Summary Basis of Decision for Afinitor

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
Afinitor

Everolimus, 5 mg and 10 mg, Tablet, Oral

Novartis Pharmaceuticals Canada Inc.

Submission control no: 125809

Date issued: 2010-06-07

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:

Afinitor

Manufacturer/sponsor:

Novartis Pharmaceuticals Canada Inc.

Medicinal ingredient:

Everolimus

International non-proprietary Name:

Everolimus

Strength:

5 mg and 10 mg

Dosage form:

Tablet

Route of administration:

Oral

Drug identification number(DIN):

  • 02339501 and 02339528

Therapeutic Classification:

Anti-neoplastic agent [mammalian target of rapamycin (mTOR) kinase inhibitor]

Non-medicinal ingredients:

Butylated hydroxytoluene (E321), magnesium stearate, lactose monohydrate, hypromellose, crospovidone, and lactose anhydrous.

Submission type and control no:

New Drug Submission, Control Number: 125809

Date of Submission:

2008-10-31

Date of authorization:

2009-12-14
AFINITOR* is a registered trademark.
2 Notice of decision

On December 14, 2009, Health Canada issued a Notice of Compliance to Novartis Pharmaceuticals Canada Inc. for the drug product Afinitor*.

Afinitor* contains the medicinal ingredient everolimus which is an antineoplastic agent.

Afinitor* is indicated for the treatment of patients with metastatic renal cell carcinoma (RCC) of clear cell morphology, after failure of initial treatment with either of the vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFr TKIs) sunitinib or sorafenib.

Renal cell carcinoma is an adenocarcinoma of the kidney that accounts for 85-90% of all kidney tumours. Afinitor* is a signal transduction inhibitor that targets the mammalian target of rapamycin (mTOR). Afinitor* has been shown to reduce cell proliferation, glycolysis, and angiogenesis in solid tumours in vivo.

The market authorization was based on quality, non-clinical, and clinical information submitted. The single pivotal study evaluating the efficacy and safety of Afinitor* in RCC was a prospective, randomized, double-blind, multicentre, placebo-controlled, parallel group Phase III study. Patients enrolled in the study had metastatic RCC that had progressed on or after either of the VEGFr TKI therapies sorafenib or sunitinib. In all, 277 patients were randomized to the Afinitor* arm and 139 patients to the matching placebo arm. The primary objective of the study was to compare progression-free survival (PFS) between the two treatment groups. The most up-to-date analyses demonstrated that patients administered Afinitor* had a median PFS time of 4.9 months compared to 1.9 months for the placebo group. Approval of Afinitor* is based on prolongation of PFS. It is not known if Afinitor* prolongs overall survival or improves quality of life in patients with metastatic RCC.

Afinitor* has important safety issues underscoring the need for this oral medication to be prescribed by a qualified healthcare professional who is experienced in the use of antineoplastic therapy. The two most clinically relevant adverse events are non-infectious pneumonitis (including interstitial lung disease) and infections.

Afinitor* (5 mg and 10 mg, everolimus) is presented in tablet form and should be administered orally once daily, at the same time every day (preferably in the morning), either in a fasting state or after no more than a light fat-free meal. Treatment should continue as long as clinical benefit is observed or until unacceptable toxicity occurs. Dosing guidelines are available in the Product Monograph.

Afinitor* is contraindicated for patients who are hypersensitive to the drug, to other rapamycin derivatives or to any of the excipients. Afinitor* 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 Afinitor* are described in the Product Monograph.

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

3 Scientific and Regulatory Basis for Decision

The New Drug Submission (NDS) for Afinitor* provides information for 5 mg and 10 mg immediate release everolimus tablets to be used as an anti-neoplastic agent for the treatment of metastatic renal cell carcinoma (mRCC).

3.1 Quality Basis for Decision

3.1.1 Drug Substance (Medicinal Ingredient)
General Information

Everolimus (a hydroxyethyl derivative of rapamycin) is the medicinal ingredient of Afinitor*. It is an anti-neoplastic agent that interacts with and inhibits signal transduction from the multi-domain serine/threonine kinase complex known as the mammalian target of rapamycin complex 1 (mTORC1). The mTORC1 plays a central role in regulating cell growth, proliferation, and survival via downstream signalling events.

Manufacturing Process and Process Controls

The drug substance is prepared semi-synthetically using rapamycin as a starting material. Rapamycin is obtained by a fermentation process. As everolimus is very sensitive to oxidative degradation, it is further processed with the antioxidant butylhydroxytoluene to produce a stabilized everolimus drug substance.

The materials used in the manufacture of the drug substance are considered to be suitable and/or meet standards appropriate for their intended use. The manufacturing process is considered to be adequately controlled within justified limits.

In-process controls performed during manufacture were reviewed and are considered acceptable. The specifications for the raw materials used in manufacturing the drug substance are also considered satisfactory.

Characterization

Detailed characterization studies were performed to provide assurance that everolimus consistently exhibits the desired characteristic structure. The structure of everolimus has been adequately elucidated and the representative spectra have been provided. Physical and chemical properties have been described and are satisfactory.

Control of Drug Substance

The sponsor has provided a summary of all drug-related and process-related impurities. The limits for impurities are the same as those approved for previous New Drug Submissions.

The drug substance specifications and analytical methods used for quality control of everolimus are considered acceptable. 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 everolimus.

Batch analysis results were reviewed and 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 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

Afinitor* tablets are supplied as white to slightly yellowish, elongated tablets with bevelled edges and no score line. The 5 mg tablets are debossed with 'NVR' on one side and '5' on the other side. The 10 mg tablets are debossed with 'NVR' on one side and 'UHE' on the other side.

The primary packaging for Afinitor* tablets are double-sided aluminium blister packs. Blisters are assembled in a cardboard-based pack.

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 drug substance with the excipients is demonstrated by the stability data presented on the proposed commercial formulation.

Pharmaceutical Development

Pharmaceutical development data, including selection of the container closure system, are considered acceptable.

Manufacturing Process and Process Controls

The drug product is formulated to contain ingredients commonly used in immediate release uncoated tablets. The manufacturing process uses conventional manufacturing techniques and consists of a two step process: wet granulation and dry blending.

All manufacturing equipment, in-process manufacturing steps and detailed operating parameters were adequately described in the submitted documentation and are acceptable. The manufacturing process is considered to be adequately controlled within justified limits.

Control of Drug Product

Afinitor* is tested to verify that its identity, appearance, content uniformity, assay, dissolution, weight, hardness, disintegration, friability, moisture content, levels of degradation products, and microbiological 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.

Stability

Based on the real-time, long-term, and accelerated stability data submitted, the proposed shelf-life at 15 to 30°C, protected from light and moisture is considered acceptable for Afinitor*.

3.1.3 Facilities and Equipment

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

All of the proposed manufacturing sites comply with the requirements of Division 2 of the Food and Drug Regulations.

3.1.4 Adventitious Agents Safety Evaluation

The magnesium stearate used in the drug product formulation is of vegetable origin and the lactose anhydrous is manufactured from milk fit for human consumption. All other 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 Afinitor* 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

The mTORC1 is part of the larger phosphoinositide 3-kinase (PI3K)/AKT/mTOR signalling pathway. Up-regulated activity of this pathway in cancer patients is often associated with worsening prognosis as a result of increased aggressiveness of neoplasms due to enhanced resistance to treatments and more rapid tumour progressions.

As a serine-threonine kinase signal-transducing protein, mTORC1 influences the synthesis of proteins involved in cell survival, growth, and proliferation. Hyperactivation of the mTOR pathway has been shown to favour cell and tissue growth. Activity of the mTOR pathway is regulated upstream by hormones and growth factors as well as directly by amino acids.

Like rapamycin, everolimus inhibits mTORC1 by binding to the immunophilin FKBP-12. This complex then binds to the FBR domain of mTORC1 and interferes with downstream signalling events including cell cycle progression at the G1 to S phase. Furthermore, inhibition of protein synthesis by everolimus has also been shown to decrease the synthesis of molecules involved in the formation of new blood vessels (that is [i.e.] vascular endothelial growth factor [VEGF]). This latter action comprises part of the proposed anti-angiogenic component of the drug.

Results of the primary pharmacodynamic studies support the proposed anti-tumour effects and mode of action (anti-angiogenic and anti-proliferative effects) of everolimus.

In vitro, everolimus binds with high affinity to the immunophilin FKBP-12, resulting in the inhibition of the mTORC1 complex. In turn, this suppresses downstream events such as S6K and 4EBP activity and cell-cycle arrest from G1 to S-phase. The effect on mTOR and S6K appeared specific; everolimus at concentrations up to 10 µM had no effect on the activities of ten different tyrosine and serine/threonine kinases tested in vitro.

Everolimus demonstrated a very broad inhibition of tumour cell lines of different tissue types in vitro with high sensitivity to anti-proliferative effects in some cells [half maximal inhibitory concentration (IC50) <1 nM and insensitivity in others (IC50 >1 µM)]. A panel of sixteen human RCC cell lines were tested in vitro for anti-proliferative activity following treatment with everolimus. Fourteen were sensitive to everolimus treatment (IC50 in the low/subnanomolar range), while two RCC cell lines were insensitive (IC50 >2500 nM).

The anti-tumour activity of everolimus was assessed in vitro in clonogenic assays. Human-derived tumour xenografts were generated by subcutaneously injecting human tumour cells into nude mice. Once tumours formed, they were extracted and the cells were re-isolated and used in clonogenic assays. Results of these tests demonstrated that everolimus inhibits tumour colony formation in a concentration-dependent manner. Sensitivity to everolimus was observed in a range of different tumour types.

An anti-angiogenic effect contributing to the anti-tumour activity is supported in vivo. Cell lines that were insensitive to everolimus in vitro responded to the drug when grown as tumours in mice. A decrease in tumour volume was noted, suggesting a significant anti-vascular/anti-angiogenic activity of everolimus consistent with the ability of this drug to decrease levels of hypoxia-inducible factor-1 (HIF-1) and VEGF in tumours in vivo. Thus, everolimus is expected to inhibit cancer cell growth by mechanisms directed against both tumour cells and the surrounding cellular milieu. Two of the human RCC cell lines were also tested for sensitivity to everolimus in vivo by growing them subcutaneously in athymic nude mice. Everolimus showed dose-dependent inhibition of growth and tumour regression in the more sensitive cell line.

The anti-angiogenic effects appeared most pronounced on VEGF-induced angiogenesis, where everolimus inhibited VEGF-induced formation of vascular tissue around growth factor-impregnated implant in rats, but had no effect on basic fibroblast growth factor (bFGF)-induced angiogenesis. It is noteworthy that everolimus had no significant effects on the baseline response to the implanted chambers, indicating that it does not impair wound healing.

At doses that were well tolerated, the efficacy of everolimus monotherapy in rat- and mouse-tissue models generally showed efficacy related to inhibition of tumour growth rather than tumour regression.

Safety Pharmacology

Safety pharmacology studies revealed no relevant effects of everolimus on vital functions of the cardiovascular system, respiratory system, or nervous system. Everolimus had no effects on QT interval prolongation in animal models as shown with conventional electrocardiogram (ECG) monitoring in minipigs and monkeys.

Although everolimus passes the blood-brain barrier, there was no indication of relevant changes in the behaviour of rodents, even after high doses.

Based on these findings, the potential for everolimus to affect vital functions in humans is considered to be low.

3.2.2 Pharmacokinetics
Absorption

The oral absorption of everolimus was low in mice (12%) and monkeys (18%) and medium in rats (39 to 43%). First-pass metabolism reduced oral bioavailability to 5% in mice, 14 to 26% in rats, and 6% in monkeys. Everolimus was cleared slowly in all species with the elimination half-life supporting a once-daily administration.

Distribution

In plasma, the free fraction of everolimus was independent of concentration and averaged 7.6% in the rat, 16% in the monkey, and only 0.1% in the mouse. With the exception of the mouse, the blood distribution of everolimus was concentration-dependent. At a concentration of 5 ng/mL, the distribution was 66% and 79% in the rat and monkey, respectively. In the mouse, the majority of everolimus (~98%) was located in plasma. The volume of distribution at steady-state (Vss) was species-dependent and ranged from high in the rat (44 to 52 L/kg) to very low in the mouse (0.37 L/kg).

The tissue distribution of everolimus was characterized in rats administered radio-labelled drug. Radioactivity was essentially extravascular with the highest levels found in the heart, lung, liver, kidney, spleen, thyroid, and adrenal gland. Unchanged everolimus was the major component in tissue samples after single oral or intravenous (IV) administration. The blood-brain passage of everolimus and/or its metabolites was found to be dose-dependent. Everolimus-related radioactivity passed the placenta of pregnant rats to a limited degree and was readily transferred into the milk of lactating rats.

Metabolism

Everolimus was extensively metabolized in the mouse, rat, and monkey. In all species, everolimus formed a large number of metabolites. Everolimus is essentially metabolized through oxidation by the cytochrome P450 (CYP) isozyme CYP3A4 in the liver and to some extent in the gut wall. Therefore, co-medications that are strong CYP3A4 inducers have the potential to reduce everolimus metabolism in vivo. Conversely, everolimus inhibited competitively the metabolism of the CYP3A4 substrate cyclosporine and was also a mixed inhibitor of the metabolism of the CYP2D6 substrate in vitro.

Excretion

Everolimus was predominantly eliminated through metabolic biliary/faecal clearance in all animal species. Excretion was essentially complete in all species. Renal excretion was a minor component (0.7 to 7%). No unchanged drug was detected in the urine or faeces.

3.2.3 Toxicology

The toxicology program included studies conducted in rodents, monkeys, and minipigs. The selection of rodent and non-rodent species is appropriate as the pharmacological target, mTOR, is highly conserved across species.

Single-Dose Toxicity

Everolimus was well-tolerated in rats and mice when administered orally as a single 2000 mg/kg dose. The only significant clinical observation was slight dyspnea (shortness of breath) in female mice.

Following single-dose IV administration in rats, 10 and 40 mg/kg were lethal, but at 2.5 mg/kg all animals survived. The estimated lethal dose for 50% of the sample population (LD50) was 6.3 mg/kg. Clinical signs included sedation, dyspnoea, and ventral and/or lateral recumbency, and convulsions in all animals.

Repeat-Dose Toxicity

Repeat-dose toxicity studies were conducted in mice for over 13 weeks, in rats up to 26 weeks, in minipigs up to 4 weeks, and in monkeys up to 52 weeks. The toxicology profile of everolimus was generally similar to that observed for rapamycin. The major target organs of toxicity were the male and female reproductive systems (testicular tubular degeneration, reduced sperm content in the epididymides, and uterine atrophy) in several species; lungs (increased alveolar macrophages) in rats and mice; and eyes (lenticular anterior suture line opacities) in rats only. Minor kidney changes were seen in rats (exacerbation of age-related lipofuscin in tubular epithelium and increases in hydronephrosis) and in mice (exacerbation of background lesions).

Everolimus exacerbated spontaneous background disease (chronic myocarditis in rats, Coxsackie virus infection of plasma and heart in monkeys, coccidian infestation of the gastrointestinal tract in minipigs, and skin lesions in mice and monkeys). These findings were generally observed at systemic exposure levels within the range of therapeutic exposure or above, with the exception of the findings in rats, which occurred below therapeutic exposure due to high tissue distribution.

Genotoxicity and Carcinogenicity

Studies covering relevant genotoxicity endpoints showed no evidence of clastogenic or mutagenic activity. Administration of everolimus to mice and rats for up to 2 years did not indicate any oncogenic potential at the highest doses, corresponding respectively to 4.3- and 0.2-times the estimated clinical exposure in humans at a dose of 10 mg/day.

Reproductive and Developmental Toxicity

In male rats, everolimus had adverse effects on the reproductive organs at doses as low as 0.5 mg/kg. Sperm motility, sperm head count, and plasma testosterone levels were diminished at 5 mg/kg, which is within the range of therapeutic exposure. This led to a reduction in male fertility; however, there was evidence of reversibility. Female fertility was not affected, but everolimus did cross the placenta. At doses as low as 0.1 mg/kg (below the therapeutic level) embryofoetal toxicity which manifested as mortality and reduced foetal weight was evident. The incidence of skeletal variations and malformations was increased at doses of 0.3 and 0.9 mg/kg.

Maternal and embryofoetal toxicity, but no teratogenicity, were seen in rabbits at doses of 0.2 and 0.8 mg/kg, respectively.

In a pre- and post-natal development study conducted with rats, doses of 0.1 and 0.3 mg/kg everolimus did not cause maternal toxicity, but were associated with reduced survival and body weight of the pups; however there was no effect on fertility and reproductive parameters of the first filial (F1) generation. Exposures in the rats and rabbits at all doses were well below maximal human therapeutic levels.

Local Tolerance

Everolimus did not cause contact hypersensitivity in guinea pigs or acute venous or primary skin irritation in rabbits. Everolimus was not antigenic in guinea pigs, mice, or rats. The value of the negative results of these studies to predict or exclude allergenic potential of everolimus is questionable as the active system anaphylaxis (ASA) and passive cutaneous anaphylactic (PCA) assays used have low sensitivity for low-molecular-weight drug.

3.2.4 Conclusion

Everolimus achieves its therapeutic effect by forming a complex with FKBP-12 that binds to and inhibits downstream signalling from mTORC1. The results of the non-clinical pharmacodynamic studies support the proposed anti-tumour effects and mode of action (anti-angiogenic and anti-proliferative effects) of everolimus. Safety pharmacology study findings suggest the potential for everolimus to affect vital organ functions in humans is low.

The pharmacokinetic (PK) profile of everolimus has been adequately characterized in the non-clinical studies. It is eliminated by metabolism and excreted mainly in the faeces. Metabolites generated appear to be pharmacologically inactive.

The toxicity profile of everolimus has been extensively characterized with findings indicating toxicity to immune system tissues, male and female reproductive organs, as well as a number of other tissues and organs. Many of these findings are likely related to the exacerbation of underlying conditions by everolimus-induced immunosuppression or are considered to be species-specific.

Overall, the non-clinical pharmacology and toxicology data submitted support the use of Afinitor* (everolimus) for the proposed indication. Clinical monitoring will clarify the safety profile of Afinitor* in the clinical setting.

3.3 Clinical basis for decision

Renal cell carcinoma is an adenocarcinoma of the kidney that originates in the renal cortex and accounts for 85-90% of all kidney tumours. Chemotherapy and radiation therapy demonstrate little efficacy in advanced disease, and the five-year survival rate is low, ranging from 5-10%.

Renal cell carcinoma is linked to different environmental factors and in some cases is due to genetic predispositions (familial forms). The strongest environmental factor remains cigarette smoking. Loss of function of the tumour-suppressor gene product Von Hippel Lindau (VHL) is the most common familial form of the disease. The increased expression of VEGF in patients with VHL syndrome has led to the supposition that angiogenesis is a key factor in RCC. This resulted in the development and market authorization of the anti-neoplastic agents sorafenib and sunitinib that block angiogenesis by inhibiting the function of the VEGF receptor (VEGFr).

Despite recent progress in treating RCC, most patients progress at some point after treatment with the VEGFr TKI therapies sorafenib and sunitinib. Everolimus is a signal transduction inhibitor targeting mTOR, or more specifically, mTORC1, a key serine-threonine kinase that plays a central role in the regulation of cell growth, proliferation and survival. Consistent with the central regulatory role of mTORC1, its inhibition by everolimus has been shown to reduce cell proliferation, glycolysis and angiogenesis in solid tumours in vivo, both through direct anti-tumour cell activity and inhibition of the tumour stromal compartment.

3.3.1 Human Pharmacology

Everolimus has been approved as an immunosuppressive drug for use in the transplant setting in many countries in Europe and elsewhere (but not in Canada or the United States of America) under the name Certican®.

Clinical studies supporting the oncological indication were used in the evaluation of Afinitor*; however, studies in support of the transplant indication were also considered.

3.3.2 Pharmacodynamics

The proposed 10 mg dose for the treatment of patients with RCC was determined in part from pre-clinical modelling studies, clinical pharmacology reports, and from published literature results. The maximum tolerated dose (MTD) was derived from kidney transplant patients receiving a 5 mg/day-dose together with Neoral® (cyclosporine A). Cyclosporine A increases everolimus exposure by 2- to 3-fold therefore it was proposed that a 10 to 15 mg dose in an oncological setting would be equivalent (in the absence of a CYP3A4 inhibitor). The dose-limiting toxicity for this study was thrombocytopaenia (three out of seven patients receiving 10 mg everolimus plus Neoral®). Results from the Phase I studies suggested that a daily dose of ≥10 mg everolimus is required to achieve inhibition of the phosphorylation of S6K1, 4E-BP1, and eIF-4G in the majority of patients.

In quiescent cells, mTOR activity is reduced and the eukaryotic initiation factor 4G (eIF-4G) protein is bound by 4E-BP1 (in its non-phosphorylated state). In cancer cells, active signalling through mTOR induces the phosphorylation of 4E-BP1, which results in the release of eIF-4G, in turn initiating CAP-dependent translation resulting in protein synthesis. In addition to its classic role in the phosphorylation of S6-kinase, mTOR has also been implicated in the direct phosphorylation of eIF-4G.

A study was conducted in adult patients with advanced solid cancers that were refractory or unsuitable for existing standard therapies. Patients who participated in the study had to be able and willing to undergo comparative tumour biopsy. One of the primary objectives of the study was to assess changes in the molecular markers within the tumours known to be most directly influenced by mTOR. Analysis of these markers was used as a means to determine the ability of Afinitor* to inhibit mTOR and its downstream effectors. The results of this study demonstrated a change in the phosphorylation status of S6, eIF-4G, 4E-BP1, and AKT in tumour and skin biopsies before and after Afinitor* treatment with the indicated doses and scheduling of the drug. In addition, the 10 mg dose was marginally better at inhibiting eIF-4G phosphorylation within the tumour; however, the 5 mg dose was nearly as effective as the 10 mg dose, therefore a dose reduction may still have a beneficial effect.

3.3.3 Pharmacokinetics
Absorption

In patients with advanced solid tumours, peak plasma everolimus concentrations (Cmax) were reached 1 to 2 hours following oral administration of a 5 to 70 mg dose under fasting conditions or with a light fat-free snack. The Cmax was dose-proportional between 5 and 10 mg in daily and weekly regimens. At doses of ≥20 mg/week, the increase in Cmax was less than dose-proportional; however, the area under the concentration time curve (AUC) showed dose-proportionality within the 5 to 70 mg dose range.

The bioavailability of everolimus is estimated to be 11% determined as the amount of remaining radioactivity of the administered radio-labelled everolimus measured at Cmax. Steady-state was achieved within two weeks with the daily dosing regimen. There was a significant correlation between AUC0-τ (the area under the curve in a dosing interval) and pre-dose trough concentration at steady-state on the daily regimen.

In healthy subjects taking 1 mg everolimus tablets, a high-fat meal reduced Cmax and AUC by 60% and 16% respectively. No data are currently available with Afinitor* 5 and 10 mg tablets, which have a quantitatively different formulation from the 1 mg everolimus tablets used in this study.

Distribution

The blood-to-plasma ratio of everolimus, which is concentration dependent within the range of 5 to 5,000 ng/mL was 17% to 73%. The amount of everolimus confined to the plasma was approximately 20% at blood concentrations observed in cancer patients given Afinitor* 10 mg/day. Plasma protein binding was approximately 74% both in healthy subjects and in patients with moderate hepatic impairment.

Metabolism

Everolimus is metabolized in the liver and is a substrate of CYP3A4 and p-glycoprotein (PgP). The major metabolites of everolimus in the blood are hydroxylation derivatives with the exception of one metabolite that was modified by the addition of a phosphatidylcholine group. All identified metabolites were >100-times less potent than everolimus in a mixed lymphocyte reaction suggesting that the parent compound is the major biologically active compound.

Excretion

Following the administration of a single dose of radio-labelled everolimus in conjunction with cyclosporine, only 5% of the total radioactivity was detected in urine whereas 80% was detected in the faeces over 10 days. The results confirm non-clinical findings and suggest that the major route of excretion is by the liver. No parent compound could be detected in either the faeces or urine.

Special Populations

The PK evaluations of everolimus in healthy subjects versus (vs.) those with moderately impaired hepatic function (Child-Pugh Class B) further demonstrate the important role of the liver in clearance of the drug. The apparent clearance of everolimus was reduced by half in individuals with moderate hepatic impairment compared with healthy, demographically-matched subjects. Dose reductions to 5 mg would be warranted in this population. No studies of patients with severe hepatic impairment (Child-Pugh Class C) have been performed. Treatment of this population should be avoided pending such investigations.

3.3.4 Clinical Efficacy

The efficacy for Afinitor* was primarily assessed in the pivotal study RECORD-1, a Phase III, prospective, randomized, double-blind, multicentre, placebo-controlled, parallel-group study. RECORD-1 was designed to evaluate the efficacy and safety of Afinitor* in patients with mRCC who failed initial treatment with either of the VEGFr TKIs sunitinib and/or sorafenib. Afinitor* fulfills an unmet medical need for these patients as no other treatment options have proven effective for this population. RECORD-1 is the only Phase III study that has addressed the efficacy of Afinitor* in patients with mRCC.

Patients who met the study criteria were randomized 2:1 to receive 10 mg/day Afinitor* (n = 277) or a matching placebo (n = 139). Prior to randomization, patients were stratified according to Memorial Sloan Kettering Cancer Centre (MSKCC) prognostic score class of good, intermediate or poor, as well as by treatment with either one or two of the VEGFr TKI therapies.

The primary objective of the study was to compare progression-free survival (PFS) in patients who received Afinitor* plus best supportive care (BSC) vs. patients who received placebo plus BSC. Secondary objectives included an assessment of benefits in overall survival (OS), objective response rates (ORR), and patient reported outcomes (disease related symptoms and quality of life [QoL]).

The study design permitted immediate cross-over from the placebo group to the treatment group upon detection of progressive disease (PD) as determined by the investigator (who became un-blinded upon progression). The primary analysis of radiological assessments was based on the Response Evaluation Criteria in Solid Tumours (RECIST) and was conducted by an independent (central) review committee (IRC) who remained blinded. RECIST is a set of criteria used to measure tumour response using X-ray, computed tomography (CT), and magnetic resonance imaging (MRI).

The results of the primary PFS data were robust. The most up-to-date analyses provided by the sponsor demonstrated that patients who were administered Afinitor* had a median PFS of 4.9 months compared to 1.9 months for those who received placebo [hazard ratio (HR): 0.33, 95% confidence interval (CI): 0.25 to 0.43, p < 0.0001].

An independent data monitoring committee (IDMC) recommended stopping the study at the second interim analysis because the pre-specified PFS endpoint had been met. Despite the PFS results, no benefit in OS was demonstrated. This may have occurred, in part, because the study protocol allowed patients in the placebo group with PD to cross-over immediately to the treatment group, which when combined with very early progression, resulted in confounding of the OS results. By the October 15th, 2007 cut-off, 79 of 98 individuals in the placebo group who had PD had accepted Afinitor* treatment. Of these 79 patients, 60 had PD within the first 8 weeks of randomization.

The Oncology Division (OD), Health Canada, felt that immediate cross-over at progression might not have been necessary despite ethical and feasibility considerations. This study was conducted to assess a second-/third-line therapy and no other treatments were available. The placebo group was therefore not necessarily disadvantaged as the effectiveness of Afinitor* had not been established at enrolment of this first and only Phase III study.

No statistically significant differences between the Afinitor* and placebo groups were observed in any of the pre-specified secondary efficacy endpoints. Only five patients (1.8%) in the Afinitor* group experienced a partial response as measured by RECIST criteria. Also, patients who received Afinitor* did not exhibit an improved QoL compared to patients who received placebo.

The superiority of Afinitor* was demonstrated in the time to definitive deterioration of Karnofsky performance status (KPS) score relative to placebo. The KPS scale classifies patients according to their functional impairment. The time to deterioration was prolonged for patients receiving Afinitor* therapy (HR: 0.66, 95% CI: 0.49 to 0.90); however, the KPS was not included as a pre-planned secondary efficacy endpoint by the sponsor. In addition, some potential for bias exists in these results as the investigator may not have been truly blinded due to the presence of rash and other adverse events (AEs) which were more commonly presented in the Afinitor* group. This may have influenced the objectivity of the assessment. Despite this potential bias, the evidence shows a modest improvement in performance status in the Afinitor* group.

Overall, the results of the pivotal study demonstrated a PFS benefit for patients receiving Afinitor* over placebo. No statistically significant differences in response rates or quality of life were demonstrated between the Afinitor* and placebo arms. Crossover to open-label Afinitor* following disease progression for patients allocated to placebo may have confounded the ability to detect treatment-related differences in overall survival. Despite the failure to show a statistically significant improvement in OS, Afinitor* is likely to provide a clinically meaningful benefit to patients whose disease has progressed after initial treatment with sunitinib and/or sorafenib. A gain of 3 months in PFS together with a compelling hazard ratio (0.33), could likely result in an OS gain for this mRCC patient population.

3.3.5 Clinical Safety

The clinical safety of Afinitor* was primarily assessed in the pivotal study RECORD-1 that is described in section 3.3.4 Clinical Efficacy.

The most up-to-date safety information provided by the sponsor (in the 90-day safety update report) reported that 13.9% of patients in the Afinitor* group discontinued due to an AE.

Mucositis (including stomatitis and aphthous stomatitis) was the most common treatment-emergent AE associated with Afinitor*. In general, these cases were manageable with minimal treatment and in some cases dose interruptions. Rarely was discontinuation from study medication required.

Other notable AEs that occurred more frequently in Afinitor*-treated patients included cardiovascular disorders (tachycardia and congestive heart failure, >1% of patients), respiratory disorders (pleural effusion, 7% of patients), and gastrointestinal disorders (abdominal pain, dry mouth, and haemorrhoids). Rash and similar events were very common and occurred in 35% of patients; however, no discontinuations resulted from these AEs.

Pneumonitis and Infections

Pneumonitis and infections were the most clinically significant treatment-emergent AEs (TEAEs), even though these were not the most commonly experienced AEs. Two deaths resulting from underlying infections were suspected to be related to Afinitor*. At the October 15th, 2007 cut-off, one Afinitor*-related death was the result of overwhelming candidal sepsis, complicated by acute respiratory failure. A second death which resulted from sepsis (on Day 112) was reported at the February 25th, 2008 cut-off date. A third patient died from an event attributed to underlying renal cancer; however, the individual also had Grade 3 pneumonitis that was suspected to be caused by Afinitor*. This event may have contributed to the fatality.

Both pneumonitis and infections must be carefully monitored during the course of treatment with Afinitor*. A Serious Warnings and Precautions box has been included in the Product Monograph highlighting pneumonitis and infections as potentially serious side effects of Afinitor*.

Thrombocytopaenia and Anaemia

Afinitor* was initially developed as an immunosuppressive drug for use in the transplant setting. During development, it was determined that a daily 5 mg dose of Afinitor* when taken with cyclosporine A produced the same systemic exposure to the drug as a daily 10 mg dose of Afinitor* taken alone; however, a 10 mg dose taken with cyclosporine A was associated with three severe cases of thrombocytopaenia in seven patients (dose-limiting toxicity).

For the current NDS, thrombocytopaenia occurred more frequently in patients receiving Afinitor* in both the pivotal and supportive studies. Based on the totality of the data, 10 mg is the near-maximum tolerated dose and thrombocytopaenia is a class effect of mTOR inhibitor drugs. As thrombocytopaenia is an important AE associated with Afinitor*, platelet counts need to be monitored closely throughout the course of treatment.

Anaemia was also common throughout the pivotal and supportive studies and in the pivotal study at least two-thirds of the cases were suspected to be directly related to Afinitor* treatment. Haemoglobin levels should be routinely measured and blood transfusions will be required for a subset of the patient population.

Hyperlipidaemia and Hyperglycaemia

Afinitor* has been shown to increase both circulating lipids (cholesterol and triglycerides) and glucose concentrations. Baseline measurements should be performed as high initial lipid and glucose concentrations were often found to be associated with exacerbation of these levels. Type 2 new-onset diabetes occurred with the use of Afinitor*, although rarely. Hyperlipidaemia can be managed with diet, statins, and fibrates as necessary. Dietary changes can usually alleviate hyperglycaemia with or without additional therapy such as metformin and possibly insulin.

Renal Considerations

Although renal failure was uncommon, the frequency increased in the 90-day safety update in the Afinitor* group indicating that it could become more prevalent with chronic use. Ten percent of the patients in the Afinitor* group experienced notable increases in serum creatinine (usually Grade 1 or 2). No increases were reported in the placebo group. Serum creatinine levels should be monitored throughout treatment.

No studies have been carried out in patients with impaired renal function. However, given that the drug is primarily metabolized by the liver, no exclusions with this population are listed.

Hepatic Considerations

Afinitor* is primarily metabolized by the liver and is a primary substrate for CYP3A4. No studies have been performed in patients with severe hepatic impairment (Child-Pugh Class C) and treatment is not recommended for this patient population. Furthermore, strong CYP3A4 inhibitors should not be taken with this medication. A dose reduction to 5 mg/day or every second day is recommended for patients that require co-medication with moderate CYP3A4 inhibitors. By contrast, although a decrease in Afinitor* systemic exposure is expected with CYP3A4 inducers, no recommendations have been made to increase the Afinitor* dose as no formal studies have been carried out in this setting.

Summary

Overall, the safety features of Afinitor* appear acceptable for the indicated population. Afinitor* has important safety issues highlighting the need for this oral medication to be prescribed by a qualified healthcare professional who is experienced in the use of anti-neoplastic therapy. The two most clinically relevant adverse drug reactions (ADRs) are pneumonitis (interstitial lung disease) and infections. Fatalities have occurred during the pivotal study with this medication due to these ADRs.

Careful monitoring of the symptoms mentioned above and appropriate use of co-medications, dose interruptions, and reductions should make the AEs manageable for the majority of patients taking this drug. Severe AEs may result in discontinuations; however, the effects are usually reversible. A few drug-related deaths have occurred in the clinical studies.

3.3.6 Additional Issues

A risk management plan (RMP) was submitted with this NDS which focuses primarily on subjects excluded from the pivotal study and potential risks to these patients if treated with Afinitor*. Hepatitis B reactivation has occurred with Afinitor* in clinical experience with the drug. Patients with active hepatitis were excluded from the pivotal study and may be more susceptible to serious illness if treated with Afinitor*.

As well, the sponsor will be requested to provide to Health Canada the final per-protocol OS analysis of the RECORD-1 study which was to be conducted two years after randomization of the last patient as a post-marketing commitment.

3.4 Benefit/Risk Assessment and Recommendation

3.4.1 Benefit/Risk Assessment

Canadian clinical experts were consulted during the assessment of the clinical efficacy and safety of Afinitor*. The advice received was taken into account in the final conclusions and decisions made by Health Canada.

Upon review of the pivotal study, it was concluded that immediate cross-over at progression was not necessary and that the presumption of efficacy of Afinitor* was perhaps premature in the pivotal Phase III study. At the same time it was felt that this drug would likely provide an OS benefit to a patient population that has no other treatment options available. It was felt that there was sufficient evidence in other studies to suggest (although no meta-analysis supports this claim) that PFS is related to OS in mRCC. In addition, the HR of 0.33 (a 67% reduction in the risk of progression) was an important factor in the overall assessment together with the increase in 3 months in median PFS (2.6-fold longer duration in median PFS).

Afinitor* is intended for patients with mRCC with few additional options for treatment. Despite the fact that a statistically significant improvement in OS may have been confounded by immediate cross-over, Afinitor* is likely to provide a clinically meaningful benefit to patients who have progressed after initial treatment with sunitinib and/or sorafenib. The evidence of efficacy is based on robust PFS data, as well as data on time to deterioration of performance status, demonstrating the superiority of Afinitor* relative to placebo, as well as QoL data demonstrating no significant difference between Afinitor* and placebo. There are safety concerns with chronic use of 10 mg/day Afinitor*, but these will be addressed through careful monitoring, secondary treatments, dose interruptions, and discontinuations if necessary. Two deaths in the pivotal study have been directly linked to Afinitor*. The drug is considered to be of moderate toxicity and will undoubtedly not be tolerated by some individuals; however, overall, Afinitor* will likely prolong the survival of patients with mRCC. Based on the totality of the data, Health Canada considers that the potential benefit of the drug outweighs the associated risks for this patient population that has no other treatment options available.

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 Afinitor* is favourable for the treatment of patients with mRCC of clear cell morphology, after failure of initial treatment with either of the VEGFr TKIs sunitinib or sorafenib.

Approval of Afinitor* is based on PFS. Prolongation of OS was not demonstrated for Afinitor* in RCC nor were QoL differences shown between patients receiving Afinitor* vs. placebo in the pivotal Phase III study.

4 Submission Milestones

Submission Milestones: Afinitor

Submission MilestoneDate
Pre-submission meeting:2008-06-12
Request for priority status
Filed:2008-09-15
Rejection/approval issued by Director, Bureau of Metabolism, Oncology and Reproductive Sciences:2008-10-16
Submission filed:2008-10-31
Screening
Screening Deficiency Notice issued:2008-12-17
Response filed:2009-01-27
Screening Acceptance Letter issued:2009-02-17
Review
Quality Evaluation complete:2009-12-11
Clinical Evaluation complete:2009-12-14
Biostatistics Evaluation complete:2009-10-02
Labelling Review complete:2009-12-08
Scientific Advisory Committee for Oncology Therapy (SAC-OT) meeting held:2009-10-15
Notice of Compliance issued by Director General:2009-12-14