Summary Basis of Decision for Movantik
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:
Summary Basis of Decision (SBD) documents provide information related to the original authorization of a product. The SBD for Movantik is located below.
Recent Activity for Movantik
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.
Post-Authorization Activity Table (PAAT) for Movantik
Updated:
The following table describes post-authorization activity for Movantik, a product which contains the medicinal ingredient naloxegol (as naloxegol oxalate). 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 Management of Drug Submissions Guidance.
Drug Identification Numbers (DINs):
- DIN 02442167 – 12.5 mg, naloxegol (as naloxegol oxalate), tablet, oral
- DIN 02442175 – 25 mg, naloxegol (as naloxegol oxalate), tablet, oral
Post-Authorization Activity Table (PAAT)
Activity/submission type, control number | Date submitted | Decision and date | Summary of activities |
---|---|---|---|
NC # 220843 | 2018-10-09 | Issued NOL2018-11-23 | Submission filed as a Level II (90 day) Notifiable Change (Risk Management Change) to update the PM with new safety information. As a result of the NC, modifications were made to the Adverse Reactions section of the PM, and corresponding changes were made to the PM Part III: Patient Medication Information. The submission was reviewed and considered acceptable, and an NOL was issued. |
SNDS # 220923 | 2018-10-12 | Cancellation Letter Received2018-10-15 | Submission filed as a Level I - Supplement to update the PM. The sponsor cancelled the submission administratively. |
SNDS # 218842 | 2018-08-02 | Issued NOC2018-10-12 | Submission filed as a Level I - Supplement to change the packaging components, and update the inner and outer labels. The information was reviewed and considered acceptable. An NOC was issued. |
Drug product (DINs 02442167 and 02442175) market notification | Not applicable | Date of first sale:2018-09-28 | The manufacturer notified Health Canada of the date of first sale pursuant to C.01.014.3 of the Food and Drug Regulations. |
NDS # 206398 | 2017-06-15 | Issued NOC2017-07-20 | Submission filed to transfer ownership of the product (that is [i.e.] drug sponsor name) from AstraZeneca Canada Inc. to Knight Therapeutics. An NOC was issued. |
SNDS # 201180 | 2016-12-14 | Issued NOC2017-06-20 | Submission filed as a Level I - Supplement to add an alternate manufacturing site for the production of the drug substance. There were no changes to the approved drug substance specifications. The information was reviewed and considered acceptable. An NOC was issued. |
SNDS # 201199 | 2016-12-15 | Cancellation Letter Received2017-02-23 | Submission filed as a Level I - Supplement to change the packaging components. The sponsor cancelled the SNDS. |
NC # 198089 | 2016-08-31 | Issued No Objection Letter2016-12-06 | Submission filed as a Level II (90 day) Notifiable Change (Risk Management Change) to update the Product Monograph (PM). As a result of the Notifiable Change, modifications were made to the Warnings and Precautions, Adverse Reactions, Drug Interactions, and Dosage and Administration sections of the PM, and corresponding changes were made to the PM Part III: Consumer Information. The submission was reviewed and considered acceptable, and a No Objection Letter was issued. |
SNDS # 193483 | 2016-03-21 | Issued NOC2016-11-08 | Submission filed as a Level I - Supplement for a new alternate mode of administration for the Movantik tablet for patients who are unable to swallow the tablets whole: crushing the tablet and mixing it in water, then direct ingestion or ingestion via nasogastric tube. The clinical benefits and risks related to the use of the tablet with the new mode of administration are expected to be comparable to those when given as a whole tablet. The data were reviewed and considered acceptable, and an NOC was issued. |
NC # 188715 | 2015-10-20 | Issued No Objection Letter2016-02-16 | Submission filed as a Level II (120 day) Notifiable Change to add a literature reference to the References section of the Product Monograph. The submission was reviewed and considered acceptable, and a No Objection Letter was issued. |
Drug product (DINs 02442167 and 02442175) market notification | Not applicable | Date of first sale:2015-08-27 | The manufacturer notified Health Canada of the date of first sale pursuant to C.01.014.3 of the Food and Drug Regulations. |
NDS # 167790 | 2013-08-30 | Issued NOC2015-06-02 | Notice of Compliance issued for New Drug Submission. |
Summary Basis of Decision (SBD) for Movantik
Date SBD issued: 2015-08-13
The following information relates to the new drug submission for Movantik.
Naloxegol (supplied as naloxegol oxalate), 12.5 mg and 25 mg, tablets, oral
Drug Identification Number (DIN):
- DIN 02442167 - 12.5 mg, tablet
- DIN 02442175 - 25 mg, tablet
AstraZeneca Canada Inc.
New Drug Submission Control Number: 167790
On June 2, 2015, Health Canada issued a Notice of Compliance to AstraZeneca Canada Inc. for the drug product Movantik.
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/risk profile of Movantik is favourable for the treatment of opioid-induced constipation (OIC) in adult patients with non-cancer pain who have had an inadequate response to laxative(s).
1 What was approved?
Movantik, a μ-opioid receptor antagonist, was authorized for the treatment of opioid-induced constipation (OIC) in adult patients with non-cancer pain who have had an inadequate response to laxative(s).
Movantik has not been studied in the pediatric (<18 years of age) population.
Movantik is contraindicated in:
- Patients who are hypersensitive to this drug or to any ingredient in the formulation or component of the container, or to any other opioid antagonist.
- Patients with known or suspected gastrointestinal obstruction or in patients at increased risk of recurrent obstruction, due to the potential for gastrointestinal perforation.
- Patients concomitantly receiving strong CYP3A4 inhibitors [for example (e.g.), ketoconazole, voriconazole, clarithromycin, and protease inhibitors such as ritonavir], due to significant increase in exposure to naloxegol.
Movantik was approved for use under the conditions stated in the Movantik Product Monograph taking into consideration the potential risks associated with the administration of this drug product.
Movantik (12.5 mg and 25 mg, naloxegol as naloxegol oxalate) is presented as a tablet. In addition to the medicinal ingredient, the tablet contains croscarmellose sodium, hypromellose, iron oxide black, iron oxide red, macrogol 400, magnesium stearate, mannitol, microcrystalline cellulose, propyl gallate, and titanium dioxide.
For more information, refer to the Clinical, Non-Clinical, and Quality (Chemistry and Manufacturing) Basis for Decision sections.
Additional information may be found in the Movantik Product Monograph, approved by Health Canada and available through the Drug Product Database.
2 Why was Movantik approved?
Health Canada considers that the benefit/risk profile of Movantik is favourable for the treatment of opioid-induced constipation (OIC) in adult patients with non-cancer pain who have had an inadequate response to laxative(s).
The binding of opioids to opioid receptors within the enteric nervous system causes physiological effects in the gastrointestinal tract including decreased motility, decreased secretions and increased absorption of fluid from intestines. These effects may cause constipation in 40-90% of individuals who take opioids.
While prescription [for example (e.g.), lactulose] or over-the-counter laxatives (e.g. senna and bisacodyl) are commonly used to treat OIC in clinical practice, they do not specifically target the opioid mediated mechanisms that cause constipation, and may fail to provide adequate treatment for many patients. Other available treatment options include methylnaltrexone (a subcutaneous injection for patients with advanced medical illness, available in the United States, the European Union, and Canada), and lubiprostone (approved in the United States and the European Union but not in Canada as an oral treatment for OIC in adult patients with chronic non-cancer pain).
The market authorization for Movantik was based on data from two 12-week, multicentre, double-blind, randomized, placebo-controlled, parallel-group studies in patients with OIC treated with opioids for at least 4 weeks for non-cancer pain. Efficacy for Movantik was not demonstrated in the overall study population from these two studies, therefore, only efficacy data from a subgroup of patients who were laxative inadequate responders (LIR) were considered. The primary efficacy endpoint was the response over the 12-week treatment period, defined by ≥3 spontaneous bowel movements (SBMs) per week and a change from baseline of ≥1 SBM per week for at least 9 out of the 12 treatment weeks including 3 out of the last 4 treatment weeks.
The results showed that in the pooled pivotal studies, the proportion of responders [that is (i.e.) patients who experienced ≥3 SBMs per week and a change from baseline of ≥1 SBM per week] in the LIR subgroup was 47.7% for those treated with Movantik 25 mg, and 30.1% for those treated with the placebo, for a statistically significant difference of 17.6%. In applying the same analysis to the more stringent and relevant endpoint of complete spontaneous bowel movement (CSBM), the proportion of responders was 20.7% for those treated with Movantik 25 mg, and 9.6% for those treated with the placebo, for a difference of 11.1%. Given the relatively stringent nature of the primary efficacy endpoint which required a sustained response in 9 out of the 12 weeks (including 3 out of the last 4 weeks), the difference in rates of responders based on SBM (17.6%) or on CSBM (11.1%) was moderate, but considered to be of clinical relevance as a second line therapy in LIR patients.
The clinical safety data from the pooled overall study populations showed a higher rate of adverse events with Movantik 25 mg (64%) as compared to placebo (51%), mainly driven by abdominal pain, diarrhea and nausea. An imbalance was reported regarding pain (abdominal, pain in extremity) and hyperhidrosis, which were more frequently reported with Movantik as compared to placebo. These may indicate some degree of opioid withdrawal syndrome, which was uncommonly reported.
A Risk Management Plan (RMP) for Movantik was submitted by AstraZeneca Canada Inc. to Health Canada. Upon review, the RMP was considered to be acceptable. 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.
Overall, the benefits of Movantik therapy are considered to outweigh the risks. Movantik has an acceptable safety profile based on the non-clinical data and clinical studies. The identified safety issues can be managed through labelling. Appropriate warnings and precautions are in place in the Movantik 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 granted 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 Movantik?
A pre-submission meeting for the current New Drug Submission (NDS) was held on April 11, 2013. The NDS was subsequently filed to Health Canada on August 30, 2013. In August 2014, a Notice of Non-Compliance (NON) was issued citing a lack of clear, clinically relevant efficacy in the study population of the two pivotal studies. The sponsor submitted a response to the NON in November 2014 and all concerns were satisfactorily addressed. After completing the review of the data package, a Notice of Compliance (NOC) was issued on June 2, 2015.
Submission Milestones: Movantik
Submission Milestone | Date |
---|---|
Pre-submission meeting: | 2013-04-11 |
Submission filed: | 2013-08-30 |
Screening 1 | |
Screening Acceptance Letter issued: | 2013-10-24 |
Review 1 | |
Biopharmaceutics Evaluation complete: | 2014-07-10 |
Quality Evaluation complete: | 2014-06-18 |
Notice of Non-Compliance (NON) issued by Director General (lack of clear, clinically relevant efficacy in study population of pivotal studies): | 2014-08-20 |
Response filed: | 2014-11-18 |
Screening 2 | |
Screening Acceptance Letter issued: | 2015-01-02 |
Review 2 | |
Clinical Evaluation complete: | 2015-06-01 |
Labelling Review complete: | 2015-05-29 |
Notice of Compliance (NOC) issued by Director General: | 2015-06-02 |
For additional information about the drug submission process, refer to the Management of Drug Submissions Guidance.
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:
- See MedEffect Canada for the latest advisories, warnings and recalls for marketed products.
- See the Notice of Compliance (NOC) Database for a listing of the authorization dates for all drugs that have been issued an NOC since 1994.
- See the Drug Product Database (DPD) for the most recent Product Monograph. The DPD contains product-specific information on drugs that have been approved for use in Canada and have been market notified (that is, the company has told Health Canada the product is being marketed).
- See the Notice of Compliance with Conditions (NOC/c)-related documents for the latest fact sheets and notices for products which were issued an NOC under the Notice of Compliance with Conditions (NOC/c) Guidance Document, if applicable. Clicking on a product name links to (as applicable) the Fact Sheet, Qualifying Notice, and Dear Health Care Professional Letter.
- See the Patent Register for patents associated with medicinal ingredients, if applicable.
- See the Register of Innovative Drugs for a list of drugs that are eligible for data protection under C.08.004.1 of the Food and Drug Regulations, if applicable.
7 What was the scientific rationale for Health Canada's decision?
7.1 Clinical basis for decision
Clinical Pharmacology
Movantik (naloxegol oxalate) is a peripherally acting μ-opioid receptor antagonist. Naloxegol is a pegylated derivative of the μ-opioid receptor antagonist naloxone and antagonizes opioid binding at the peripheral μ-opioid receptors. As a pegylated derivative of naloxone, the ability of naloxegol to cross the blood brain barrier is limited. Naloxegol acts primarily on the gut μ-opioid receptors and counteracts opioid-induced constipation (OIC) with limited impact on the opioid-mediated analgesic effects on the central nervous system (CNS) at the intended therapeutic doses.
The primary route of naloxegol elimination is via hepatic metabolism, with renal excretion playing a minimal role. In clinical studies, six metabolites were found in feces, urine, or plasma, none of which have been identified as unique or disproportionate human metabolites. The major plasma circulating species is naloxegol.
In vitro data indicate that naloxegol is a substrate for cytochrome P450 (CYP) 3A4 and that CYP3A is the major CYP enzyme responsible for the metabolism of naloxegol. Naloxegol is also a substrate of the P-glycoprotein (P-gp) transporter, which likely plays a significant role in limiting CNS exposure to naloxegol and in its disposition.
Drug-drug interaction studies established that concomitant use of strong CYP3A4 inhibitors lead to a significant increase in naloxegol plasma levels, which prompted the inclusion of a corresponding contraindication in the Product Monograph. Concomitant use of moderate and to a lesser extent weak CYP3A4 inhibitors also resulted in a significant increase in naloxegol plasma levels. A statement was included in the Warnings and Precautions section of the Product Monograph to not use Movantik with moderate inhibitors unless absolutely necessary and with careful risk-benefit considerations along with halving the daily dose to 12.5 mg. The use of weak CYP3A4 inhibitors also resulted in some increase in plasma levels and thus required a reduction of daily dosing to 12.5 mg. Finally, the use of strong CYP3A4 inducers resulted in a significant decrease of naloxegol exposure. This was also labelled accordingly such that concomitant use is discouraged.
A randomized, double-blind, placebo-controlled, open-label positive-controlled, 4-way single dose crossover electrocardiogram study in healthy subjects [number (n) = 51] revealed no effects at the therapeutic dose (25 mg), but a mild (approximately 4 ms) QTc prolongation effect was predicted on the basis of pharmacokinetic/pharmacodynamic modelling when strong CYP3A4 and/or P-gp inhibitors are co-administered.
For more information, refer to the Movantik Product Monograph, approved by Health Canada and available through the Drug Product Database.
Clinical Efficacy
The efficacy of Movantik (naloxegol oxalate) in the treatment of opioid-induced constipation (OIC) was assessed in two replicate, Phase III, multicentre, randomized, double-blind, placebo-controlled studies (KODIAC 4 and KODIAC 5). The studies were 12 weeks in duration and enrolled patients with OIC and non-cancer related pain. Patients were eligible to participate if they were taking a minimum of 30 morphine equivalent units of opioids per day for at least 4 weeks before enrolment and had self-reported OIC. Opioid-induced constipation was confirmed through a 2-week run-in period and defined as <3 spontaneous bowel movements (SBMs) per week on average with at least one or more symptoms of straining, hard/lumpy stools, and or sensation of incomplete evacuation/anorectal obstruction in at least 25% of bowel movements. Patients with possible clinically important disruption of the blood-brain barrier were excluded. Patients who had a QTcF >500 ms at screening, a recent history of myocardial infarction within six months before randomization, symptomatic congestive heart failure, or who had any other overt cardiovascular disease were also excluded from the clinical studies. In addition, patients with moderate or severe hepatic insufficiency were excluded.
The response to Movantik was evaluated over 12 weeks in patients who had an inadequate response to laxatives in the 2 weeks prior to enrolment. The laxative response status was determined at the screening visit using an investigator administered questionnaire (Stool Symptom Screener) about previous laxative use and constipation symptoms over the past 2 weeks. Patients categorized as laxative inadequate responder (LIR) reported using a minimum of one class of laxative for at least 4 days in the 14 days preceding the first study visit. In addition, LIR patients had at least one of the following OIC symptoms, described as moderate, severe, or very severe in intensity: incomplete bowel movements, hard stools, straining, or sensation of needing to pass a bowel movement but unable to do so.
Throughout the study, patients were prohibited from using laxatives with the exception of bisacodyl rescue laxative which was permitted if they had not had a bowel movement for 72 hours. An SBM was defined as a bowel movement without rescue laxative taken within the past 24 hours.
A total of 652 patients in KODIAC 4 and 700 patients in KODIAC 5 were randomized to receive 12.5 mg or 25 mg of Movantik or placebo once daily for 12 weeks. The mean age of the patients was 52 years, 62% were women, 79% were white, and 11% were 65 years of age or older. The mean daily opioid morphine equivalent dose was 138 mg/day and patients had been taking opioids for an average of 3.6 years for the current episode of pain. Back pain (57%) and arthritis (10%) were the most common reasons for pain. Laxative use within the 2 weeks prior to enrolment was reported by 71% of patients. Eighty-four percent (84%) of patients reported using laxatives within 6 months prior to enrolment. Patients were stratified based on their response to laxative use (LIR, Laxative Adequate Responders, and Laxative Unknown Responders) and were randomly assigned to one of the three treatment groups in a 1:1:1 ratio.
The primary efficacy endpoint was the response over the 12-week treatment period, defined by ≥3 SBMs per week and a change from baseline of ≥1 SBM per week for at least 9 out of the 12 treatment weeks including 3 out of the last 4 treatment weeks.
Issuance of a Notice of Non-Compliance and Response
The efficacy of Movantik was not supported in the overall study population by the efficacy data from the two pivotal studies submitted by the sponsor. The rate of response over 12 weeks was 42% in the Movantik-treatment arm versus (vs.) 29% in the placebo-treatment arm. This resulted in a statistically significant difference of 13% between groups based on data pooled from both studies; however, the magnitude of this difference was of unclear clinical relevance, especially given the nature of the efficacy endpoint that was chosen [that is (i.e.) SBM] and given the fact that the sponsor had pre-specified the studies to detect a significant difference of 25%. Furthermore, using the more appropriate, and internationally recommended clinical endpoint of complete spontaneous bowel movement (CSBM) for the definition of responders, the difference in responders' rates was even lower at only 6.2%. Such a treatment effect is considered to be of little clinical significance since 16 patients would have to be treated to expect one patient to benefit from Movantik i.e., number needed to treat (NNT) of 16.
Given the availability of other drugs in the peripherally acting μ-opioid receptor antagonist class for treating OIC, and given the minimal efficacy demonstrated by Movantik in the overall population, the risk-benefit profile of Movantik was not considered to be favourable based on the data submitted and a Notice of Non-Compliance (NON) was issued to the sponsor on August 20, 2014 citing a lack of clear, clinically relevant efficacy in the study population of the two pivotal studies.
In the response to the NON, the sponsor proposed to restrict the indication to the pre-specified sub-group of LIR patients based on the 14-day period prior to study treatment. The screening tool used to determine the status of response to laxative was not adequately validated for this purpose. However, in response to the NON, the sponsor showed that LIR and non-LIR patients were most likely different populations and the risk of misclassification of LIR as non-LIR was remote. In a clinical practice setting, LIR patients would likely be an important target population in whom Movantik will be prescribed (i.e., those who have failed common laxative treatments). Therefore, after careful consideration, it was concluded that restricting the indication to the LIR subgroup would be acceptable.
Overall Efficacy Analysis
The primary efficacy endpoint was the response over the 12-week treatment period, defined by ≥3 SBMs per week and a change from baseline of ≥1 SBM per week for at least 9 out of the 12 treatment weeks including 3 out of the last 4 treatment weeks. When taking into consideration only the LIR patients in the pooled pivotal studies (Movantik 25 mg, n = 241, and placebo, n = 239), the results for the primary efficacy endpoint using SBM showed that the proportion of responders was 48% with Movantik 25 mg, and 30% with the placebo, for a statistically significant difference of 18%. This difference in rates corresponds to a NNT of 5.7 patients.
Using pooled responder rates from KODIAC 4 and KODIAC 5 based on the more stringent CSBM, the proportion of responders was 21% with Movantik 25 mg, and 10% with the placebo, for a difference of 11%. Given the relatively stringent nature of the primary efficacy endpoint which required a sustained response for 9 out of the 12 weeks (including 3 out of the last 4 weeks), the difference in rates of responders based on SBM (18%) or on CSBM (11%) was moderate but was considered to be of clinical relevance as a second line therapy in LIR patients.
For more information, refer to the Movantik Product Monograph, approved by Health Canada and available through the Drug Product Database.
Clinical Safety
There are two main known potential safety issues with opioid antagonists:
- The possible inhibition of the centrally mediated effects of opioids drugs thus reversing the analgesic effect of opioids, and/or
- Eliciting an opioid withdrawal syndrome (OWS) which can lead to serious safety outcomes.
The safety profile of Movantik was based primarily on data from the overall study population of the two 12-week pivotal placebo-controlled Phase III studies described in the Clinical Efficacy section. In these studies, a combined total of 441 patients received Movantik 12.5 mg and 446 patients received Movantik 25 mg.
The clinical safety data showed a higher rate of adverse events (AEs) with Movantik 25 mg (64%) as compared to Movantik 12.5 mg (52%) and placebo (51%). This was mainly driven by abdominal pain (reported in 16% of patients receiving Movantik 25 mg), diarrhea (reported in 9% of patients receiving Movantik 25 mg), and nausea (reported in 8% of patients receiving Movantik 25 mg). Back pain, pain in extremity, and hyperhidrosis were more frequently reported with Movantik 25 mg (4.3%, 2.2%, 2.9%, respectively) as compared to placebo (2.0%, 0.7%, 0.2%, respectively). This may indicate some degree of opioid withdrawal syndrome, however, opioid withdrawal syndrome as an adverse event (AE) was uncommonly reported (≤1%).
Serious adverse events (SAEs) were reported in 3.4% (n = 15) of patients administered Movantik 25 mg, 5.7% (n = 25) of patients receiving Movantik 12.5 mg, and 5.2% (n = 23) of patients administered placebo. The most frequently reported SAEs were infections (n = 12) followed by neurological (n = 6) and gastrointestinal (n = 6) events across treatment groups. Overall, no clear significant trend or safety signals emerged for SAEs, except possibly for few cases of changes in blood pressure. Two cases of severe hypertension (malignant or accelerated) were reported in the Movantik 25 mg treatment group. One case of hypertension and one case of hypotension were reported in the Movantik 12.5 mg treatment group. No cases were reported in the placebo treatment group. Due to the potential for gastrointestinal perforation, Movantik is contraindicated in patients with (or at increased risk of) gastrointestinal obstruction.
Treatment-related AEs (as judged by the investigator) were also reported more frequently with Movantik 25 mg (32.5%) as compared to Movantik 12.5 mg (20.6%) and placebo (16.2%). This difference was mainly due to more treatment-related events of the gastrointestinal tract in the Movantik 25 mg group. The proportion of patients who discontinued due to AEs in KODIAC 4 and KODIAC 5 was higher in the Movantik 25 mg group (10.3%) than in the Movantik 12.5 mg (4.8%) and placebo (5.4%) groups, driven predominantly by differences in the incidence of diarrhea and abdominal pain.
The pooled data from studies KODIAC 4, KODIAC 5, and KODIAC 7 (supportive extension study) suggested a possible small imbalance in the rates of AEs related to blood pressure changes, such as increased blood pressure (2.9% with Movantik 25 mg vs. 2.3% with Movantik 12.5 mg vs. 1.1% with placebo), decreased blood pressure (1.3% vs. 0.5% vs. 0.7%), and syncope (0.4% vs. 0.5% vs. 0%). These findings were not replicated in the 52-week study, except possibly for syncope, and therefore, these few cardiovascular findings are not considered to be a clinically significant issue at this time. Patients who had a QTcF >500 ms at screening, a recent history of myocardial infarction within six months before randomization, symptomatic congestive heart failure, or who had any other overt cardiovascular disease were excluded from the clinical pivotal studies. Therefore, as a mitigating measure; a warning was included in the Product Monograph to limit the use of Movantik in patients with pre-existing cardiovascular conditions.
The sponsor did not report any significant differences in the safety profile of Movantik in the LIR population as compared to the overall study population.
As per the Canadian Risk Management Plan, the sponsor was requested to provide results of any incoming post-market safety data, especially, but not limited to, data from a planned cardiovascular safety study required by the United States Food and Drug Administration (FDA) as a post-market commitment.
Supportive Studies
One additional double-blind, placebo-controlled, randomized, 12-week safety extension study (KODIAC 7) included 291 patients who were rolled over from the KODIAC 4 study (97 treated with Movantik 25 mg, 94 with Movantik 12.5 mg, and 100 with placebo). Overall 188 patients were exposed to >24-weeks of treatment (63 patients with Movantik 25 mg, 56 with Movantik 12.5 mg, and 63 with placebo groups).
In addition, KODIAC 8 was a Phase III, 52-week, multicentre, open-label, randomized parallel group, safety and tolerability study of Movantik 25 mg taken daily versus usual care (flexible laxative treatment regimen determined by the investigator according to his/her best clinical judgement) in the treatment of OIC in patients with non-cancer-related pain. The overall population was comparable to that enrolled in the pivotal studies. A total of 804 patients were included in the safety set (534 treated with Movantik 25 mg), and 270 treated with ‘usual care' at the discretion of the investigator.
Frequencies and patterns of AEs in the 12-week blinded safety extension study (KODIAC 7) and in the 52-week long-term open-label safety study (KODIAC 8) were generally consistent with those in the pivotal studies, as were the patients' reasons for requiring opioid medication.
For the KODIAC 8 Study, the incidence and pattern of discontinuations due to AEs in the Movantik 25 mg group was comparable to those seen in the pivotal studies.
For more information, refer to the Movantik Product Monograph, approved by Health Canada and available through the Drug Product Database.
7.2 Non-Clinical Basis for Decision
The non-clinical profile of naloxegol was established in a comprehensive investigational program that included studies of in vitro and in vivo pharmacodynamics, safety pharmacology, pharmacokinetics, toxicology, genotoxicity, carcinogenicity and reproductive and developmental toxicity.
In vitro non-clinical drug-drug interaction studies indicated that naloxegol is a substrate for cytochrome P450 (CYP) 3A4 and P-glycoprotein (P-gp), thus co-administration of naloxegol with drugs modulating CYP3A4 and P-gp may result in overexposure or under exposure to naloxegol.
Severe renal impairment increased the plasma levels of naloxegol, therefore, adjustment of doses should be considered; in patients with moderate renal impairment there was some increase in plasma levels that may necessitate some dose adjustment.
Although naloxegol is unlikely teratogenic at the proposed clinical dose levels, naloxegol was shown in non-clinical studies to pass the placental barrier, and it is also excreted in the milk of rats. In addition, there was a lack of studies conducted in juvenile animals. As a result, the use of naloxegol in pregnant and nursing mothers and juveniles should be avoided.
A cardiovascular telemetry study in dogs showed small, transient decreases in blood pressure, left ventricular systolic pressure, cardiac contractility and relaxation indices, as well as increases in heart rate, at supra-therapeutic doses; however, a thorough QT clinical study in healthy human male subjects revealed that naloxegol did not have an effect on QTc interval at the 25 mg therapeutic dose.
For more information, refer to the Movantik 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 Movantik 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 expiration period of 24 months is acceptable.
Proposed limits of drug-related impurities are considered adequately qualified [that is (i.e.) within International Conference on Harmonisation (ICH) limits and/or qualified from toxicological studies].
All sites involved in production are compliant with Good Manufacturing Practices.
All non-medicinal ingredients (described earlier) found in the drug product are acceptable for use in drugs according to the Food and Drug Regulations.
None of the excipients used in the manufacture of Movantik are of human or animal origin.
Related Drug Products
Product name | DIN | Company name | Active ingredient(s) & strength |
---|---|---|---|
MOVANTIK | 02442167 | KNIGHT THERAPEUTICS INC. | NALOXEGOL (NALOXEGOL OXALATE) 12.5 MG |
MOVANTIK | 02442175 | KNIGHT THERAPEUTICS INC. | NALOXEGOL (NALOXEGOL OXALATE) 25 MG |