Summary Basis of Decision for Incivek ™

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
IncivekTM

Telaprevir, 375 mg, Tablet, Oral

Vertex Pharmaceuticals Canada Inc.orporated

Submission control no: 142482

Date issued: 2012-03-23

Foreword

Health Canada's Summary Basis of Decision (SBD) documents outline the scientific and regulatory considerations that factor into Health Canada regulatory decisions related to drugs and medical devices. SBDs are written in technical language for stakeholders interested in product-specific Health Canada decisions, and are a direct reflection of observations detailed within the evaluation reports. As such, SBDs are intended to complement and not duplicate information provided within the Product Monograph.

Readers are encouraged to consult the 'Reader's Guide to the Summary Basis of Decision - Drugs' to assist with interpretation of terms and acronyms referred to herein. In addition, a brief overview of the drug submission review process is provided in the Fact Sheet entitled 'How Drugs are Reviewed in Canada'. This Fact Sheet describes the factors considered by Health Canada during the review and authorization process of a drug submission. Readers should also consult the 'Summary Basis of Decision Initiative - Frequently Asked Questions' document.

The SBD reflects the information available to Health Canada regulators at the time a decision has been rendered. Subsequent submissions reviewed for additional uses will not be captured under Phase I of the SBD implementation strategy. For up-to-date information on a particular product, readers should refer to the most recent Product Monograph for a product. Health Canada provides information related to post-market warnings or advisories as a result of adverse events (AE).

For further information on a particular product, readers may also access websites of other regulatory jurisdictions. The information received in support of a Canadian drug submission may not be identical to that received by other jurisdictions.

Other Policies and Guidance

Readers should consult the Health Canada website for other drug policies and guidance documents. In particular, readers may wish to refer to the 'Management of Drug Submissions Guidance'.

1 Product and submission information

Brand name:

IncivekTM

Manufacturer/sponsor:

Vertex Pharmaceuticals Canada Inc.orporated

Medicinal ingredient:

Telaprevir

International non-proprietary Name:

Telaprevir

Strength:

375 mg

Dosage form:

Tablet

Route of administration:

Oral

Drug identification number(DIN):

  • 02371553

Therapeutic Classification:

Antiviral agent

Non-medicinal ingredients:

Colloidal silicon dioxide, croscarmellose sodium, D&C Red Number 40, dibasic calcium phosphate (anhydrous), FD&C Blue Number 2, hypromellose acetate succinate, microcrystalline cellulose, polyethylene glycol, polyvinyl alcohol, sodium lauryl sulfate, sodium stearyl fumarate, talc, and titanium dioxide.

Submission type and control no:

New Drug Submission, Control Number: 142482

Date of Submission:

2011-01-14

Date of authorization:

2011-08-16
2 Notice of decision

On August 16, 2011, Health Canada issued a Notice of Compliance to Vertex Pharmaceuticals (Canada) Incorporated for the drug product Incivek.

Incivek contains the medicinal ingredient telaprevir, which is an antiviral agent. Incivek is a specific hepatitis C virus protease inhibitor.

Incivek, in combination with peginterferon (Peg-IFN) alfa and ribavirin (RBV), is indicated for the treatment of genotype 1 chronic hepatitis C in adult patients with compensated liver disease, including cirrhosis, who are treatment naïve or who have previously been treated with interferonbased treatment, including prior null responders, partial responders, and relapsers.

The market authorization was based on quality, non-clinical, and clinical information submitted. The efficacy and safety of Incivek were primarily based on the analyses of Phase 2 and 3 studies conducted with Incivek in combination with Peg-IFN-alfa/RBV in either treatment-naïve patients or previously treated patients (null responders, partial responders, relapsers). The sustained virologic response (SVR: 24 weeks after the last planned dose) was evaluated for all patients who received at least one dose of the treatment within each study. Results from these studies, especially Phase 3 studies, showed that Incivek, when used for 12 weeks in combination with either 24 or 48 weeks of treatment with Peg-IFN-alfa/RBV, produced sustained virologic response rates that were superior compared to 48 weeks of treatment with Peg-IFN-alfa/RBV alone. In addition, this result was irrespective of whether patients were treatment-naïve, or patients who were previously treated with Peg-IFN-alfa/RBV, including prior null responders, partial responders, and relapsers.

Incivek (375 mg, telaprevir) is supplied in a tablet form. The recommended dose of Incivek is 750 mg (two 375 mg tablets) taken orally three times a day (7-9 hours apart) with food. The total daily dose is six tablets (2,250 mg). Consultation of the Incivek Product Monograph prior to initiation of therapy is highly recommended for further dosing guidelines. Incivek must not be administered as a single drug therapy, but only in combination with Peg-IFN-alfa/RBV. The Product Monographs of Peg-IFN-alfa/RBV must be consulted prior to initiation of therapy with Incivek.

Incivek, in combination with Peg-IFN-alfa/RBV is contraindicated for:

  • patients who are hypersensitive to telaprevir or to any ingredient in the formulation or component of the container;
  • women who are pregnant or men whose female partners are pregnant;
  • when combined with drugs that are highly dependent on cytochrome P450 (CYP) 3A for clearance and for which elevated plasma concentrations are associated with serious and/or life-threatening events (narrow therapeutic index). Incivek is also contraindicated when combined with drugs that strongly induce CYP3A and thus may lead to lower exposure and loss of efficacy of Incivek.

The contraindications to Peg-FN-alfa/RBV also apply to Incivek combination treatment. Refer also to the prescribing information for Peg-IFN-alfa/RBV.

Incivek should be administered under the conditions stated in the Product Monograph taking into consideration the potential risks associated with the administration of this drug product. Detailed conditions for the use of Incivek are described in the Product Monograph.

Priority Review Status was granted for the evaluation of Incivek as it appeared to provide substantial evidence of increased clinical efficacy such that the overall benefit/risk profile is improved over existing therapies.

Based on the Health Canada review of data on quality, safety, and efficacy, Health Canada considers that the benefit/risk profile of Incivek is favourable for the indication stated above.

3 Scientific and Regulatory Basis for Decision

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)

General Information

Telaprevir, the medicinal ingredient of Incivek, is a direct-acting agent against the hepatitis C virus (HCV). Incivek is a specific inhibitor of an enzyme called HCV NS3/4A protease which is essential for viral replication.

Manufacturing Process and Process Controls

Telaprevir is manufactured via a multi-step synthesis. Each step of the manufacturing process is considered to be controlled within acceptable limits:

  • The sponsor has provided information on the quality and controls for all materials used in the manufacture of the drug substance;
  • The drug substance specifications are found to be satisfactory. Impurity limits meet International Conference on Harmonisation (ICH) requirements; and
  • The processing steps have been evaluated and the appropriate ranges for process parameters have been established.
Characterization

The structure of telaprevir has been adequately elucidated and the representative spectra have been provided. Physical and chemical properties have been described and are found to be satisfactory.

Impurities and degradation products arising from manufacturing and/or storage were reported and characterized. These products were found to be within ICH-established limits and/or were qualified from batch analysis or toxicological studies and therefore, are considered to be acceptable.

Control of Drug Substance

Copies of the analytical methods and where appropriate, validation reports were provided and are considered satisfactory for all analytical procedures used for release and stability testing of telaprevir.

Batch analysis results were reviewed. All results comply with the specifications and demonstrate consistent quality of the batches produced.

The drug substance packaging is considered acceptable

Stability

Based on the long-term, real-time, and accelerated stability data submitted, the proposed retest period, shelf-life, and storage conditions for the drug substance were supported and are considered to be satisfactory.

3.1.2 Drug Product

Description and Composition

Incivek contains the medicinal ingredient, telaprevir. The drug product is supplied as purple film-coated capsule-shaped tablets for oral administration. Each tablet is debossed with the characters "V 375" on one side.

Each Incivek tablet contains 375 mg of telaprevir and the following inactive ingredients: colloidal silicon dioxide; croscarmellose sodium; D&C Red Number 40; dibasic calcium phosphate (anhydrous); FD&C Blue Number 2; hypromellose acetate succinate; microcrystalline cellulose; polyethylene glycol; polyvinyl alcohol; sodium lauryl sulphate; sodium stearyl fumarate; talc; and titanium dioxide.

Incivek tablets are packaged in one of two formats: a 28-day pack; or a 28-day hospital supply.

  • 28-day pack contains 4 weekly cartons of 7 blister stripes each (6 tablets per blister strip); or
  • 28-day hospital supply contains 168 tablets in a bottle.

All non-medicinal ingredients (excipients) found in the drug product are acceptable for use in drugs according to the Food and Drug Regulations. The compatibility of telaprevir with the excipients is demonstrated by the stability data presented on the proposed commercial formulation.

Pharmaceutical Development

Changes to the manufacturing process and formulation made throughout the pharmaceutical development are considered acceptable upon review.

Manufacturing Process and Process Controls

The method of manufacturing is considered acceptable and the process is considered adequately controlled within justified limits.

Control of Drug Product

Incivek is tested to verify that its identity, appearance, content uniformity, assay, dissolution, moisture content, and levels of degradation products and drug-related impurities are within acceptance criteria. The test specifications and analytical methods are considered acceptable; the shelf-life and the release limits for individual and total degradation products are within acceptable limits.

The validation process is considered to be complete. Data from final batch analyses were reviewed and are considered to be acceptable according to the specifications of the drug product.

Stability

Based on the real-time, long-term, and accelerated stability data submitted, the proposed 24-month shelf-life for Incivek is considered acceptable when the product is stored at 25°C.

The compatibility of the drug product with the container closure system was demonstrated through the stability studies. The container closure system met all validation test acceptance criteria.

3.1.3 Facilities and Equipment

The design, operations, and controls of the facilities and equipment that are involved in the production of Incivek are considered suitable for the activities and products manufactured.

All sites are compliant with Good Manufacturing Practices (GMP).

3.1.4 Adventitious Agents Safety Evaluation

Not applicable. The excipients used in the drug product formulation are not from animal or human origin.

3.1.5 Conclusion

The Chemistry and Manufacturing information submitted for Incivek 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.

3.2 Non-Clinical Basis for Decision

3.2.1 Pharmacodynamics

Telaprevir is a potent inhibitor of HCV NS3/4A proteases derived from genotype 1 hepatitis C-infected patients. The interaction of telaprevir with the HCV NS3/4A protease occurs as a two step process, with formation of a weaker complex with an inhibitory constant (Ki) of 44 nM, followed by the formation of a tightly bound complex with a Ki of 7-10 nM. The effects of interferon (INF)-alpha and telaprevir, and also ribavirin (RBV) and telaprevir, both appeared to be additive thereby suggesting the combination of telaprevir with INF-alpha and RBV may result in a much stronger antiviral effect than administration of telaprevir alone. Telaprevir demonstrated no effect on the antiviral activity of the human immunodeficiency virus-1 (HIV-1) protease, or on the antiviral activity of hepatitis B.

Safety Pharmacology

A series of safety pharmacology studies were conducted to assess telaprevir's effects on vital organ systems. Results from in vivo safety pharmacology studies showed a low potential for effects on the central nervous system (CNS), respiratory, and cardiovascular systems. In vitro studies with telaprevir did indicate inhibition of the human ether-a-gogo- related gene (hERG) tail current in a concentration-dependent manner with 10 to 15% inhibition of hERG tail current at levels achieved in plasma at the recommended human dose. Although cardiovascular parameters in dogs were not affected with telaprevir, the inhibition of hERG tail current at human dose levels did suggest the need for thorough QT studies in human healthy volunteers to ensure cardiac safety of the drug.

3.2.2 Pharmacokinetics

Absorption

Following administration, telaprevir absorption was generally rapid with mean peak plasma concentrations occurring between 0.5 to 2 hours post-dose. The absolute bioavailability of telaprevir was approximately 33% for male rats, 52% for female rats, and less than 22% for rabbits. Apparent bioavailability for dogs in the fasted-state ranged from 43% to 67%. Exposure increased between 1.5- to 4-fold in the presence of food in dogs with bioavailability ranging from 70% to 95%. Repeated administration resulted in decreases in systemic exposure in pregnant mice by 2-fold and in rats by 1.5- to 2.5-fold, whereas increases in exposure were noted in dogs by 1.5-fold. No apparent genderdifferences in exposures were noted in mice and dogs, whereas exposures in female rats were markedly higher, up to 2-fold, than those observed in male rats.

Distribution

Telaprevir had moderate plasma protein binding and was widely distributed to tissues, predominately in the gastrointestinal tract, liver, pancreas, and kidneys. After repeated administration, telaprevir was mainly distributed in the liver in rats and dogs. Telaprevir was also shown to be excreted in the milk of lactating rats.

Metabolism

The metabolism of telaprevir involved oxidation, reduction, and hydrolysis pathways to produce numerous metabolites and isomers. Telaprevir is mainly metabolized by the cytochrome P450 (CYP) isozyme 3A4 (substrate of CYP3A4) and is an inhibitor of CYP3A4. Telaprevir is also a substrate for efflux pump protein P-glycoprotein (P-gp). Metabolite profiling conducted in plasma samples from rats and dogs identified VRT-127394 (epimer of telaprevir), pyrazinoic acid (PZA), and M3 isomer (reduction product, designated VRT-0922061) as the predominant circulating metabolites.

Excretion

Telaprevir is excreted predominantly though the faeces.

3.2.3 Toxicology

Single-Dose Toxicity

Single-dose studies conducted in rats and mice at doses up to 1,000 mg/kg suggested a low potential for acute oral toxicity for a spray dried dispersion (SDD) formulation of telaprevir. For instance, under the conditions tested, no early deaths, no clinical signs, no changes in body weight, and no gross lesions at necropsy were noted following treatment with telaprevir.

Repeat-Dose Toxicity

Telaprevir SDD formulations were evaluated in repeat-dose toxicity studies up to six and nine months in duration in rats and dogs, respectively, at doses ranging from 1 to 1,000 mg/kg/day in rats and 15 to 500 mg/kg/day in dogs. These evaluations identified toxicities (described below) which precluded determination of no observed adverse effect levels (NOAELs). For instance, the studies identified the bone marrow/haematopoetic system (accompanied splenic changes), the liver, and the male reproductive system as primary target organs of telaprevir-related toxicity in rats. Repeat-dose studies conducted in dogs identified similar target organs of telaprevir-related toxicity with exception of the vascular system being a primary target organ rather than the male reproductive system. The findings appear to be reversible within three months upon stopping treatment with exception of the liver effects in rats, organ weight changes in the spleen and testes in rats, and increased cholesterol and myelofibrosis in bone marrow of the sternum in dogs.

Genotoxicity

Under the conditions tested, evidence of genotoxicity was not observed with telaprevir in a bacterial mutagenicity assay, in an in vitro mammalian chromosomal aberration assay, or in an in vivo micronucleus study in mouse.

Carcinogenicity

Based on the proposed duration of treatment of 12 weeks and the negative genotoxicity findings, telaprevir was not tested for its carcinogenic potential.

Reproductive and Developmental Toxicity

Telaprevir (spray dried dispersion formulation) was evaluated in a fertility and early embryonic development study in rats, with male rats receiving doses up to 300 mg/kg/day and females receiving doses up to 500 mg/kg/day. The no observed adverse effect level (NOAEL) for degenerative testicular toxicity was established at exposures 0.17-fold the human exposures at the recommended clinical dose. Potential effects on sperm, [for example (e.g.) decreased percentage of motile sperm and increased non-motile sperm count] were observed in a rat fertility study at levels 0.30-fold the human exposure at the recommended clinical dose. Additional effects on fertility included an increased percentage of the following: pre-implantation loss; dams with nonviable embryos; and nonviable conceptuses per litter. These effects are likely associated with testicular toxicity in male rats, but contributions of the female cannot be ruled out.

Telaprevir SDD formulation was also evaluated in embryo-foetal development studies in pregnant mice at doses up to 1,000 mg/kg/day and in pregnant rats at doses up to 500 mg/kg/day. No gross external, soft tissue or skeletal foetal alterations (malformations or variations) were associated with telaprevir treatment.

Telaprevir SDD formulation was also administered to female rats in a prenatal and postnatal development study at doses up to 500 mg/kg/day. Telaprevir had no apparent effects on the natural delivery, on Caesarean-sectioning, or in the first generation offsprings. Decreases in pup bodyweights were noted during lactation.

Local Tolerance

Under the conditions tested, dermal and ocular studies demonstrated that telaprevir was non-irritating for either skin or eyes. The M11 metabolite of telaprevir, VRT-841125, did show skin sensitizing potential and revealed that a telaprevir metabolite can act as an antigen in a delayed type hypersensitivity reaction.

3.2.4 Summary and Conclusion

Safety pharmacology studies conducted with telaprevir suggest a low potential for possible effects on CNS, respiratory and cardiovascular systems. In vitro studies with telaprevir indicate inhibition of the hERG tail current in a concentration-dependent manner with 10 to 15% inhibition at levels achieved in plasma with clinical doses.

Overall, the primary organs of toxicity in non-clinical studies were the bone marrow/haematopoetic system (accompanied splenic changes), the liver, and the male reproductive system. Under the conditions tested, telaprevir did not appear to cause damage to deoxyribonucleic acid (DNA) when tested in a standard battery of mutagenesis assays, in the presence and absence of metabolic activation. In reproductive studies, male rats did develop gross and histologic findings in the testes. In addition, male reprotoxicity findings appeared to affect pre-implantation and post-implantation, but the contribution of female reproductive system toxicity could not be ruled out.

The non-clinical studies for this drug submission are considered acceptable. The nonclinical pharmacology and toxicology program for Incivek (telaprevir) demonstrated that the compound is relatively safe for humans. No significant issues within the non-clinical studies have been identified which would preclude approval of this drug product for the proposed indication.

3.3 Clinical basis for decision

3.3.1 Pharmacodynamics

A QT/QTc study was conducted in healthy volunteers to thoroughly evaluate the effect of telaprevir on QT/QTc intervals and cardiac safety. Incivek was tested at a dose of 750 mg every eight hours (q8h) for Days 1 to 4, and a single 750 mg morning dose on Day 5, as well as a 1,875 mg 8qh on Days 1 to 4, with a single 1,875 mg morning dose on Day 5. The 1,875 mg dose in healthy volunteers closely matched the plasma concentrations of the target patient population receiving the 750 mg dose (in combination with Peg-IFNalpha/ RBV). According to the optimal study population-specific non-linear heart rate correction, 1,875 mg q8h was associated with statistically significant QTc prolongation from 1 to 6 hours on Day 5, with a maximum difference compared to placebo of mean 8.0 milliseconds at 5 hours post-dosing. The maximum magnitude of QTc prolongation with telaprevir 1,875 mg was comparable to that observed with the positive control treatment, moxifloxacin 400 mg which produced a peak effect of 9.6 milliseconds (mean values) at 6 hours post-dosing. Statistically significant QRS prolongation was also evident from 1 to 24 hours, with a maximum difference compared to placebo of 2.6 milliseconds (mean values) at 6 hours post-dosing. Additionally, statistically significant PR interval prolongation was observed from 4 to 6 hours with a maximum difference compared to placebo of mean 5.0 milliseconds (mean values) at 6 hours postdosing. At the 1,875 mg q8h dose, all respiration rate derived heart rate was not significantly increased at any time post-dose time point, with a maximum effect of 7.9 beats per minute observed at 8 hours post-dosing. Electrocardiogram results collected from a Phase 2 study and Phase 3 study also confirmed similar observations for QTc prolongation during telaprevir treatment.

3.3.2 Pharmacokinetics

Absorption

Maximum plasma concentrations following a single dose of Incivek were generally achieved within 4 to 5 hours post-dose. In vitro studies performed on human Caco-2 cells revealed that telaprevir is a substrate of P-gp. Exposure to telaprevir is higher during coadminstration of Peg-IFN-alfa/RBV than after administration of Incivek alone.

A food study conducted in healthy volunteers revealed that the fat content of a meal significantly affects exposure to telaprevir. When compared with administration following a standard meal (21 grams of fat), exposure to telaprevir decreased by 19%, 42%, and 73% when administered with meals that were successively less in fat [that means (i.e.) 9 grams of fat, 3.6 grams of fat, and fasting condition]. Following this trend, telaprevir exposure increased by 36% when administered with a high-fat (56 grams of fat) meal, compared to a standard meal. Similarly, the maximum concentration of telaprevir followed this trend with the change in amount of fat in meals. Based on this study, to achieve optimal telaprevir exposure, Incivek should be taken with food.

Distribution

Telaprevir is approximately 59% to 76% bound to human plasma proteins. Telaprevir binds primarily to alpha 1-acid glycoprotein and albumin and the binding is concentration dependent, decreasing with increasing concentrations of telaprevir. Following oral administration, the typical apparent volume of distribution of telaprevir was approximately 252 litres, with an inter-individual variability of 72%.

Metabolism

Telaprevir is extensively metabolized in the liver, involving hydrolysis, oxidation, and reduction. Multiple metabolites were detected in the faeces, plasma, and urine; but no new unique human metabolites were observed other than those noted in the non-clinical species. Following repeated-oral administration, R-diastereomer of telaprevir, pyrazinoic acid, and a metabolite that underwent reduction at the alpha-ketoamide bond of telaprevir (not active) were found to be the predominant metabolites of telaprevir.

In vitro studies using recombinant human CYP isoforms indicated that CYP3A4 was the major isoform responsible for telaprevir metabolism. However, non-CYP mediated metabolism likely plays a role after multiple dosing of telaprevir.

Excretion

Incivek is predominantly eliminated through the faeces. Following administration of a single dose of radiolabelled Incivek, the median recovery of the administered radioactive dose was approximately 82% in faeces, 9% in exhaled air and 1% in urine.

Biopharmaceutics

A comparative bioavailability study was conducted in healthy volunteers to evaluate the bioequivalence of an uncoated telaprevir tablet (used within the Phase III clinical studies) and a film-coated telaprevir tablet proposed for market. Results from this study showed the rate and extent of telaprevir absorption from the uncoated (clinical) 375 mg tablet was comparable to the coated 375 mg tablet under fed conditions. Therefore, the comparative data was determined to be acceptable.

3.3.3 Clinical Efficacy

The efficacy of Incivek was evaluated primarily in three Phase III studies in genotype 1 hepatitis C-infected patients. Among these three studies, two were conducted in treatment-naïve patients, and the third study was conducted in previously treated patients (relapsers, partial responders, and null responders). All patients enrolled had compensated liver disease, detectable hepatitis C virus ribonucleic acid (HCV-RNA) particles, and liver histopathology consistent with chronic hepatitis C. The methodology used to assess efficacy was that of plasma HCV-RNA measurements through the use of the COBAS® TaqMan® HCV test (version 2.0). The assay had a lower limit of quantitation of 25 IU/mL. The primary outcome measure for all three studies was the sustained virologic response (SVR). SVR was defined as undetectable HCV RNA 24 weeks after the end of treatment and is considered a virologic cure. The lower limit of detection was 10 IU/mL. Undetectable HCV RNA was used to determine SVR.

Treatment-naïve Patients

The first Phase III study, herein known as "Advance", was a randomized, double-blind, parallel-group, placebo-controlled study conducted in treatment-naïve patients who had not received any prior hepatitis C treatment, including INF or Peg-INF monotherapy. This study sought to explore whether Incivek, in combination with Peg-IFN-alfa/RBV would be superior in efficacy compared to Peg-IFN-alfa/RBV treatment alone. In addition, this study also sought to explore whether a shortened dosing regimen for Incivek (8-week treatment) would offer an improved tolerability as compared to the proposed 12-week Incivek dosing regimen while not compromising efficacy.

The study design consisted of either administering Incivek for 8 weeks in combination with Peg-IFN-alfa/RBV (T8/PR regimen) for either 24 or 48 weeks; or administering Incivek for 12 weeks in combination with Peg-IFN-alfa/RBV (T12/PR regimen) for either 24 or 48 weeks. The duration of Peg-IFN-alfa/RBV administration was determined through a response-guided therapy determined by the presence or absence of an extended Rapid Virologic Response (eRVR). Patients who demonstrated presence of an eRVR (eRVR+); that being an undetectable HCV-RNA at either Week 4 or Week 12, received a shortened 24-week Peg-IFN-alfa/RBV dosing regimen. Patients who demonstrated absence of an eRVR (eRVR-); that being who did not have undetectable HCV-RNA at either Week 4 or Week 12, received a 48-week Peg-IFN-alfa/RBV dosing regimen. The control group (Pbo/PR48 regimen) received a matched-placebo for the first 12 weeks along with a Peg-IFN-alfa/RBV dosing for 48 weeks.

Results from the Advance study demonstrated significant improvement in SVR rates in both Incivek treatment groups (T12/PR = 74.7% and T8/PR = 68.7%) compared to the placebo treatment group (Pbo/PR48 = 43.8%). Furthermore, relapse rates were lower in both Incivek treatment groups compared to the placebo group, as were on-treatment virologic failure rates. Incivek-treated patients in both groups achieved comparable eRVR rates which were much higher than in the placebo group. However, for patients which achieved eRVR+, the SVR rates were higher in the placebo-treated patients compared to the Incivek-treated patients, suggesting no additional efficacy was achieved with Incivek treatment among eRVR+ patients. For patients who failed to achieve an eRVR (i.e., eRVR-), SVR rates in the Incivek treatment groups were also higher compared to the placebo group.

When study results were compared between T12/PR and T8/PR treatment regimens, T12/PR achieved favourable efficacy outcomes, namely higher SVR rates, lower relapse rates and lower on-treatment virologic failure rates. Although eRVR rates were comparable, the eRVR to SVR rates were higher in T12/PR. In conclusion, results from the Advance study showed that Incivek administration, in combination with Peg-IFNalfa/RBV treatment, demonstrated superiority in efficacy when compared to Peg-IFNalfa/RBV treatment alone (placebo + Peg-IFN-alfa/RBV) in treating chronic hepatitis C treatment-naïve patients. Furthermore, the 12-week Incivek dosing regimen was more favourable than the 8-week Incivek dosing regimen.

A second Phase III study, herein known as "Illuminate", was a randomized, activecontrolled, open-label study also conducted in treatment-naïve patients. This study was designed as a supportive study to determine whether a 24-week Peg-IFN/RBV duration was non-inferior to a 48-week Peg-IFN/RBV duration in patients who achieved eRVR (i.e., eRVR+). Patients received 12 weeks of Incivek in combination with Peg-IFN/RBV (T12/PR). Patients with eRVR+ were randomized at Week 20 to complete either 24 or 48 weeks of treatment. Patients who did not achieve eRVR (i.e., eRVR-) were assigned to receive a total of 48 weeks of Peg-IFN/RBV.

In the Illuminate study, SVR rates were 92% in T12/PR24/eRVR+ patients and 87.5% in T12/PR48/eRVR+ patients, demonstrating that the T12/PR24 regimen was non-inferior to the T12/PR48 regimen among eRVR+ patients. Moreover, relapse rates were lower among eRVR+ patients randomized to T12/PR48 compared to all other treatment groups. Although the number of cirrhotic patients was limited, eRVR+ cirrhotic patients randomized to the T12/PR48 regimen achieved a much higher SVR rate (91.7%) than those eRVR+ cirrhotic patients randomized to the shorter T12/PR24 regimen (SVR rate 66.7%). This leads to suggest eRVR+ cirrhotic patients may benefit from a longer Peg- IFN-alfa/RBV dosing (i.e., 48 weeks). In conclusion, the efficacy results of the Illuminate study were consistent with the results seen within the Advance study. In addition, results from the Illuminate study also demonstrated overall non-inferior efficacy in eRVR+ patients with genotype 1 chronic hepatitis C who received the T12/PR24 treatment regimen as apposed to the extended Peg-IFN-alfa/RBV 48-week dosing regimen (T12/PR48). Moreover, cirrhotic patients with eRVR+ also showed a clinical benefit from the 48-week Peg-IFN-alfa/RBV dosing treatment as opposed to the 24-week Peg-IFNalfa/RBV dosing treatment.

Treatment-failure Patients

The third Phase III study, herein known as "Realize", was a randomized, double-blind, placebo-controlled, three-arm study conducted in patients who did not achieve SVR through prior treatment with either Peg-IFN-alfa-2a/RBV or Peg-IFN-alfa-2b/RBV. This study sought to demonstrate the superiority of Incivek treatment in combination with Peg-IFN-alfa/RBV therapy compared to a placebo and Peg-IFN-alfa/RBV therapy alone in achieving SVR in prior relapsers, prior partial responders, and prior null responders. In addition, this study also sought to evaluate whether a delayed start (DS) of Incivek offered an efficacy advantage by reducing the emergence of Incivek-resistant variants and thus on-treatment virologic failures.

The study enrolled prior relapsers (patients with HCV-RNA undetectable at end of treatment with a Peg-IFN-based regimen, but HCV-RNA detectable within 24 weeks of treatment follow-up) and prior non-responders (patients who did not have undetectable HCV-RNA levels during or at the end of a prior course of at least 12 weeks of treatment). The non-responder population included 2 sub-groups: prior partial responders (greater than or equal to 2-log10 reduction in HCV-RNA at week 12, but not achieving HCV-RNA undetectable at end of treatment with Peg-IFN-alfa/RBV); and prior null responders (less than 2-log10 reduction in HCV-RNA at week 12 of prior treatment with Peg-IFNalfa/RBV).

Patients were randomized in a 2:2:1 ratio to one of three treatment groups. In two of the treatment groups patients received Incivek for 12 weeks in combination with Peg-IFNalfa/RBV for 48 weeks; one group started the treatment regimens simultaneously (T12/PR48), and the other had a 4-week delayed start of Incivek (T12-DS/PR48) to examine the impact on relapse and resistance. The third treatment group received a placebo for 16 weeks in combination with Peg-IFN-alfa/RBV dosing for 48 weeks (Pbo16/PR48).

The proportion of patients achieving SVR was significantly higher in each of the Incivek treatment groups compared to placebo. For the prior relapser population, in the Incivek treatment groups, SVR rates were 83.4% to 87.9%, respectively compared to 23.5% for the placebo group. For prior non-responder population, SVR rates were 41.3% and 41.5% respectively, in the Incivek treatment groups compared to 9.4% in the placebo group. Sustained virologic rates were also significantly higher in each of the prior non-responder subgroups: prior null; and prior partial responders. For the prior null responder population, SVR rates were 29.2% and 33.3%, respectively for the Incivek groups, compared to 5.4% in the placebo group. For prior partial responders, SVR rates were 59.2% and 54.2%, respectively, for the Incivek groups compared to 14.8% for the placebo group. In conclusion, Incivek met its primary objective of demonstrating superior efficacy when combined with Peg-IFN-alfa/RBV compared to Peg-IFNalfa/RBV therapy alone (standard treatment) in patients with genotype 1 chronic hepatitis C infection who previously failed treatment with Peg-IFN-alfa/RBV. Further, the delayed start of Incivek treatment did not result in an added clinical benefit relative to simultaneous start of Incivek and Peg-IFN-alfa/RBV.

It is worthy to note that response-guided therapy was not evaluated in the Realize clinical study in the treatment-failure population. However, data from the Phase II studies indicated that the treatment-failure population was not uniform. Prior relapsers and prior non-responders are quite distinct, and the prior relapse population appears to have more similarities to the treatment-naïve population than to the prior non-responders (particularly prior null responders), based on rapidity of the antiviral response and on SVR rates. The primary benefit of response-guided therapy is that there is a shortened duration of therapy from 48-weeks to 24-weeks which is likely to improve the safety profile due to a reduced exposure to Peg-IFN-alfa/RBV.

3.3.4 Clinical Safety

The overall safety profile of Incivek was based mainly on the three Phase III studies previously described in section 3.3.3 Clinical Efficacy, in addition to supportive safety data derived from two Phase II studies.

The most frequent adverse events (AEs) in patients who received Incivek in combination with Peg-IFN-alfa/RBV were fatigue; pruritus; nausea; influenza-like illness; rash; anaemia; insomnia; diarrhoea; and pyrexia. The most frequent adverse drug reactions (ADRs) in patients who received Incivek in combination with Peg-IFN-alfa/RBV were pruritus; rash; nausea; anaemia; diarrhoea; vomiting; hemorrhoids; and proctalgia (i.e., pain in anal or rectal region). Serious ADRs occurred in 3.9% of patients in the T12/PR treatment group compared to 0.7% in the Pbo/PR group. The most frequent serious adverse events (SAEs) were anaemia and rash. A total of 10.4% of patients discontinued the study due to ADRs; with rash, anaemia, pruritus, nausea, and vomiting being the most frequent ADRs leading to discontinuation.

Pooled safety data from the Phase II and Phase III studies identified a total of nine deaths. Among these deaths, five occurred in patients randomized to Incivek therapy and four occurred in patients who received Peg-IFN-alfa/RBV therapy alone. None of the deaths in the Incivek treatment group occurred while administering Incivek. One death, confirmed to be malignant neoplasm of the lung, was diagnosed 96 days after the last dose of Incivek was administered and was considered by the site investigator as possibly related to Incivek. This patient had however, a history of smoking 15 to 20 cigarettes per day for over a 40-year period.

Anorectal ADRs (e.g., hemorrhoids, anorectal discomfort, anal pruritus and rectal burning) were reported more frequently in the T12/PR group than in the Pbo/PR group [26.2% versus (vs.) 5.4%]. Serious anorectal events, events of at least Grade 3 or leading to discontinuation were infrequent (<1.0%) in the T12/PR group and did not occur in the Pbo/PR group.

Safety Issues of Interest
QT, QRS and PR Prolongation

Healthy volunteers that received Incivek at a dose concentration of 1,875 mg every 8 hours demonstrated a modest effect on QTc, QRS and PR interval prolongation. Exposure at this dose was comparable to the exposure in hepatitis C-infected patients dosed at 750 mg every 8 hours in combination with Peg-IFN-alfa/RBV. Caution is therefore recommended when prescribing Incivek concurrently with drugs known to induce QT prolongation and which are CYP3A substrates (such as erythromycin, ketoconazole, haloperidol, tacrolimus, and salmeterol). Incivek may increase concentrations of the co-administered drug which may result in an increased risk of their associated cardiac AEs. In the event that co-administration of such drugs with Incivek is judged strictly necessary, clinical monitoring, including electrocardiogram (ECG) assessments, should be considered. Additionally, monitoring and correction of electrolyte disturbance (e.g., hypokalemia, hypomagnesemia, and hypocalcemia) should be considered prior to initiation and during Incivek therapy.

Use of Incivek should be avoided in patients with: congenital QT prolongation; or a family history of congenital QT prolongation; or sudden death. In the event that treatment with Incivek in such patients is judged clinically necessary, consideration should be given to monitoring, including ECG assessments. Incivek should be used with caution in patients with: a history of acquired QT prolongation; a history of arrhythmias (especially ventricular arrhythmias or atrial fibrillation); a history of heart failure with reduced leftventricular ejection fraction; myocardial ischemia or infarction; cardiomyopathy; conduction system disease; or a requirement for drugs known to prolong the QT interval without CYP3A4 involvement by telaprevir (e.g., methadone).

Syncope

Administration of Incivek was observed to be associated with an increased incidence of syncope. In the pooled safety data from the Phase II and III placebo-controlled studies, the incidence of syncope, syncope vasovagal, and loss of consciousness was three-times higher for patients treated with Incivek 750 mg every 8 hours than the placebo. The possibility does exist that either the QT prolongation, PR interval prolongation, or increased heart rate may be related to the syncope. In the absence of further ECG data, it is difficult to assess whether electrocardiographic effects contributed to this event, but the possibility cannot be ruled out. Other electrocardiographic AEs which occurred at a higher rate in Incivek-treated patients compared to placebo included atrial/cardiac flutter and bundle branch block.

Anaemia

In the Phase II and III studies, Peg-IFN-alfa/RBV therapy has been observed to be a cause of decreased haemoglobin concentrations, thereby leading to anaemia. The noted frequency of anaemia ADRs reported during Incivek treatment compared to placebo was higher within the T12/PR group (31.8%) than in the Pbo/PR group (14.8%). The severity of anaemia events was also higher in the T12/PR treatment group than in the Pbo/PR group. Anaemia events of at least Grade 3 occurred in 4.9% (T12/PR) versus 0.8% (Pbo/PR), of patients. Serious anaemia events were reported in 1.6% of patients in the T12/PR group, respectively, compared to <1.0% of patients in the placebo/PR group. Anaemia events leading to permanent discontinuation of the study drug alone and all study drugs were reported in 2.7% and <1.0% of patients in the T12/PR group, respectively, compared to <1.0% in each group, respectively, in patients in the placebo/PR group.

Skin Rash

The overall incidence and severity of skin rash increased when Incivek was coadministered with Peg-IFN-alfa/RBV. During the Incivek treatment phase, rash ADRs (all grades) were reported in 48.7% of patients who received Incivek in combination Peg- IFN-alfa/RBV compared to 28.0% of patients who received Peg-IFN-alfa/RBV treatment alone. More than 90% of skin rashes were of mild or moderate severity and were typically pruritic or eczematous in nature and involved less than 30% of the body surface area. The most common time for a rash to begin was during the first 4 weeks following initiation of Incivek treatment, however rashes could also occur at any other time during therapy. Improvement of the rash normally occurred after Incivek dosing was completed or discontinued; however, some rashes may take longer for complete resolution.

Due to the incidence of severe rash, the sponsor created a dermatology assessment panel to assess the cases of severe rash. The dermatology assessment panel reviewed a total of 221 cases, and found a number of serious skin reactions, including drug rash with eosinophilia and systemic symptoms (DRESS) and Stevens-Johnson Syndrome (SJS). The DRESS and SJS events were reported in the clinical development programme, at a rate of <1% of patients who received Incivek in combination with Peg-INF-alfa/RBV, compared to none who received Peg-INF-alfa/RBV alone. There were no deaths, and no cases of toxic epidermal necrolysis; however all serious skin reactions did require hospitalization for resolution.

The incidence of pruritus ADRs was noted to be higher in Incivek-treated patients compared to placebo-treated patients, 51.5% vs. 26.4% respectively. Discontinuation of Incivek or the placebo due to pruritus was infrequent in both groups, but higher in the T12/PR group than in the placebo group (1.0% vs. 0.1%). The incidence of pruritus was highest during the first 4 weeks of treatment and remained higher among patients receiving Incivek therapy. After Week 12, the incidence of pruritus was comparable between groups.

Hepatic Impairment

Incivek is not recommended for use in patients with moderate or severe hepatic impairment (Child-Pugh B or C, score ≥7) or patients with decompensated liver disease. However, no dose adjustment is necessary for patients with mild hepatic impairment (Child-Pugh A, score 5-6).

Pregnancy

Ribavirin is a known teratogen. Therefore, Incivek treatment in combination with Peg-INF-alfa/RBV should not be started unless a negative pregnancy test has been obtained immediately prior to initation of therapy. Extreme care must be taken to avoid pregnancy in female patients and in female partners of male patients. Female patients of childbearing potential and their male partners and male patients and their female partners of childbearing potential must use two effective methods of contraception during treatment, and for six months afterwards. Female patients should have monthly pregnancy tests during treatment. Hormonal contraceptives may not be reliable during Incivek treatment. Therefore, during this time, and for two months following treatment, female patients of childbearing potential should use two non-hormonal methods of contraception.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

The drug submission for Incivek was reviewed under the Priority Review Policy given Incivek was shown to provide substantial evidence of increased clinical efficacy when added to the current standard of care (Peg-INF-alfa/RBV), such that the overall benefit/risk profile is improved over existing therapies.

The efficacy of treatment of Incivek for 12 weeks in combination with Peg-INFalfa/RBV was shown to be statistically superior in Phase II and Phase III studies when compared to Peg-INF-alfa/RBV treatment alone. This included treatment-naïve patients, as well as patients who were previously treated with Peg-INF-alfa/RBV, including prior relapsers, prior partial responders and prior null responders.

There are some safety risks associated with Incivek treatment which include rash and in some instances serious skin reactions requiring hospitalization for resolution. These serious skin reactions occurred at a rate of <1% in patients treated with Incivek during clinical studies, and in no patients treated with Peg-INF-alfa/RBV alone. In addition, patients treated with Incivek experienced anaemia at a higher incidence (and greater severity) than patients treated with Peg-INF-alfa/RBV alone. Patients taking Incivek were also more prone to anorectal ADRs.

Other safety risks observed in patients treated with Incivek were prolongation of the QT, QRS and PR intervals, increased incidence of hypokalemia, and increased incidence of syncope. Inicivek was also noted to be a CYP3A4 inhibitor and substrate, as well as a Pgp inhibitor and substrate.

Incivek has shown to be a benefit in the treatment of chronic hepatitis C, genotype 1, in adult patients with compensated liver disease, including cirrhotics, when combined with Peg-INF-alfa/RBV therapy. In addition, clinical studies conducted did demonstrate that Incivek was well-tolerated and associated with a manageable safety profile. Based on the safety and efficacy profile, the benefits of Incivek in combination with Peg-INFalfa/RBV therapy seem to outweigh the risks. Restrictions to manage risks associated with the identified safety concerns have been incorporated into the Incivek Product Monograph.

3.4.2 Recommendation

Based on the Health Canada review of data on quality, safety and efficacy, Health Canada considers that the benefit/risk profile of Incivek is favourable in the treatment of genotype 1 chronic hepatitis C in adult patients with compensated liver disease, including cirrhosis, who are treatment-naïve or who have previously been treated with interferonbased treatment, including prior null responders, partial responders, and relapsers. The 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.

4 Submission Milestones

Submission Milestones: IncivekTM

Submission MilestoneDate
Pre-submission meeting:2010-10-13
Request for priority status
Filed:2010-12-09
Approval issued by Bureau of Gastroenterology, Infection and Viral Diseases:2010-01-13
Submission filed:2011-01-14
Screening
Screening Acceptance Letter issued:2011-02-18
Review
Biopharmaceutics Evaluation complete:2011-07-04
Quality Evaluation complete:2011-08-02
Clinical Evaluation complete:2011-08-10
Labelling Review complete:2011-08-10
Notice of Compliance issued by Director General:2011-08-16