Summary Basis of Decision for Givlaari

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 Givlaari is located below.

Recent Activity for Givlaari

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.

Summary Basis of Decision (SBD) for Givlaari

Date SBD issued: 2021-01-21

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

Givosiran (supplied as givosiran sodium)

Drug Identification Number (DIN):

  • DIN 02506343 - 189 mg/mL givosiran, solution, subcutaneous injection

Alnylam Netherlands B.V.

New Drug Submission Control Number: 237194

On October 9, 2020, Health Canada issued a Notice of Compliance to Alnylam Netherlands B.V. for the drug product Givlaari.

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 Givlaari is favourable for the treatment of acute hepatic porphyria in adults.

1 What was approved?

Givlaari was authorized for the treatment of acute hepatic porphyria in adults. Givosiran, the medicinal ingredient in Givlaari, is a double-stranded small interfering ribonucleic acid (siRNA) that specifically targets 5'-aminolevulinate synthase 1 (ALAS1) messenger RNA (mRNA).

Based on the data submitted to and reviewed by Health Canada, the safety and efficacy of Givlaari in pediatric patients have not been established. Therefore, Health Canada has not authorized an indication for pediatric use.

Clinical studies with Givlaari did not include sufficient numbers of patients to determine if use in geriatric patients is associated with differences in safety or effectiveness.

Givlaari is contraindicated in patients with known severe hypersensitivity (e.g., anaphylaxis) to givosiran or to any ingredient in the formulation, including any non-medicinal ingredient, or component of the container.

Givlaari was approved for use under the conditions stated in its Product Monograph taking into consideration the potential risks associated with the administration of this drug product.

Givlaari (189 mg/mL givosiran, supplied as givosiran sodium) is presented as a solution. In addition to the medicinal ingredient, the solution contains water for injection, phosphoric acid and/or sodium hydroxide (for pH adjustment).

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

2 Why was Givlaari approved?

Health Canada considers that the benefit-risk profile of Givlaari is favourable for the treatment of acute hepatic porphyria (AHP) in adults.

Porphyrias constitute a group of rare, serious, and debilitating disorders caused by deficiencies in the enzymes involved in the production of the heme molecule. Heme is an essential component of the hemoproteins, which include hemoglobin and cytochrome P450 enzymes. When a step in the heme synthetic pathway is blocked, one or more toxic intermediate molecules, known as porphyrins or porphyrin precursors, accumulate. In AHP, the overproduction and initial accumulation of porphyrin precursors or porphyrins occurs first in the liver.

The AHP family consists of four subtypes, each involving a defect in a distinct heme pathway enzyme. Acute intermittent porphyria (AIP) is the most common AHP subtype, representing 80% of all cases. The other three subtypes include aminolevulinic acid (ALA) dehydratase deficiency porphyria (also known as "ALAD deficiency porphyria" or "ADP"), hereditary coproporphyria (HCP) and variegate porphyria (VP). All subtypes of AHP share common pathophysiology, clinical manifestations, and options for treatment. The disease presentation is similar in pediatric and adult patients, although in the pediatric population boys are more commonly affected than girls. In the adult population, there appears to be an increased incidence in females. The estimated overall global prevalence of symptomatic AHP is approximately 1 to 5 per 100,000 individuals.

In AHP patients, the overproduction of toxic heme intermediates in the liver leads to injury of the central, peripheral, and autonomic nervous systems. Clinical manifestations are considered to be primarily due to the accumulation of ALA and porphobilinogen (PBG) as a result of the induced expression of hepatic aminolevulinate acid synthase-1 (ALAS1). Patients with AHP may experience acute, incapacitating, and potentially life-threatening neurovisceral attacks. The signs and symptoms are variable and reflect damage to the autonomic nervous system (severe abdominal pain, nausea, vomiting, constipation, tachycardia, labile hypertension, and postural hypotension), peripheral nervous system (diffuse muscle weakness, pain in back and limbs, cranial neuropathies, and respiratory muscle weakness), and/or central nervous system (anxiety, insomnia, confusion, agitation, hallucinations, and seizures). Other manifestations may include hyponatremia, transaminase elevations, lipase elevations, and altered renal function (increased creatinine and decreased estimated glomerular filtration rate).

Acute porphyria attacks typically require urgent medical attention in a healthcare setting. Some attacks with progressive motor weakness, paralysis, or respiratory failure require prolonged hospitalization and rehabilitation, and neurologic damage can be permanent. Each attack is serious, highly morbid, and carries potential for permanent disability. Where specific treatment is delayed or not available, attacks can be life threatening. Porphyria disease activity negatively affects quality of life and physical functioning. The goal of therapy is supportive and to promptly terminate the attack to avoid permanent neurologic injury. Due to the unpredictability and severity of porphyria disease manifestations, many patients are unable to work or attend school, have decreased socialization, and increased rates of depression and anxiety.

Current treatment options for acute AHP attacks are limited and include supportive care with intravenous fluids and glucose, a high carbohydrate diet, opioid analgesics, antiemetics, and anti-seizure medications as indicated, as well as the removal of known precipitating triggers. In Canada, treatment for AHP is not adequately managed by the current available treatments. Panhematin (hemin), a human blood-derived heme formulation for intravenous administration, is often used as a treatment. However, in Canada its authorization is limited to the treatment of recurrent attacks of AIP temporally related to the menstrual cycle, after initial carbohydrate therapy is known or suspected to be inadequate. It is not authorized for the chronic treatment of AHP attacks. Hemin infusion has a short duration of action that limits its effectiveness. This treatment also requires intravenous administration with infusions lasting at least 30 minutes and carries a long-term risk of iron overload and venous inflammation (e.g., phlebitis). Liver transplantation may be the only treatment available for patients with refractory disease that includes recurrent attacks, poor response to heme therapy, and neurologic progression. Although transplantation can be curative for these patients, it is highly invasive and not widely used due to limited organ availability and the risks associated with life-long immunosuppression.

The medicinal ingredient in Givlaari, givosiran, is a double-stranded small interfering ribonucleic acid (siRNA) that causes degradation of ALAS-1 messenger RNA (mRNA) in hepatocytes through RNA interference, thereby reducing the elevated levels of liver ALAS-1 mRNA. This leads to reduced circulating levels of the neurotoxic intermediates ALA and PBG, factors associated with attacks and other disease manifestations of AHP.

The clinical efficacy of Givlaari for the treatment of AHP was primarily demonstrated in a randomized, double-blind, placebo-controlled, pivotal Phase III study (ENVISION), which was followed by an open-label extension study. The study enrolled 94 adults, randomized in a 1:1 ratio to receive monthly subcutaneous injections of Givlaari 2.5 mg/kg (48 patients) or placebo (46 patients) over a 6-month double-blind period. Treatment groups were stratified by AHP type. Patients with AIP were further stratified by each patient's use of a hemin prophylaxis regimen at the time of screening and by each patient's historical annualized attack rate.

The primary endpoint of ENVISION was the annualized rate of composite porphyria attacks over the 6-month double-blind period in mutation-confirmed patients with AIP. These patients constituted 89 of the 94 AHP patients. The composite porphyria attacks consisted of attacks requiring hospitalization, an urgent healthcare visit, or intravenous hemin administration at home.

Key secondary endpoints included the composite annualized attack rate for all AHP patients during the 6-month double-blind period, measurement of urinary ALA and PBG at various times, as well as hemin doses administered, pain, fatigue and nausea scores, and other parameters.

During the 6-month double-blind period, patients with AIP receiving Givlaari demonstrated a statistically significant reduction of 74% in the composite annualized attack rate compared to placebo-treated patients. The mean annualized attack rate was 3.2 (95% confidence interval [CI], 2.3-4.6) for patients receiving Givlaari compared to 12.5 (95% CI, 9.4-16.8) for patients receiving placebo. During the study period, 50.0% of patients with AIP receiving Givlaari had no composite attacks versus 16.3% receiving placebo. In addition, a 73% reduction in the annualized attack rate was observed for all AHP patients, a key secondary endpoint. Results for each of the three individual components of the composite primary efficacy endpoint were consistent with the treatment benefit of Givlaari observed in patients with AIP. A continued benefit in attack reduction was evident in patients who remained on Givlaari in the open-label extension study. Patients randomized to placebo in the double-blind period who crossed over to Givlaari in the open-label extension period displayed a similar reduction in attacks compared to patients who were originally randomized to Givlaari in the double-blind period.

Among the secondary endpoints, Givlaari demonstrated statistically significant benefits in patients with AIP for mean annualized days of intravenous hemin use, reduced urinary ALA and PBG levels, and reduced worst daily pain scores.

The clinical safety of Givlaari was assessed using data from the pivotal ENVISION study, the open-label extension study, a supportive study (Study 002), and the clinical development plan. The open-label extension study assessed the long-term safety and efficacy of Givlaari and was ongoing at the time of authorization.

In the pivotal study, adverse reactions were defined as adverse events that occurred at least 5% more frequently in patients treated with Givlaari compared to the placebo. The most frequently occurring adverse reactions reported in at least 10% of patients in patients treated with Givlaari vs. placebo-treated patients were nausea (27% vs. 11%) and injection site reactions (25% vs. 0%). Other frequent (experienced by at least 5%) adverse events reported in patients treated with Givlaari versus placebo, respectively, included fatigue (10% vs. 4%), increased transaminases (13% vs. 2%), increased blood creatinine (15% vs. 4 %) and rash (17% vs. 4%). One adverse reaction, elevated transaminases, resulted in the discontinuation of Givlaari (one patient, 2.1%).

Overall, exposure to Givlaari was limited in duration, but the data provided were considered acceptable. In general, the study population is considered representative of the target population for treatment with Givlaari. Clinical data were not provided for adolescents (12 to 18 years old) due to the rarity of the disease expression in this age group. Data in the geriatric population (65 years or older) or in patients with various stages of hepatic impairment and severe renal impairment were either not provided or were limited. The use of Givlaari in patients with moderate or severe hepatic impairment and in patients with end stage renal disease or on dialysis were included as missing information in the Risk Management Plan (RMP).

An increased incidence of chronic kidney disease (increased creatinine and/or decreased estimated glomerular filtration rate) was observed. Most cases were transient in nature. These cases raise a safety concern for patients with pre-existing severe chronic kidney disease, as no dedicated severe renal impairment study was conducted. Furthermore, serious adverse events of worsening of chronic kidney disease and abnormal liver function tests occurred in some patients. The serious adverse event of abnormal liver function tests led to discontinuation of Givlaari in one patient. In addition, one adverse drug reaction of anaphylaxis also occurred in a patient who subsequently withdrew from the study.

The interpretation of the safety data is confounded by the associated chronic kidney disease and liver dysfunction/damage that are seen as comorbid diseases in patients with AHP. However, it is possible that these disorders may be exacerbated by Givlaari treatment and this will need to be monitored closely in the RMP.

The efficacy and safety data regarding the use of Givlaari in patients with AHP subtypes other than AIP (i.e., HCP, VP, and ADP) are limited. This paucity of data should be taken into consideration when assessing the risk-benefit profile of Givlaari therapy in patients with these rare subtypes of AHP.

An RMP for Givlaari was submitted by Alnylam Netherlands B.V. 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.

The submitted inner and outer labels, package insert and Patient Medication Information section of the Givlaari Product Monograph meet the necessary regulatory labelling, plain language and design element requirements. A review of the submitted brand name assessment, including testing for look‑alike sound‑alike attributes, was conducted and the proposed name Givlaari was accepted.

Based on the non-clinical and clinical studies, Givlaari has been shown to have a favourable benefit-risk profile for the target patient population. Considering the nature of the disease and the scarcity of approved targeted treatments, the clinical benefits of Givlaari outweigh the currently known risks associated with its use. The identified safety issues can be managed through labelling and adequate monitoring. Appropriate warnings and precautions are in place in the Givlaari Product Monograph to address the identified safety concerns. The collection and analysis of long-term data will be necessary along with the RMP to better characterize the short- and long-term effects of Givlaari.

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 Givlaari?

The drug submission for Givlaari was reviewed under the Priority Review Policy. The sponsor submitted substantial evidence of clinical effectiveness demonstrating that Givlaari provides a significant increase in efficacy and has an improved benefit-risk profile compared to existing therapies for acute hepatic porphyria, a serious, life-threatening, and severely debilitating disease not adequately managed by a drug marketed in Canada.

Submission Milestones: Givlaari

Submission MilestoneDate
Pre-submission meeting2019-11-19
Request for priority status
Filed2020-01-24
Approval issued by Director, Bureau of Medical Sciences2020-02-24
Submission filed2020-03-19
Screening
Screening Acceptance Letter issued2020-04-15
Review
Review of Risk Management Plan complete2020-09-15
Quality Evaluation complete2020-10-05
Labelling Review complete2020-10-06
Clinical/Medical Evaluation complete2020-10-08
Notice of Compliance issued by Director General, Therapeutic Products Directorate2020-10-09

The Canadian regulatory decision on the review of Givlaari was based on a critical assessment of the data package submitted to Health Canada. The foreign review completed by the United States Food and Drug Administration (FDA) was used as an added reference.

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

The acute hepatic phorphyria (AHP) family consists of four subtypes, each involving a genetic mutation in a distinct heme pathway enzyme. The subtypes are as follows:

  • acute intermittent porphyria (AIP) - caused by mutations in the hydroxymethylbilane synthase (also known as porphobilinogen [PBG] deaminase) gene;
  • hereditary coproporphyria (HCP) - caused by mutations in the coproporphyrinogen oxidase gene;
  • variegate porphyria (VP) - caused by mutations in the protoporphyrinogen oxidase gene; and
  • aminolevulinic acid (ALA) dehydratase-deficient porphyria (ADP) - caused by mutations in the ALA dehydratase gene.

Several lines of evidence indicate that ALA and PBG are central to the pathophysiology of AHP disease and are causal mediators of injury. Both ALA and PBG have been shown to directly induce oxidative stress, inflammation, vasoconstriction, and cell death in vitro in multiple cell types and tissues, including the kidney, liver and nervous system. In patients who received a liver transplantation due to severe recurring porphyria attacks, ALA and PBG levels normalized within a few days of surgery and porphyria attacks and chronic porphyria-related symptoms resolved.

Givosiran, the medicinal ingredient in Givlaari, is a double-stranded small interfering ribonucleic acid (siRNA) that causes degradation of aminolevulinate synthase 1 (ALAS-1) messenger ribonucleic acid (mRNA) in hepatocytes through RNA interference, reducing the elevated levels of liver ALAS-1 mRNA. This leads to reduced circulating levels of the elevated toxic heme intermediates ALA and PBG thereby preventing or reducing neurovisceral attacks and ongoing symptoms in all AHP disease subtypes.

The clinical pharmacology program consisted of biomarker, pharmacokinetic, pharmacodynamics, and dose-titration studies to assess the subcutaneous administration of givosiran 2.5 mg/kg once monthly in healthy volunteers and in AHP patients.

Clinical data showed that following subcutaneous administration, givosiran was rapidly absorbed into the plasma and was primarily distributed to the liver where it was metabolized by nucleases to shorter oligonucleotides and an equipotent active metabolite, AS (N-1)3' givosiran that represents 45% of givosiran exposures.

In givosiran dose ranges of 0.035 to 5 mg/kg, dose-dependent reductions in urinary ALAS-1 mRNA, ALA and PBG levels were observed. In AHP patients receiving monthly doses of 2.5 mg/kg givosiran, maximal reductions of ALA and PBG levels were achieved by 3 months. Modelling of the urinary ALA reduction and clinical activity, as measured by the incidence of porphyria attack (i.e., the annualized attack rate [AAR]) demonstrated that givosiran 2.5 mg/kg monthly dosing reduced the AAR to 2.9 attacks/year compared to 4.2 attacks/year with 2.5 mg/kg quarterly dosing and 12.5 attacks/year with placebo. These data support givosiran 2.5 mg/kg monthly dosing as the optimal dose regimen.

Given its mechanism of action, givosiran can lower hepatic heme levels and could reduce the activity of heme-dependent proteins in the liver such as drug metabolizing cytochrome P450 (CYP450) enzymes. The review of data showed that givosiran treatment had an important impact on some CYP450 enzyme activity. In vitro studies indicated that givosiran does not directly inhibit or induce CYP enzymes. However, clinical drug interaction studies demonstrated that the concomitant use of a single subcutaneous dose of givosiran 2.5 mg/kg:

  • increased caffeine (sensitive CYP1A2 substrate) blood concentration with time [area under the concentration versus time curve (AUC)] by 3.0‑fold and maximum concentration (Cmax) by 1.3-fold, and
  • increased dextromethorphan (sensitive CYP2D6 substrate) blood concentration with time (AUC) by 2.4‑fold and maximum concentration (Cmax) by 2.0‑fold.

This is important as multi-drug therapy is common in AHP patients due to multiple comorbidities, such as chronic pain, depression, and hypertension. As such, patients should avoid concomitant use of givosiran with CYP1A2 or CYP2D6 substrates for which minimal concentration changes may lead to serious or life-threatening toxicities. If concomitant use is unavoidable, a decrease in CYP1A2 or CYP2D6 substrate drug dosage is recommended.

No dosage adjustment of CYP2C9, CYP2C19 and CYP3A4 substrates is recommended when coadministered with givosiran.

The population pharmacokinetics and the pharmacokinetic/pharmacodynamic analysis did not reveal any clinically meaningful differences in the pharmacokinetics or pharmacodynamics of givosiran based on age, sex, or race/ethnicity.

For adolescent (≥12 to <18 years old) patients, predictive pharmacokinetic and pharmacodynamic simulations were conducted assuming a body weight of 40 kg (the median body weight for 12-year old boys and girls based on Centers for Disease Control and Prevention [CDC] growth charts) and compared with adults (≥18 years old; a body weight of 66 kg) for monthly doses of givosiran 2.5 mg/kg. Predicted mean plasma exposures (Cmax and AUC) of givosiran in adolescents were within 22% of that in adults. The predicted mean urinary ALA level in adolescents after 2.5 mg/kg once-monthly doses of givosiran was 1.26 mmol/mol creatinine which was comparable to 1.18 mmol/mol creatinine in adults.

While age is not a covariate in population pharmacokinetic analysis, clinical studies of givosiran did not include a sufficient number of patients to determine the effectiveness of the drug in the geriatric population.

Based on the population pharmacokinetic and the pharmacokinetic/pharmacodynamic (percent reduction in urinary ALA and PBG) analyses, caution is recommended in patients with severe renal impairment. No dosage adjustment is necessary in patients with mild or moderate renal impairment. Population pharmacokinetic data indicated that the exposures of givosiran were predicted to increase by 14% for givosiran and by 43% for AS(N-1)3' givosiran (an active metabolite with equal potency as givosiran) in patients with mild-to-severe renal impairment compared to patients with normal renal function, which is not considered clinically significant. The effect of end-stage renal disease (estimated glomerular filtration rate <15 mL/min/1.73 m2) on givosiran pharmacokinetics is unknown.

No dosage adjustment is necessary in patients with mild hepatic impairment based on the population pharmacokinetics and pharmacokinetic/pharmacodynamic analysis. Hepatic impairment was not a significant covariate in the population pharmacokinetic analysis. The effect of moderate to severe hepatic impairment on the pharmacokinetics of givosiran has not been evaluated.

A dedicated thorough QT study has not been conducted with givosiran. The effect of givosiran on the corrected QT interval (QTc) was evaluated in a double blind, placebo-controlled study and the open-label extension study. No large mean increase in QTc (i.e., >20 ms) was detected at the 2.5 mg/kg once monthly dose level. The administration of single or multiple doses of givosiran was not associated with clinically relevant changes in the QTc interval, including at plasma concentrations approximately 2- to 3-fold higher than the expected plasma Cmax at the therapeutic dose of 2.5 mg/kg once monthly in AHP patients.

The clinical pharmacology data support the use of Givlaari for the recommended indication. For further details, please refer to the Givlaari Product Monograph, approved by Health Canada and available through the Drug Product Database.

Clinical Efficacy

The clinical efficacy of Givlaari was evaluated in the pivotal study ENVISION, a randomized, double-blind, placebo-controlled, multicentre, Phase III study with a subsequent open-label extension period.

The ENVISION study enrolled 94 adult patients with acute hepatic porphyria. These patients were randomized in a ratio of 1:1 to receive 2.5 mg/kg Givlaari (48 patients) or a placebo (46 patients) over a 6-month double-blind period.

Key inclusion criteria for the study were: patients aged 12 years and older, who had experienced a minimum of two porphyria attacks requiring hospitalization, an urgent healthcare visit, or intravenous hemin administration at home in the 6 months prior to study entry. Patients had to be willing to discontinue and/or not initiate prophylactic hemin. Hemin use was permitted during the study for the treatment of acute porphyria attacks. Patients were required to have a diagnosis of AHP (AIP, HCP, VP or ADP) based on clinical features, at least one documented urinary or plasma PBG or ALA value greater than or equal to the upper limit of normal within the past year prior to or during screening, and documented genetic evidence of a mutation in a porphyria-related gene. For patients whose genetic testing did not identify a mutation in a porphyria-related gene (<5% of cases), the patient could be eligible for inclusion if they had both clinical features and diagnostic biochemical criteria consistent with AHP.

The primary endpoint of the study was the effect of subcutaneously administered Givlaari as compared to placebo in AIP patients for the composite annualized porphyria attack rate (AAR) during the 6-month double-blind period. The composite annualized porphyria attack rate was defined as the annualized rate of porphyria attacks requiring hospitalization, an urgent healthcare visit, or intravenous hemin administration at home. Porphyria attacks were defined as:

  • an acute episode of neurovisceral pain in the abdomen, back, chest, extremities and/or limbs, with no other medically determined cause, and
  • requiring treatment with intravenous dextrose or hemin, carbohydrates, analgesics, or other medications such as antiemetics at a dose or frequency beyond the participant's usual daily porphyria management.

A key secondary endpoint was the effect of Givlaari as compared to placebo in all AHP patients for the composite AAR during the 6-month double-blind period. Other secondary endpoints included an evaluation of the effects of Givlaari compared to placebo with respect to urinary ALA and PBG levels and hemin usage in AIP patients. The rate of porphyria attacks requiring hospitalization, an urgent healthcare visit, or intravenous hemin administration at home in patients was also evaluated in AHP patients.

Other patient-reported outcomes analyzed as secondary endpoints were the change from baseline in score on the Physical Component Summary (PCS) of the 12-item Short-Form Health Survey, version 2 (SF-12).

The primary endpoint of the study was met as the results demonstrated a statistically significant reduction of 74% in the composite AAR in AIP patients compared to placebo (rate ratio [RR] = 0.26, p <0.0001). These results were clinically meaningful and consistently produced significant reductions in all three composite components of the composite AAR (hospitalization, urgent care visit, and intravenous hemin use). In addition, a 73% reduction was observed in the composite AAR among all AHP patients, a key secondary endpoint (RR = 0.27, p <0.0001).

Decreases in the composite AAR were observed after the first dose of Givlaari and were maintained throughout the study. The statistically significant difference of treatment with Givlaari versus placebo was observed for each non-overlapping component of the composite AAR endpoint, pre-specified sensitivity analyses, and all pre-specified subgroup analyses (by age, sex, race, region, baseline body mass index, prior hemin prophylaxis status, historical attack rate, prior chronic opioid use, and the presence of prior chronic symptoms when not having attacks). Of the patients treated with Givlaari, 50% had no attacks that met the criteria for the endpoint of composite porphyria attacks versus 16.3% who received placebo.

Further, treatment with Givlaari led to rapid and sustained reductions in elevated baseline levels of ALA and PBG without evidence of recovery between dosing. For median urinary ALA levels, a clinically meaningful and statistically significant reduction was demonstrated with Givlaari compared with placebo in AIP patients, with median of treatment differences of 14.6 mmol/mol creatinine at Month 3 (p <0.0001) and of 12.8 mmol/mol creatinine at Month 6 (p <0.0001). For median urinary PBG levels, a clinically meaningful and statistically significant reduction was demonstrated for Givlaari compared with placebo in AIP patients at Month 6, with a median of treatment difference of 27.5 mmol/mol creatinine (p <0.0001).

For AIP patients, a clinically meaningful and statistically significant reduction in the annualized days of hemin use was observed in patients treated with Givlaari (RR = 0.23, p <0.0001). Overall, 54.3% of Givlaari-treated patients with AIP and 23.3% of placebo-treated patients with AIP had 0 days of hemin use.

With respect to the SF-12, the results across all the domains showed a consistent effect favouring Givlaari over placebo with the largest differences seen with bodily pain, social functioning, and role limitations due to physical problems.

Supportive Studies

The preliminary results from the open-label extension period are consistent with the results from the double-blind period from the pivotal ENVISION study and have demonstrated continued benefit from treatment with Givlaari. Following the completion of treatment and assessments in the 6-month double-blind period, patients enrolled in the ENVISION study were allowed to continue in the open-label extension period. This period was ongoing at the time of the review, therefore, the full data collection was not available to Health Canada. Patients from the placebo arm of the ENVISION study were permitted to cross over to receive Givlaari 2.5 mg/kg every month or Givlaari 1.25 mg/kg every month. Patients who were in the treatment arm of the ENVISION study receiving Givlaari 2.5 mg/kg every month were permitted to reduce their dose to 1.25 mg/kg due to transaminase elevations. All patients were eligible to receive treatment with Givlaari for up to 29 months in the open-label extension period.

Although the study was ongoing at the time of this submission, the available data for AIP and AHP patients who received long-term treatment with Givlaari (median of 7.61 months of exposure) demonstrated:

  • maintenance of the decreased porphyria attack composite endpoint AAR,
  • maintenance of the stable and durable lowering of urinary ALA and PBG levels,
  • maintenance of decreased days of intravenous hemin administration,
  • maintenance of the improvements in the weekly pain score, the PCS of the SF-12, and the EQ-5D-5L score.

Placebo crossover patients demonstrated rapid and sustained improvements in the porphyria attack composite endpoint AAR, urinary ALA and PBG levels, hemin use, and PCS of the SF-12 on Givlaari in the open-label extension period, with effects and a time course consistent with those observed for Givlaari during the 6‑month double blind period. Patients who crossed over from placebo in the 6‑month double blind period to 1.25 mg/kg or 2.5 mg/kg Givlaari in the open-label extension period had substantial reductions in ALA at Month 7. Lower reductions were observed with the 1.25 mg/kg Givlaari dose.

Indication

The New Drug Submission for Givlaari was filed by the sponsor with the following indication:

  • Givlaari (givosiran) is indicated for the treatment of acute hepatic porphyria (AHP) in adults and adolescents 12 years and older.

Based on the review of the data submitted, Health Canada determined that the safety and efficacy of Givlaari in pediatric (<18 years of age) patients have not been established. Accordingly, Health Canada approved the following indication:

  • Givlaari (givosiran) is indicated for the treatment of acute hepatic porphyria (AHP) in adults.

In addition, the clinical studies did not included sufficient numbers of patients to determine if the use of Givlaari in geriatric patients is associated with differences in safety or effectiveness.

Overall Analysis of Efficacy

The clinical efficacy of Givlaari has been demonstrated pharmacodynamically and clinically through significant reductions in the frequency of severe porphyria attacks. The pivotal study was adequately powered and showed consistent results for the majority of the sub-analyses and multiple secondary outcomes favouring Givlaari over placebo.

Overall, until the present time, treatment of acute porphyria attacks in patients with AHP has been limited. The development of Givlaari fulfills a therapeutic need for these patients as clinically significant benefits have been demonstrated including reductions in acute attacks, reduced pain and analgesic use (including opioid analgesics), reduced intravenous hemin use, reduced time in hospital or off work, and improved quality of life measures.

One of the limitations of the development program and the ENVISION study with open-label extension study was the brief duration of therapy for a chronic disease (6 months for the double-blind period). Secondly, the study did not provide a large representative sample for every subtype of AHP as patients with AIP represented 95% of the population in the pivotal study. It is unclear if the benefits outweigh the uncertainty of potential long-term safety in patients with lower frequencies of attacks (i.e., adolescents); however, it is likely an important therapeutic in patients with a high frequency (>4) of attacks over the course of a calendar year.

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

Clinical Safety

The clinical safety of Givlaari was primarily evaluated in the double-blind period in the pivotal study (ENVISION) and the subsequent open-label extension period described in the Clinical Efficacy section. Additional data were obtained from a supportive study (Study 002).

Safety data were obtained from patients receiving monthly doses of Givlaari 2.5 mg/kg as well as patients receiving monthly doses of Givlaari 1.25 mg/kg, a reduced dose for patients in the open-label extension period who had experienced elevated transaminases. As porphyria attacks were evaluated as a measure of efficacy for Givlaari, symptoms/signs consistent with acute porphyria attacks were not treated as adverse events (AEs) or serious adverse events (SAEs) in the safety evaluations.

Data from the double-blind period in the pivotal ENVISION study demonstrated that in the AHP population, 89.6% of patients in the Givlaari group and 80.4% of patients in the placebo group reported at least one AE. Serious adverse events were reported in 20.8% of patients in the Givlaari-treatment group and 8.7% of patients in the placebo-treatment group. There were no patient deaths during the study.

The most common AEs reported in >10% of patients in the Givlaari group vs. the placebo group were nausea (27% vs. 11%), injection site reactions (25% vs. 0%), headache (12.5% vs 15.2%), chronic kidney disease (10.4% vs. 0%), fatigue (10% vs. 4%), transaminase increased (13% vs 2%) blood creatinine increase (15% vs 4%) and rash (17% vs 4%).

Adverse events reported with ≥5% higher frequency in the Givlaari group included injection site reactions, nausea, fatigue, transaminase increased, increased blood creatinine (includes chronic kidney disease and decreased estimated glomerular filtration rate) and rash (includes pruritus, eczema, erythema, and urticarial rash). All injection site reactions were mild or moderate in severity. Nausea experienced by patients in this study was mainly mild in severity. Of the five Givlaari-treated patients with adverse events of increased chronic kidney disease (increased creatinine and/or decreased estimated glomerular filtration rate), four had a pre-existing medical history of chronic kidney disease, renal impairment, and/or hypertension, and the AEs reported were a worsening of the condition. None of these AEs led to treatment discontinuation or study withdrawal.

In the Givlaari-treatment group, 16.7% of patients experienced a severe AE vs. 10.9% of patients in the placebo group. The only severe AE reported in more than one patient was a device-related infection, which was reported in 4.2% of Givlaari-treated patients and 2.2% of patients receiving placebo.

Adverse events considered as related to the study drug occurred in 45.8% of patients in the Givlaari group versus 26.1% of placebo-treated patients. Study drug-related AEs reported in ≥5% of patients in the Givlaari-treated group vs. the placebo-treated group included nausea (16.7% vs. 4.3%), injection site reactions (14.6% vs. 0%), alanine aminotransferase (ALT) elevation (8.3% vs. 2.2%), headache (6.3% vs. 2.2%), asthenia (6.3% vs. 4.3%), aspartate aminotransferase (AST) elevation (6.3% vs. 2.2%) and chronic kidney disease (6.3% vs. 0%). The frequency of AEs seemed to remain stable over the study for both groups. The exception to this was the hepatic AEs (ALT/AST elevations) where there appeared to be a cluster in the Givlaari-treated group at the 3- to 5‑month period.

Two serious adverse events were reported in two patients: one patient experienced worsening chronic kidney disease and one patients discontinued treatment due to over 8‑times the upper limit of normal value for ALT.

In Phase I/II studies, one case of anaphylaxis was reported in Study 002 and this patient was adequately treated and withdrawn from the study. Another patient in Study 002 with low-titer antibodies developed injection site erythema that was moderate in intensity and related to the study drug. Following this event, based on a physician and patient decision, Givlaari treatment was discontinued due to a lack of marked response from the study drug.

Data from pediatric patients and adults 65 years of age and older were absent in the submission. Pharmacokinetic data suggested age was not a significant covariate in the pharmacokinetics of givosiran. Given that AHP is a life-long disease, Health Canada found it relevant to have a larger sample of patients followed for a longer period to authorize an indication in adolescents. No pediatric patients were enrolled in any stage of the development program or pivotal studies due to the low frequency of attacks and the rarity of this condition in the pediatric population. The identified potential risks and uncertainties of Givlaari treatment paired with the lack of pediatric data did not support authorization of an adolescent indication. Overall, the rather short follow-up and small sample size bring some uncertainties as to whether all the potential side effects of the treatment are captured within the study. Further, data on patients with various stages of renal or hepatic impairment are either absent or very limited.

Overall Analysis of Safety

Data from the double-blind period of the pivotal ENVISION study, the open-label extension period, the supportive study (Study 002) and the clinical development plan comprised the reviewed safety database. Overall exposure to Givlaari is limited in duration but this is considered acceptable, considering the rarity of these diseases. In general, the study population is representative of the target population for which treatment will be authorized. Data on adolescents (≥12 years of age and ≤18) were absent due to the extreme rarity of the condition presenting in that age group that, when identified, typically presents with infrequent attacks. Data in the elderly or in patients with various stages of hepatic impairment and severe renal impairment were either absent or limited. The use of Givlaari in patients with moderate or severe hepatic impairment and in patients with end stage renal disease or on dialysis were included as missing information in the risk management plan (RMP).

The main safety concerns with the use of Givlaari within the timeframe studied included a higher frequency of nausea, injection site reactions, rash, fatigue, increased creatinine and hepatic transaminases compared to placebo. Increased incidences of chronic kidney disease (increased creatinine and/or decreased estimated glomerular filtration rate), that in most cases were transient in nature are a concern for those with severe chronic kidney disease, as no dedicated study in this group was conducted. Furthermore, serious adverse events including worsening of chronic kidney disease and abnormal liver function tests occurred in some patients with the latter leading to discontinuation of Givlaari in one patient. In addition, one adverse drug reaction of anaphylaxis also occurred in a patient that was subsequently withdrawn from the study.

The potential risks of treatment with Givlaari are confounded by comorbid diseases that may exist in patients with AHP, including chronic kidney disease and liver dysfunction as long-term sequelae. Within the clinical studies, it appears that treatment with Givlaari may have exacerbated these conditions. The follow-up duration of 6 months during the double-blind period is relatively short for a life-long chronic disease. As such, an RMP will include a continued monitoring of patients for potential safety concerns and AEs that might reoccur or persist for longer periods of time than the study period.

Appropriate warnings and precautions are in place in the approved Givlaari Product Monograph to address the identified safety concerns.

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

7.2 Non-Clinical Basis for Decision

The review of the non-clinical data for givosiran, the medicinal ingredient in Givlaari, revealed no special hazards for humans based on conventional studies of safety pharmacology and genotoxicity.

In silico and in vitro non-clinical studies demonstrated that givosiran is highly specific for ALAS-1 mRNA and is unlikely to have off-target, sequence-related effects. In vitro transfection assays in human liver carcinoma cell lines showed dose-dependent inhibition of endogenous ALAS-1 mRNA levels.

In multiple in vivo studies across species, givosiran has demonstrated potent and dose-dependent pharmacologic activity when administered subcutaneously, resulting in a consistent reduction of ALAS-1 mRNA in the liver.

In all repeated-dose studies (performed in mice, rats, and monkeys), assessment of toxicity was based on mortality, clinical observations, clinical pathology, anatomic pathology, and toxicokinetic evaluations. A consistent in-life finding across studies was liver toxicity. Givosiran-related microscopic findings were observed in the liver, kidney, pancreas (rats only), lymph nodes (monkeys only) and at the injection site. Mild increases in liver enzyme (alkaline phosphatase [ALP], aspartate aminotransferase [AST]), and gamma-glutamyl transferase [GGT]) levels were also observed. Comparisons between plasma, liver and kidney exposures to repeated givosiran treatment showed that the liver concentrations were highest followed by the kidney and plasma concentrations for each dose groups. Additionally, the half-life values of givosiran in the kidney were more prolonged than in the liver. The studies did not address the difference in dosing regimens between human (once monthly dose) and non-clinical species (once weekly dose).

A two-year carcinogenicity study conducted in rats was ongoing at the time of the review of the submission. Upon completion of the study, the results will be submitted to Health Canada. The evaluation of givosiran in a 26‑week carcinogenicity study in Tg-rasH2 mice showed no evidence of carcinogenicity at dose levels of up to 1,500 mg/kg/month.

In pregnant rabbits, givosiran produced maternal toxicity starting at a 1.5 mg/kg/day dose. Increased incidences of skeletal variations were observed with the administration of a single dose of 20 mg/kg. The 1.5 mg/kg/day dose in rabbits is 5‑times the maximum recommended human dose (MRHD) of 2.5 mg/kg/month normalized to account for a daily dose schedule. There were no givosiran-related adverse effects upon the male and/or female fertility endpoints evaluated. Available data in animals demonstrates the excretion of givosiran in milk, therefore a risk to the newborns/infants cannot be excluded.

A dose of ≤10 mg/kg/week was determined to be the no-observed-adverse-effect level (NOAEL) by the repeated-dose toxicity studies in rats, with impurities corresponding to values of ≤3.4% by anion-exchange high-performance liquid chromatography) and ≤8.7% by ion-pair reversed-phase partition high-performance liquid chromatography. Due to a difference in the amount of drug administered in the repeated-dose toxicity study as compared to the proposed amount, the impurities can be considered qualified at 3‑fold the amount used in the repeated-dose toxicity study. These limits are equivalent to or higher than the proposed specification limits.

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

Appropriate warnings and precautionary measures are in place in the Givlaari Product Monograph to address the identified safety concerns.

For more information, refer to the Givlaari 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 Givlaari has demonstrated that the drug substance and drug product can be consistently manufactured to meet the approved specifications. Proper development and validation studies were conducted, and adequate controls are in place for the commercial processes. Changes to the manufacturing process and formulation made throughout the pharmaceutical development are considered acceptable upon review. Based on the stability data submitted, the proposed shelf life of 36 months is acceptable when the drug product is stored at 2ºC to 25ºC.

Proposed limits of drug-related impurities are considered adequately qualified, i.e., within International Council for Harmonisation limits and qualified from toxicological studies.

All sites involved in production are compliant with Good Manufacturing Practices.

All non-medicinal ingredients (described earlier) found in the drug product are acceptable for use in drugs according to the Food and Drug Regulations.

None of the excipients used in the formulation of Givlaari are of human or animal origin.