Summary Basis of Decision for Korsuva

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 Korsuva is located below.

Recent Activity for Korsuva

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 Korsuva, a product which contains the medicinal ingredient difelikefalin (supplied as difelikefalin acetate). 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: 2023-08-28

Drug Identification Number (DIN):

DIN 02529688 – 50 mcg/mL difelikefalin, solution, intravenous administration

Post-Authorization Activity Table (PAAT)

Activity/submission type, control number Date submitted Decision and date Summary of activities
Drug product (DIN 02529688) market notification Not applicable Date of first sale: 2023-02-06 The manufacturer notified Health Canada of the date of first sale pursuant to C.01.014.3 of the Food and Drug Regulations.
NDS # 254548 2021-09-10 Issued NOC 2022-08-16 NOC issued for New Drug Submission.
Summary Basis of Decision (SBD) for Korsuva

Date SBD issued: 2022-12-16

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

Difelikefalin (supplied as difelikefalin acetate)

Drug Identification Number (DIN):

  • DIN 02529688 - 50 mcg/mL difelikefalin, solution, intravenous administration

Vifor Fresenius Medical Care Renal Pharma Ltd.

New Drug Submission Control Number: 254548

 

On August 16, 2022, Health Canada issued a Notice of Compliance to Vifor Fresenius Medical Care Renal Pharma Ltd for the drug product Korsuva.

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 Korsuva is favourable for the treatment of moderate to severe pruritus associated with chronic kidney disease in adult patients on hemodialysis (HD).

 

1 What was approved?

 

Korsuva, a kappa opioid receptor agonist, was authorized for the treatment of moderate to severe pruritus associated with chronic kidney disease in adult patients on hemodialysis (HD).

No data are available to Health Canada regarding use in pediatric patients (younger than 18 years of age). Therefore, Health Canada has not authorized an indication for pediatric use.

Evidence from clinical studies suggests that use in the geriatric population (65 years of age or older) is associated with differences in safety.

Korsuva (50 mcg/mL difelikefalin, supplied as difelikefalin acetate) is presented as a solution. In addition to the medicinal ingredient, the solution contains acetic acid, sodium acetate trihydrate, sodium chloride, and water for injection.

The use of Korsuva is contraindicated in patients who are hypersensitive to this drug or to any ingredient in the formulation, including any non-medicinal ingredient, or component of the container.

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 Korsuva 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 Korsuva approved?

 

Health Canada considers that the benefit-harm-uncertainty profile of Korsuva is favourable for the treatment of moderate to severe pruritus associated with chronic kidney disease in adult patients on hemodialysis (HD).

Chronic kidney disease-associated pruritus (CKD‑aP), also known as uremic pruritus, is a common systemic condition that presents with localized or generalized pruritus (itching) in patients with chronic kidney disease (CKD) that is not attributable to another cause. The pathophysiology of CKD‑aP is not fully understood but is believed to be due to a combination of systemic inflammation, imbalance in the endogenous opioid system, toxin deposition, and/or peripheral neuropathy.

The clinical presentation of CKD‑aP varies between patients and may fluctuate over time. Severity of the condition may range from mild itching with no to minimal impact on daily activities, to extreme itching that leads to restlessness, sleep disruption, fatigue, symptoms of depression, reduction in quality of life, and missed dialysis sessions. Results from observational studies have suggested that moderate-to-severe pruritus is associated with a higher mortality risk among hemodialysis patients, even after adjusting for other risk factors. This relationship is thought to be at least partly due to poor sleep quality.

At the time of authorization of Korsuva, there were no approved drug therapies in Canada for its proposed indication. Current approaches to the treatment of CKD‑aP include optimization of dialysis prescription; optimal treatment of hyperparathyroidism, hyperphosphatemia, and hypermagnesemia; topical emollients; topical analgesics oral antihistamines; and corticosteroids. Additional approaches to treatment include off-label use of medications that act on the central nervous system or immune system; however, such medications are associated with significant side effect profiles and limited evidence for efficacy in treatment of CKD‑aP. Ultraviolet B (UVB) phototherapy is a treatment option for refractory CKD‑aP, but is associated with an increased risk of carcinogenesis, can be difficult to access, and is not recommended in some circumstances (e.g., photosensitivity, immunosuppressive therapy).

Difelikefalin, the medicinal ingredient in Korsuva, is a new active substance and first‑in-class drug. It is a targeted kappa opioid receptor (KOR) agonist with low central nervous system (CNS) penetration and with no binding or functional activity at mu opioid receptors. Opioid receptors are known to modulate itch signals and inflammation, with KOR activation reducing itch and producing immunomodulatory effects.

The results of two pivotal Phase III studies, KALM‑1 (CLIN3102; 378 patients) and KALM‑2 (CLIN3103; 473 patients), provided evidence of the clinical efficacy of Korsuva. In both studies, patients received either Korsuva (0.5 mcg/kg of dry body weight) or placebo three times per week during a 12‑week double‑blind phase. In the KALM‑1 study, the double‑blind phase was followed by a 2‑week discontinuation period. Patients did not receive any study drug during this period and were monitored for signs or symptoms of physical dependence. In both studies, patients who received at least 30 doses of either Korsuva or placebo during the 12‑week double‑blind phase had the option to receive Korsuva three times per week for an additional 52 weeks.

The primary efficacy endpoint was a responder analysis of the proportion of patients who self‑reported an improvement of ≥3 points on the 24‑hour Worst Itching Intensity Numerical Rating Scale (WI‑NRS) at Week 12 in the Korsuva group versus (vs.) the placebo group. This endpoint was met in both pivotal studies, and statistically significant differences were identified in the intent‑to‑treat (ITT) analyses in both pivotal studies (851 patients in total). The least‑squares mean percentages of patients reporting an improvement from baseline of ≥3 points on the 24‑hour WI‑NRS at Week 12 were greater in the group treated with Korsuva than in the group that received placebo. In the KALM‑1 study, the least‑squares mean percentages were 51.0% in the Korsuva group vs. 27.6% in the placebo group (odds ratio of 2.72, p<0.001). In the KALM‑2 study, the least‑squares mean percentages were 54.0% in the Korsuva group vs. 42.2% in the placebo group (odds ratio of 1.61, p = 0.020). In both studies, a treatment effect was noted by Weeks 2 to 3, and was maintained throughout the 12‑week double‑blind treatment period.

The clinical safety of Korsuva was assessed mainly through data obtained from the primary safety pool, which includes data from 848 patients from the 12‑week placebo‑controlled phases of the KALM‑1 and KALM‑2 studies who were treated with either Korsuva or placebo (424 patients each). Results from a larger safety pool of 1,306 patients were also evaluated, which included the KALM‑1 and KALM‑2 studies, their respective open‑label extension periods, and two additional open‑label supportive Phase III studies (CLIN3101 and CLIN3105). Patients in this pool were treated with Korsuva at the recommended dose of 0.5 mcg/kg of dry body weight, with 711 patients, 533 patients, and 400 patients having been continuously exposed for at least 6 months, 9 months, and 12 months, respectively.

In pooled placebo-controlled analyses, the main safety concerns identified for Korsuva included somnolence, dizziness, mental status changes (including confusional state), gait disturbances including falls (possibly a consequence of concurrent dizziness and somnolence in some patients), headache, hyperkalemia (with greater incidence in patients who took concomitant opioids), paraesthesia (pooled term including paraesthesia, hypoesthesia, paresthesia oral and hypoesthesia oral), nausea, and diarrhea. A greater incidence of somnolence was observed in patients 65 years of age and older compared to younger patients. Additionally, there is a potential increase in the risk of somnolence and dizziness in patients using concomitant sedating antihistamines, opioid analgesics, or other CNS depressants.

The somnolence, dizziness, and mental status changes observed in the Phase III clinical studies are particularly notable, as the non‑clinical data indicated minimal CNS penetration of difelikefalin. The identification of these events in the clinical studies suggests that there is some penetration of difelikefalin into the CNS at clinical doses. Although the pathogenesis of the CNS-related adverse events was not known at the time of authorization, based on the results from dedicated abuse, dependence, and withdrawal studies, difelikefalin is anticipated to have no meaningful abuse and dependence potential and was not associated with opioid withdrawal symptoms. In addition, across all clinical studies of difelikefalin, there were no adverse event reports related to misuse, abuse, diversion, or dependence with intravenously administered difelikefalin at doses up to 80‑fold higher than the proposed clinical dose. Evaluation of the available longer-term safety data did not raise any additional major safety concerns.

An intact blood-brain barrier (BBB) is important for minimizing difelikefalin uptake into the CNS. As such, patients with clinically important disruptions to the BBB (e.g., primary brain malignancies, CNS metastases or other inflammatory conditions, active multiple sclerosis or advanced Alzheimer’s disease) may be at risk for difelikefalin entry into the CNS. The Product Monograph therefore states that Korsuva should be prescribed with caution in such patients, taking into account whether the benefits of Korsuva outweigh the potential risks for the individual, and ensuring observation for potential CNS effects.

A Risk Management Plan (RMP) for Korsuva was submitted by Vifor Fresenius Medical Care Renal Pharma Ltd 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 Korsuva 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 Korsuva was accepted.

Korsuva 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 Korsuva 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 Korsuva?

 

The New Drug Submission (NDS) for Korsuva was reviewed as part of the New Active Substance Work-Sharing Initiative, a work-sharing initiative of the national health regulatory agencies of Canada, Australia, Singapore, Switzerland, and the United Kingdom (the Access Consortium). This partnership aims to promote collaboration, optimize the use of resources, reduce duplication, and enhance the ability of each agency to ensure consumers have timely access to safe, effective, and high‑quality therapeutic products.

Health Canada completed the review of the quality components of the NDS for Korsuva, while the Swiss Agency for Therapeutic Products (Swissmedic) completed the review of the non‑clinical components, and Australia's Therapeutic Goods Administration (TGA) completed the review of the clinical components. Singapore’s Health Sciences Authority (HSA) participated as a peer reviewer. The review of the submission was collaborative, with each regulatory agency sharing the outcome of its review with the others. However, each agency made its regulatory decision independently.

The review of the NDS for Korsuva was also based on a critical assessment of the data package submitted to Health Canada and of foreign reviews as described in the Draft Guidance Document: The Use of Foreign Reviews by Health Canada. Method 3 was used for the review of the quality components of the submission, while Method 2 was used for the review of the clinical and non‑clinical components.

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

 

Submission Milestones: Korsuva

Submission Milestone Date
New Drug Submission filed 2021-09-10
Screening  
Screening Acceptance Letter issued 2021-10-21
Review  
Review of Risk Management Plan completed 2022-07-20
Quality evaluation completed 2022-07-22
Non-clinical evaluation completed 2022-08-04
Labelling review completed 2022-08-04
Clinical/medical evaluation completed 2022-08-05
Notice of Compliance issued by Director General, Pharmaceutical Products Directorate 2022-08-16

 

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

 

As described above, the New Drug Submission for Korsuva was reviewed as part of the New Active Substance Work Sharing Initiative. Australia’s Therapeutic Goods Administration (TGA) completed the review of the clinical components of the NDS for Korsuva. Although the agencies collaborated on the review of the submission, each agency made its regulatory decision independently. Additionally, the clinical component of the NDS for Korsuva was reviewed as per Method 2 described in the Draft Guidance Document: The Use of Foreign Reviews by Health Canada .

Clinical Pharmacology

The pathophysiology of chronic kidney disease‑associated pruritus is thought to be multifactorial, including systemic inflammation and an imbalance in the endogenous opioid system (e.g., overexpression of mu opioid receptors and concomitant downregulation of kappa opioid receptors [KORs]). Opioid receptors are known to modulate itch signals and inflammation, with KOR activation reducing itch and producing immunomodulatory effects.

Difelikefalin (the medicinal ingredient in Korsuva) is a targeted KOR agonist. It has low central nervous system (CNS) penetration and no binding or functional activity at mu opioid receptors, which mostly avoids mu-associated side effects such as constipation, euphoria, dependence, and respiratory depression. The physicochemical properties of difelikefalin (hydrophilic, synthetic D‑amino acid peptide with high polar surface area and charge at physiological pH) minimize its passive diffusion (permeability) and active transport across membranes (blood‑brain barrier [BBB]), thus limiting penetration into the CNS.

The activation by difelikefalin of KORs on peripheral sensory neurons and immune cells is considered mechanistically responsible for the antipruritic and anti‑inflammatory effects.

Pharmacokinetics

Difelikefalin exhibited dose‑proportional pharmacokinetics following administration of single and multiple doses ranging from 0.5 mcg/kg to 2.5 mcg/kg (1 to 5 times the recommended dosage) in patients with chronic kidney disease and who were undergoing hemodialysis. Steady‑state was reached after the second dose and the mean accumulation ratio was up to 1.6.

Plasma protein binding of difelikefalin is low to moderate (24% to 32%) and remains unaffected by renal impairment. Following the intravenous administration of difelikefalin ranging from 1 mcg/kg to 6 mcg/kg in patients undergoing hemodialysis, plasma protein binding ranged from 23% to 28%. Difelikefalin is not metabolized by cytochrome P450 (CYP) enzymes, human hepatic microsomes or hepatocytes, or kidney microsomes. In an in vivo absorption, distribution, metabolism, and elimination (ADME) study in humans, difelikefalin was the predominant component in plasma samples, accounting for more than 99% of the circulating radioactivity in both healthy volunteers and patients undergoing hemodialysis.

Hemodialysis reduced plasma concentrations of difelikefalin by 70% to 80%, and difelikefalin was not detectable in plasma after two dialysis cycles. Following administration of difelikefalin to hemodialysis patients, 11.2% of the dosage was excreted in urine, 58.8% in feces, and 19.5% in dialysate fluid.

Difelikefalin is not an inhibitor or substrate of clinically relevant enzymes or transporters based on the results of in vitro studies. Additionally, difelikefalin has minimal to no potential for induction of human CYP1A2, CYP2B6, or CYP3A4 enzymes. These findings suggest that difelikefalin is not likely to interact with co‑administered drugs.

The results of population pharmacokinetic analyses did not reveal any clinically significant differences in the pharmacokinetics of difelikefalin based on age, sex, and race/ethnicity.

The available data in hemodialysis patients indicated that no dosage adjustment is needed in patients with mild to moderate hepatic impairment. However, the clinical data in patients with moderate hepatic impairment is limited. Difelikefalin is not recommended for patients with severe hepatic impairment, as the effects of this condition on the pharmacokinetics of difelikefalin have not been evaluated.

Patients with mild renal impairment have a comparable pharmacokinetic profile to that of healthy subjects based on the available data. Total body clearance of difelikefalin in subjects with severe renal impairment is reduced relative to healthy, matched, control subjects and plasma concentrations of difelikefalin in subjects undergoing hemodialysis remain relatively constant until cleared during dialysis.

Pharmacodynamics

The penetration of difelikefalin into the CNS (brain) is restricted by its physicochemical properties, which minimize passive diffusion or active transport across membranes.

A thorough QT study was conducted in which healthy volunteers were administered difelikefalin as a single intravenous 0.5 mcg/kg dose (the clinical dose) or as a single 3.0 mcg/kg dose (6 times the clinical dose). The outcomes of this study indicated that difelikefalin did not prolong the corrected QT (QTc) interval to a clinically relevant extent.

A dose‑dependent increase in serum prolactin concentrations was observed in Phase I and II studies. Although it was not assessed in Phase III studies, one treatment‑emergent adverse event (TEAE) of increased blood prolactin was observed in the pooled Phase III safety analyses in a patient treated with Korsuva. However, no TEAEs were reported which were potentially indicative of clinical effects of hyperprolactinaemia in hemodialysis patients exposed to Korsuva for up to one year. The clinical relevance of these observations remain unknown.

Dose‑dependent decreases in serum thyroid stimulating hormone (TSH) and free thyroxine levels were observed in a Phase II study in Japanese patients, with a return to baseline levels following treatment discontinuation. Decreased TSH and decreased free thyroxine levels were both observed as TEAEs in patients following intravenous administration of difelikefalin, while these events were not observed in patients administered the placebo. Although TSH and free thyroxine were not assessed in Phase III studies, one TEAE of decreased free thyroxine was detected in the pooled Phase II/III safety analyses in a patient treated with Korsuva. This TEAE was considered by the investigator to be related to the study treatment. However, there was no overall imbalance in thyroid‑related adverse events between patients treated with Korsuva or placebo in the Phase III studies. The clinical relevance of these observations remain unknown.

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

Clinical Efficacy

Evidence of the clinical efficacy of Korsuva (difelikefalin) was provided through two pivotal Phase III placebo‑controlled studies, KALM‑1 (CLIN3102; 378 patients) and KALM‑2 (CLIN3103; 473 patients). Both studies included a 12‑week double‑blind phase in which patients received either Korsuva at a dose of 0.5 mcg/kg of dry body weight or placebo three times per week. In the KALM‑1 study, the double‑blind treatment period was followed by a 2‑week discontinuation period designed to assess potential signs or symptoms of drug withdrawal. Patients did not receive any study drug during this period and were monitored for signs or symptoms of physical dependence. In both studies, patients who received at least 30 doses of either Korsuva or placebo during the 12‑week double‑blind phase had the option to receive open‑label Korsuva at a dose of 0.5 mcg/kg of dry body weight three times per week for an additional 52 weeks.

Patients included in the two pivotal studies were required to be at least 18 years of age, be undergoing hemodialysis three times per week for end stage renal disease (ESRD), and have a mean baseline score of >4 (in KALM‑1) or ≥5 (in KALM‑2) on the patient‑reported daily 24‑hour Worst Itching Intensity Numerical Rating Scale (WI‑NRS). The WI‑NRS is a validated numerical rating scale for the evaluation of the intensity of chronic itch, ranging from 0 (no itching) to 10 (worst itching imaginable). Patients were permitted to continue their current antipruritic therapies, provided there had been no change within 14 days prior to screening. Patients receiving ongoing ultraviolet B therapy were excluded.

The primary efficacy endpoint, which was met in both pivotal studies, was a responder analysis of the proportion of patients who self‑reported an improvement of ≥3 points on the 24‑hour WI‑NRS at Week 12 in the Korsuva group versus (vs.) the placebo group. Justification for this endpoint as a clinically meaningful threshold was based on a published psychometric analysis and results from the supportive Phase II safety study, CR845‑CLIN2101. Statistically significant differences were identified in the intent‑to‑treat (ITT) analyses in both pivotal studies (851 patients in total). The least‑squares mean percentages of patients reporting an improvement from baseline of ≥3 points on the 24‑hour WI‑NRS at Week 12 were greater in the group treated with Korsuva than in the group that received placebo. In the KALM‑1 study, the least‑squares mean percentages were 51.0% in the Korsuva group vs. 27.6% in the placebo group (odds ratio of 2.72, p<0.001). In the KALM‑2 study, the least‑squares mean percentages were 54.0% in the Korsuva group vs. 42.2% in the placebo group (odds ratio of 1.61, p = 0.020). In both studies, a treatment effect was noted by Weeks 2 to 3, and was maintained throughout the 12‑week double‑blind treatment period.

The key secondary efficacy endpoints included a WI‑NRS responder analysis based on the proportion of patients achieving an improvement of ≥4 points (a more conservative threshold than the primary endpoint), and other validated patient‑reported outcomes evaluating the impact on quality of life. The least‑squares mean percentages of patients reporting an improvement from baseline of ≥4 points on the WI‑NRS at Week 12 were greater in the group treated with Korsuva than in the group that received placebo. In both studies, this difference was statistically significant. In the KALM‑1 study, the least‑squares mean percentages were 38.9% in the Korsuva group vs. 18.0% in the placebo group (odds ratio of 2.89, p<0.001). In the KALM‑2 study, the least‑squares mean percentages were 41.2% in the Korsuva group vs. 28.4% in the placebo group (odds ratio of 1.77, p = 0.010). Results of the additional validated quality of life measures (i.e., change from baseline in 5‑D Itch Scale and total Skindex‑10 Scale scores at Week 12) were also generally supportive of the efficacy of Korsuva in the target patient population.

Indication

Health Canada approved the following indication:

  • Korsuva (difelikefalin) is indicated for the treatment of moderate to severe pruritus associated with chronic kidney disease in adult patients on hemodialysis.

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

Clinical Safety

Data from three randomized Phase II studies (CR845-CLIN2005, CR845-CLIN2101, and PR-13A9-P2-A) were submitted and evaluated as supportive safety studies. Based on observations from these studies, the 0.5 mcg/kg dose of difelikefalin was determined to have the most favourable benefit risk profile and was the dose used for further evaluation in Phase III studies. The results of the supportive studies did not raise any efficacy concerns. The safety data were consistent with safety findings from the pivotal Phase III studies and did not raise any additional safety concerns.

The main evidence of the clinical safety of Korsuva was obtained from the primary safety pool, which includes data from 848 patients who were treated with either Korsuva or placebo (424 patients each) during the 12‑week placebo‑controlled phases of the pivotal Phase III studies, KALM‑1 and KALM‑2 (described in the Clinical Efficacy section). Patients in the primary safety pool were undergoing hemodialysis and had moderate‑to-severe pruritus as defined by baseline WI‑NRS scores.

Results from a larger safety pool were also evaluated, which included the exposure provided by the KALM‑1 and KALM‑2 studies and their respective open‑label extension periods, along with the exposure provided by two additional open‑label supportive Phase III studies, CLIN3101 and CLIN3105. This larger safety pool included 1,306 patients undergoing hemodialysis who had moderate‑to‑severe pruritus. Patients in this pool were treated with Korsuva at the recommended dose of 0.5 mcg/kg of dry body weight, with 711 patients having been continuously exposed to Korsuva for at least 6 months, and 400 patients having been continuously exposed for at least 12 months.

In pooled placebo-controlled analyses, the main safety concerns identified for Korsuva included somnolence, dizziness, mental status changes (including confusional state), gait disturbances including falls (possibly a consequence of concurrent dizziness and somnolence in some patients), headache, hyperkalemia (with greater incidence in patients who took concomitant opioids), paraesthesia (pooled term including paraesthesia, hypoesthesia, paresthesia oral and hypoesthesia oral), nausea, and diarrhea. A greater incidence of somnolence was observed in patients 65 years of age and older who took Korsuva compared to younger patients. Additionally, there is a potential increase in the risk of somnolence and dizziness in patients using concomitant sedating antihistamines, opioid analgesics, or other CNS depressants.

The somnolence, dizziness, and mental status changes observed in the Phase III clinical studies are particularly notable, as the non‑clinical data indicated minimal CNS penetration of difelikefalin (the medicinal ingredient in Korsuva). The identification of these events in the clinical studies suggests that there is some penetration of difelikefalin into the CNS at clinical doses. Although the pathogenesis of the CNS‑related adverse events was not known at the time of authorization, based on the results from dedicated abuse, dependence, and withdrawal studies, difelikefalin is anticipated to have no meaningful abuse and dependence potential and was not associated with opioid withdrawal symptoms. In addition, across all clinical studies of difelikefalin, there were no adverse event reports related to misuse, abuse, diversion, or dependence with intravenously administered difelikefalin at doses of up to 80‑fold higher than the proposed clinical dose. Evaluation of the available longer‑term safety data did not raise any additional major safety concerns.

An intact blood-brain barrier is important for minimizing difelikefalin uptake into the CNS. As such, patients with clinically important disruptions to the blood‑brain barrier (e.g., primary brain malignancies, CNS metastases or other inflammatory conditions, active multiple sclerosis, advanced Alzheimer’s disease) may be at risk for difelikefalin entry into the CNS. The Product Monograph therefore states that Korsuva should be prescribed with caution in such patients, taking into account whether the benefits of Korsuva outweigh the potential risks for the individual, and ensuring observation for potential CNS effects.

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

 

 

 

7.2 Non-Clinical Basis for Decision

 

As described above, the New Drug Submission (NDS) for Korsuva was reviewed as part of the New Active Substance Work Sharing Initiative. The Swiss Agency for Therapeutic Products (Swissmedic) completed the review of the non‑clinical component of the NDS for Korsuva. Although the agencies collaborated on the review of the submission, each agency made its regulatory decision independently. Additionally, the review of the non‑clinical component of the New Drug Submission for Korsuva was conducted as per Method 2 described in the Draft Guidance Document: The Use of Foreign Reviews by Health Canada.

Single‑dose toxicology studies were conducted in mice, rats, and monkeys. Difelikefalin was administered intravenously to mice at doses from 50 mg/kg to 200 mg/kg. Mortality was noted within 30 minutes in all animals administered the 200 mg/kg dose. The single‑dose maximum tolerated dose (MTD) was determined to be 100 mg/kg. A dose range‑finding study was conducted in rats, in which lethargy, decreased body weight gain, and reduced food consumption were observed at doses ranging from 50 mg/kg to 100 mg/kg. Due to concerns about the tolerability of repeat dosing at higher doses, 25 mg/kg/day was chosen for the repeat‑dose, dose‑finding phase. In monkeys, single doses of difelikefalin ranging from 0.5 mg/kg to 16 mg/kg were administered intravenously. Clinical signs were observed starting at 4 mg/kg and increased in severity and duration at higher doses. Veterinary intervention was required in one animal that received the 16 mg/kg dose. The single‑dose MTD in monkeys was determined to be 8 mg/kg.

Multiple repeat‑dose toxicology studies were conducted in rats, including a 28‑day study in which animals were administered doses of difelikefalin ranging from 1 mg/kg/day to 25 mg/kg/day and 13‑week and 26‑week studies in which doses ranged from 0.25 mg/kg/day to 25 mg/kg/day. At the high dose of 25 mg/kg/day, the mean exposure margin (based on the area under the concentration‑time curve [AUC]) exceeded the 50‑fold clinical exposure at the proposed therapeutic dose of 0.5 mcg/kg/dose. The adverse effects consistently observed across all studies and across all dose levels included clinical observations and effects on body weight and food consumption. These findings were transient and attributed to exaggerated pharmacodynamic and secondary effects. Difelikefalin‑related testicular effects were observed at a generally low incidence in rats, and predominantly at the high dose of 25 mg/kg/day. Due to the observation of testicular effects at high exposure margins, they are expected to pose low clinical risk.

Repeat‑dose toxicology studies in monkeys included a 28‑day study in which animals were administered doses of difelikefalin ranging from 0.25 mg/kg/day to 4 mg/kg/day, and 13‑week and 39‑week studies in which doses ranged from 0.06 mg/kg/day to 1 mg/kg/day. At the high dose of 1 mg/kg/day, the mean exposure margin (based on the AUC) exceeded the 50‑fold clinical exposure at the proposed therapeutic dose of 0.5 mcg/kg/dose. As observed in the rat studies, the most consistent difelikefalin‑related effects in monkeys included clinical observations and effects on body weight and food consumption. Clinical signs were observed at doses of 0.25 mg/kg/day and higher, were transient in nature, and were generally dose‑dependent with respect to incidence, severity, and/or duration. In one of the 39‑week studies, a one‑day dosing holiday was required for animals in the 1 mg/kg/day group due to the severity of the clinical signs observed. Additionally, a single female in the 1 mg/kg/day group died prematurely, although the relationship to difelikefalin was uncertain. A dose‑dependent body weight loss was observed at dose levels of 0.25 mg/kg/day and higher following the first dose, which was associated with a reduction in food consumption. However, mean body weights in these groups were similar to control values within 3 days to 2 weeks after the initiation of dosing.

The outcomes of developmental and reproductive toxicity studies did not identify concerns with respect to male or female fertility and mating, or for embryofetal, prenatal, and postnatal development at clinically relevant exposures. One notable finding with respect to maternal toxicity was an association between difelikefalin and fewer pregnancies in rabbits.

Difelikefalin was detected in fetal plasma, indicating that placental transfer of difelikefalin occurs. However, based on the results of reproductive toxicity studies, it was not associated with embryofetal toxicity. Difelikefalin was also shown to be transferred into milk, but there were no quantifiable levels of the test article in the plasma of nursing pups.

No concerns regarding genotoxicity, carcinogenicity, phototoxicity, immunotoxicity or local tolerance emerged from the studies performed.

The results of dedicated studies indicate that difelikefalin poses a low risk for abuse potential or dependence.

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

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

 

 

7.3 Quality Basis for Decision

 

As described above, the New Drug Submission (NDS) for Korsuva was reviewed as part of the New Active Substance Work Sharing Initiative. Health Canada completed the review of the quality component of the NDS for Korsuva. Although the agencies collaborated on the review of the submission, each agency made its regulatory decision independently. Additionally, the quality component of the NDS for Korsuva was reviewed as per Method 3 described in the Draft Guidance Document: The Use of Foreign Reviews by Health Canada.

The chemistry and manufacturing information submitted for Korsuva 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. The formulation and manufacturing process used for the clinical lots have been found to be representative of those proposed for the commercial lots. Based on the stability data submitted, the proposed shelf life of 30 months is acceptable when the drug product is stored at room temperature (15 ºC to 30 ºC).

Proposed limits of drug-related impurities are considered adequately qualified (i.e. within International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use [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.

None of the excipients used in the formulation of Korsuva is of human or animal origin.