Summary Basis of Decision for Sohonos

Review decision

The Summary Basis of Decision explains why the product was approved for sale in Canada. The document includes regulatory, safety, effectiveness and quality (in terms of chemistry and manufacturing) considerations.


Product type:

Drug

Summary Basis of Decision (SBD) documents provide information related to the original authorization of a product. The SBD for Sohonos is located below.

Recent Activity for Sohonos

SBDs written for eligible drugs approved after September 1, 2012 will be updated to include post-authorization information. This information will be compiled in a Post-Authorization Activity Table (PAAT). The PAAT will include brief summaries of activities such as submissions for new uses of the product, and whether Health Canada's decisions were negative or positive. PAATs will be updated regularly with post-authorization activity throughout the product's life cycle.

The following table describes post-authorization activity for Sohonos, a product which contains the medicinal ingredient palovarotene. For more information on the type of information found in PAATs, please refer to the Frequently Asked Questions: Summary Basis of Decision (SBD) Project: Phase II and to the list of abbreviations that are found in PAATs.

For additional information about the drug submission process, refer to the AnchorAnchorManagement of Drug Submissions and Applications Guidance.

Updated: 2023-07-17

Drug Identification Number (DIN):

  • DIN 02524627 - 1.0 mg palovarotene, capsule, oral administration
  • DIN 02524635 - 1.5 mg palovarotene, capsule, oral administration
  • DIN 02524643 - 2.5 mg palovarotene, capsule, oral administration
  • DIN 02524651 - 5.0 mg palovarotene, capsule, oral administration
  • DIN 02524678 - 10.0 mg palovarotene, capsule, oral administration

Post-Authorization Activity Table (PAAT)

Activity/submission type, control number Date submitted Decision and date Summary of activities
Drug product (DINs 02524627, 02524635, 02524643, 02524651, 02524678) market notification Not applicable Date of first sale: 2022-06-20 The manufacturer notified Health Canada of the date of first sale pursuant to C.01.014.3 of the Food and Drug Regulations.
NDS # 252065 2021-04-23 Issued NOC 2022-01-21 NOC issued for New Drug Submission.
Summary Basis of Decision (SBD) for Sohonos

Date SBD issued: 2022-06-30

The following information relates to the New Drug Submission for Sohonos.

Palovarotene

Drug Identification Number (DIN):

  • DIN 02524627 - 1.0 mg palovarotene, capsule, oral administration
  • DIN 02524635 - 1.5 mg palovarotene, capsule, oral administration
  • DIN 02524643 - 2.5 mg palovarotene, capsule, oral administration
  • DIN 02524651 - 5.0 mg palovarotene, capsule, oral administration
  • DIN 02524678 - 10.0 mg palovarotene, capsule, oral administration

Ipsen Biopharmaceuticals Canada Inc.

New Drug Submission Control Number: 252065

On January 21, 2022, Health Canada issued a Notice of Compliance to Ipsen Biopharmaceuticals Canada Inc. for the drug product Sohonos.

The market authorization was based on quality (chemistry and manufacturing), non-clinical (pharmacology and toxicology), and clinical (pharmacology, safety, and efficacy) information submitted. Based on Health Canada’s review, the benefit-harm-uncertainty profile of Sohonos is favourable for reducing the formation of heterotopic ossification in adults and children aged 8 years and above for females and 10 years and above for males with fibrodysplasia (myositis) ossificans progressiva.

1 What was approved?

Sohonos, a retinoid/retinoic acid receptor gamma (RARγ) selective agonist, was authorized for the reduction of the formation of heterotopic ossification in adults and children aged 8 years and above for females and 10 years and above for males with fibrodysplasia (myositis) ossificans progressiva.

The safety and efficacy of Sohonos have been established in female pediatric patients 8 years of age and older, and male pediatric patients 10 years of age and older. Health Canada has therefore authorized an indication for pediatric use in these patient populations.

The safety and efficacy of Sohonos have not been established in females less than 8 years of age and males less than 10 years of age. Health Canada has not authorized an indication for pediatric use in these patient populations.

Clinical studies of Sohonos did not include sufficient numbers of patients 65 years of age and older to determine whether they respond differently than younger patients.

Sohonos (1 mg, 1.5 mg, 2.5 mg, 5 mg, and 10 mg palovarotene) is presented as a capsule. In addition to the medicinal ingredient, the capsule contains croscarmellose sodium, gelatin, lactose monohydrate, magnesium stearate, microcrystalline cellulose, pharmaceutical grade printing ink, povidone, sodium lauryl sulfate, and titanium dioxide.

The use of Sohonos is contraindicated in:

  • Patients who are pregnant or breastfeeding.
  • Patients of childbearing potential unless all the conditions for pregnancy prevention are met, or they are not at risk of pregnancy due to physical limitations as assessed by the physician.
  • Patients with a history of allergy or hypersensitivity to retinoids, or to any component of this product.

The drug product was approved for use under the conditions stated in its Product Monograph taking into consideration the potential risks associated with its administration. The Sohonos Product Monograph is available through the Drug Product Database.

For more information about the rationale for Health Canada's decision, refer to the Clinical, Non-clinical, and Quality (Chemistry and Manufacturing) Basis for Decision sections.

2 Why was Sohonos approved?

Health Canada considers that the benefit-harm-uncertainty profile of Sohonos is favourable for the reduction of the formation of heterotopic ossification in adults and children aged 8 years and above for females and 10 years and above for males with fibrodysplasia (myositis) ossificans progressiva.

Fibrodysplasia ossificans progressiva (FOP) is an ultra-rare congenital disease with approximately 800 known cases worldwide. It is characterized by uncontrolled, progressive, and abnormal growth of bone in non-skeletal tissues through a process known as heterotopic ossification (HO). Heterotopic ossification is cumulative throughout life, resulting in segments, sheets, or ribbons of extra bone developing throughout the body, progressively restricting movement. It presents within the first decade of life, typically beginning in the head, neck and shoulders, resulting in considerable deformity. The majority of patients with FOP are confined to a wheelchair in their twenties, and the average lifespan is 40 years. The most common cause of death in patients with FOP is cardiorespiratory failure or pneumonia secondary to thoracic insufficiency syndrome.

Heterotopic ossification is episodic, and is usually preceded by flare ups which can occur spontaneously or following trauma such as intramuscular injections or viral infections. The earliest indication of a flare up is edema at the site. The remaining nodule consolidates and hardens with fibrosis, and ultimately with ossification. On average, patients with FOP have two flare ups per year. At the time of authorization, there were no authorized treatments for FOP available in Canada. To date, the management of FOP has been supportive, focusing on the prevention of flare ups, education/counseling, and improvement of quality of life.

Evidence of the clinical efficacy of Sohonos was provided through data from the Phase II Studies 201 and 202, and the pivotal Phase III study, MOVE (Study 301). All three studies were conducted in patients with FOP. The Natural History Study (NHS) was an observational study of 114 non-treated patients with FOP. Many subjects from the NHS were rolled over into the pivotal and supportive clinical studies, while some remained in the NHS. Several dosing regimens were evaluated in the Phase II and III studies, and are described in the Clinical Efficacy section.

In the Phase II program, which included Studies 201 and 202, multiple dosing regimens were evaluated for efficacy with respect to reducing the formation of HO. Altogether, the results obtained from the Phase II studies demonstrated greater efficacy with higher-dose regimens compared to lower-dose regimens, confirmed the need for a longer duration of treatment beyond six weeks, and established the efficacy of the chronic/flare-up regimen in the treatment of FOP (described in the Clinical Efficacy section).

The pivotal Phase III study, MOVE (Study 301), provided the main evidence of the clinical efficacy of Sohonos. In total, 107 patients with FOP 4 years of age and older were treated according to the chronic/flare-up regimen with weight-based dose adjustments. Patients were treated for 24 months, followed by a 24-month extension period. The efficacy of this regimen in preventing new HO was evaluated by whole body computed tomography (CT) imaging (excluding the head), and compared to data from untreated patients from the NHS. All whole body CT images from the MOVE study and the NHS were read in a blinded manner.

The efficacy analysis was based on data from patients in the target population, which included female patients 8 years of age and older, and male patients 10 years of age and older. Several statistical analyses of the mean annualized new HO volume were conducted, which ultimately demonstrated the efficacy of Sohonos. Efficacy data from the weighted linear mixed effect (wLME) model, which accounts for the fitted weight of the different lengths of observed patients’ follow-up in the analysis of new HO, showed a 56% reduction (nominal p = 0.0292) in the overall population. The mean annualized new HO was 62% lower in patients treated with Sohonos compared to untreated patients in the overall population (nominal Wilcoxon rank-sum test p = 0.0006). Collectively, the results demonstrate the clinical efficacy of Sohonos in reducing the formation of HO in adults, in female children 8 years of age and older, and in male children 10 years of age and older with FOP.

The most common adverse reactions (reported at frequencies higher than 10%) in female patients 8 years of age and older and male patients 10 years of age and older are listed in the Clinical Safety section. The majority of adverse reactions were mild or moderate in severity across all clinical trials. Treatment-emergent adverse events (TEAEs) were generally similar between chronic and flare-up treatments. The incidence of dose reduction was higher during flare-up treatments (40%) than during chronic treatments (11.1%), which was attributed to higher incidences of mucocutaneous TEAEs.

Night blindness (nyctalopia) has been identified as a potentially dangerous effect associated with systemic retinoids, the class of drugs to which Sohonos belongs. This may be dose-dependent, and can make driving a vehicle at night hazardous during treatment. Night blindness is generally reversible after cessation of treatment, but can also persist in some cases. A single mild case of night blindness was identified during the Sohonos development program, which resolved after 134 days without treatment discontinuation or dose reduction.

A Serious Warnings and Precautions box in the Sohonos Product Monograph highlights the risk of premature physeal (growth plate) closure (PPC) in growing children with FOP, as well as the teratogenic potential of Sohonos. Due to the risk of PPC, Sohonos is not indicated in female children less than 8 years of age or male children less than 10 years of age and X-ray surveillance is recommended every 3 months. Additionally, Sohonos must not be used by patients who are pregnant or intend to become pregnant due to the risk of teratogenicity. To minimize the risk of fetal exposure, Sohonos is to be administered only if all conditions for pregnancy prevention are met.

Sohonos is started at different ages in females (8 years) and males (10 years) due to physiological gender differences in puberty and rapid bone growth. Female children enter puberty at a younger age than male children. Subgroup analysis based on the following age ranges were assessed: male children under 10 years of age and female children under 8 years of age (pre-pubertal), male children between the ages of 10 and 14 and female children between the ages of 8 and 14 (children at the onset of puberty to an age which would estimate a 90% skeletal maturity), children/adolescents between the ages of 14 and 18, and subjects over the age of 18.

Safety signals in the pediatric population prompted a temporary clinical hold on all subjects <14 years of age due to reports of PPC. The Data Safety Monitoring Board analysis revealed an unacceptably high PPC rate in the <8/10 age subgroup (56%) despite the efficacy results (93% based on the wLME analysis normalized volume of HO to subject weight). The benefit-risk profile for this age group was not favorable for Sohonos. The 8/10-14 age subgroup had a lower PPC rate (23%) and a high efficacy rate (62% based on the wLME analysis normalized volume of HO to subject weight), while the >14 and >18 groups both had low PPC rates (<1%) with good efficacy (61% and 19%, respectively).

A Risk Management Plan (RMP) for Sohonos was submitted by Ipsen Biopharmaceuticals Canada Inc. to Health Canada. The RMP is designed to describe known and potential safety issues, to present the monitoring scheme and when needed, to describe measures that will be put in place to minimize risks associated with the product. Upon review, the RMP was considered to be acceptable.

The submitted inner and outer labels, package insert and Patient Medication Information section of the Sohonos Product Monograph meet the necessary regulatory labelling, plain language and design element requirements.

The sponsor submitted a brand name assessment that included testing for look-alike sound-alike attributes. Upon review, the proposed name Sohonos was accepted.

Sohonos has an acceptable safety profile based on the non-clinical data and clinical studies. The identified safety issues can be managed through labelling and adequate monitoring. Appropriate warnings and precautions are in place in the Sohonos Product Monograph to address the identified safety concerns.

This New Drug Submission complies with the requirements of sections C.08.002 and C.08.005.1 and therefore Health Canada has issued the Notice of Compliance pursuant to section C.08.004 of the Food and Drug Regulations. For more information, refer to the Clinical, Non-clinical, and Quality (Chemistry and Manufacturing) Basis for Decision sections.

3 What steps led to the approval of Sohonos?

The New Drug Submission for Sohonos was subject to an expedited review process under the Priority Review Policy. The sponsor presented substantial evidence of clinical effectiveness to demonstrate that Sohonos provides effective treatment of a serious, life-threatening and severely debilitating condition for which no drug is presently marketed in Canada.

For additional information about the drug submission process, refer to the Management of Drug Submissions and Applications Guidance.

Submission Milestones: Sohonos

Submission Milestone Date
Pre-submission meeting 2019-07-30
Request for priority status filed 2021-03-15
Request for priority status approved by Director, Bureau of Medical Sciences 2021-04-15
New Drug Submission filed 2021-04-23
Screening  
Screening Deficiency Notice issued 2021-05-20
Response to Screening Deficiency Notice filed 2021-07-01
Screening Acceptance Letter issued 2021-07-26
Review  
Biostatistics evaluation completed 2021-12-23
Review of Risk Management Plan completed 2022-01-12
Quality evaluation completed 2022-01-12
Non-clinical evaluation completed 2022-01-14
Clinical/medical evaluation completed 2022-01-20
Labelling review completed 2022-01-20
Notice of Compliance issued by Director General, Therapeutic Products Directorate 2022-01-21
4 What follow-up measures will the company take?

Requirements for post-market commitments are outlined in the Food and Drugs Act and Regulations.

6 What other information is available about drugs?

Up-to-date information on drug products can be found at the following links:

7 What was the scientific rationale for Health Canada's decision?
7.1 Clinical basis for decision

Clinical Pharmacology

Fibrodysplasia ossificans progressiva (FOP) is a genetic condition caused by a gain-of-function mutation in the gene which encodes activin receptor type 1A/activin receptor-like kinase 2 (the ACVR1/ALK2 gene), a bone morphogenetic protein (BMP) type I receptor. The gain-of-function mutation in ALK2 aberrantly activates the BMP-Smad1/5/8 signalling pathway, which diverts normal soft tissue injury repair mechanisms away from tissue regeneration by promoting chondrogenesis and heterotopic bone formation.

Palovarotene, the medicinal ingredient in Sohonos, is an orally bioavailable retinoic acid receptor gamma (RARγ) selective agonist. Retinoic acid receptor gamma is expressed in chondrogenic cells and chondrocytes operating as an unliganded transcriptional repressor. Through binding to RARγ, palovarotene decreases BMP signaling and inhibits Smad1/5/8 signaling, which are deeply involved in the pathogenesis of myositis ossificans, and therefore in the pathogenesis of FOP. Palovarotene prevents chondrogenesis and ultimately heterotopic ossification (HO) by interfering with these pathways, which enables normal muscle tissue repair or regeneration to take place, thereby reducing damage to muscle tissue.

In vitro, palovarotene inhibited BMP-mediated Smad1/5 signalling in a human FOP fibroblast cell line carrying the gain-of-function ALK2 mutation, R206H. In animal models of FOP and injury-induced HO, palovarotene reduced new HO and maintained joint mobility by decreasing mast cell infiltration and fibroproliferative response at the site of injury.

The pharmacokinetics of palovarotene have been well characterized through single and multiple dose studies in healthy subjects as well as in patients with FOP. A study was conducted in which healthy adult subjects received 20 mg palovarotene once daily for 14 days after a standard breakfast. The mean elimination half-life of palovarotene was 8.7 hours, the median time to maximum concentration (Tmax) was 4.6 hours, the average maximum concentration (Cmax) was 140 ng/mL, and the average exposure as measured by the area under the concentration-time curve (AUC[0-τ]) was 942 ng*hr/mL. Little or no accumulation was detected following once-daily dosing. The mean steady-state trough plasma concentration was 3.5 ng/mL. Palovarotene was constrained to plasma, with 99.0% mean protein binding observed in vitro. The mean apparent volume of distribution at steady-state (Vss/F) was 319 L. Palovarotene was eliminated mainly through the feces, with minimal excretion in urine.

The results of an ascending single-dose study demonstrated that the pharmacokinetics of palovarotene was linear and dose-proportional in the range of 0.1 mg to 50 mg in fed, non-smoking subjects. The oral absorption of palovarotene increased when administered with food in healthy subjects. Therefore, palovarotene was administered with food in all studies conducted in patients with FOP, and the Product Monograph states that Sohonos should be taken with food. Plasma AUC and Cmax values were comparable when palovarotene was consumed either by swallowing the capsule whole or by sprinkling the contents of the capsule onto applesauce prior to consumption. Both methods of administration were evaluated following a high-fat, high-calorie breakfast.

Palovarotene is metabolized extensively by cytochrome P450 (CYP) 3A4, and to a lesser extent by CYP2C8 and CYP2C19. Five metabolites of palovarotene were observed: M1 (6,7-dihydroxy), M2 (6-hydroxy), M3 (7-hydroxy), M4a (6-oxo), and M4b (7-oxo). These metabolites reached steady-state by Day 4, with high variability in plasma levels. Palovarotene and its four known major metabolites (M2, M3, M4a, and M4b) were found to collectively represent 40% of the total exposure in plasma, based on data collected after the administration of [14C]-radiolabelled palovarotene. Results of an in vitro RARγ transactivation assay indicated that the M3 and M4b metabolites have 1.7% and 4.2% of the pharmacological activity of the parent drug, palovarotene.

The results of non-clinical drug interaction studies indicated that palovarotene had low potential to induce or inhibit major CYP enzymes and transporters. However, the results of a dedicated drug-drug interaction study showed that the co-administration of palovarotene with a CYP3A4 inhibitor (ketoconazole) significantly increased palovarotene exposure, and co-administration with a CYP3A4 inducer (rifampicin) significantly decreased palovarotene exposure. Based on these outcomes, the Product Monograph states that the co-administration of palovarotene with strong CYP3A4 inhibitors and inducers should be avoided. Palovarotene had no clinically significant effect on midazolam exposure (a CYP3A4 substrate), and was not a perpetrator or victim drug when co-administered with prednisone, a glucocorticoid used in the clinical management of FOP.

A randomized, double-blind, placebo- and positive-controlled, 4-way crossover electrocardiogram assessment study was conducted in which 31 healthy adult subjects received single 20 mg (therapeutic) and 50 mg (supratherapeutic) doses of palovarotene. No pharmacodynamic effects were observed on the corrected QT (QTc) interval, QRS duration, PR interval, or heart rate.

The results of a population pharmacokinetic analysis showed no evidence that age, sex, race, smoking status, or health status affected the pharmacokinetics of palovarotene. Body weight had a significant effect on the pharmacokinetics of palovarotene, as increasing exposure was observed with decreasing weight at the same dose. Based on these findings, weight-adjusted doses were used for skeletally immature children in the clinical studies in order to provide exposure levels similar to those in adolescents and adults receiving 5 mg to 20 mg doses. Therefore, weight-adjusted doses have also been recommended for children under 14 years of age in the Product Monograph.

No dedicated studies were conducted in subjects with renal or hepatic impairment. A population pharmacokinetic analysis showed no clinically significant differences in subjects with mild hepatic impairment, and in subjects with mild or moderate renal impairment. The effects of moderate to severe hepatic impairment and severe renal impairment on the pharmacokinetics of palovarotene have not been evaluated. Therefore, recommendations have been included in the Product Monograph to use caution in patients with moderate hepatic impairment, and to avoid the use of palovarotene in patients with severe hepatic or renal impairment.

For further details, please refer to the Sohonos Product Monograph, approved by Health Canada and available through the Drug Product Database.

Clinical Efficacy

The clinical efficacy of Sohonos was established based on results from the Phase II Studies 201 and 202, and the pivotal Phase III study, MOVE (Study 301). All three studies were conducted in patients with FOP.

Several dosing regimens were evaluated in these studies:

  • The 5/2.5 regimen (flare-up treatment): Patients received 5 mg Sohonos once daily for two weeks, followed by 2.5 mg once daily for four weeks.
  • The 10/5 regimen (flare-up treatment): Patients received 10 mg Sohonos once daily for two weeks, followed by 5 mg once daily for four weeks.
  • The 20/10 regimen (flare-up treatment): Patients received 20 mg Sohonos once daily for four weeks, followed by 10 mg once daily for eight weeks, for a total of 12 weeks, even if symptoms resolved earlier.
  • The c/20/10 regimen (chronic/flare-up regimen): Patients received 5 mg Sohonos once daily for up to 24 months. In the event of a flare-up or substantial high-risk traumatic event likely to lead to a flare-up, the dose was escalated to 20 mg once daily for four weeks, followed by 10 mg once daily for eight weeks. If needed, additional flare-up treatment was administered at a dose of 10 mg once daily and extended in four-week increments. Patients returned to a 5 mg dose once daily following resolution of the flare-up.

Data from a non-interventional natural history study (NHS) were also submitted, which included 114 untreated patients with FOP between 4 and 56 years of age.

Phase II program: Studies 201 and 202

Study 201

Study 201 involved 40 patients with FOP, who were randomized in a 3:3:2 ratio to receive treatment according to the 10/5 regimen, the 5/2.5 regimen, or placebo for six weeks. This study evaluated the ability of different doses of Sohonos to prevent HO at the flare-up site in patients with FOP. The six-week treatment period was followed by a six-week observation period. The primary endpoint was the proportion of responders, which was defined as patients who had no or minimal new HO at the flare-up site at Week 6. A 62% decline was observed in the incidence of new HO in patients treated with the 10/5 regimen. No reductions were observed in HO volume, incidence, or joint range of motion in patients treated with the 5/2.5 regimen. Although the results were not statistically significant for either regimen, a trend was observed in patients treated with the 10/5 regimen towards reduced volume, incidence of HO, and joint range of motion.

Study 202

Study 202 was an open-label study that evaluated the long-term safety and efficacy of different dosing regimens of Sohonos treatment in adult and pediatric patients with FOP.

Part A evaluated the efficacy of a six-week 10/5 regimen in pediatric and adult patients with FOP. Forty patients were enrolled, including 20 patients with 28 treated flare-ups, and 20 untreated subjects. Doses were adjusted based on weight. The primary endpoints and key secondary endpoints were the amount of reduction in each of the following: HO volume, frequency of flare-ups, and long-term post-exposure persistence of effect. Patients were monitored through measurements of the volume and incidence of HO and other clinical parameters, and through assessments of physical symptoms, responses to patient questionnaires, and suicidal ideation/mental health. For flare-ups with new HO, a slight improvement in HO volume was observed with the six-week 10/5 regimen, which was negated by an increase from 5,204 mm3 to 7,506 mm3 in the six weeks following withdrawal of Sohonos. These outcomes indicate that a higher dose and a longer flare-up regimen may be more efficacious.

Part B involved 54 patients with 79 treated flare-ups, and included pediatric and adult patients. Adult patients and pediatric patients with at least 90% skeletal maturity were treated according to the chronic/flare-up regimen. Pediatric patients with less than 90% skeletal maturity were treated based on the 20/10 regimen (treated for flare-ups without chronic dosing), and received weight-based dose adjustments.

Part C was ongoing at the time of authorization, and included 48 adult and pediatric patients receiving treatment according to the chronic/flare-up regimen, with weight-based dose adjustments. The data from Part C were integrated with the data from Part B. The primary endpoint was the annualized change in new HO volume. The incidence of flare-ups with new HO was 41.2% in patients treated with the 20/10 regimen, compared to 20.6% in flares treated with the chronic/flare-up (c/20/10) regimen. This demonstrated the effectiveness of chronic dosing. Additionally, the c/20/10 regimen was found to be more effective than the 10/5 regimen in reducing the incidence of HO, with incidences of 20.6% and 35.7% observed in patients treated with each of these regimens, respectively. The volume of HO was higher in patients treated with the 20/10 regimen than in patients treated with the 10/5 regimen, although the outcomes were not statistically significant. However, edema prior to the treatment of flare-ups, known to predict more severe HO episodes, was more prevalent in patients assigned to the 20/10 regimen, which could have skewed the results.

The results obtained from the Phase II studies demonstrated a persistence of HO formation after a six-week treatment regimen was completed. This suggested that the six-week 10/5 regimen was not optimal for all flare-ups. The greater efficacy of higher-dose regimens compared to lower-dose regimens, as well as the efficacy of the chronic/flare-up regimen in the treatment of FOP was demonstrated in Study 202 parts B and C.

Pivotal Study: MOVE (Study 301)

The clinical efficacy of Sohonos was evaluated in the pivotal, single-arm Phase III study, MOVE (Study 301), which enrolled 107 patients with FOP 4 years of age and older. Patients in this study were treated according to the chronic/flare-up regimen, with weight-based dose adjustments. The efficacy of this regimen in preventing new HO was assessed by low-dose, whole body computed tomography (CT) imaging (excluding the head), and compared to data from untreated patients from the NHS. The primary endpoint was the annualized change in new HO volume.

The duration of treatment was 24 months (Part A), followed by a 24-month extension (Part B). All whole body CT images from treated patients in the MOVE study and untreated patients from the NHS were read in a blinded manner. The R206H mutation was identified in 99 of the 107 patients in the MOVE study, and 8 patients had other FOP mutations. Ninety-seven of the 99 patients with the R206H mutation had at least one post-baseline HO volume measurement, and were included in the overall population analysis.

The efficacy analysis was based on data from patients in the overall population (which included all enrolled patients who had a baseline HO volume measurement and at least one post-baseline HO volume measurement) and in the target population (which included female patients 8 years of age and older, and male patients 10 years of age and older). Seventy-seven patients in this target population were treated with Sohonos and had a median age of 14 years (range: 8 to 61 years), and 76 patients were from the NHS (untreated) and had a median age of 18 years (range: 9 to 56 years).

Prespecified analyses of the mean annualized new HO volume were conducted using a Bayesian compound Poisson model with square root transformation. The outcome of these analyses demonstrated that the futility boundary had been crossed. Post hoc analyses were then performed, and the results revealed that using the square root transformation of the data in the Bayesian model moved the statistical conclusion from significant therapeutic benefit of Sohonos to showing futility. Additional analyses were conducted using the Bayesian and weighted linear mixed effect (wLME) models of annualized new HO volume, without square root transformation and including all raw data, which demonstrated the efficacy of Sohonos. Efficacy data from the wLME model, which accounts for the fitted weight of the different lengths of observed patients’ follow-up in the analysis of new HO, showed a 56% reduction (nominal p = 0.0292) in the overall population. The mean annualized new HO was 62% lower in patients treated with Sohonos compared to untreated patients in the overall population (nominal Wilcoxon rank-sum test p = 0.0006).

When the data from the group for patients between 8 (females) or 10 (males) and 14 years of age (defined as the ≥8/10-14 year age group in the Product Monograph) are combined with data from the patients in the >14 year age group (defined as the ≥8/10 year age group), the statistical significance and power are lost. In the ≥8/10 year age group, the mean volume reduction is 11,045 mm3, which represents a 57% reduction in the mean (nominal p = 0.0953). By comparison, there was a 61% reduction in new HO in the >14 year age group, with a reduction of 47% (nominal p<0.05) using a wLME analysis which normalizes for weight. The loss of statistical power is due to extreme variability in HO values in this very small cohort of patients.

The incidence of catastrophic HO was overall lower in patients treated with Sohonos, although one case in this group was severe, with an HO volume exceeding 230,000 mm3. The affected individual was in the ≥8/10-14 year age group and affected the statistical power of the data in the age-based subgroup analysis.

There were no statistically or clinically significant differences identified across treatment groups with regards to functional or patient reported outcomes across groups, including surveys (the FOP Physical Function Questionnaire [FOP-PFQ], the Patient-Reported Outcomes Measurement Information System [PROMIS] Global Health Scale, Cumulative Analogue Joint Involvement Scale [CAJIS] scores, suicide risk scores), range of motion at flare-up location, laboratory values, and clinical examinations.

Collectively, the results demonstrate the clinical efficacy of Sohonos in reducing the formation of HO in adults, in female children 8 years of age and older, and in male children 10 years of age and older with FOP.

Indication

Health Canada approved the following indication:

Sohonos (palovarotene capsules) is indicated to reduce the formation of heterotopic ossification in adults and children aged 8 years and above for females and 10 years and above for males with fibrodysplasia (myositis) ossificans progressiva.

For more information, refer to the Sohonos Product Monograph, approved by Health Canada and available through the Drug Product Database.

Clinical Safety

The most common adverse reactions (reported at frequencies higher than 10%) in female patients 8 years of age and older and male patients 10 years of age and older with FOP were dry skin (78%), pruritus (55%), alopecia (41%), rash (39%), erythema (32%), skin exfoliation (31%), skin reaction (24%), drug eruption (17%), skin irritation (12%), dry lip (55%), chapped lips (17%), dry mouth (13%), cheilitis (11%), nausea (11%), paronychia (14%), arthralgia (14%), dry eye (26%), skin abrasion (21%), epistaxis (12%), and headache (17%). The majority of adverse reactions were mild or moderate in severity across all clinical trials.

Treatment-emergent adverse events (TEAEs) were generally similar between chronic and flare-up treatments. The incidence of dose reduction was higher during flare-up treatments (40%) than during chronic treatments (11.1%), which was attributed to higher incidences of mucocutaneous TEAEs.

Night blindness (nyctalopia) has been identified as a potentially dangerous effect associated with systemic retinoids, the class of drugs to which Sohonos belongs. This may be dose-dependent, and can make driving a vehicle at night hazardous during treatment. Night blindness is generally reversible after cessation of treatment, but can also persist in some cases. A single mild case of night blindness was identified during the Sohonos development program, which resolved after 134 days without treatment discontinuation or dose reduction.

A Serious Warnings and Precautions box in the Sohonos Product Monograph highlights the risk of premature physeal (growth plate) closure (PPC) in growing children with FOP, as well as the teratogenic potential of Sohonos.

Premature physeal closure was identified as an important risk associated with Sohonos treatment in growing children. Initially, patients enrolled in the studies were 4 years of age and older. A partial clinical hold was implemented on patients less than 14 years of age following the detection of a high rate of PPC, which was later amended to a permanent hold on female patients less than 8 years of age and male patients below 10 years of age. Premature physeal closure was reported in 24 patients (24%) less than 18 years of age who were treated with Sohonos, which included 10 of 39 patients aged 8 (females) or 10 (males) to less than 14 years (26%), and 14 of 25 patients aged less than 8 (females) or 10 (males) years (56%). Overall, 8 patients had severe PPC, including 5 patients 7 years of age or younger. Therefore, Sohonos is not indicated in female children less than 8 years of age or male children less than 10 years of age. Additionally, close monitoring including X-ray surveillance, is recommended every 3 months.

Sohonos is started at different ages in females (8 years) and males (10 years) due to physiological gender differences in puberty and rapid bone growth. Female children enter puberty at a younger age than male children. Subgroup analysis based on the following age ranges were assessed: male children under 10 years of age and female children under 8 years of age (pre-pubertal), male children between the ages of 10 and 14 and female children between the ages of 8 and 14 (children at the onset of puberty to an age which would estimate a 90% skeletal maturity), children/adolescents between the ages of 14 and 18, and subjects over the age of 18.

Safety signals in the pediatric population prompted a temporary clinical hold on all subjects <14 years of age due to reports of PPC. The Data Safety Monitoring Board analysis revealed an unacceptably high PPC rate in the <8/10 age subgroup (56%) despite the efficacy results (93% based on the wLME analysis normalized volume of HO to subject weight). The benefit-risk profile for this age group was not favourable for Sohonos. The 8/10-14 age subgroup had a lower PPC rate (23%) and a high efficacy rate (62% based on the wLME analysis normalized volume of HO to subject weight), while the >14 and >18 groups both had low PPC rates (<1%) with good efficacy (61% and 19%, respectively).

The retinoid class of drugs, to which Sohonos belongs, is associated with birth defects in humans (teratogenicity). There have been no reports of pregnancy or in utero exposure to Sohonos reported in clinical studies. Sohonos must not be used by patients who are pregnant or intend to become pregnant due to the risk of teratogenicity. To minimize the risk of fetal exposure, Sohonos is to be administered only if all conditions for pregnancy prevention are met.

Health Canada has determined that appropriate risk management measures are in place to address the safety concerns identified for Sohonos, and to support its safe and effective use. Overall, the benefit-harm-uncertainty profile of Sohonos is favourable for the approved indication. For more information, refer to the Sohonos Product Monograph, approved by Health Canada and available through the Drug Product Database.

7.2 Non-Clinical Basis for Decision

The toxicity profile of palovarotene was evaluated in mice, rats, rabbits, and dogs in several non-clinical studies, including single-dose, chronic repeat-dose, reproductive toxicity, genotoxicity and phototoxicity studies.

The toxicity of palovarotene was evaluated in repeat-dose daily oral toxicity studies lasting up to 6 weeks in juvenile rats, up to 26 weeks in adult rats, up to 4 weeks in adult rabbits, and up to 39 weeks in adult dogs. The toxicity profile observed with repeated daily doses of palovarotene was similar to those of vitamin A and other retinoids. The dose-limiting toxicities of palovarotene were a subset of typical retinoid toxicities, primarily mucocutaneous toxicity in all non-clinical species and skeletal effects in rats.

In rats, the effects of palovarotene were similar regardless of age. A wide range of skeletal effects were observed in juvenile rats, juvenile mouse models of fibrodysplasia ossificans progressiva (FOP), and adult rats. These effects were consistent with the intended pharmacodynamic activity of palovarotene (to inhibit chondrogenesis, and thereby inhibit osteogenesis), and showed signs of reversing after dosing stopped in rats receiving 0.5 mg/kg/day, but not in rats receiving 1.2 mg/kg/day. In adult rats, chondrodystrophy was observed in the epiphyseal growth plate, which remains open into adulthood in this species. The incidence and intensity of chondrodystrophy was found to increase with dose level, but not with duration of dosing. Additionally, chondrodystrophy was reversible within 4 weeks in mild or moderate cases, but not in severe cases.

No microscopic findings were observed in any species which would indicate changes in osteoblast or osteoclast activity. This is consistent with the understanding that palovarotene-related effects target cartilage, rather than bone. These observations highlight the challenge of inhibiting pathological bone formation prior to skeletal maturation. Taking this risk into account, Sohonos is not indicated for female children less than 8 years of age and male children less than 10 years of age.

Signs of epithelial toxicity were detected in repeat-dose studies in rats, rabbits, and dogs, which are characteristic of changes induced by retinoic acid receptor gamma (RARγ) agonists. Skin lesions in the epidermis (characterized as erythema, edema, epithelial hyperplasia, hyperkeratosis, and/or hypergranulosis) were detected in all tested species given daily oral doses of palovarotene. In dogs, additional histopathologic findings included minimal hyperplasia, decrease in the absolute monocyte count, swollen ears, aural discharge, ocular discharge, and red conjunctivae.

No effects were observed on reproductive function, fertility, or early embryonic development when palovarotene was administered to female rats for two weeks prior to mating and up to Day 7 postcoitum at doses of 0.3 and 1.0 mg/kg/day. Prolonged periods of diestrous and a slightly lower ovulation rate were noted following doses of 3.0 mg/kg/day (high dose). Consequently, the 1.0 mg/kg/day dose, which is below the range of clinically relevant exposure, was considered as the no-observed-adverse-effect level (NOAEL).

Evidence of testicular degeneration, severe toxicity, and deaths were observed in rats after 4 weeks of dosing at 5.0 mg/kg/day, a level which exceeds the maximum tolerated dose (MTD). No testicular findings were observed at lower doses (0.04, 0.2, and 1.0 mg/kg/day) in this study. The relevance of these observations for human subjects remains unclear, as other retinoids have not been found to affect sperm count or motility at therapeutic dose levels. No effects were observed on reproductive function, fertility, or early embryonic development when palovarotene was administered to male rats for 9 weeks (beginning prior to cohabitation through two weeks after mating) at doses of 0.3 and 1.0 mg/kg/day. Consequently, the 1.0 mg/kg/day dose, which is below the range of clinically relevant exposure, was considered as the NOAEL.

In an embryo-fetal developmental study in rats, malformations and embryo-lethality were observed following the administration of palovarotene at doses below the human exposure at the chronic clinical dose. Signs of slight maternal toxicity (reduced maternal body weight gain and/or decreased food consumption) were observed when palovarotene was orally administered to pregnant rats at doses of 0.25 and 1.25 mg/kg/day from gestation days 6 to 17. Fetal malformations typical of retinoids (e.g., cleft palate, misshapen skull bones, and short long bones) were identified following daily oral administration of palovarotene to pregnant rats in the second and third trimester. At higher dose levels, these effects resulted in reduced fetal survival. The NOAEL for both maternal and fetal toxicity is considered to be 0.01 mg/kg/day, which is below the range of clinically relevant exposure. Warnings have been included in the labelling to highlight these risks, and women of childbearing potential are required to follow strict guidelines regarding contraception and pregnancy testing.

No significant behavioural or cardiovascular findings were detected in safety pharmacology studies conducted with clinically relevant doses of palovarotene. Additionally, palovarotene was highly protein-bound and did not partition to red blood cells in all species tested in vitro and in vivo. In non-clinical mass balance studies, palovarotene was moderately to well absorbed. Palovarotene was mainly eliminated in the feces, with minimal excretion in urine.

Palovarotene and its metabolites were not found to be mutagenic in Ames assays. Clastogenic effects were not observed in an in vivo mouse micronucleus study in which two doses of palovarotene up to 25 mg/kg each were administered on two consecutive days. The concentrations at which clastogenic activity was observed for palovarotene and its human metabolites were orders of magnitude higher than the anticipated human maximal serum concentration at the highest proposed clinical dose (138-202 ng/mL at 20 mg/day). Based on these results, palovarotene and its metabolites are not expected to be genotoxic to human subjects at the proposed clinical dose. Carcinogenicity studies have not been conducted with palovarotene. However, palovarotene is not expected to be carcinogenic based on the results indicating that it is not genotoxic, along with non-clinical chronic histopathology data and literature review.

The results of the non-clinical studies as well as the potential risks to humans have been included in the Sohonos Product Monograph. Considering the intended use of Sohonos, there are no pharmacological or toxicological issues within this submission which preclude authorization of the product.

For more information, refer to the Sohonos Product Monograph, approved by Health Canada and available through the Drug Product Database.

7.3 Quality Basis for Decision

The chemistry and manufacturing information submitted for Sohonos has demonstrated that the drug substance and drug product can be consistently manufactured to meet the approved specifications. Proper development and validation studies were conducted, and adequate controls are in place for the commercial processes. Changes to the manufacturing process and formulation made throughout the pharmaceutical development are considered acceptable upon review. Based on the stability data submitted, the proposed shelf life of 24 months is acceptable when the drug product is stored at room temperature (15 ºC to 30 ºC) and protected from light.

Proposed limits of drug-related impurities are considered adequately qualified (i.e. within International Council for Harmonisation [ICH] limits and/or qualified from toxicological studies).

All sites involved in production are compliant with good manufacturing practices.

None of the non-medicinal ingredients (excipients, described earlier) found in the drug product are prohibited by the Food and Drug Regulations.

The biologic raw materials used during manufacturing originate from sources with no or minimal risk of transmissible spongiform encephalopathy (TSE) agents or other human pathogens.