Summary Basis of Decision for Giotrif

Review decision

The Summary Basis of Decision explains why the product was approved for sale in Canada. The document includes regulatory, safety, effectiveness and quality (in terms of chemistry and manufacturing) considerations.


Product type:

Drug

Summary Basis of Decision (SBD) documents provide information related to the original authorization of a product. The SBD for Giotrif is located below.

Recent Activity for Giotrif

SBDs written for eligible drugs approved after September 1, 2012 will be updated to include post-authorization information. This information will be compiled in a Post-Authorization Activity Table (PAAT). The PAAT will include brief summaries of activities such as submissions for new uses of the product, and whether Health Canada's decisions were negative or positive. PAATs will be updated regularly with post-authorization activity throughout the product's life cycle.

Post-Authorization Activity Table (PAAT) for Giotrif

Updated:

2021-11-02

The following table describes post-authorization activity for Giotrif, a product which contains the medicinal ingredient afatinib (as afatinib dimaleate). For more information on the type of information found in PAATs, please refer to the Frequently Asked Questions: Summary Basis of Decision (SBD) Project: Phase II and to the list of abbreviations that are found in PAATs.

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

Drug Identification Number (DIN):

  • DIN 02415666 - 20 mg afatinib as afatinib dimaleate, tablet, oral
  • DIN 02415674 - 30 mg afatinib as afatinib dimaleate, tablet, oral
  • DIN 02415682 - 40 mg afatinib as afatinib dimaleate, tablet, oral

Post-Authorization Activity Table (PAAT)

Activity/submission type, control numberDate submittedDecision and dateSummary of activities
NC # 2257312019-03-14Issued NOL
2019-06-07
Submission filed as a Level II (90 day) Notifiable Change to update the PM with new safety information. As a result of the NC, modifications were made to the Serious Warnings and Precautions Box, Warnings and Precautions, and Adverse Reactions sections of the PM, and corresponding changes were made to the PM Part III: Patient Medication Information. The submission was reviewed and considered acceptable, and an NOL was issued.
SNDS # 2097852017-09-29Issued NOC
2018-09-20
Submission filed as a Level I - Supplement to expand the indication to include three new rare Epidermal Growth Factor Receptor (EGFR) mutations in the indication and the Clinical trials section of the Product Monograph (PM): mutations in exon 21 L861Q, exon 18 G719X, and exon 20 S768I. Following review, the information was added to the Clinical trials section only and an NOC was issued.
NC # 2048162017-04-18Issued NOL
2017-07-14
Submission filed as a Level II (90 day) Notifiable Change to update the PM. As a result of the NC, modifications were made to the Action and Clinical Pharmacology, section of the PM, and corresponding changes were made to the PM Part III: Patient Medication Information. The submission was reviewed and considered acceptable, and an NOL was issued.
SNDS # 1966312016-07-14Issued NOC
2017-06-22
Submission filed as a Level I - Supplement to provide a safety and efficacy update to the PM. As a result of the SNDS, modifications were made to the Adverse Reactions, and Dosage and Administration sections of the PM, and corresponding changes were made to the PM Part III: Patient Medication Information. The benefit/risk profile for Giotrif remains positive when used for its approved indication. An NOC was issued.
SNDS # 1876562015-09-14Issued NOC
2016-08-25
Regulatory Decision Summary published
NC # 1743482014-05-06Issued No Objection Letter
2014-08-05
Submission filed as a Level II (90 day) Notifiable Change (Safety Change) to update the Warnings and Precautions section of the Product Monograph with new safety information to show that Giotrif combined with vinorelbine should not be used in patients with HER 2 positive metastatic breast cancer. In addition, two minor revisions were also proposed under the Dosage and Administration section and in Part III. The submission was reviewed and a No Objection Letter was issued.
Drug product (DINs 02415666, 02415674, 02415682) market notificationNot applicableDate of first sale:
2014-01-17
The manufacturer notified Health Canada of the date of first sale pursuant to C.01.014.3 of the Food and Drug Regulations.
NDS # 1587302012-09-19Issued NOC
2011-11-01
Notice of Compliance issued for New Drug Submission.
Summary Basis of Decision (SBD) for Giotrif

Date SBD issued: 2014-05-12

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

Afatinib (as afatinib dimaleate), 20, 30, and 40 mg,  tablets, oral

Drug Identification Number (DIN):

  • DIN 02415666 - 20 mg tablet
  • DIN 02415674 - 30 mg tablet
  • DIN 02415682 - 40 mg tablet

Boehringer Ingelheim Canada Ltd.

New Drug Submission Control Number: 158730

On November 1, 2013, Health Canada issued a Notice of Compliance to Boehringer Ingelheim (Canada) Ltd. for the drug product, Giotrif.

The market authorization was based on quality (chemistry and manufacturing), non clinical (pharmacology and toxicology), and clinical (pharmacology, safety, and efficacy) information submitted. Based on Health Canada's review, the benefit/risk profile of Giotrif is favourable as monotherapy for the treatment of Epidermal Growth Factor Receptor (EGFR) tyrosine kinase inhibitor-naïve patients with metastatic (including cytologically proven pleural effusion) adenocarcinoma of the lung with activating EGFR mutation(s).

1 What was approved?

Giotrif, a protein kinase inhibitor, was authorized as monotherapy for the treatment of Epidermal Growth Factor Receptor (EGFR) tyrosine kinase inhibitor-naïve patients with metastatic (including cytologically proven pleural effusion) adenocarcinoma of the lung with activating EGFR mutation(s).

  • A validated test is required to identify EGFR mutation status.
  • The clinical effectiveness was based on progression-free survival and objective response. No overall survival benefit was demonstrated. Safety and efficacy of Giotrif have not been established in patients with EGFR mutations other than exon 19 deletions (DEL19) and the exon 21 L858R point mutation (see the Product Monograph Clinical Trials section).
  • Close monitoring and proactive management of diarrhea is essential for successful Giotrif treatment (see the Product Monograph Warnings and Precautions section).

Giotrif is contraindicated in patients with known hypersensitivity to afatinib or to any of the ingredients of the product. Giotrif was approved for use under the conditions stated in the Giotrif Product Monograph taking into consideration the potential risks associated with the administration of this drug product.

Giotrif (20, 30, and 40 mg, afatinib, as afatinib dimaleate) is presented as film-coated tablets. In addition to the medicinal ingredient, the film-coated tablets contain colloidal anhydrous silica, crospovidone, lactose monohydrate, magnesium stearate, and microcrystalline cellulose.

For more information, refer to the Clinical, Non-Clinical, and Quality (Chemistry and Manufacturing) Basis for Decision sections.

Additional information may be found in the Giotrif Product Monograph, approved by Health Canada and available through the Drug Product Database.

2 Why was Giotrif approved?

Health Canada considers that the benefit/risk profile of Giotrif is favourable as monotherapy for the treatment of Epidermal Growth Factor Receptor (EGFR) tyrosine kinase inhibitor naïve-patients with metastatic (including cytologically proven pleural effusion) adenocarcinoma of the lung with activating EGFR mutation(s).

Metastatic adenocarcinoma of the lung (including malignant pleural effusion) is a serious, life-threatening disease with poor 5-year survival. There is an unmet medical need for treatment options that improve clinical outcome of the patients, including prolonging survival and/or quality of life.

In the pivotal study LUX-Lung 3, Giotrif has demonstrated a significant improvement in progression-free survival (PFS) compared with the cisplatin/pemetrexed chemotherapy doublet in patients with metastatic (including cytologically proven pleural effusion)  adenocarcinoma of the lung and not having been treated with a prior EGFR TKI [that is (i.e.), EGFR TKI-naïve patients]. The median PFS was 11.1 months in the Giotrif arm (N = 230), versus (vs.) 6.9 months in the chemotherapy arm (N = 115), corresponding to a hazard ratio (HR) of 0.58 (95% CI: 0.43, 0.78, p = 0.0004). Objective response rate was higher in the Giotrif arm (56.1%) than in the chemotherapy arm (22.6%). This magnitude of tumor control is considered clinically meaningful. Efficacy results of the LUX-Lung 2 study supported the tumor response data in the pivotal study.

In LUX-Lung 3 there were 37 patients with uncommon EGFR mutation subtypes (10 different molecular subtypes with unequal distribution between the two study arms) in the pivotal study. Among 26 patients treated with Giotrif, 9 achieved a partial response (N = 4) or prolonged stable disease (N = 5) with prolonged PFS of longer than 6 months, although the activity of Giotrif appeared lower in those with the T790M mutation (only one response in 11 patients). This level of tumor response may be clinically meaningful, taking into consideration limited therapeutic options for these patients and the rarity of the disease [a variety of uncommon EGFR mutations represent approximately 10% of all EGFR mutations in non-small cell lung cancer (NSCLC)]. Therefore, this patient subpopulation is not explicitly excluded from the authorized indication. However, it is prominently captured in the Giotrif PM that safety and efficacy of Giotrif have not been established in patients with uncommon EGFR mutations; and that data are more limited in patients with tumors with the T790M mutation.

Giotrif was associated with a number of safety concerns. Clinically significant (based on frequency, severity and seriousness) adverse events (AEs) reported in Giotrif-treated patients include diarrhea which can result in acute renal insufficiency and severe electrolyte imbalance, severe skin toxicities, interstitial lung disease (ILD) or ILD-like events including fatalities, and hepatotoxicity including uncommon events of hepatic failure with fatalities. With pre-defined, AE severity-based Giotrif dose adjustment in combination with proactive AE management, Giotrif-related AEs appeared to be largely manageable. Giotrif-related death was uncommon in the pivotal study.

Overall, the clinical benefit/risk profile of Giotrif seen in the pivotal and supportive studies is considered positive for EGFR TKI-naïve patients with metastatic adenocarcinoma of the lung harboring common EGFR activating mutation(s). However, data supporting use in other uncommon mutations are limited, particularly in de novo T790M mutations; this is adequately captured in the Giotrif Product Monograph (PM). Giotrif has an acceptable safety profile based on the non-clinical data and clinical studies. The identified safety issues can be managed through labelling. Appropriate warnings and precautions are in place in the Giotrif PM to address the identified safety concerns. A Risk Management Plan (RMP) for Giotrif was considered acceptable. The RMP describes known and potential safety issues, presents the monitoring scheme, and describes measures that will be put in place to minimize risks associated with authorized use of Giotrif.

Clinical evidence provided in the submission was not sufficient to support Giotrif use in patients who have failed prior treatment with an EGFR TKI or use of a 50 mg dose either as a starting dose or in dose escalation; these usages are not included in the approved PM (see Question 7: What was the scientific rationale for Health Canada's decision? for more details).

This New Drug Submission complies with the requirements of sections C.08.002 and C.08.005.1 and therefore Health Canada has granted the Notice of Compliance pursuant to section C.08.004 of the Food and Drug Regulations. For more information, refer to the Clinical, Non-Clinical, and Quality (Chemistry and Manufacturing) Basis for Decision sections.

3 What steps led to the approval of Giotrif?

On November 8, 2012, the Giotrif submission received a Screening Deficiency Notice due to a lack of case report forms for all studies as per section C.08.005.1. The sponsor addressed this deficiency, as well as other screening clarifax issues; the submission was subsequently accepted for review as a New Drug Submission on January 7, 2013.

Submission Milestones: Giotrif

Submission MilestoneDate
Pre-submission meeting:2012-07-19
Submission filed:2012-09-19
Screening
Screening Deficiency Notice issued:2012-11-08
Response filed:2012-11-23
Screening Acceptance Letter issued:2012-01-07
Review
On-Site Evaluation:2013-06-17 - 2013-06-20
Biopharmaceutics Evaluation complete:2013-07-26
Quality Evaluation complete:2013-09-30
Clinical Evaluation complete:2013-11-03
Labelling Review complete:2013-11-03
Notice of Compliance issued by Director General2013-11-01

The Canadian regulatory decision on the non-clinical and clinical review of Giotrif was based on a critical assessment of the Canadian data package. The United States Food and Drug Administration's (US FDA) review reports as well as the review reports of the European Medicines Agency (EMA) were used as an added reference in addition to the EMA's 120 day questions and sponsor's response.

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

4 What follow-up measures will the company take?

Requirements for post-market commitments are outlined in the Food and Drugs Act and Regulations.

6 What other information is available about drugs?

Up to date information on drug products can be found at the following links:

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

Clinical Pharmacology

Metastatic adenocarcinoma of the lung is a life-threatening, incurable disease with poor long-term survival.

Afatinib (Brand name: Giotrif) is an, irreversible inhibitor of, human epidermal growth factor receptor (ErbB1), HER 2 (ErbB2), and HER 4.

The clinical pharmacology program included human pharmacodynamics and pharmacokinetic studies.

Giotrif exhibited non-linear pharmacokinetics; more than proportional increases in exposure were observed with increasing dose in the clinical dose range. Afatinib was excreted primarily unchanged via the biliary/fecal route, consistent with in vitro studies that demonstrated an absence of interaction with CYP 450 enzymes.

Patients with moderate renal impairment may have increased toxicities due to higher systemic exposure to afatinib at the recommended dose of 40 mg once daily. Conversely, administration of afatinib with food significantly decreased exposure. The effects of severe renal and hepatic impairment on the PK parameters of afatinib have not been studied. This data deficiency is captured in the PM.

Together, the clinical and pharmacological data support the use of Giotrif for EGFR TKI-naïve patients with metastatic adenocarcinoma of the lung harboring activating EGFR mutations. Definition of special populations for which exposure is significantly altered or in whom exclusion is appropriate have been considered and adequately addressed in the Giotrif Product Monograph.

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

Clinical Efficacy

The clinical efficacy of Giotrif was primarily evaluated in four key studies: LUX-Lung 3 (pivotal), LUX-Lung 2, LUX-Lung 1, and LUX-Lung 5.

The LUX-Lung 3 and LUX-Lung 2 studies enrolled patients with EGFR mutation-positive adenocarcinoma who were not previously treated with an EGFR TKI (i.e., EGFR TKI-naïve patients). In contrast, LUX-Lung 1 and LUX-Lung 5 enrolled patients who had received previous EGFR TKI (i.e., EGFR TKI-treated patients).

The pivotal LUX-Lung 3 study was a global, randomized, multicenter, open-label Phase III study comparing efficacy and safety of Giotrif monotherapy with standard cisplatin/pemetrexed doublet chemotherapy in patients with locally advanced (Stage IIIB) or metastatic (Stage IV) adenocarcinoma of the lung harboring activating EGFR mutations. A total of 345 patients were randomized (2:1) to receive once daily oral Giotrif 40 mg until disease progression or intolerance, or standard cisplatin/pemetrexed chemotherapy up to 6 cycles. The study was conducted in the first-line setting, i.e., eligible patients could have not received prior systemic chemotherapy for advanced disease or prior EGFR TKI for any disease stage. The primary study endpoint was progression-free survival (PFS) and key secondary endpoints included objective response rate (ORR) and overall survival (OS).

Among the patients randomised, 65% were female, the median age was 61 years, 26% were Caucasian and 72% were Asian, the baseline ECOG performance status was 0 in 39% and 1 in 61%,. The majority of the patients (89%) had the two common EGFR mutations (del19 and L858R) with 11% having other less common mutations. Patients with active brain system metastases (i.e., stable <4 weeks and/or symptomatic and/or requiring treatment with anticonvulsants or steroids and/or leptomeningeal disease) were excluded from the study.

Results from the LUX-Lung 3 study demonstrated a statistically significant improvement in PFS (the primary efficacy endpoint) in the Giotrif treatment group compared to the control chemotherapy treatment group. The median PFS was 11.1 months for Giotrif-treated patients vs. 6.9 months for chemotherapy-treated patients. Objective response rate was higher in the Giotrif group (56.1%) compared to the chemotherapy group (22.6%). This magnitude of PFS benefit is considered clinically meaningful.

Based on an updated OS analysis when 175 patients had died, median OS was 28.1 months and 28.2 months in the Giotrif and chemotherapy groups, respectively. The OS results neither demonstrated a significant OS benefit nor indicated an apparent detrimental effect on OS by Giotrif therapy vs. control chemotherapy. Due to the study design, OS results may be limited by a lack of statistical power and confounded by uncontrolled use of post-progression anti-neoplastic therapies, including other EGFR TKIs and chemotherapy agents. The lack of OS benefit was captured prominently in the Giotrif Product Monograph to inform prescribers of this limitation of the efficacy dataset supporting the authorized indication and clinical use.

A total of 4 patients met the pre-defined criteria (displaying minimum treatment-related toxicities after first treatment cycle) for a dose-escalation of Giotrif from 40 mg to 50 mg. The patient number was considered insufficient to permit meaning efficacy assessment for the dose escalation scheme. Taking into consideration the potential of increased toxicities with increasing Giotrif dosage, such a dose escalation scheme is not included in the Giotrif PM.

In a pre-specified subgroup analysis based on EGFR mutation subtypes (common mutations vs. uncommon mutations), PFS and OS were in favor of the chemotherapy treatment among patients whose tumor harbored uncommon EGFR mutations [Hazard Ratio (HR) 1.9 and 3 for PFS and OS, respectively]. This subgroup was small [Number of patients (N) = 37 patients] and genetically heterogeneous (10 different molecular subtypes with unequal distribution between the two study groups), which limits the value and interpretation of pooled analyses based on the entire subgroup. Therefore, the clinical efficacy of Giotrif was assessed by each mutation subtype in this subgroup. Among 26 patients treated with Giotrif, 4 achieved a partial response (all of 6 months or greater duration), although the antitumor activity of Giotrif appeared to be more limited in those with the T790M mutation, with only one response in a total of 11 patients. This level of tumor response in those with uncommon mutations may be clinically meaningful, taking into consideration limited therapeutic options for these patients and the rarity of the disease (uncommon EGFR mutations represent approximately 10% of all EGFR mutations in NSCLC). Therefore, Health Canada did not recommend that this patient subpopulation be explicitly excluded from the authorized indication. In the Giotrif PM, it is prominently captured that safety and efficacy of Giotrif have not been established in patients with uncommon EGFR mutations; and that data are more limited in patients with tumors with the T90M mutation. Mutation-specific objective tumor responses are also listed.

The LUX-Lung 2 study was an open label, single arm, Phase II study which investigated the efficacy and safety of Giotrif as a monotherapy in EGFR TKI-naïve patients with metastatic adenocarcinoma of the lung harboring EGFR-activating mutations.

A total of 129 patients entered the LUX-Lung 2 study and were enrolled into four key cohorts defined by the starting dose of Giotrif (40 mg or 50 mg daily) and line of therapy [first-line (N = 61) or second-line setting (N = 68) (i.e., after failure of one prior chemotherapy regimen)] to receive Giotrif starting dose of 40 mg or 50 mg daily.

The primary efficacy endpoint was overall response rate (ORR). Results of this study demonstrated anti-tumor activity of Giotrif in patients with EGFR mutations in the first-line or second-line setting. Study data indicated that the 40 mg Giotrif starting dose is associated with comparable best overall response and better tolerability compared with the 50 mg Gitorif starting dose for protracted treatment with Giortif in a population of EGFR TKI-naïve patients. Efficacy results of the LUX-Lung 2 study supported the tumor response data in the pivotal study.

LUX-Lung 1 was an international, multicentre, randomized, double-blind Phase IIb/III trial comparing the efficacy and safety of Giotrif (50 mg daily) plus best supportive care (BSC) vs. placebo plus BSC in patients with metastatic adenocarcinoma of the lung failing previous reversible EGFR TKIs, [for example (e.g.), erlotinib, gefitinib]. A total of 585 patients were randomized (2:1) to receive Giotrif or placebo treatment. Confirmation of EGFR mutation status was not required before study treatment. All patients had undergone at least 12 weeks of prior therapy with a reversible EGFR TKI and a prior systemic chemotherapy.

This study showed similar survival between treatment groups, with a median OS of 12.0 months for placebo and 10.8 months for Giotrif  (HR 1.08; 95% CI 0.86, 1.35) and did not meet its pre-specified primary endpoint. Despite this, secondary endpoints were analyzed; based on the independent assessment, median PFS was 3.3 months in the Giotrif arm vs. 1.1 months in the placebo arm (HR 0.38, p<0.0001). While this HR was considered promising, the absolute magnitude 2-3 months improvement in median PFS was considered modest. The great majority of best overall tumor response was stable disease, in contrast to the high ORR in EGFR TKI-naïve patients in the pivotal LUX Lung 3 study.

LUX-Lung 5 study was a Phase III randomised trial of Giotrif plus weekly paclitaxel vs. the investigator's choice of chemotherapy following Giotrif monotherapy in patients with metastatic adenocarcinoma of the lung failing previous erlotinib or gefitinib treatment. Only the interim report of the LUX-Lung 5 study was submitted including data from Part A of the study in which all patients received Giotrif monotherapy (starting dose 50 mg). Enrolled patients were not required to have analysis of tumour samples for EGFR mutation status.

The primary endpoint was PFS based on investigator assessment. A total of 1,154 patients were entered into the study and received Giotrif treatment. Results for mutation status were available for only 84 patients, 49 patients (58.3%) were mutation positive and 35 patients (41.7%) were mutation negative. At the interim analysis, 872 PFS event had occurred (76%). The median PFS was 3.3. Median PFS was longer in patients whose tumors harbored EGFR mutations (4.2 months) than in mutation negative patients (2.6 months).

Overall, the clinical benefit of Giotrif among EGFR TKI-naïve patients is considered significant, based on the PFS and ORR results in the pivotal LUX-Lung 3 and supportive LUX-Lung 2 study. The clinical benefit of Giotrif among patients who failed a prior EGFR TKI (i.e., EGFR TKI-treated patients) is unclear. This is because the LUX-Lung 1 did not meet its primary efficacy endpoint, i.e., it did not demonstrate a significant improvement in OS in the Giotrif group compared with the placebo group. The level of tumor control (based on PFS and ORR) in the LUX-Lung 1 study in the context of a negative primary efficacy analysis, is not considered substantial evidence of clinical benefit. Giotrif was not authorized for use in EGFR TKI-treated patients with metastatic adenocarcinoma of the lung harboring activating EGFR TKIs. This is reflected in the indication in the approved PM.

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

Clinical Safety

The overall safety profile of Giotrif is based on results of four key studies [LUX-Lung 3 (pivotal), LUX-Lung 2, LUX-Lung 1, and LUX-Lung 5], as well as pooled safety results from different cohorts, e.g., those received 40 mg Giotrif monotherapy, 50 mg monotherapy, 40 or 50 mg monotherapy, and Giotrif at any dose as either monotherapy or in combination.

The adverse event (AE) analyses in the pivotal study were primarily based on treatment-emergent AEs (i.e., events with an onset between the start of study drug and 28 days after stopping it).

Virtually all patients in the pivotal LUX-Lung 3 study (described in Clinical Efficacy) experienced at least one AE during the study. Giotrif demonstrated a different AE profile compared to control chemotherapy: the most common AEs of any Common Terminology Criteria Adverse Events (CTCAE) Grade in the Giotrif arm were diarrhea (96.1% of patients), rash/acne (90.0%)1, stomatitis (73.4%)1, and nail effect (61.6%)1. These AEs were also the most frequent CTCAE Grade 3 events (rash1 16.2%, diarrhoea 14.8%, nail effect1 11.8% and stomatitis1 8.3%). In the chemotherapy treatment group, the most frequently reported AEs were nausea (67.6% of patients), decreased appetite (55.0%), and fatigue1 (49.5%). The most frequent AEs of CTCAE Grade 3 in this control group were neutropenia (16.2% of patients), fatigue (12.6%), and leukopenia (8.1%).

Serious AEs were reported in 28.8% patients in the Giotrif treatment group, compared with 22.5% in the chemotherapy treatment group. The most frequent serious AEs in Giotrif-treated patients were: diarrhea; vomiting; dyspnea; fatigue; dehydration; pneumonia; and stomatitis. Fatal adverse events related to Giotrif included one event each of dyspnea, acute respiratory distress syndrome (ARDS) [interstitial lung disease (ILD)-like], sepsis, and death (not otherwise specified).

A pre-defined, AE severity (CTCAE Grade) -based Giotrif dose adjustment scheme was implemented in the pivotal study. Dose reductions due to AEs occurred in 57.2% of Giotrif-treated patients versus 16.2% in chemotherapy-treated patients. The very common AEs (>10%) leading to dose reduction in patients treated with Giotrif included diarrhea, rash/acne, paronychia, and stomatitis. Permanent study treatment discontinuation occurred in 14.0% of patients in the Giotrif treatment group and 15.3% in the chemotherapy treatment group. The common AEs which led to discontinuation in the Giotrif were: diarrhea, dyspnea; and ILD. This Giotrif dose adjustment scheme, in addition to other risk mitigation strategies (e.g., proactive treatment of diarrhea) appeared to be effective in managing severe and/serious AEs, as dose reduction of Giotrif was associated with a lower frequency of common adverse events, e.g., after first-dose reduction, the frequency for diarrhea decreased from 96% to 52%. The median treatment duration at reduced dose levels (30 mg - 140 days; 20 mg - 277 days) were longer than the starting 40 mg dose (85 days), indicating that many patients tolerated Giotrif at reduced doses and likely benefited from the dose reduction by staying on the treatment at lower doses longer.

Afatinib inhibits HER2 and left ventricular dysfunction has been associated with HER2 inhibition. In the pivotal trial routine left ventricular ejection fraction (LVEF) monitoring was not compulsory in the chemotherapy treated patients and therefore only approximately 14% had an on treatment LVEF assessment compared to 91% of Giotrif-treated patients. With a mean total treatment time of 11 months, 31% of Giotrif- treated patients had a decrease of LVEF of >10% from baseline; and with mean total treatment time 2.8 months 13% of pemetrexed/cisplatin-treated patients had such a decrease. Giotrif has not been studied in patients with an abnormal LVEF or those with a significant cardiac history. In patients with cardiac risk factors and those with conditions that can affect left ventricular function, cardiac monitoring, including an assessment of LVEF at baseline and during Giotrif treatment, should be considered.

The safety profile of Giotrif at a starting dose of 40 mg daily in the supportive LUX-Lung 2 study (described in Clinical Efficacy) was generally consistent with that in the pivotal study. However, severe and serious AEs appeared to be more frequent in the 50 mg cohort compared with the 40 mg cohort. Adverse events of CTCAE Grade 3 were 43.3% in the 40 mg starting dose group and 58.6% in the 50 mg starting dose group. Adverse events of CTCAE Grade 5 were reported in 9.3% of patients overall, including 6.7% of patients in the 40 mg starting dose group and 10.1% of patients in the 50 mg. These observations raised the safety concern for the dose escalation scheme in the pivotal study (described in Clinical Efficacy). Considering limited efficacy evidence supporting Giotrif dose escalation from 40 mg to 50 mg and this safety concern, such a dosage scheme is not included in the Giotrif PM.

Compared to the placebo group, AEs and serious AEs in the LUX-Lung 1 study (described in Clinical Efficacy) were substantially increased in the Giotrif treatment group. Adverse events considered related to the study medication were reported by 95.4% of patients in the Giotrif group and 37.9% of patients in the placebo group. Adverse events of CTCAE grade 3 or higher were reported for 56.7% of patients in the Giotrif group and 24.6% of patients in the placebo group. Study drug was permanently discontinued due to AEs in 17.9% of patients in the Giotrif group and 6.2% of patients in the placebo group. Drug-related serious AEs were reported for 39 patients (10.0%) in the Giotrif group and one patient (0.5%) in the placebo group. More patients (78.9%) in the placebo arm received non-study anti-cancer therapy following their disease progression, compared with 67.6% in the Giotrif group.

Almost all patients (99.0%) experienced at least one treatment-emergent AE in the LUX-Lung 5 study (described in Clinical Efficacy). The most frequent AE was diarrhea (85.2%), followed by rash (69.5%) and stomatitis (50.8%). The highest CTCAE Grade of AEs was grade 3 in 39.9% of patients; grade 4 in 5.2% of patients; and grade 5 in 15.9%. Giotrif-related AEs leading to discontinuation occurred in 11.7% of patients, with diarrhea and rash as the most frequent events. Serious AEs occurred in 38.9% of patients, including 10.4% with related serious AEs. In 11 patients (1.0%), AEs leading to death were considered drug-related.

More adverse events ≥ CTCAE Grade 3 were reported for patients ≥ 65 years than patients < 65 years in clinical trials.

Overall, Giotrif monotherapy was associated with significant risks of adverse events, some of which were serious and fatal. However, with pre-defined, AE severity-based Giotrif dose adjustment in combination with proactive AE management and proper patient selection (especially for diarrhea), Giotrif-related AEs appeared to be largely manageable. Giotrif-related deaths were uncommon in the pivotal study.

Successful management of AEs is essential for the success of Giotrif treatment (maintaining an acceptable benefit/risk profile for Giotrif). The following are highlights of the measures undertaken by Health Canada to improve the risk communication and mitigation with regard to Giotrif-related toxicities:

  1. The proposed PM provided by the sponsor was revised per Health Canada's recommendations to improve communication of significant risks identified in clinical trials, including creating a Serious Warnings and Precautions Box for diarrhea, severe skin toxicities, interstitial lung disease and hepatotoxicity.
  2. The approved PM contains an explicit warning against Giotrif use in patient subgroups that may be at particular risk of severe and/or serious AEs related to Giotrif, including those with recent or ongoing gastrointestinal disorders, those with severe renal and/or hepatic impairment, and those who develop cardiac signs/symptoms during Giotrif treatment.
  3. Risk management recommendations, based on the protocol of pivotal study, were enhanced in the PM, especially for diarrhea. Giotrif-related diarrhea occurred in virtually all patients and was the leading Giotrif-related severe AE, serious AE, and AE resulting in dose reduction. Close monitoring and proactive management of diarrhea is essential for successful Giotrif treatment. Accordingly, diarrhea was managed in clinical trials by proactive anti-diarrheal treatment and by following a severity-based dose reduction scheme. In the clinical trials, patients played an important role in managing diarrhea by recognizing signs of diarrhea, implementing anti-diarrheal treatment (e.g., medications, hydration, adjusting anti-diarrheal dosage according to the protocol), and reporting signs and symptoms to investigators.

    Per Health Canada's recommendation, the proposed PM was revised to more effectively communicate the risk and risk mitigation recommendations for treatment-related diarrhea. The key changes included:

    1. addition of the statement "close monitoring and proactive management of diarrhea is essential for successful Giotrif treatment" in the Indications and Clinical Use section;
    2. addition of a detailed description of the diarrhea management protocol employed in the pivotal study in the Warnings and Precautions section;
    3. addition of the recommendation that "prescribers should ensure that patients are well informed of the risk of diarrhea and are able to proactively manage the side effect" and "patients should be provided with contact information of a physician experienced in cancer treatment and seek advice on diarrhea management" and
    4. an revisions to the table that outlines Giotrif dose adjustment recommendations to be consistent with those in the pivotal study.
  4. A Risk Management Plan (RMP) for Giotrif was submitted and reviewed by Health Canada. The RMP describes known and potential safety issues, presents the monitoring scheme, and describes measures that will be put in place to minimize risks associated with Giotrif under the authorized use. A sponsor initiated drug-distribution scheme will be assessed by Health Canada as an Annex under the RMP. Health Canada considers that such a scheme may offer additional benefit for managing diarrhea and other Giotirf-related risk.

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

7.2 Non-Clinical Basis for Decision

Afatinib covalently binds to the kinase domains of EGFR (ErbB1), HER2 (ErbB2), and HER4 (ErbB4) and irreversibly inhibits tyrosine kinase autophosphorylation, resulting in downregulation of ErbB signaling. Afatinib reduced tumour size of human xenografts in mice expressing either EGFR or HER-2. Afatinib is not a substrate for, or an inhibitor of, the cytochrome P450 family of enzymes. The maximum decrease in left ventricular contractility was 20% in pigs treated with afatinib at a single dose concentration of 20 mg/kg.

A non-clinical formulation of afatinib demonstrated moderate oral bioavailability of approximately 45% in rats and 11% in mini-pigs. According to quantitative whole body autoradiography studies in rats, afatinib distributed into most tissues. Trough levels of afatinib increased in blood, plasma and tissues over time following repeated dosing. The accumulation factors were in the range of 6.7 (brain) to 23.8 (skin). Exposure to the central nervous system was low upon single oral dosing, yet accumulation in the brain was noted upon repeat dosing of afatinib.

In vitro studies demonstrate that afatinib is not a significant cytochrome 450 (CYP) substrate, nor an inhibitor or inducer of CYP 450 enzymes. Therefore, drug-drug interactions (DDIs) of Giotrif with medications that are substrates for and/or modulate CYP 450 enzyme activity are considered unlikely to occur.

Afatinib is both a substrate and an inhibitor of both P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP). The potential for drug interactions, particularly DDIs, is significant when afatinib is administered with other medications that are P-gp or BCRP substrates or those that alter P-gp or BCRP function. These interactions could lead to increased toxicities or altered clinical effectiveness of afatinib or concomitant medications.

Afatinib binds covalently to plasma proteins and hemoglobin. This characteristic of the drug may constitute a risk factor for idiosyncratic, immune-based drug reactions.

In oral repeated-dose studies, the main target organs for toxicity were skin, gastrointestinal tract, and kidneys. Epithelial atrophy of the upper respiratory tract, prostate, seminal vesicles, uterus, vagina and the cornea of the eyes was observed. Total systemic exposure (AUC) at the no-observed-adverse-effect level (NOAEL) in animals was consistently lower than that observed in patients.

Long-term animal studies have not been conducted to evaluate the carcinogenic potential of afatinib. A marginal response to afatinib was observed in a single tester strain of a bacterial (Ames) mutagenicity assay; however, no mutagenic or genotoxic potential could be identified in other in vitro or in in vivo studies.

In embryo-fetal development studies in animals, notable changes included skeletal alterations consisting of incomplete ossifications/unossified elements and abortions, reduced fetal weights as well as mainly visceral and dermal variations. The respective total systemic exposures (AUC) in these studies were below expected human exposure in both rats (0.4 times) and rabbits (0.22 times). Women of child-bearing should be advised to avoid becoming pregnant while receiving treatment with Giotrif.

Radiolabelled afatinib was excreted into milk of the dams following a single oral administration during lactation. The average concentrations in milk at time points 1 hour and 6 hours post dose were approximately 80 and 150-fold above the respective concentration in plasma. Breastfeeding during Giotrif treatment and up to 2 weeks after the last dose is not recommended.

Giotrif may be phototoxic as studies have shown that afatinib absorbs light in the range of natural sunlight and demonstrated phototoxic potential in the in vitro 3T3 NRU assay. Furthermore, studies with dosing of radiolableled afatinib in rats demonstrated distribution of radioactivity to sun-exposed tissues (retina and skin). Patients should be advised to avoid sun exposure. This is captured in the Giotrif Product Monograph.

The nonclinical pharmacology and toxicology programs conducted with afatinib support the recommended indication of Giotrif for EGFR TKI-naïve patients with metastatic adenocarcinoma of the lung with activating EGFR mutation(s). The results of the non-clinical studies as well as the potential risks to humans have been included in the Giotrif Product Monograph. In view of the intended use of Giotrif, there are no non-clinical issues which preclude authorization of the product.

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

7.3 Quality Basis for Decision

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

The proposed limits of drug-related impurities are considered adequately qualified from toxicological studies.

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

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

Lactose monohydrate is the only material of animal origin used in the manufacture of Giotrif film-coated tablets. The manufacturer of lactose monohydrate has provided a written assurance that the lactose is produced from milk sourced from healthy animals under the same conditions as milk collected for human consumption.

1 Includes similar pooled Medical Dictionary for Regulatory Activities (MedDRA) preferred terms.