Summary Basis of Decision for Adempas

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 Adempas is located below.

Recent Activity for Adempas

The SBDs written for eligible drugs (as outlined in Frequently Asked Questions: Summary Basis of Decision [SBD] Project: Phase II) 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. The PAATs will be updated regularly with post-authorization activity throughout the product life cycle.

Post-Authorization Activity Table (PAAT) for Adempas

Updated: 2024-07-08

The following table describes post-authorization activity for Adempas, a product which contains the medicinal ingredient riociguat. 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 found in Post-Authorization Activity Tables (PAATs).

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

Drug Identification Number (DIN):
  • DIN 02412764 - 0.5 mg riociguat, tablet, oral administration
  • DIN 02412772 - 1.0 mg riociguat, tablet, oral administration
  • DIN 02412799 - 1.5 mg riociguat, tablet, oral administration
  • DIN 02412802 - 2.0 mg riociguat, tablet, oral administration
  • DIN 02412810 - 2.5 mg riociguat, tablet, oral administration

Post-Authorization Activity Table (PAAT)

Activity/Submission Type, Control Number

Date Submitted

Decision and Date

Summary of Activities

SNDS # 262975

2022-03-31

Issued NOC 2022-10-13

Submission filed as a Level II – Supplement (Safety) to update the PM with new safety information, and migrate it to the 2020 format. The submission was reviewed and considered acceptable. As a result of the SNDS, modifications were made to the Contraindications and Drug Interactions sections of the PM, and corresponding changes were made to Patient Medication Information and to the package insert. An NOC was issued.

SNDS # 226431

2019-04-17

Issued NOC 2020-03-31

Submission filed as a Level I – Supplement to update the PM with new safety and efficacy information. The submission was reviewed and considered acceptable. As a result of the SNDS, modifications were made to the Warnings and Precautions; Drug Interactions; Dosage and Administration; and Clinical Trials, sections of the PM, and corresponding changes were made to Part III: Consumer Information and to the package insert. An NOC was issued.

NC # 198385

2016-09-14

Issued NOL 2016-12-08

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, additions were made to the Warnings and Precautions, and Drug Interactions sections of the PM, and corresponding changes were made to the PM Part III: Consumer Information. The submission was reviewed and considered acceptable, and an NOL was issued.

Dear Healthcare Professional Letter posted

Not applicable

Posted 2016-08-15

Dear Healthcare Professional Letter posted, containing important safety information for healthcare professionals and the general public.

NDS # 163446

2013-03-26

Issued NOC 2014-03-06

This NDS was submitted, prior to the initial approval of Adempas, to propose an additional new indication. The data were reviewed and the benefit/risk profile of Adempas was considered favorable in the new indication. An NOC was issued for the use of Adempas for the treatment of pulmonary arterial hypertension [(PAH) World Health Organization (WHO) Group 1] as monotherapy or in combination with endothelin receptor antagonists in adult patients (≥18 years of age) with WHO Functional Class II or III pulmonary hypertension.

Drug product (DINs 02412764, 02412772, 02412799, 02412802, 02412810) market notification

Not applicable

Date of first sale: 2013-09-25

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

NDS # 162761

2013-02-25

Issued NOC 2013-09-19

Notice of Compliance issued for New Drug Submission.

 
Summary Basis of Decision (SBD) for Adempas

Date SBD issued: 2013-12-10

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

Riociguat, 0.5 mg, 1.0 mg, 1.5 mg, 2.0 mg and 2.5 mg, tablets, oral

Drug Identification Number (DIN):

  • DIN 02412764 - 0.5 mg
  • DIN 02412772 - 1.0 mg
  • DIN 02412799 - 1.5 mg
  • DIN 02412802 - 2.0 mg
  • DIN 02412810 - 2.5 mg

Bayer Inc.

New Drug Submission Control Number: 162761

 

On September 19, 2013, Health Canada issued a Notice of Compliance for the drug product Adempas.

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 Adempas was considered favorable for the management of inoperable chronic thromboembolic pulmonary hypertension (CTEPH) [World Health Organization (WHO) Group 4] and persistent or recurrent CTEPH after surgical treatment in adult patients (≥18 years of age) with WHO Functional Class II or III pulmonary hypertension.

 

1 What was approved?

 

Adempas, a soluble guanylate cyclase (sGC) stimulator, was authorized for the management of inoperable chronic thromboembolic pulmonary hypertension [CTEPH, World Health Organization (WHO) Group 4], or persistent or recurrent CTEPH after surgical treatment, in adult patients (≥18 years of age) with WHO Functional Class II or III pulmonary hypertension.

Adempas is contraindicated for use concomitantly with other drugs affecting the nitric oxide-soluble guanylate cyclase-cyclic guanosine monophosphate (NO-sGC-cGMP) pathway, due to the risk of developing potentially life-threatening episodes of hypotension or syncope. These drugs include phosphodiesterase type 5 (PDE5) inhibitors [for example (e.g.,) sildenafil, tadalafil, vardenafil], nitrates taken either regularly or intermittently, in any form (e.g. oral, sublingual, transdermal, inhalation), and nitric oxide donors, such as amyl nitrate. Adempas is also contraindicated during pregnancy and nursing and in patients with a known hypersensitivity to Adempas or to any ingredient in the formulation. Adempas was approved for use under the conditions stated in the Adempas Product Monograph taking into consideration the potential risks associated with the administration of this drug product.

Adempas (0.5 mg, 1.0 mg, 1.5 mg, 2.0 mg and 2.5 mg riociguat) is presented as a film-coated tablet. In addition to the medicinal ingredient, the film-coated tablet contains cellulose microcrystalline, crospovidone, hypromellose 5cP, lactose monohydrate, magnesium stearate, sodium laurilsulfate. The film-coating is composed of hydroxypropylcellulose, hypromellose 3cP, propylene glycol, titanium dioxide. The Adempas 1.0 mg, 1.5 mg, 2.0 mg and 2.5 mg tablet film coating contain in addition ferric oxide yellow and the Adempas 2.0 mg and 2.5 mg tablet film coating contain in addition ferric oxide red.

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 Adempas Product Monograph, approved by Health Canada and available through the Drug Product Database.

 

2 Why was Adempas approved?

 

Health Canada considers that the benefit/risk profile of Adempas is favourable for the management of inoperable chronic thromboembolic pulmonary hypertension [CTEPH, World Health Organization (WHO) Group 4], or persistent or recurrent CTEPH after surgical treatment, in adult patients (≥18 years of age) with WHO Functional Class II or III pulmonary hypertension.

Chronic thromboembolic pulmonary hypertension (CTEPH), one of the leading causes of severe pulmonary hypertension (PH) and right heart failure, is primarily caused by venous thromboembolism (VTE), and as such is associated with significant morbidity and mortality. Although CTEPH can present at any age, more than half the adults are diagnosed during their peak productive years (ages 18-45 years). The treatment of choice is a surgical treatment by pulmonary endarterectomy (PEA), a procedure to physically remove pulmonary arterial thrombi. However, a substantial proportion of patients are not suitable for surgery. These patients, plus patients who present with persistent or recurrent pulmonary hypertension after PEA (approximately 30% of patients after PEA) are in need of disease modifying therapies. There are currently no approved pharmacotherapies for CTEPH.

Adempas has been shown to be efficacious for the management of inoperable CTEPH (WHO Group 4) or persistent or recurrent CTEPH after surgical treatment in adult patients (≥18 years of age) with WHO Functional Class II or III pulmonary hypertension.

The market authorization was based on data from one Phase III pivotal study (CHEST-1), one supportive Phase III study (PATENT-1) and their respective on-going-extension studies (CHEST-2 and PATENT-2).

The CHEST-1 study was a randomised, double-blind, placebo-controlled, multicentre, multi-national study to evaluate the efficacy and safety of Adempas [1.0 mg, 1.5 mg, 2.0 mg and 2.5 mg three times daily (TID)] in 261 patients with CTEPH. These patients were considered not to be eligible for PEA, or had recurrent CTEPH after an unsuccessful PEA. Data from CHEST-1 showed a statistically significant improvement in the primary efficacy variable - the standardized 6-minute walk test (6MWT). The Adempas-treated patients improved their walking distance in the 6MWT of 39 ± 79 m [standard deviation (SD)] at Week 16 compared to a deterioration of -6 ± 84 m in patients treated with placebo. Statistically significant improvements (p<0.001) in relevant secondary hemodynamic parameters [cardiac output (CO), pulmonary artery pressure, pulmonary vascular resistance (PVR)] were also noted. Also, relatively more patients on Adempas ended the study in a lower WHO functional class than patients on placebo.

Supportive evidence regarding efficacy came from study PATENT-1, a randomized, double-blind, placebo-controlled Phase III study in patients with pulmonary arterial hypertension (PAH). In PATENT-1, a total of 254 patients administered Adempas significantly improved their walking distance in the 6MWT compared to 127 patients on placebo.

Long-term maintenance of efficacy of Adempas was confirmed by two ongoing long-term studies (CHEST-2 and PATENT-2). At 18 months, the improvements in the 6MWT, pulmonary hemodynamic variables and clinical scales seen at Weeks 12 and 16 of the short-term controlled studies (PATENT-1 and CHEST-1) are maintained.

The safety evaluation for Adempas was conducted in more than 650 patients in Phase III clinical studies. The identified safety concerns include hypotension and its sequelae [that is (i.e.,) dizziness, loss of consciousness, or syncope], gastrointestinal disorders (i.e., dyspepsia, nausea, diarrhea, and vomiting), risk of bleeding and the over-additive blood pressure-lowering effects during co-administration with other drugs affecting the nitric oxide-soluble guanylatecyclase-cyclic guanosine monophosphate (NO-sGC-cGMP) pathway, such as phosphodiesterase type 5 (PDE5) inhibitors. These issues have been addressed through appropriate labelling in the Adempas Product Monograph.

The most common adverse events (AEs) occurring in ≥10% of patients treated with Adempas [up to 2.5 mg TID] were headache, dizziness, dyspepsia, peripheral edema, nausea, diarrhea, and vomiting. Frequencies of AEs were comparable between the CHEST-1 and PATENT-1 studies, with the exception of a lower rate of anemia in the CTEPH patients and a higher rate of dizziness.

There were 11 deaths in the pooled controlled studies; 5 in the Adempas groups and 6 in the placebo groups. In both groups, most fatal events were due to progression of the disease.

A Risk Management Plan (RMP) for Adempas was submitted by Bayer 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 therapeutic benefits seen in the pivotal study, the supportive study and the two extension studies are favourable and the benefits of therapy with Adempas are considered to outweigh the potential risks. Adempas 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 patient monitoring. Appropriate warnings and precautions are included in the Adempas 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 Adempas?

 

The drug submission for Adempas was reviewed and accepted under the Priority Review Policy. Sufficient evidence was submitted demonstrating Adempas provided an effective treatment for the management of inoperable chronic thromboembolic pulmonary hypertension (CTEPH) and persistent or recurrent CTEPH after surgical treatment, an insidious and debilitating disease for which no pharmaceutical treatment is presently marketed.

 

Submission Milestones: Adempas

Submission Milestone Date
Pre-submission meeting: 2012-11-01
Request for priority status  
Filed: 2012-12-20
Approval issued by the Bureau of Cardiology, Allergy and Neurological Sciences, Cardio-Renal Division: 2013-01-18
Submission filed: 2013-02-25
Screening  
Screening Acceptance Letter issued: 2013-03-25
Review  
Biopharmaceutics Evaluation complete: 2013-06-21
Quality Evaluation complete: 2013-09-12
Clinical Evaluation complete: 2013-09-17
Labelling Review complete: 2013-09-13
Notice of Compliance issued by Director General 2013-09-19

 

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:

 

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

 

Clinical Pharmacology

Adempas (riociguat) is a highly specific stimulator of the soluble guanylate cyclase (sGC), an enzyme in the cardiopulmonary system and the receptor for nitric oxide (NO). When NO binds to sGC, the enzyme enhances synthesis of the signaling molecule cyclic guanosine monophosphate (cGMP), which plays an important role in regulating vascular tone, proliferation, fibrosis, and inflammation.

Adempas has a dual mode of action: it sensitizes sGC to endogenous NO by stabilizing the NO-sGC binding. It also directly stimulates sGC via a different binding site, independently of NO. Adempas, as a stimulator of sGC, addresses NO deficiency by restoring the NO-sGC-cGMP pathway, leading to increased generation of cGMP. There is a direct relationship between the concentration of Adempas in the plasma and hemodynamic parameters, such as systemic and pulmonary vascular resistance, systolic blood pressure, and cardiac output.

The comprehensive clinical pharmacology program included studies in patients with chronic thromboembolic pulmonary hypertension (CTEPH), or symptomatic pulmonary arterial hypertension (PAH) and in hepatically impaired, renally impaired and healthy subjects.

A total of four safety concerns have been identified with the use of Adempas, namely, hypotension, co-administration with other drugs affecting the nitric oxide-soluble guanylatecyclase-cyclic guanosine monophosphate (NO-sGC-cGMP) pathway, gastro-intestinal disorders and risk of bleeding. As a sGC stimulator, Adempas acts as a vasodilatator, lowering the pulmonary and systemic blood pressures which can cause hypotension. Concomitant use of Adempas with nitrates or phosphodiesterase type 5 (PDE5) inhibitors can also cause hypotension by having a synergistic effect on the NO-sGC-cGMP pathway. Adempas has smooth muscle relaxation properties not only in the arterial wall but also in the gastrointestinal system, which can cause gastrointestinal disorders, such as dyspepsia, nausea, diarrhea and vomiting. Finally, the risk of bleeding with the use of Adempas was a very common adverse event (AE), particularly in patients receiving anticoagulation therapy. Idiopathic bleeding events were observed in 58 (11.8%) of the Adempas treated patients compared to 18 (8.4%) in the placebo group. There were 101 adverse bleeding events in the two controlled trials (CHEST-1 and PATENT-1), 12 of which were serious events (all in the Adempas group). Appropriate warnings and precautions are included in the approved Adempas Product Monograph.

Overall, the clinical pharmacological data support the use of Adempas for the specified indication. Appropriate warnings and precautions are in place in the approved Adempas Product Monograph to address the identified safety concerns.

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

Clinical Efficacy

The clinical efficacy of Adempas was evaluated in one pivotal Phase III study (CHEST-1) and one supportive study (PATENT-1).

The pivotal Phase III study was a randomized, double-blind, placebo-controlled, multicentre, multi-national study to evaluate the efficacy and safety of oral Adempas [1.0 mg, 1.5 mg, 2.0 mg, or 2.5 mg three times daily (TID)] in 261 patients aged from 19 to 80 years old, who were considered not eligible for pulmonary endarterectomy (PEA) (72%), or who had recurrent chronic thromboembolism pulmonary hypertension (CTEPH) (28%) after an unsuccessful PEA. The majority of patients had a World Health Organization (WHO) Functional Class II (31%) or III (64%) CTEPH at baseline and were mostly Caucasians (71%), female (66%), and middle-aged [59 ± 14 standard deviation (SD) years]. The mean baseline 6-minute walking distance (6MWD) for these patients was 347 m.

In total, 173 patients received Adempas and 88 patients received placebo for 16 weeks. During the first 8 weeks, patients were uptitrated to the highest tolerable dose. The dose of Adempas was titrated every two weeks based on the patient's systolic blood pressure and signs or symptoms of hypotension. The 8-week titration phase was followed by an 8-week main study phase.

The primary efficacy endpoint was a measure of exercise tolerance, described as the distance covered in a 6-minute walking test (6MWT) performed after 16 weeks in accordance with the American Thoracic Society guideline. This test reflects an individual's ability to perform daily physical activities The secondary endpoints were the change from baseline in pulmonary vascular resistance (PVR) after 16 weeks, change from baseline in N-terminal prohormone B-type natriuretic peptide (NT-proBNP) after 16 weeks [(NT-proBNP is a neurohormone release by the heart and thought to be a reliable indicator for cardiac failure)], change from baseline in WHO functional class after 16 weeks, time to clinical worsening (TTCW), change from baseline in Borg CR 10 Scale or Modified Borg Dyspnoea Scale (measured at the end of the 6MWT) after 16 weeks, change from baseline in European-quality of life 5-dimensions instrument (EQ-5D) questionnaire after 16 weeks and change from baseline in "Living with Pulmonary Hypertension (LPH)" questionnaire after 16 weeks.

Results at the 6MWT, which measures the distance a patient can walk on a flat, hard surface in 6 minutes, showed improvements from week 2 onward. At week 16, the mean increase in the 6MWD within the Adempas group was 46 m (least-squares) [95% Confidence Interval (CI): 25 m to 67 m; p<0.0001] compared to placebo. Improvements of Adempas over placebo were observed in all sub-groups evaluated, such as the inoperable patients [number of patients (n) = 189], who demonstrated an increase in 6MWD of 54 m (95% CI: 29 m to 79 m), and patients with recurrent or persisting CTEPH following PEA (n = 72), who demonstrated an increase in 6MWD of 27 m (95% CI: -10 m to 63 m). In patients with a WHO functional class of III/IV at baseline, Adempas led to a 53 m (95% CI: 27 m to 79 m) improvement in the 6MWD from baseline to week 16. In patients with a WHO functional class of I/II at baseline, the treatment effect was 26 m (95% CI: -9 m to 59 m). A larger proportion of patients in the Adempas group (1.0 to 2.5 mg) had an improvement in 6MWD of at least 30 m by week 16 compared to the placebo group: [63% versus (vs.) 30%].

Improvements across all the secondary efficacy endpoints were observed in the Adempas treatment group. A reduction of PVR against placebo of 246 dyn·sec/cm5(95% CI: -303 to -190, p<0.0001) was observed in the intent-to-treat (ITT) population. The baseline for PVR was 791 ± 432 dyn·sec/cm5 in the Adempas group and 779 ± 401 dyn·sec/cm5 in the placebo group. Levels of NT-proBNP were significantly reduced in the Adempas group; placebo-corrected mean change from baseline was -444 pg/mL (95% CI -843 to -45, p<0.0001). A total of 32.9% of patients in the individual dose titration group (riociguat 1.0 to 2.5 mg) compared to 14.9% patients in the placebo group had an improvement of at least one WHO functional class (p = 0.0026). The secondary endpoint of Time To Clinical Worsening (TTCW) was a combined endpoint of death (all-cause mortality) and events reflective of residual clinical worsening. The sum of events leading to clinical worsening had a lower event rate in the Adempas group (2.3%) than in the placebo group (5.7%), but the difference was not considered statistically significant (p = 0.1724). Benefit was observed in both inoperable and operable CTEPH patients. Baseline values for the Borg CR 10 Scale were comparable in both treatment groups. Between baseline and week 16, the mean Borg CR 10 Scale decreased in the Adempas group (mean change of -0.83), but remained almost unchanged in the placebo group (mean change of 0.17). The treatment difference between the Adempas group and the placebo group was statistically significant (p = 0.0035). However, since the Borg Scale was below clinical worsening in the hierarchical testing order for secondary variables, statistical significance cannot be formally claimed for this variable. Also for EQ-5D, although statistical significance in the improvement of an individual's perceived quality of life cannot be formally claimed, the self-reported score improved for the Adempas group while it worsened for the placebo group (+0.0615 ± 0.2768 vs. -0.0819 ± 0.3446 placebo; p<0.0001). The last secondary variable, the LPH questionnaire, is a self-report measuring the effects of pulmonary hypertension (PH) and PH-specific treatments on an individual's quality of life. The results showed that, at the last visit, patients in the Adempas group had a statistically nonsignificant improvement in perceived quality of life compared to patients in the placebo group (-6.72 ± 18.62. vs. -2.09 ± 19.31 placebo; p = 0.1220).

Supportive Studies

Supportive evidence regarding efficacy comes from a Phase III, randomised, double-blind, placebo-controlled, multicentre, multinational study (PATENT-1) to evaluate the efficacy and safety of Adempas in patients with symptomatic pulmonary arterial hypertension (PAH). A total of 586 patients were enrolled in the 12 week study. Of the 586 patients enrolled, 445 were randomized and 443 received study medication (254 in the Adempas individual titration (IDT) group, 126 in the placebo group and 63 in the Adempas 1.0-1.5 mg group).

In PATENT-1, the patients on Adempas significantly improved their walking distance in the 6MWT from baseline to week 12 compared to the patients on placebo. The estimated, placebo-corrected overall treatment effect was 35.78 m (95% CI: 20.06 m to 51.51 m; p<0.0001). Pulmonary hemodynamic and clinically important secondary endpoint variables (CO, PVR, Borg dyspnea scale and change in WHO functional class) also pointed at statistically significant improvements of patients on Adempas.

The long-term maintenance of efficacy was confirmed by 2 ongoing long-term studies (CHEST-2 and PATENT-2). At 18 months, the improvements in the 6MWD, pulmonary hemodynamic variables and clinical scales seen at Weeks 12 and 16 of the short-term controlled studies were maintained (data cut-off April/May 2012).

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

Clinical Safety

The clinical safety evaluation was based primarily on data from one pivotal study (CHEST-1) and one supportive study (PATENT-1) previously described in the Clinical Efficacy section. The safety of Adempas was also assessed in two long term open-label extension studies (CHEST-2 and PATENT-2).

CHEST-2 was a long-term extension, multicentre, multinational study to evaluate the safety and tolerability of Adempas (1.0 mg, 1.5 mg, 2.0 mg or 2.5 mg TID) in patients with CTEPH (n = 237 patients from CHEST-1). The mean treatment duration at the cut-off date was 388 days with a median duration of 336 days (range 15 to 989 days) and a total treatment exposure of 206 person years. PATENT-2 was a long-term extension, multi-centre, multi-national study to evaluate the safety and tolerability of Adempas (1.0 mg, 1.5 mg, 2.0 mg or 2.5 mg TID) in patients with symptomatic PAH (n = 396 patients from PATENT-1).

The safety analyses included evaluation of adverse events (AE), serious adverse events (SAE), and deaths. In the pooled controlled studies (CHEST-1 and PATENT-1), 490 patients were exposed to Adempas and 214 to placebo. In the pooled extension studies (CHEST-2 and PATENT-2), 557 patients were treated with Adempas.

Considering Adempas is a vasodilator, common AEs observed in the Phase III clinical studies were dizziness (19.8% of patients in the Adempas group compared to 13.1% in the placebo group), (pre)syncope and hypotension (10% of patients in the Adempas group compared to 3.7% in the placebo group). Bleeding was also a very common AE. Idiopathic bleeding events were observed in 58 (11.8%) of the Adempas-treated patients compared to 18 (8.4%) in the placebo group. Of the 58 cases, 10 cases were considered serious adverse events (SAE) and 1 case was fatal. Pulmonary hemorrhage was reported in less than 1% of patients treated during long-term extension studies with Adempas. Anemia also occurred commonly in the pooled Phase III studies. Anemia (or respective changes in laboratory values reported as an AE) was noted in 33 (6.7%) of the Adempas-treated patients, in 2 of these cases as an SAE. Anemia occurred in 5 (2.3%) of the patients on placebo, once as an SAE.

The overall rates of discontinuation due to an AE in the pooled Phase III studies (CHEST 1 and PATENT 1) were 2.9% for Adempas and 5.1% for placebo.

Treatment-emergent adverse events (TEAEs) were defined as any AE occurring after the first dose administration until 48 hours after the last dose administration. The most frequent TEAEs (> 10%) in each treatment group of CHEST-1 and PATENT-1 studies were headache (26.9%), dizziness (19.8%), dyspepsia (18.6%), peripheral edema (17.3%), nausea (14.1%), diarrhea (12.0%), bleeding (11.8%), and vomiting (10.2%).

There were 11 deaths in the pooled controlled studies (CHEST-1 and PATENT-1); 5 in the Adempas groups and 6 in the placebo groups. In both groups, most fatal events were due to progression of the disease. Sepsis occurred in one fatal case, and death from hemorrhage or hemoptysis occurred in 2 patients on Adempas. There were 23 deaths in the pooled extension studies (CHEST-2 and PATENT-2). Most fatalities (n = 15) were due to progression of the disease: pulmonary arterial hypertension (PAH), cardiac arrest, right ventricular failure, cardiac failure and cardiogenic shock. Hemorrhage was the cause of 3 deaths. Other causes of death were: sudden cardiac death, sepsis, shock, and lung cancer.

Frequencies of adverse events were comparable between the CHEST-1 and PATENT-1 studies, with the exception of a lower rate of anemia in the CTEPH patients and a higher rate of dizziness.

Overall, Adempas is well-tolerated. The submitted data support the safety of Adempas for use in patients with CTEPH who are not considered candidates for surgical treatment, or who have persistent or recurrent pulmonary hypertension post-operatively. There were no signs of cardiac, renal or hepatic toxicity. The risk of hypotension is minimized by a gradual individual dose titration to the highest tolerated dose, and by contraindicating concomitant use with other drugs affecting the NO-sGC-cGMP pathway. Gastrointestinal disorders occurred very commonly, but seemed to be manageable, as few subjects discontinued from the clinical studies. The apparent increased risk of bleeding has been addressed in the label by means of a prominent warning and a description of bleeding events in the adverse reactions section of the Product Monograph. The risk of pulmonary bleeding and hemoptysis may be avoided by contraindicating its use in patients with a history of hemoptysis. As stated in the Product Monograph, patients with a history of serious hemoptysis should avoid the use of Adempas. For more information, refer to the Adempas Product Monograph, approved by Health Canada and available through the Drug Product Database.

 

 

 

7.2 Non-Clinical Basis for Decision

 

The non-clinical pharmacology and toxicology studies support the use of riociguat (the medicinal ingredient in Adempas) for the management of inoperable chronic thromboembolic pulmonary hypertension (CTEPH) [World Health Organization (WHO) Group 4], or persistent or recurrent CTEPH after surgical treatment, in adult patients (≥18 years of age) with WHO Functional Class II or III pulmonary hypertension.

In order to support the human use of Adempas, which acts as a stimulator of soluble guanylatecyclase (sGC), a comprehensive set of non-clinical studies were conducted in in vitro assays and in vivo with various animal models. As such, the potential toxicity of riociguat was comprehensively evaluated in a program of: oral and intravenous single-dose studies in rodents, and oral repeat-dose general toxicity studies in rodents (up to 13 weeks in mice; up to 26 weeks in rats) and dogs (up to 52 weeks); in vitro and in vivo genotoxicity studies; 2-year carcinogenicity studies in mice and rats; and reproductive and developmental studies. The majority of the findings in oral repeat-dose studies were attributed to effects related to the pharmacological properties of riociguat, notably reduced gastrointestinal tract motility due to smooth muscle relaxation, as well as reduced blood pressure and increased heart rate secondary to vasodilation.

Cardiovascular pathological findings such as endocarditis, perivascular edema and tunica media hypertrophy of coronary vessels related to treatment with Adempas were observed in 13-and 26-week repeat-dose toxicity studies in dogs, without clear overall correlation between the morphological lesions and hemodynamic changes on an individual animal basis. Pathological changes in bone and/or growth plates were observed in young rats, but not in mature rats and dogs. Riociguat was not genotoxic in a battery of in vitro and in vivo tests and was not carcinogenic in rats at exposures up to 2 times the maximum recommended human dose. Adempas is contraindicated in women who are or may become pregnant due to significant maternal and fetal toxicity observed in rats and rabbits. The main metabolite of riociguat, M1, demonstrated a comparable but less potent pharmacological profile and a similar general toxicology profile as riociguat. Overall, the non-clinical pharmacodynamics, pharmacokinetics, safety pharmacology and toxicology support the clinical use of Adempas.

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

For more information, refer to the Adempas 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 Adempas has demonstrated that the drug substance and drug product can be consistently manufactured to meet the approved specifications. Proper development and validation studies were conducted, and adequate controls are in place for the commercial processes. Changes to the manufacturing process and formulation made throughout the pharmaceutical development are considered acceptable upon review. Based on the stability data submitted, the proposed shelf-life is considered acceptable.

Proposed limits of drug-related impurities are considered adequately qualified [that is (i.e.) within International Conference on Harmonisation (ICH)] and are limited to 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.