec experienced a retinal disorder (foveal thinning and a loss of central vision) related to the subretinal injection of the treatment.

In the voretigene neparvovec group during the first year after the randomization period, 3 patients experienced a cataract, 2 patients experienced a retinal tear, and 1 patient developed an asymptomatic full-thickness macular hole. During the follow-up, 1 patient who was originally in the control group and crossed over to voretigene neparvovec experienced a retinal disorder (foveal thinning and a loss of central vision). Another patient experienced 1 SAE of retinal detachment.

Table 2. Summary of Key Results From Study 301.

Table 2

Summary of Key Results From Study 301.

Critical Appraisal

Study 301 used accepted methods to conceal allocation where a randomization list was generated by an independent biostatistician. However, because it was an open-label trial, patients were aware of the treatment allocation following randomization. Therefore, the evaluation of patient-reported outcomes (such as those measured by VFQ) and adverse events may be biased by treatment knowledge. The treatment effect on these subjective outcomes can potentially be overestimated as a consequence of the patient’s expectation of the efficacy of a new drug. Certain steps have been taken to ensure appropriate blinding of the assessment of the primary outcome measure (MLMT) and it seems there is potential for an unbiased outcome assessment with the MLMT, despite the open-label design. On the other hand, while the trial states that orientation and mobility assessors were masked, there is insufficient detail provided in the CSR to judge if adequate blinding of the outcome assessment was performed for all secondary outcome measures (e.g., VF and VA).

Imbalances in the baseline patient characteristics between the voretigene neparvovec and control groups included age and visual performance. It is unclear whether differences in baseline age between the voretigene neparvovec and control groups may introduce a risk of bias; on the one hand, the clinical experts indicated that age on its own should not be a criterion for treating patients but rather the presence of sufficient viable retinal cells. The imbalance at baseline in MLMT performance following assignment to the voretigene neparvovec or control group may bias the observed treatment effect estimate; however, the direction of the bias is unclear. As differences in MLMT, VA, and VF were noted at baseline in Study 301 — and as these outcomes represent the key clinical data — there is uncertainty associated with the effect estimate for these outcomes given that patients in the voretigene neparvovec treatment group had better MLMT scores at baseline (a larger percentage of patients with a baseline MLMT performance < 125 lux were enrolled in the voretigene neparvovec group [57%] than in the control group [40%]). Also, at baseline, patients randomized to the voretigene neparvovec group had better acuity, on average (less VA loss), and better VF than patients randomized to the control group. However, there is no evidence to indicate how visual performance at baseline could affect the treatment effect and, if bias exists, it is not possible to judge the direction of the bias.

Scores on the MLMT may underestimate the treatment effect of voretigene neparvovec due to the potential ceiling effect, where patients who passed the test at the second-lowest light level at baseline were able to achieve a maximum increase of only 1 unit. Although 62% of patients in the voretigene neparvovec group achieved the maximum possible increase following administration, compared with none in the control group, the observed mean increase in MLMT score of 1.8 may be an underestimate of the treatment effect because of this ceiling effect. The MLMT also potentially has a floor effect, where 1 patient in the voretigene neparvovec group did not pass the MLMT at the highest light level at baseline and also failed at year 1. It is worth noting that of the 5 patients who did not pass screening, 2 (40%) were not eligible based on mobility test performance, 1 was excluded due to ceiling effect, and 1 was excluded due to floor effect. In addition, it is not clear whether the 12 unique MLMT course configurations were of equivalent difficulty.

While patients in Study 301 were required to have a diagnosis of LCA due to RPE65 mutations to be enrolled in the pivotal trial of voretigene neparvovec, the clinical experts indicated the results reported for these patients should be generalizable to patients with RP as long as they have confirmed biallelic RPE65 mutations. Voretigene neparvovec is designed to deliver a normal copy of the RPE65 gene to cells of the retina in patients with a reduced level of or no biologically active RPE65; it is therefore intended to treat the underlying mechanism of the disease, which is the same in patients with confirmed RPE65 mutations regardless of clinical phenotype.13

The clinical experts indicated that measuring viable retinal cells is not a straightforward procedure, and that the methods used in the pivotal trial to determine whether a patient has sufficient viable retinal cells do not give a complete picture of the health of the retina or the number of viable photoreceptors. They also indicated that the technology to assess the structure and function of the retina has evolved since the trial. The clinical experts indicated that OCT can measure the thickness of the retina; however, it may not inform the treating physician whether there are viable retinal cells.

Indirect Comparisons

No indirect comparisons were submitted by the sponsor or identified by CADTH.

Other Relevant Evidence

Description of Studies

The sponsor submitted 2 supportive trials, Study 101 and Study 102. Study 101 (N = 12) was a phase I, open-label, single-arm, dose-escalation study that assessed the safety and tolerability of 3 different doses of voretigene neparvovec administered via subretinal administration to 1 eye (first-treated eye) of patients with LCA due to RPE65 mutations. Study 101 also evaluated the clinical efficacy; however, no formal hypothesis testing was conducted. Study 102 was a follow-on to Study 101 in which patients received voretigene neparvovec treatment in the previously uninjected eye (second-treated eye). In Study 101, 12 patients, who were 8 years of age or older at the time of administration, received unilateral subretinal injection in the eye with the worse function (first-treated eye). Three doses of voretigene neparvovec were tested sequentially: 1.5 × 1010 vg, 4.8 × 1010 vg, and 1.5 × 1011 vg. The dose of voretigene neparvovec under review is 1.5 × 1011 vg. Study 102 was a follow-on study to Study 101. Eleven of the 12 treated patients in Study 101 received a subretinal injection in the contralateral eye (second-treated eye) consisting of 1 dose of 1.5 × 1011 vg of voretigene neparvovec in a total volume of 300 μL.

Efficacy Results

All efficacy data are descriptive, and no inferences can be made because there were no hypotheses being tested.

In Study 101, 4 patients were considered non-evaluable, given the inconsistent use of patching as well as the variability of lighting conditions and test-course difficulty. Following vector administration, follow-up mobility testing using the injected eye indicated that 4 of the 8 evaluable patients were able to complete the mobility test at a light level that was at least 1 level darker than baseline.

In Study 102, monocular mobility testing was assessed for change from baseline for the eye injected in Study 102. Of the 11 patients who received voretigene neparvovec, 8 were considered evaluable for mobility testing, while three were not and, therefore, were not included for presentations of this parameter. All 8 evaluable patients whose eyes had been injected in Study 102 completed the mobility test 1 year after injection at a light level that was at least 1 level darker than baseline. Five of the 8 (63%) evaluable patients received the maximum attainable score. Mobility testing results continued to show that, through year 4, all 8 evaluable patients completed the mobility test at a light level that was at least 1 level darker than baseline.

For the eye injected in Study 101 at year 1, 4 of 7 (57%) evaluable patients showed a decrease of 10 dB or more in FST compared with baseline, with a change from baseline in all 7 evaluable patients ranging from −8.1 dB to −33.7 dB. At year 2, 3 of 7 (43%) evaluable patients showed a decrease of 10 dB or more in FST, with a change from baseline ranging from −5.7 dB to −29.1 dB in 6 of the 7 evaluable patients.

At the time of database lock for the CSR for Study 102 (May 19, 2019), the available FST data indicated that the light sensitivity of the eye injected in Study 102 increased after injection for 8 of 11 patients; this increase was greater than the 10 dB cut-off, was considered clinically important in 7 of 11 patients, and was below this cut-off for 1 patient. Light sensitivity remained stable in the other 3 patients.

In Study 101 at year 1, for the eye injected in that study, 7 of the 12 (58%) evaluated patients had a change in LogMAR score of 0.3 or more compared with baseline (corresponding to an improvement of at least 3 lines [15 letters] on the eye chart). At year 2, 4 of 11 (36%) patients had a change in LogMAR score of 0.3 or more compared with baseline. At year 3, 5 of 9 (56%) patients had a change in LogMAR score of 0.3 or more compared with baseline.

In the Study 102 LogMAR scores, based on Holladay scale for off-chart results, 1 (9%) of the 11 patients assessed had a change in LogMAR score of 0.3 or more compared with baseline 1 year after receiving treatment.

Harms Results

All patients in Study 101 and Study 102 experienced at least 1 TEAE.

One SAE was reported in Study 101, where 1 patient experienced an anal fistula that required hospitalization. The SAE was mild in severity and considered related to an underlying diagnosis of inflammatory bowel disease. The event was recovered and/or resolved with no sequelae.

One SAE was reported in Study 102, which was elevated intraocular pressure (grade 4) in the patient’s right eye resulting in hospitalization. The SAE, which was moderate in severity, was deemed related to the use of a depo-steroid injection for a known rare complication of vitrectomy (endophthalmitis).

No deaths or discontinuations from the study due to adverse events were reported in either study.

Critical Appraisal

Studies 101 and 102 are phase I, single-arm, open-label, non-randomized studies with a small sample size and are not considered to provide high-quality evidence to support the efficacy of voretigene neparvovec.

No conclusions can be drawn regarding the clinical efficacy of voretigene neparvovec. Study 101 was a dose-escalating study, and neither Study 101 nor Study 102 were designed or powered to assess the clinical efficacy of voretigene neparvovec. No formal hypothesis testing was conducted, and the within-group changes were not designed to be inferential. Only descriptive statistics were presented.

During the course of Study 101, the mobility test was further refined and standardized, which affected both the number of patients considered evaluable and the interpretation of the results.

The dosage of voretigene neparvovec used in 9 of the 12 patients enrolled in Study 101 was not the dosage approved by Health Canada; hence, the generalizability of the study results to the Canadian patient population is unclear.

Compared with Study 101, Study 301 included a broader population, including younger patients (4 years of age and older in Study 301 versus 8 years of age and older in Study 101) and patients with less advanced disease (VA no better than 20/60 in Study 301 versus VA no better than 20/160 in Study 101). The more stringent criteria introduced in Study 102 for determining the number of viable retinal cells (≥ 3 disc areas of retina without atrophy or pigmentary degeneration within the posterior pole) was also used in Study 301, whereas in Study 101, patients were eligible if they had one or more disc areas of retina that were not involved in complete retinal degeneration. Finally, periocular injection of the various corticosteroids used in Study 101 and Study 102 was discontinued in Study 301 to decrease the incidence and severity of elevated intraocular pressure and cataract formation or progression.

Conclusions

Currently, there is no pharmacologic treatment available for the treatment of patients with vision loss due to IRD caused by confirmed biallelic RPE65 mutations; the current standard of care is supportive in nature and focuses on monitoring, psychological support, mobility training, and visual rehabilitation to maintain the patients’ ability to perform ADL and improve HRQoL. Based on the results of 1 phase III study (Study 301), voretigene neparvovec, compared with the control group, demonstrated a statistically significant improvement in functional vision under dim light conditions, as measured by the MLMT one year post treatment. Voretigene neparvovec also resulted in a statistically significant improvement in FST one year post treatment. No improvement was observed in VA and the effect of voretigene neparvovec on HRQoL is unknown. The improvements observed with voretigene neparvovec after one year appear to be maintained for up to 4 years; however, further data are required and there is uncertainty about the duration of treatment effect. Harms, although present, were related to the surgical aspects of administration; the most common ocular adverse events related to the administration procedure were conjunctival hyperemia, cataract, increased intraocular pressure, retinal tear, dellen, macular hole, subretinal deposits, eye inflammation, eye irritation, eye pain, and maculopathy; 2 ocular SAEs had severe consequences.

Copyright © 2021 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: NBK569039

Views

  • PubReader
  • Print View
  • Cite this Page
  • PDF version of this title (2.2M)

Other titles in this collection

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...