Summary Basis of Decision for Kerendia

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 Kerendia is located below.

Recent Activity for Kerendia

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 Kerendia

Updated:

2023-02-27

The following table describes post-authorization activity for Kerendia, a product which contains the medicinal ingredient finerenone. 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 and Applications Guidance.

Drug Identification Number (DIN):

DIN 02531917 – 10 mg finerenone, tablet, oral administration

DIN 02531925 – 20 mg finerenone, tablet, oral administration

Post-Authorization Activity Table (PAAT)

Activity/submission type, control numberDate submittedDecision and dateSummary of activities
Drug product (DINs 02531917, 02531925) market notificationNot applicableDate of first sale:
2022-11-22

The manufacturer notified Health Canada of the date of first sale pursuant to C.01.014.3 of the Food and Drug Regulations.

NDS # 2582312021-11-01Issued NOC
2022-10-14

NOC issued for New Drug Submission.

Summary Basis of Decision (SBD) for Kerendia

Date SBD issued: 2023-02-27

The following information relates to the new drug submission for Kerendia.

Finerenone

Drug Identification Number (DIN):

  • DIN 02531917 - 10 mg finerenone, tablet, oral administration
  • DIN 02531925 - 20 mg finerenone, tablet, oral administration

Bayer Inc.

New Drug Submission Control Number: 258231

On October 14, 2022, Health Canada issued a Notice of Compliance to Bayer Inc. for the drug product Kerendia.

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 Kerendia is favourable as an adjunct to standard of care therapy in adults with chronic kidney disease and type 2 diabetes to reduce the risk of:

  • end-stage kidney disease and a sustained decrease in estimated glomerular filtration rate,
  • cardiovascular death, non‑fatal myocardial infarction and hospitalization for heart failure.

1 What was approved?

Kerendia, an aldosterone antagonist, was authorized as an adjunct to standard of care therapy in adults with chronic kidney disease and type 2 diabetes to reduce the risk of:

  • end-stage kidney disease and a sustained decrease in estimated glomerular filtration rate,
  • cardiovascular death, non‑fatal myocardial infarction and hospitalization for heart failure.

Kerendia is not authorized for use in pediatric patients (<18 years of age), as no data are available for this population.

Evidence from clinical studies suggests that use in the geriatric population (≥65 years of age) is not associated with significant differences in safety or effectiveness. However, greater sensitivity of some older individuals cannot be ruled out.

Kerendia (10 mg and 20 mg finerenone) is presented as a tablet. In addition to the medicinal ingredient, the tablet contains cellulose microcrystalline, croscarmellose sodium, ferric oxide red (10 mg film‑coated tablet), ferric oxide yellow (20 mg film‑coated tablet), hypromellose 5 cP, lactose monohydrate, magnesium stearate, sodium laurilsulfate, talc, and titanium dioxide.

The use of Kerendia 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,
  • who are receiving concomitant systemic treatment with medications that are strong cytochrome P450 (CYP) 3A4 inhibitors (e.g., itraconazole, ketoconazole, ritonavir, nelfinavir, cobicistat, clarithromycin, telithromycin, and nefazodone),
  • with Addison’s disease.

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 Kerendia 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 Kerendia approved?

Health Canada considers that the benefit-harm-uncertainty profile of Kerendia is favourable as an adjunct to standard of care therapy in adults with chronic kidney disease (CKD) and type 2 diabetes (T2D) to reduce the risk of:

  • end-stage kidney disease and a sustained decrease in estimated glomerular filtration rate,
  • cardiovascular death, non‑fatal myocardial infarction and hospitalization for heart failure.

Type 2 diabetes and chronic kidney disease are major public health concerns both nationally and internationally. Diabetes mellitus is a metabolic disorder characterized by impaired insulin secretion or action, resulting in hyperglycemia. Diabetes can result in a range of long‑term complications such as cardiovascular disease, retinopathy, nephropathy, neuropathy, amputations, and decreased life expectancy. Chronic kidney disease is defined as abnormalities of kidney structure or function, present for 3 months or longer, with implications for health. Individuals with T2D are at an increased risk of developing diabetic kidney disease, and individuals with diabetes are over 12 times more likely to be hospitalized with end‑stage kidney disease. Approximately 40% of patients with T2D are likely to develop CKD during their lifetime. In Canada, an estimated 935,000 people have T2D and CKD.

A comprehensive treatment strategy is needed for patients with T2D and CKD in order to reduce the risks of kidney disease progression and cardiovascular morbidity and mortality. The development and progression of renal damage in patients with diabetes can be reduced and slowed through intensive glycemic control, management of hypertension, and lifestyle changes. The progression of CKD in diabetic patients can also be treated using medications that disrupt the renin‑angiotensin‑aldosterone system (RAAS). Sodium‑glucose co‑transporter 2 (SGLT2) inhibitors and glucagon‑like peptide 1 (GLP‑1) receptor agonists have also recently shown benefit as treatments for patients with CKD associated with T2D.

Finerenone, the medicinal ingredient in Kerendia, is a nonsteroidal antagonist of the mineralocorticoid receptor (MR). Finerenone binds to the MR and blocks the binding of aldosterone, a component of the RAAS. Potential mechanisms of action for Kerendia which have been reported under certain experimental conditions include:

  • attenuation of inflammation and fibrosis that are thought to be mediated by MR overactivation,
  • in vitro, a dose‑dependent inhibition of MR interaction with transcriptional coactivators implicated in the expression of pro‑inflammatory and pro‑fibrotic mediators, and
  • blocking of MR by finerenone in vivo, which counteracts sodium retention in the kidneys and hypertrophic processes in the kidneys, heart, and blood vessels.

The clinical efficacy and safety of Kerendia were evaluated in two pivotal Phase III studies, FIDELIO‑DKD and FIGARO‑DKD. In both studies, the effect of Kerendia on kidney and cardiovascular outcomes was evaluated in adults with CKD and T2D as compared to treatment with placebo. The results from these studies provided sufficient evidence to support the safe and efficacious use of Kerendia in the target population.

In the FIDELIO‑DKD study, 5,674 patients were randomized to receive either 10 mg or 20 mg Kerendia (2,833 patients in total for both doses) or placebo (2,841 patients) once daily for a mean duration of treatment of 2.2 years, with a median study follow‑up time of 2.6 years.

The primary endpoint was a composite of time to first occurrence of one of the following:

  • kidney failure (defined as chronic dialysis or kidney transplantation, or a sustained decrease in estimated glomerular filtration rate [eGFR] to <15 mL/min/1.73 m2 over at least 4 weeks),
  • a sustained decline in eGFR of 40% or more compared to baseline over at least 4 weeks, or
  • renal death.

Kerendia was found to be superior to placebo, as it significantly reduced the risk of the primary composite endpoint compared to placebo in a time‑to‑event analysis (hazard ratio [HR] 0.82; 95% confidence interval [CI]: 0.73, 0.93; p = 0.0014).

The key secondary endpoint was a composite of time to first occurrence of cardiovascular death, non‑fatal myocardial infarction, non‑fatal stroke, or hospitalization for heart failure. Kerendia significantly reduced the risk of the key secondary composite endpoint compared to placebo (HR 0.86; 95% CI: 0.75, 0.99; p = 0.0339).

The treatment effect for the primary and key secondary endpoints was generally consistent across subgroups regardless of baseline characteristics. The treatment effect for the primary endpoint was mainly driven by an effect on a reduction in sustained decrease in eGFR, though kidney failure also contributed to the treatment effect. There were too few renal deaths during the study to allow for statistical inference, and the incidence observed was the same in the Kerendia and placebo groups (two events each).

In the FIGARO‑DKD study, 7,352 patients were randomized to received either 10 mg or 20 mg Kerendia (3,686 patients in total for both doses) or placebo (3,666 patients) once daily for a mean duration of treatment of 2.9 years, with a median study follow‑up time of 3.4 years.

The primary endpoint was a composite of time to first occurrence of one of the following: cardiovascular death, non‑fatal myocardial infarction, non‑fatal stroke, or hospitalization for heart failure.

Kerendia significantly reduced the risk of the primary composite endpoint compared to placebo in a time‑to‑event analysis (hazard ratio [HR] 0.87; 95% CI: 0.76, 0.98; p = 0.0264). Hospitalization for heart failure was the major driver for these results, though cardiovascular death also contributed to the treatment effect. The other components were not statistically significant. The treatment effect for the primary endpoint was consistent across subgroups (regardless of baseline characteristics).

The key secondary endpoint was a composite of time to first occurrence of kidney failure, a sustained decline in eGFR of 40% or more compared to baseline over at least 4 weeks, or renal death. Although a lower incidence rate was also observed of the secondary composite outcome, this difference did not achieve statistical significance (HR 0.87, 95% CI: 0.76, 1.01; p = 0.0689).

The clinical safety of Kerendia was evaluated in the 2,827 patients who received Kerendia in the FIDELIO‑DKD study and the 3,683 patients who received Kerendia in the FIGARO‑DKD study. The mean treatment durations in the studies were 2.2 years and 2.9 years, respectively.

Serious adverse reactions were reported in both the FIDELIO‑DKD study (32% of Kerendia‑treated patients versus [vs.] 34% of placebo‑treated patients) and the FIGARO‑DKD study (31% of Kerendia‑treated patients vs. 33% of placebo‑treated patients). Adverse reactions led to permanent discontinuation in 7% of patients receiving Kerendia and 6% of patients receiving placebo in the FIDELIO‑DKD study, and 6% of patients receiving Kerendia and 5% of patients receiving placebo in the FIGARO‑DKD study.

Adverse reactions that were reported at a higher frequency in Kerendia‑treated patients and that occurred in 5% or more of patients receiving Kerendia were:

  • FIDELIO‑DKD study:
    • hyperkalemia (18.3% ‑ Kerendia vs. 9.0% ‑ placebo)
    • decreased eGFR (6.3% ‑ Kerendia vs. 4.7% ‑ placebo)
  • FIGARO‑DKD Study:
    • hyperkalemia (10.8% ‑ Kerendia vs. 5.3% ‑ placebo).

Adverse reactions that were reported in 1% or more of patients receiving Kerendia in the Phase III studies (pooled FIDELIO‑DKD and FIGARO‑DKD) and that were reported more frequently in patients receiving Kerendia than in patients receiving placebo included anemia (6.5% vs. 6.1%), hyperuricemia (5.1% vs. 3.9%), hyponatremia (1.3% vs. 0.7%), hypotension (4.6% vs. 3.9%), and pruritus (2.9% vs. 2.2%).

The safety of Kerendia during pregnancy and breastfeeding and in patients with severe hepatic impairment or end‑stage kidney failure is uncertain, since these populations were excluded from clinical studies. Patients with severe renal impairment (eGFR <25 mL/min/1.73 m2) were under‑represented. The long‑term safety of Kerendia is also uncertain. Appropriate cautionary statements, additional patient monitoring, dosage adjustments or interruptions, and descriptions of the findings and safety concerns are included in the Kerendia Product Monograph to guide health professionals and patients on the safe and efficacious use of Kerendia.

A Risk Management Plan (RMP) for Kerendia was submitted by Bayer 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 Kerendia 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 Kerendia was accepted.

Kerendia 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 Kerendia 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 Kerendia?

The Canadian regulatory decision on the Kerendia New Drug Submission (NDS) was made independently. The review of the NDS was based on a critical assessment of the data package submitted to Health Canada and of the reviews completed by the United States Food and Drug Administration as described in the Draft Guidance Document: The Use of Foreign Reviews by Health Canada. Various methods were used for the review of the clinical data, and Method 3 was used for the review of the non‑clinical and quality components of the submission.

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

Submission Milestones: Kerendia

Submission MilestoneDate
New Drug Submission filed2021-11-01
Screening
Screening Deficiency Notice issued2021-12-14
Response to Screening Deficiency Notice filed2021-12-15
Screening Acceptance Letter issued2021-12-22
Review
Biostatistics evaluation completed2022-09-16
Review of Risk Management Plan completed2022-09-22
Quality evaluation completed2022-10-03
Non-clinical evaluation completed2022-10-04
Labelling review completed2022-10-12
Clinical/medical evaluation completed2022-10-13
Notice of Compliance issued by Director General, Pharmaceutical Products Directorate2022-10-14

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 clinical review of the New Drug Submission for Kerendia was conducted using various methods described in the Draft Guidance Document: The Use of Foreign Reviews by Health Canada.

Clinical Pharmacology

Finerenone, the medicinal ingredient in Kerendia, is a nonsteroidal, selective antagonist of the mineralocorticoid receptor (MR). Finerenone binds to the MR and blocks the binding of aldosterone, a component of the renin-angiotensin-aldosterone system (RAAS). Aldosterone plays a central role in the homeostatic regulation of blood pressure and plasma sodium and potassium levels. The mechanisms of action of Kerendia which contribute to the reduction of renal and cardiovascular events are not completely understood and may be due to multi-factorial effects in different tissues. Potential mechanisms of action which have been reported under certain experimental conditions include:

  • attenuation of inflammation and fibrosis that are thought to be mediated by MR overactivation,
  • in vitro, inhibition of MR interaction with transcriptional coactivators implicated in the expression of pro‑inflammatory and pro‑fibrotic mediators in a dose‑dependent manner,
  • blocking of MR by finerenone in vivo, which counteracts sodium retention in the kidneys and hypertrophic processes in the kidneys, heart, and blood vessels.

Finerenone has no relevant affinity for androgen, progesterone, estrogen and glucocorticoid receptors and therefore is unlikely to cause sex hormone‑related adverse events (e.g., gynecomastia).

The pharmacokinetics and pharmacodynamics of finerenone were evaluated in a series of Phase I, II and III studies. The absolute oral bioavailability of finerenone is estimated as 43.5%. Finerenone is eliminated almost exclusively in the form of metabolites, with approximately 80% and 20% of the administered dose excreted in urine and feces, respectively. The pharmacokinetic parameters of finerenone are linear across the investigated dose range of 1.25 mg to 20 mg.

An increase in finerenone exposure was observed in special populations of interest, including elderly subjects, subjects with moderate and severe renal impairment (defined as an estimated glomerular filtration rate [eGFR] of 15 to <60 mL/min/1.73 m2), and subjects with moderate hepatic impairment (Child Pugh B). As the dosage of finerenone is based on multiple factors, including levels of serum potassium, eGFR, and pre‑existing conditions, dose adjustment is not necessary for elderly subjects and subjects with moderate hepatic impairment. Subjects with moderate to severe renal impairment should initiate finerenone treatment at a lower dose (10 mg) compared to subjects with mild renal impairment and normal kidney function (20 mg), depending on baseline eGFR. There was no clinical experience with subjects with end‑stage renal disease (eGFR of <15 mL/min/1.73 m2) or severe hepatic impairment (Child Pugh C) in the Phase I studies.

Cytochrome P450 (CYP) 3A4 is a major contributor to the metabolic clearance of finerenone. Therefore, CYP3A4 inhibitors or inducers were identified as potentially relevant extrinsic factors. Concomitant use of finerenone with strong CYP3A4 inhibitors is contraindicated due to an expected increase in finerenone exposure (as measured by the area under the concentration‑time curve [AUC]) by more than 350%. Interactions between finerenone and the moderate CYP3A4 inhibitors erythromycin and verapamil were identified through clinical drug‑drug interaction studies. Caution and additional monitoring of potassium levels are advised when finerenone is administered concomitantly with moderate CYP3A4 inhibitors, as an increase in finerenone exposure (as measured by the AUC) of 170‑248% has been observed. Due to an expected decrease in finerenone exposure by more than 80%, concomitant use of finerenone with moderate and strong CYP3A4 inducers should be avoided.

Pharmacodynamics studies evaluated the effects of finerenone on healthy subjects, patients with chronic kidney disease (CKD) and type 2 diabetes (T2D), and on the QT interval.

In healthy subjects, multiple dose regimens of finerenone (daily doses of 20 mg or 40 mg over 10 days) led to activation of the RAAS. This was based on reversible increases of plasma renin activity and serum aldosterone concentrations, which returned to baseline values within 48 hours after the last dose. Following activation of the MR with the agonist fludrocortisone, single doses of finerenone up to 20 mg showed natriuretic effects while decreasing urinary potassium excretion compared to placebo. The highest single dose of 80 mg and the highest multiple dose of 40 mg of finerenone did not affect vital signs parameters in healthy participants.

Based on data from the Phase III studies in adult patients with CKD and T2D (described in the Clinical Efficacy section), the placebo‑corrected relative reduction in urinary albumin‑to‑creatinine ratio (UACR) in patients randomized to finerenone at Month 4 were 31% and 32% for the FIDELIO‑DKD and FIGARO‑DKD studies, respectively. In both studies, the reduction in UACR was maintained for at least 48 months. In a Phase IIb dose‑finding study in adult patients with CKD and T2D, the placebo‑corrected relative reductions in the UACR at Day 90 were 25% and 38% in patients treated once daily with finerenone 10 mg and 20 mg, respectively.

In a dedicated QT study in 57 healthy participants, there was no indication of a clinically relevant prolonging effect of finerenone on the QT/corrected QT (QTc) interval after single doses of 20 mg (therapeutic) or 80 mg (supratherapeutic).

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

Clinical Efficacy

The clinical efficacy of Kerendia was evaluated in two randomized, double‑blind, placebo‑controlled, multicenter pivotal Phase III studies, FIDELIO‑DKD and FIGARO‑DKD. In both studies, the effect of Kerendia (a once‑daily 10 mg or 20 mg dose) on kidney and cardiovascular outcomes was evaluated in adults with chronic kidney disease (CKD) and type 2 diabetes (T2D) as compared to treatment with placebo. The results from these two studies provided sufficient evidence to support the safe and efficacious use of Kerendia in the target population.

FIDELIO‑DKD Study

The FIDELIO‑DKD study was conducted in adult patients with CKD and T2D, with either a UACR of 30 to <300 mg/g, an eGFR of 25 to <60 mL/min/1.73 m2 and diabetic retinopathy, or a UACR of ≥300 mg/g and an eGFR of 25 to <75 mL/min/1.73 m2, and a serum potassium concentration of ≤4.8 mmol/L at screening. Additionally, patients were required to be receiving standard of care, including a maximum tolerated labeled dose of an angiotensin‑converting enzyme inhibitor (ACEi) or an angiotensin receptor blocker (ARB). Patients with diagnosed heart failure with reduced ejection fraction and persistent symptoms (New York Heart Association II‑IV) were excluded.

In total, 5,674 patients were randomized to receive either 10 mg or 20 mg Kerendia (2,833 patients in total for both doses) or placebo (2,841 patients) once daily for a mean duration of treatment of 2.2 years, with a median study follow‑up time of 2.6 years.

The primary endpoint was a composite of time to first occurrence of one of the following:

  • kidney failure (defined as chronic dialysis or kidney transplantation, or a sustained decrease in eGFR to <15 mL/min/1.73 m2 over at least 4 weeks),
  • a sustained decline in eGFR of 40% or more compared to baseline over at least 4 weeks, or
  • renal death.

Kerendia was found to be superior to placebo, as it significantly reduced the risk of the primary composite endpoint compared to placebo in a time‑to‑event analysis using the Cox proportional hazards model and log‑rank test (hazard ratio [HR] 0.82; 95% confidence interval [CI]: 0.73, 0.93; p = 0.0014).

The key secondary endpoint was a composite of time to first occurrence of cardiovascular death, non‑fatal myocardial infarction, non‑fatal stroke, or hospitalization for heart failure. Kerendia significantly reduced the risk of the key secondary composite endpoint compared to placebo (HR 0.86; 95% CI: 0.75, 0.99; p = 0.0339).

The treatment effect for the primary kidney and key secondary cardiovascular endpoints was generally consistent across subgroups regardless of baseline characteristics. The treatment effect for the primary endpoint was mainly driven by an effect on a reduction in sustained decrease in eGFR, though kidney failure also contributed to the treatment effect. There were too few renal deaths during the study to allow for statistical inference, and the incidence observed was the same in the Kerendia and placebo groups (two events each).

FIGARO‑DKD Study

The FIGARO‑DKD was conducted in adult patients with CKD and T2D, with either a UACR of 30 to <300 mg/g and an eGFR of 25 to 90 mL/min/1.73 m2, or a UACR of ≥300 mg/g and an eGFR of ≥60 mL/min/1.73 m2 at screening. Additionally, patients were required to have a serum potassium concentration of ≤4.8 mmol/L at screening and had received standard of care background therapy, including a maximum tolerated labeled dose of a renin angiotensin system (RAS) inhibitor (either an ACEi or an ARB). Patients with diagnosed heart failure with reduced ejection fraction and persistent symptoms (New York Heart Association II‑IV) were excluded.

In total, 7,352 patients were randomized to receive either 10 mg or 20 mg Kerendia (3,686 patients in total for both doses) or placebo (3,666 patients) once daily for a mean duration of treatment of 2.9 years, with a median study follow‑up time of 3.4 years.

The primary endpoint was a composite of time to first occurrence of one of the following: cardiovascular death, non‑fatal myocardial infarction, non‑fatal stroke, or hospitalization for heart failure.

Kerendia significantly reduced the risk of the primary composite endpoint compared to placebo in a time‑to‑event analysis using the Cox proportional hazards model and log‑rank test (hazard ratio [HR] 0.87; 95% CI: 0.76, 0.98; p = 0.0264).

Among the components of the composite primary endpoint, hospitalization for heart failure was the major driver for the results, though cardiovascular death also contributed to the treatment effect. The other components were not statistically significant. There was a substantial number of events in each component of the primary composite, including 408 cardiovascular deaths in total and 280 total hospitalizations for heart failure across both treatment groups. The treatment effect for the primary endpoint was consistent across subgroups (regardless of baseline characteristics).

The key secondary endpoint was a composite of time to first occurrence of kidney failure, a sustained decline in eGFR of 40% or more compared to baseline over at least 4 weeks, or renal death. Although a lower incidence rate was also observed of the secondary composite outcome compared to placebo, this difference did not achieve statistical significance (HR 0.87, 95% CI: 0.76, 1.01; p = 0.0689).

Indication

Sponsor's proposed indication

Health Canada-approved indication

Kerendia (finerenone) is indicated to delay progression of kidney disease and to reduce the risk of major adverse cardiovascular events (cardiovascular death, non‑fatal myocardial infarction, non‑fatal stroke) and hospitalization for heart failure in adults with chronic kidney disease (CKD) and type 2 diabetes (T2D).

Kerendia (finerenone) is indicated as an adjunct to standard of care therapy in adults with chronic kidney disease (CKD) and type 2 diabetes (T2D) to reduce the risk of:

· end‑stage kidney disease and a sustained decrease in estimated glomerular filtration rate,

· cardiovascular death, non‑fatal myocardial infarction and hospitalization for heart failure.

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

Clinical Safety

The clinical safety of Kerendia in patients with CKD and T2D was evaluated in the Phase III FIDELIO‑DKD and FIGARO‑DKD studies, described in the Clinical Efficacy section. In the FIDELIO‑DKD study, 2,827 patients received Kerendia with a mean treatment duration of 2.2 years. In the FIGARO‑DKD study, 3,683 patients received Kerendia with a mean treatment duration of 2.9 years.

Serious adverse reactions were reported in both the FIDELIO‑DKD study (32% of Kerendia‑treated patients versus [vs.] 34% of placebo‑treated patients) and in the FIGARO‑DKD study (31% of Kerendia‑treated patients vs. 33% of placebo‑treated patients). Adverse reactions led to permanent discontinuation in 7% of patients receiving Kerendia and 6% of patients receiving placebo in the FIDELIO‑DKD study, and 6% of patients receiving Kerendia and 5% of patients receiving placebo in the FIGARO‑DKD study.

Adverse reactions that were reported at a higher frequency in Kerendia‑treated patients and that occurred in 5% or more of patients receiving Kerendia were:

  • FIDELIO‑DKD study:
    • hyperkalemia (18.3% ‑ Kerendia vs 9.0% ‑ placebo)
    • decreased eGFR (6.3% ‑ Kerendia vs. 4.7% ‑ placebo)
  • FIGARO‑DKD study:
    • hyperkalemia (10.8% ‑ Kerendia vs. 5.3% ‑ placebo).

Adverse reactions that were reported in 1% or more of patients receiving Kerendia in the Phase III studies (pooled FIDELIO‑DKD and FIGARO‑DKD) and that were reported more frequently in patients receiving Kerendia than in patients receiving placebo included anemia (6.5% vs. 6.1%), hyperuricemia (5.1% vs. 3.9%), hyponatremia (1.3% vs. 0.7%), hypotension (4.6% vs. 3.9%), and pruritus (2.9% vs. 2.2%).

The safety of Kerendia during pregnancy and breastfeeding and in patients with severe hepatic impairment or end‑stage kidney failure is uncertain, since these populations were excluded from clinical studies. Patients with severe renal impairment (<25 mL/min/1.73 m2) were under‑represented. The long‑term safety of Kerendia is also uncertain. Appropriate cautionary statements, additional patient monitoring, dosage adjustments or interruptions, and descriptions of the findings and safety concerns are in place in the Kerendia Product Monograph to guide health professionals and patients on the safe and efficacious use of Kerendia.

Health Canada has determined that appropriate risk management measures are in place to address the safety concerns identified for Kerendia, and to support its safe and effective use. Overall, the safety profile of Kerendia was found to be acceptable for the approved indication.

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

7.2 Non-Clinical Basis for Decision

As described above, the review of the non‑clinical component of the New Drug Submission for Kerendia was conducted as per Method 3 described in the Draft Guidance Document: The Use of Foreign Reviews by Health Canada.

The non‑clinical program for Kerendia (finerenone) included evaluations of pharmacology, pharmacokinetics, general toxicology, carcinogenicity, and genetic toxicology. A complete set of reproductive toxicology studies was conducted, along with studies in juvenile rats and studies evaluating local tolerance and phototoxicity. Studies were conducted in rats, mice, rabbits, dogs, and monkeys. Finerenone was administered orally to reflect the intended route of administration in humans.

The effects observed in repeat‑dose toxicity studies were mainly due to exaggerated pharmacodynamic activities of finerenone and secondary adaptive responses. Reduced body weights were noted in rats receiving a dose equivalent to 17 times the exposure of the drug in humans as measured by the area under the concentration‑time curve of unbound (AUCunbound) 20 mg finerenone. Reduced prostate size and weight were reported in dogs starting with a dose equivalent to 10 times the human exposure based on AUCunbound of 20 mg finerenone. Histopathological changes in the adrenal glands were identified in both species. Additionally, changes in plasma electrolytes (sodium, potassium and calcium) were observed throughout rat toxicity studies. These findings were attributed to the pharmacological mode of action of finerenone and are ascribed to the compensatory activation of the adrenal glands following mineralocorticoid receptor blockade by finerenone.

Finerenone did not show carcinogenic potential in two‑year carcinogenicity studies in male and female rats and in female mice at doses representing 19 to 28 times the AUCunbound in humans. In male mice, an increase in Leydig cell adenoma was observed at doses representing 26 times the AUCunbound in humans. A dose representing 17 times the AUCunbound in humans did not cause any tumours. Based on the known sensitivity of rodents to develop these tumours, the pharmacology‑based mechanism at supratherapeutic doses, and adequate safety margins, the increase in Leydig cell tumours in male mice is not considered clinically relevant.

Finerenone was not found to be genotoxic based on results from a bacterial reverse mutation study, an in vitro chromosome aberration assay, and a micronucleus study in male mice. Results from in silico assessments indicated that finerenone and its metabolites are not mutagenic.

In rats, male fertility was not affected by finerenone at levels up to 16 times the AUCunbound in humans. Signs of reduced female fertility (decreased number of corpora lutea and implantation sites) and early embryonic toxicity (increased post‑implantational loss and decreased number of viable fetuses) were detected at doses of finerenone approximately 21 times the AUCunbound in humans. In addition, reduced ovarian weights were observed at approximately 17 times the AUCunbound in humans. No effects were observed on female fertility and early embryonic development at 10 times the AUCunbound in humans.

Signs of embryo‑fetal toxicity were observed in rats at the maternal toxic dose of 10 mg/kg/day, corresponding to 19 times the AUCunbound in humans. At 30 mg/kg/day, the incidence of visceral and skeletal variations was increased. Complex malformations were observed in one fetus, including a rare malformation (double aortic arch) at a dose corresponding to 25 times the AUCunbound in humans. The no‑observed-adverse‑effect level (NOAEL) (low dose in rats, high dose in rabbits) provided safety margins of 10 to 13 times for AUCunbound.

Finerenone had very low blood/brain penetration in rats, but penetration of the placental barrier in pregnant rats was evident. In lactating rats, finerenone and its metabolites were found to be excreted in milk. Following the administration of finerenone during pregnancy and lactation in the pre‑ and postnatal developmental toxicity study in rats, increased pup mortality and an increase in pup locomotor activity were observed starting at a dose equivalent to 4 times the AUCunbound in humans. Additional adverse events (e.g., lower pup weight, delayed pinna unfolding) were observed at a dose equivalent to 13 times the AUCunbound in humans. The NOAEL set by the study provided a safety margin of only 2‑fold for the proposed dose of finerenone in humans. Although the relevance of these findings for humans is unclear, finerenone is not recommended for use during pregnancy and breastfeeding. The increased locomotor activity in offspring may indicate a potential risk for the fetus. Additionally, due to the findings in pups, a risk for the nursing infant cannot be excluded.

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

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

7.3 Quality Basis for Decision

As described above, the review of the quality component of the New Drug Submission for Kerendia was conducted 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 Kerendia 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 submitted stability data, the proposed shelf life of 36 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.

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