Summary Basis of Decision for Tepmetko
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:
Summary Basis of Decision (SBD) documents provide information related to the original authorization of a product. The SBD for Tepmetko is located below.
Recent Activity for Tepmetko
The SBDs written for eligible drugs (as outlined in Frequently Asked Questions: Summary Basis of Decision [SBD] Project: Phase II) approved after September 1, 2012 will be updated to include post-authorization information. This information will be compiled in a Post-Authorization Activity Table (PAAT). The PAAT will include brief summaries of activities such as submissions for new uses of the product, and whether Health Canada's decisions were negative or positive. The PAATs will be updated regularly with post-authorization activity throughout the product life cycle.
The following table describes post-authorization activity for Tepmetko, a product which contains the medicinal ingredient tepotinib (supplied as tepotinib hydrochloride). For more information on the type of information found in PAATs, please refer to the Frequently Asked Questions: Summary Basis of Decision (SBD) Project: Phase II and to the List of abbreviations found in Post-Authorization Activity Tables (PAATs).
For additional information about the drug submission process, refer to the Guidance Document: The Management of Drug Submissions and Applications.
Updated: 2024-09-05
Drug Identification Number (DIN):
DIN 02516322 - 225 mg tepotinib, tablet, oral administration
Post-Authorization Activity Table (PAAT)
Activity/Submission Type, Control Number |
Date Submitted |
Decision and Date |
Summary of Activities |
SNDS-C # 274967 |
2023-05-08 |
Issued NOC 2024-03-04 |
Submission filed as a Level I – Supplement in response to commitments made as per the provisions of the Guidance Document: Notice of Compliance with Conditions (NOC/c). The submission provided the final report from the VISION clinical study to further characterize the clinical benefit of Tepmetko for the treatment of patients with non-small cell lung cancer harbouring MET exon 14 skipping alterations who are treatment-naïve and who have previously received systemic therapy. The data were reviewed and considered acceptable. The PM was updated based on the most recent clinical study data. As a result of the submission, the conditions were removed from the NOC that had been issued 2021-05-27 (NDS # 242300). |
PBRER-C # 275795 |
2023-05-29 |
Filed 2024-01-24 |
Submission filed in response to commitments made as per the provisions of the Guidance Document: Notice of Compliance with Conditions (NOC/c). PBRER-C for the period 2022-03-25 to 2023-03-24. The information was reviewed and found acceptable. No further action was required. |
SNDS-C # 270905 |
2022-12-21 |
Issued NOC under NOC/c Guidance 2023-08-25 |
Submission filed as a Level I – Supplement in response to commitments made as per the provisions of the Guidance Document: Notice of Compliance with Conditions (NOC/c) (NDS # 242300). The submission provided clinical reports from two clinical drug-drug interaction studies in healthy volunteers. The PM was updated to reflect the results of these studies and an NOC/c was issued. |
PBRER-C # 264597 |
2022-05-26 |
Filed 2022-10-06 |
Submission filed in response to commitments made as per the provisions of the Guidance Document: Notice of Compliance with Conditions (NOC/c). PBRER-C for the period 2021-03-25 to 2022-03-24. The information was reviewed and found acceptable. No further action was required. |
Drug product (DIN 02516322) market notification |
Not applicable |
Date of first sale: 2021-10-07 |
The manufacturer notified Health Canada of the date of first sale pursuant to C.01.014.3 of the Food and Drug Regulations. |
NDS # 242300 |
2020-07-31 |
Issued NOC under NOC/c Guidance 2021-05-27 |
NOC issued under the NOC/c Guidance for New Drug Submission. |
Summary Basis of Decision (SBD) for Tepmetko
Date SBD issued: 2021-10-21
The following information relates to the New Drug Submission for Tepmetko.
Tepotinib (supplied as tepotinib hydrochloride)
Drug Identification Number (DIN):
- DIN 02516322 - 225 mg tepotinib, tablet, oral administration
EMD Serono, a Division of EMD Inc. Canada
New Drug Submission Control Number: 242300
On May 27, 2021, Health Canada issued a Notice of Compliance under the Notice of Compliance with Conditions (NOC/c) Guidance to EMD Serono, a Division of EMD Inc. Canada, for the drug product Tepmetko. The product was authorized under the NOC/c Guidance on the basis of the promising nature of the clinical evidence, and the need for further follow-up to confirm the clinical benefit. Patients should be advised of the fact that the market authorization was issued with conditions.
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-harm-uncertainty profile of Tepmetko is favourable for the treatment of adult patients with locally advanced unresectable or metastatic non-small cell lung cancer harbouring mesenchymal-epithelial transition tyrosine kinase receptor exon 14 skipping alterations.
1 What was approved?
Tepmetko, a protein kinase inhibitor, was authorized for the treatment of adult patients with locally advanced unresectable or metastatic non-small cell lung cancer (NSCLC) harbouring mesenchymal-epithelial transition (MET) tyrosine kinase receptor exon 14 (METex14) skipping alterations (METex14-altered NSCLC).
Documentation of METex14 skipping alteration status based on a validated METex14 assay is required prior to treatment with Tepmetko.
Tepmetko is not authorized for use in pediatric patients (less than 18 years of age) as no clinical safety or efficacy data are available for this population.
No clinically important differences in safety or efficacy were observed between patients aged 65 years or older and younger patients in the pivotal VISION study. Of the 255 patients with advanced NSCLC with METex14 skipping alterations in the VISION study who received 450 mg Tepmetko once daily, 79% were 65 years or older, 43% were 75 years or older, and 8% were 85 years or older.
Tepmetko (225 mg tepotinib, supplied as 250 mg tepotinib hydrochloride) is presented as a tablet. In addition to the medicinal ingredient, each tablet contains colloidal silicon dioxide, crospovidone, hypromellose, lactose monohydrate, magnesium stearate, mannitol, microcrystalline cellulose, polyethylene glycol, red iron oxides (E172), titanium dioxide, and triacetin.
Tepmetko is contraindicated in patients who are hypersensitive to this drug or to any ingredient in the formulation, including any non-medicinal ingredient, or component of the container.
Tepmetko was approved for use under the conditions stated in its Product Monograph, taking into consideration the potential risks associated with the administration of this drug product. A Serious Warnings and Precautions box describing the risk of hepatotoxicity, interstitial lung disease/pneumonitis, and embryo-fetal toxicity has been included in the Tepmetko Product Monograph.
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 Tepmetko Product Monograph, approved by Health Canada and available through the Drug Product Database.
2 Why was Tepmetko approved?
Health Canada considers that the benefit-harm-uncertainty profile of Tepmetko is favourable for the treatment of adult patients with locally advanced unresectable or metastatic non-small cell lung cancer (NSCLC) harbouring mesenchymal-epithelial transition (MET) tyrosine kinase receptor exon 14 (METex14) skipping alterations (METex14-altered NSCLC). Tepmetko was authorized under the Notice of Compliance with Conditions (NOC/c) Guidance on the basis of the promising nature of the clinical evidence, and the need for further follow-up to confirm the clinical benefit.
In Canada, lung cancer was the most common cancer diagnosis in 2020, with most new diagnoses made at an advanced or metastatic stage. Lung cancer is responsible for 25% of cancer deaths among men and women. Approximately 85% of lung cancers are NSCLCs. Roughly 3% of NSCLCs harbour METex14 skipping alterations; the incidence is 3 to 4% in patients with adenocarcinoma histology and 1 to 2% in patients with other NSCLC histologies, with the exception of sarcomatoid carcinomas in which the incidence is higher (8% to 32%). At diagnosis, the median age of patients with METex14-altered NSCLC is over 65 years.
In Canada, there is no approved therapy that specifically targets advanced METex14-altered NSCLC. The current standard of care for patients with NSCLC in whom a therapy-predictive biomarker conferring sensitivity to targeted inhibition has been identified is targeted therapy, when possible. Some patients are enrolled into clinical trials for investigational treatments, while others receive chemotherapy with or without immunotherapies or bevacizumab (a vascular endothelial growth factor inhibitor), in accordance with guidelines for oncogene driver-negative disease. While the efficacy of chemotherapy, including platinum doublet chemotherapy, has not been established for METex14-altered NSCLC, limited data suggest unsatisfactory responses. Checkpoint inhibitors also appear to be less efficacious in the treatment of METex14-altered NSCLC. Currently available therapies are also associated with significant toxicity profiles and administrational burden.
The MET gene directs the production of the MET transmembrane tyrosine kinase receptor. Under normal physiological conditions, MET regulates invasive growth and morphogenesis in multiple embryonic tissues, and is involved in wound healing and the regeneration of damaged organs, in particular the liver. Exon 14 of the MET gene encodes the juxtamembrane region of the receptor that contains several negative regulatory sites of activity. Skipping of exon 14 results in the oncogenic activation of MET. Diverse alterations in multiple locations of the gene result in METex14 skipping.
Tepmetko (tepotinib, supplied as tepotinib hydrochloride) is a kinase inhibitor that targets MET, including variants with exon 14 skipping alterations. The results of the pivotal Phase II VISION study support the market authorization of Tepmetko for the treatment of adult patients with locally advanced unresectable (Stage IIIb) or metastatic (Stage IV) METex14-altered NSCLC. The VISION study was a single-arm, open-label, multicentre, non-randomized, multicohort study with Tepmetko 450 mg administered orally once daily until progression of disease or unacceptable toxicity occurred. The primary efficacy endpoint of the study was the confirmed objective response (complete response or partial response) according to the Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 guideline and based on a blinded independent review. In the efficacy analysis population (number of patients [n] = 146) the objective response rate (ORR) was 45.2% (95% confidence interval [CI]: 37.0, 53.6). The median duration of response (DOR), a secondary endpoint, was 11.1 months (95% CI: 8.4, 18.5). Overall, the benefit with Tepmetko was observed to be stable and independent of whether patients were treatment naive or previously treated. Based on the current understanding of oncogene-driven NSCLC, an extrapolation from the metastatic stage (98% of patients) to the locally advanced unresectable stage was considered reasonable.
The most common (≥20%) treatment-emergent adverse events (TEAEs) in the VISION safety analysis set (n = 255) were edema (69.0%), mainly peripheral edema (60.0%); fatigue (27.5%); nausea (26.7%); diarrhea (26.3%); increase in creatinine (25.9%); musculoskeletal pain (24.3%); hypoalbuminemia (23.9%); and dyspnea (20.4%). The primary serious risks were identified as hepatotoxicity and interstitial lung disease and pneumonitis. Based on its mechanism of action and findings from animal studies, Tepmetko can cause fetal harm when administered to a pregnant woman. In addition, there were observed increases in serum creatinine that were potentially caused by Tepmetko’s inhibition of renal tubular transporter proteins: organic cation transporter 2 and multidrug and toxin extrusion 2. Consequently, creatinine alone is not a reliable biomarker to assess renal function in patients treated with Tepmetko.
Concomitant use of Tepmetko with strong cytochrome P450 (CYP) 3A4 or permeability glycoprotein (P-gp) inducers and with dual strong CYP3A inhibitors and P-gp inhibitors should be avoided. Monitoring of the clinical effects of P-gp substrates with a narrow therapeutic index is recommended during co-administration with Tepmetko. A study which assessed the effect of food on the pharmacokinetics of tepotinib tablets supports the recommendation to take the daily dose of Tepmetko with food.
A Risk Management Plan (RMP) for Tepmetko was submitted to Health Canada by EMD Serono, a Division of EMD Inc. Canada. Upon review, the RMP was considered acceptable. The RMP is designed to describe known and potential safety issues, to present the monitoring scheme, and when needed, to describe measures that will be put in place to minimize risks associated with the product.
Appropriate warnings and precautions are in place in the approved Tepmetko Product Monograph to address the identified safety concerns, including a Serious Warnings and Precautions box describing the risk of hepatotoxicity, interstitial lung disease/pneumonitis, and embryo-fetal toxicity. In addition, treatment with Tepmetko should be initiated and supervised by a qualified physician experienced in the use of anticancer therapies.
The submitted inner and outer labels, package insert, and Patient Medication Information section of the Tepmetko Product Monograph meet the necessary regulatory labelling, plain language, and design element requirements. A review of the submitted brand name assessment, including testing for look-alike sound-alike attributes, was conducted and the proposed name Tepmetko was accepted.
Based on the benefit-harm-uncertainty profile of the product, Health Canada issued a Notice of Compliance with conditions (NOC/c) for Tepmetko under the NOC/c Guidance, pending the results of trials to verify its clinical benefit. EMD Serono, a Division of EMD Inc. Canada, has committed to provide the final report for the confirmatory VISION study to verify the clinical benefit of Tepmetko by providing a more precise estimation of the blinded independent central review-assessed ORR and DOR. EMD Serono, a Division of EMD Inc. Canada, has also committed to conduct two additional drug interaction studies.
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 Tepmetko?
The new drug submission (NDS) for Tepmetko was subject to advance consideration under the Notice of Compliance with Conditions (NOC/c) Guidance. The sponsor presented promising evidence of clinical effectiveness to demonstrate that Tepmetko provides effective treatment of advanced (Stage IIIb/IV) non-small cell lung cancer harbouring mesenchymal-epithelial transition tyrosine kinase receptor exon 14 skipping alterations, a serious, life-threatening disease for which no drug is presently marketed in Canada.
Subsequent review led to the decision to issue the sponsor market authorization under the NOC/c Guidance, in recognition of the promising but unconfirmed evidence of clinical effectiveness in the submission. In keeping with the provisions of the NOC/c Guidance, the sponsor agreed to provide additional information to confirm the clinical benefit.
The NDS for Tepmetko was reviewed under Project Orbis. Project Orbis provides a framework for concurrent submission and review of oncology products as well as information sharing among regulators from multiple jurisdictions. Health Canada, the United States Food and Drug Administration (FDA), the Australian Therapeutic Goods Administration, and Switzerland’s Swissmedic participated in the review of the NDS for Tepmetko. Although the review of the submission was collaborative, each jurisdiction made their regulatory decision independently. The Canadian regulatory decision on the review of Tepmetko was based on a critical assessment of the data package submitted to Health Canada. The foreign reviews completed by the FDA were used as an added reference.
Submission Milestones: Tepmetko
Submission Milestone | Date |
---|---|
Pre-submission meeting | 2020-04-22 |
Acceptance of Advance Consideration under the Notice of Compliance with Conditions (NOC/c) Guidance | 2020-06-30 |
Submission filed | 2020-07-31 |
Screening | |
Screening Deficiency Notice issued | 2020-08-25 |
Response filed | 2020-09-30 |
Screening Acceptance Letter issued | 2020-10-05 |
Review | |
Biopharmaceutics Evaluation complete | 2020-12-14 |
Quality Evaluation complete | 2021-01-28 |
Review of Risk Management Plan complete | 2021-01-31 |
Non-Clinical Evaluation complete | 2021-04-16 |
Labelling Review complete | 2021-04-21 |
Clinical/Medical Evaluation complete | 2021-04-23 |
Notice of Compliance with Conditions Qualifying Notice (NOC/c-QN) issued | 2021-04-23 |
Review of Response to NOC/c-QN: | |
Response filed (Letter of Undertaking) | 2021-04-30 |
Clinical/Medical Evaluation complete | 2021-05-26 |
Notice of Compliance (NOC) issued by Director General, Therapeutic Products Directorate under the Notice of Compliance with Conditions (NOC/c) Guidance | 2021-05-27 |
For additional information about the drug submission process, refer to the Management of Drug Submissions and Applications Guidance.
4 What follow-up measures will the company take?
In addition to requirements outlined in the Food and Drugs Act and Regulations, and in keeping with the provisions outlined in the Notice of Compliance with Conditions (NOC/c) Guidance, the sponsor has agreed to submit, as a Supplemental New Drug Submission-Confirmatory (S/NDS-c), a final report including datasets from the pivotal VISION clinical study to confirm and further characterize the clinical benefit of Tepmetko for the treatment of patients with non-small cell lung cancer harbouring mesenchymal-epithelial transition exon 14 skipping alterations, who are treatment naive and who have previously received systemic therapy, by providing a more precise estimation of the blinded independent central review-assessed overall response rate and duration of response. It is anticipated that the S/NDS-c will be filed around April 2023.
As part of the marketing authorization for Tepmetko, the sponsor agreed to additional conditions, which include, but are not limited to, conducting two additional studies. One study is a drug interaction study to evaluate the single dose pharmacokinetics of Tepmetko, when administered concomitantly with a strong cytochrome P450 (CYP) 3A4 inducer and moderate CYP2C8 inducer, to assess the magnitude of decreased drug exposure and to determine appropriate dosing recommendations. The S/NDS-c for this study is estimated to be filed around July 2022. The second study is a drug interaction study evaluating the effect of itraconazole on the single dose pharmacokinetics of Tepmetko to assess the magnitude of increased drug exposure and to determine appropriate dosing recommendations when Tepmetko is administered concomitantly with a strong CYP3A4 and permeability glycoprotein inhibitor. The S/NDS-c for this study is estimated to be filed around September 2022.
6 What other information is available about drugs?
Up-to-date information on drug products can be found at the following links:
- See MedEffect Canada for the latest advisories, warnings and recalls for marketed products.
- See the Notice of Compliance (NOC) Database for a listing of the authorization dates for all drugs that have been issued an NOC since 1994.
- See the Drug Product Database (DPD) for the most recent Product Monograph. The DPD contains product-specific information on drugs that have been approved for use in Canada.
- See the Notice of Compliance with Conditions (NOC/c)-related documents for the latest fact sheets and notices for products which were issued an NOC under the Notice of Compliance with Conditions (NOC/c) Guidance Document, if applicable. Clicking on a product name links to (as applicable) the Fact Sheet, Qualifying Notice, and Dear Health Care Professional Letter.
- See the Patent Register for patents associated with medicinal ingredients, if applicable.
- See the Register of Innovative Drugs for a list of drugs that are eligible for data protection under C.08.004.1 of the Food and Drug Regulations, if applicable.
7 What was the scientific rationale for Health Canada's decision?
7.1 Clinical Basis for Decision
Clinical Pharmacology
The mesenchymal-epithelial transition (MET) tyrosine kinase receptor is a cell surface receptor. Mesenchymal-epithelial transition and its ligand, hepatocyte growth factor (HGF), are involved in carcinogenesis and tumour progression. Oncogenic activation of MET has been shown to promote cancer cell proliferation, survival, migration, invasion, and tumour angiogenesis, as well as to mediate resistance to cancer therapies.
Tepotinib is a kinase inhibitor that targets MET, including variants with exon 14 skipping alterations. Tepotinib inhibits HGF-dependent and HGF-independent MET phosphorylation and MET-dependent downstream signaling pathways. Tepotinib also inhibits the melatonin 2 and imidazoline 1 receptors at clinically achievable concentrations.
The proposed clinical dose of tepotinib is 450 mg (500 mg as tepotinib hydrochloride) taken once daily in patients with advanced non-small cell lung cancer (NSCLC) harbouring MET tyrosine kinase receptor exon 14 (METex14) skipping alterations (METex14-altered NSCLC).
Pharmacodynamic (PD) profiling in KP-4 xenograft tumours suggest that near-complete inhibition of MET kinase activity (≥95% reduction in phospho-MET) is required to achieve tumour regression. Therefore, this level of MET inhibition was used as the target to achieve in humans. Human pharmacokinetic (PK)/PD model simulations suggest that a tepotinib dose of 450 mg (500 mg as tepotinib hydrochloride) once daily is sufficient to achieve sustained inhibition of phospho-MET at a level >95% in more than 90% of a mixed solid tumour population. According to a translational PK/PD model, a dose reduction to 250 mg is predicted to maintain biologically meaningful target inhibition with ≥95% phospho-MET inhibition in more than 80% of the population.
No clinical maximum tolerable dose was reached in a dose-escalation administration up to 1,400 mg once daily. Tepotinib exposure increased in the presence of food, with an increase in the area under the plasma concentration-time curve (AUC) by about 1.6-fold and an increase in the maximum observed plasma concentration (Cmax) by 2-fold, compared with the fasted state. It is recommended that tepotinib be taken with food. In human plasma, tepotinib is highly protein-bound (98%). Following a single intravenous dose of carbon 14-labelled tepotinib, the volume of distribution during the terminal phase was shown to be large, with a geometric mean (geometric coefficient of variation) of 574 L.
Tepotinib is primarily metabolized by cytochrome P450 (CYP) 3A4 and 2C8 enzymes. One major circulating plasma metabolite, M506, has been identified and found to be a minor contributor to the overall efficacy of tepotinib.
Tepotinib is mainly excreted in the feces (approximately 85% of administered radioactivity, adjusted for total recovery), with urine being a minor excretion pathway. Metabolites account for 48% of the tepotinib-related radioactivity recovered in feces and urine. Following a single oral administration of a radiolabelled tepotinib dose of 500 mg, M506 accounted for only about 3% of the total radioactivity in feces.
Following a single intravenous dose of tepotinib, the geometric mean (geometric coefficient of variation) clearance was estimated to be 12.8 L/hour. The effective half-life for tepotinib was approximately 32 hours, based on the population PK analysis. After multiple daily administrations of tepotinib at a dose of 500 mg, median accumulation was 3.3-fold for AUC from time 0 to 24 hours (AUC0-24h) and 2.5-fold for Cmax.
Co-administration of Tepmetko with a strong CYP3A or permeability-glycoprotein (P-gp) inhibitor may increase tepotinib exposure. Co-administration with Tepmetko also increased the concentration of certain P-gp substrates. Tepotinib had no clinically relevant effect on the PK of the CYP3A4 substrate, midazolam. No clinically significant differences in tepotinib were observed when co-administrated with omeprazole. No clinically significant differences in glucose levels were observed when metformin (a multidrug and toxin extrusion 2 and organic cation transporter 2 substrate) was co-administered with tepotinib. No clinically significant difference in the adverse events were observed when rosuvastatin (a substrate to the drug transporter breast cancer resistance protein) was co-administered with tepotinib.
Exposure-safety analyses revealed that the risk of peripheral oedema increased with increasing tepotinib exposure. An analysis of pooled integrated exposure-corrected QT interval (QTc) data from four studies and an analysis of data from the pivotal VISION study (see Clinical Efficacy section) concluded that tepotinib at the proposed clinical dose of 450 mg (500 mg as tepotinib hydrochloride) once daily is not associated with prolongation of the QTc interval to a clinically relevant extent. The QTc effect of supratherapeutic tepotinib exposure was not evaluated.
Overall, the clinical pharmacological data support the use of Tepmetko for the recommended indication.
For further details, please refer to the Tepmetko Product Monograph, approved by Health Canada and available through the Drug Product Database.
Clinical Efficacy
The clinical efficacy of Tepmetko was evaluated in the pivotal VISION study, an ongoing, single-arm, open-label, multicentre, non-randomized, multicohort, two-part, Phase II study. The VISION study assessed the antitumour activity and tolerability of Tepmetko monotherapy (450 mg tepotinib [as 500 mg tepotinib hydrochloride] orally once daily) in adult patients with confirmed locally advanced unresectable (Stage IIIb) or metastatic (Stage IV) METex14-altered NSCLC. Patients with confirmed advanced METex14-altered NSCLC or MET amplification status were assigned after screening to either Cohort A (METex14 skipping alterations, regardless of MET amplification status), Cohort B (MET amplification and negative for METex14 skipping alterations), or Cohort C (METex14 skipping alterations, regardless of MET amplification status). The clinical efficacy of Tepmetko was assessed in Cohort A. Cohort C was a confirmatory cohort intended to extend and confirm the existing results for Cohort A. Interim results were obtained using a data cut-off date (DCO) of January 1, 2020 (DCO Jan 01 2020). These results were updated on July 1, 2020 (DCO Jul 01 2020).
The primary efficacy outcome measure of the VISION study was a confirmed objective response (OR), evaluated according to the Response Evaluation Criteria in Solid Tumours (RECIST) Version 1.1 guideline and based on an evaluation by a blinded independent review committee (BIRC). Patients were identified as having an OR if they achieved either a confirmed complete response or partial response from the first administration of Tepmetko to the first observation of progressive disease. The confirmed objective response rate (ORR) was evaluated. The duration of response (DOR; as per the BIRC), a secondary efficacy endpoint, was the time from when the complete response or partial response criteria were first met until progressive disease or death due to any cause within 84 days of the last evaluable tumour assessment, whichever occurred first. This endpoint was considered a clinically relevant antitumour activity endpoint for this study.
The efficacy analysis population comprised 146 patients from Cohort A who had received at least one dose of Tepmetko prior to October 2, 2019 and who had undergone at least two post baseline assessments or had discontinued study treatment for any reason (intention-to-treat [ITT]-02 Oct 2019 [DCO Jul 01 2020]). Further, these patients had at least 9 months of follow-up from the start of treatment (which was expected to yield 6 months of follow-up beyond the onset of response in responders), and were regarded as adequate to support stable OR and meaningful DOR results. Of note, within the ITT-02 Oct 2019 (DCO Jul 01 2020) population, 99 patients received their first dose prior to April 2, 2019 and had at least 15 months of follow-up from the start of treatment (ITT-02 Apr 2019 [DCO Jul 01 2020]).
At baseline, patients in the ITT-02 Oct 2019 (DCO Jul 01 2020) population had a median age of 73 years and were distributed equally with respect to gender. In addition, there was a higher proportion of smokers than would be expected from an NSCLC population whose tumours are driven by other oncogenes. Ninety-eight percent of patients had metastatic disease and 87% had adenocarcinoma histology. Patients were either treatment naive (n = 65) or had received up to two lines of prior systemic anticancer therapies (n = 81). Among previously treated patients, 89% had received prior platinum-based chemotherapy.
Patients in Cohort A were enrolled based on a positive test for METex14 by either liquid biopsy (65%) or tissue biopsy (58%). Thirty-three patients (23%) were identified as METex14-positive by both liquid biopsy and tissue biopsy. To account for potential differences in the detection of METex14 by different testing methodologies, three primary analysis sets were derived from the ITT analysis set: liquid biopsy positive (L+ analysis set; n = 95), tumour biopsy positive (T+ analysis set; n = 84), and all patients who tested positive for METex14 skipping alterations, irrespective of testing methodology (combined analysis set; n = 146). Patients in the combined analysis set included patients who were tested with only one test.
The study aimed to show an ORR (as per the BIRC) in the range of 40% to 50% and to demonstrate that the lower limit of the corresponding exact two-sided 95% confidence interval (CI) for ORR exceeded 20% across lines of therapy. Given the lack of published evidence documenting treatment outcomes in the METex14-altered NSCLC population at the time of study planning, the threshold of 20% was chosen taking into consideration the ORRs achieved with available therapies in patients with advanced NSCLC (i.e., platinum-based chemotherapy in first-line therapy, immune-checkpoint inhibitor monotherapy in second-line therapy, and single-agent chemotherapy in third-line therapy).
In the ITT-02 Oct 2019 (DCO Jul 01 2020) population, the ORR was 45.2% (95% CI: 37.0, 53.6). All responders (n = 66) achieved a best overall response of partial response; no patients achieved a best overall response of complete response. Most responses were achieved within three months of the initial dose, and the majority (44 of 66 responders; 66.7%) of responses were deep (i.e., a shrinkage of target lesions of 50% or more). The depth of response was an exploratory outcome measure. While the ORR was consistent between the primary L+ and T+ analysis sets, clinical data were very limited for patients who tested positive via liquid biopsy but negative via tumour tissue biopsy (L+/T-). Overall, the ORR (as per the BIRC) was consistent between the ITT-02 Oct 2019 (DCO Jul 01 2020) population and the ITT-02 Apr 2019 (DCO Jul 01 2020) population and between the DCO Jan 1 2020 and the DCO Jul 01 2020. Additionally, the ORR as per the investigator evaluation was supportive of the primary BIRC analysis.
The median DOR (as per the BIRC) for the ITT-02 Oct 2019 (DCO Jul 01 2020) population was 11.1 months (95% CI: 8.4, 18.5). The proportion of responders with DOR ≥6, ≥9, and ≥12 months was 74.2%, 43.9%, and 21.2%, respectively. While the proportion of responders with sustained DOR ≥6 months and ≥9 months had increased considerably at the update compared with the DCO Jan 01 2020, a smaller difference was observed for the proportion with DOR ≥12 months.
Overall, the benefit with Tepmetko was observed to be stable and independent of whether patients were treatment naive or previously treated. In treatment-naive patients compared with previously treated patients, there were 72.4% and 75.7% responders, respectively, with DOR ≥6 months; 34.5% and 51.4%, respectively, with DOR ≥9 months; and 17.2% and 24.3%, respectively, with DOR ≥12 months. The enrolment of treatment-naive patients occurred via a protocol amendment approximately six months after the first patient was enrolled, resulting in a shorter observation period for first-line therapy compared with later lines. Based on the current understanding of oncogene-driven NSCLC, an extrapolation from the metastatic stage (98% of patients) to the locally advanced unresectable stage was considered reasonable.
Efficacy results placed in the context of 1) intra-study comparisons in the VISION study before and after initiation of Tepmetko and 2) published evidence on available therapies for NSCLC used to treat METex14-altered NSCLC suggested increased clinical benefit with Tepmetko, primarily based on response rates. However, definitive conclusions could not be drawn. Real-world effectiveness outcomes were also inconclusive.
Taken together, the observed ORR and median DOR are considered promising evidence supporting clinical benefit in patients with METex14-altered NSCLC.
Indication
The New Drug Submission for Tepmetko was filed by the sponsor with the following indication:
Tepmetko (tepotinib) is indicated for the treatment of adult patients with advanced non-small cell lung cancer (NSCLC) harbouring MET tyrosine kinase receptor exon 14 (METex14) skipping alterations.
To ensure safe and effective use of the product, Health Canada approved the following indication:
Tepmetko (tepotinib) is indicated for the treatment of adult patients with locally advanced unresectable or metastatic non-small cell lung cancer (NSCLC) harbouring mesenchymal-epithelial transition (MET) tyrosine kinase receptor exon 14 skipping alterations.
Documentation of MET tyrosine kinase receptor exon 14 (METex14) skipping alteration status based on a validated METex14 assay is required prior to treatment with Tepmetko.
Efficacy in patients with NSCLC harbouring METex14 skipping alterations was based on objective response rate and duration of response in a single-arm study.
For more information, refer to the Tepmetko Product Monograph, approved by Health Canada and available through the Drug Product Database.
Clinical Safety
The clinical safety of Tepmetko was primarily evaluated in data from the Safety Analysis Set (SAF) from the ongoing pivotal VISION study (see Clinical Efficacy section).
At the DCO 01 Jul 2020, the SAF comprised a pooled cohort of 255 patients with advanced METex14-altered NSCLC from Cohort A (n = 152) and Cohort C (n = 103), all of whom had received at least one dose of Tepmetko monotherapy at the recommended dose of 450 mg tepotinib (as tepotinib hydrochloride) once daily.
At the DCO 01 Jul 2020, the median duration of therapy in the SAF was 22.3 weeks (range: 0 to 188 weeks), with 42% and 18% of patients exposed for ≥6 months and >12 months, respectively. Long-term safety data are therefore considered limited. The median age was 72 years (range: 41 to 94 years), with 79% of patients ≥65 years of age and 43% of patients ≥75 years of age. Gender was well-balanced (52% female). The most common race was White (67%), followed by Asian (28%). Study treatment was ongoing in 40% of patients, with disease progression, adverse events, and death representing the most common reasons for permanent treatment discontinuation.
Treatment-emergent adverse events (TEAEs) occurred in 96.5% of patients with the most common (≥20%) TEAEs being edema (69.0%), mainly peripheral edema (60.0%); fatigue (27.5%); nausea (26.7%); diarrhea (26.3%); increase in creatinine (25.9%); musculoskeletal pain (24.3%); hypoalbuminemia (23.9%); and dyspnea (20.4%). Treatment-related TEAEs, based on investigator assessment, were reported in 86.3% of patients.
Grade ≥3 TEAEs occurred in 52.9% of patients. Grade ≥3 TEAEs that occurred in ≥5% of patients included peripheral edema (7.8%), hypoalbuminemia (5.5%), and pleural effusion (5.1%). Treatment-related TEAEs of Grade ≥3 were reported in 25.1% of patients; peripheral edema (7.5%) was the only event occurring in ≥5% of patients.
Serious TEAEs occurred in 45.1% of patients. Serious TEAEs in ≥2% of patients included pleural effusion (6.7%), pneumonia (4.7%), dyspnea (3.9%), general health deterioration (3.5%), peripheral edema (2.4%), generalized edema (2.0%), pulmonary embolism (2.0%), and musculoskeletal pain (2.0%).
The most frequent TEAEs (≥1%) leading to permanent discontinuations of Tepmetko in 20.4% of patients were peripheral edema (3.5%), pleural effusion (2.0%), dyspnea (1.6%), general health deterioration (1.6%), pneumonitis (1.2%), and genital edema (1.2%). Temporary treatment discontinuations of Tepmetko due to a TEAE occurred in 43.9% of patients; the maximum permitted period of continuous treatment interruption was 21 days. Dose reductions of Tepmetko due to a TEAE occurred in 29.8% of patients.
Grade 5 TEAEs occurred in 11.8% of patients, and overall, seemed typical for an advanced NSCLC population. A fatal adverse reaction occurred in one patient (0.4%) due to interstitial lung disease. A case of fatal hepatic failure occurred in one patient (0.4%). While challenges with causality assessment in the case of fatal hepatic failure are acknowledged, a potential relationship with Tepmetko could not be ruled out. Additional considerations included the limited long-term safety data of Tepmetko and supportive pooled data (POOL) from five open-label, single-arm studies in which 448 patients with various solid tumours, including 255 patients from the SAF, received Tepmetko monotherapy at the proposed dose of 450 mg once daily. The median duration of exposure was 16.9 weeks, reflecting the short duration of exposure of Phase I studies or study parts. Data from the POOL studies were supportive of the hepatotoxicity observed in the VISION study. Furthermore, MET is involved in regeneration of damaged organs, in particular the liver. A case of fatal dyspnea from possible fluid overload occurred in one patient (0.4%). The causality of fatal dyspnea remained indeterminate as multifactorial events could have contributed, including fluid overload and progression of the underlying cancer with bilateral pleural effusions (cytology unknown).
The primary serious risks were identified as hepatotoxicity and interstitial lung disease/pneumonitis. Based on Tepmetko’s mechanism of action and findings from animal studies, it can cause fetal harm when administered to a pregnant woman.
Edema and hypoalbuminemia were very commonly reported and might be considered class effects. While acknowledging the potential for confounding by the underlying NSCLC, and possibly by the presence of METex14 skipping alterations, a contributory role of Tepmetko in the development of pleural effusion could not be excluded.
A median increase in serum creatinine of 31% was computed 21 days after initiation of Tepmetko in patients with and without renal failure TEAEs. One component of the observed increases in creatinine may be the inhibition of active tubular secretion by Tepmetko. Therefore, creatinine alone is not a reliable biomarker to assess renal function. Tepmetko did not appear to have a direct kidney toxicity. As the pathophysiology of pre-renal disease is multifactorial, the potential for Tepmetko indirectly contributing to acute kidney injury remains. While renal failure-type TEAEs were more commonly observed in patients with baseline renal impairment, many factors could have contributed to, and could explain, these observations.
Overall, the TEAE profile was similar between the DCO dates of January 1, 2020 and July 1, 2020 and the safety profile of the SAF was generally consistent with, and supported by, the data from the POOL studies.
Based on the information reviewed, Tepmetko has an acceptable and manageable safety profile, taking into consideration the intended patient population with advanced METex14-altered NSCLC. The identified safety issues can be managed through labelling, adequate monitoring, and dose modifications.
Appropriate warnings and precautions are in place in the approved Tepmetko Product Monograph to address the identified safety concerns, including a Serious Warnings and Precautions box describing the risk of hepatotoxicity, interstitial lung disease/pneumonitis, and embryo-fetal toxicity. Treatment with Tepmetko should be initiated and supervised by a qualified physician experienced in the use of anticancer therapies.
A Risk Management Plan (RMP) for Tepmetko was submitted to Health Canada by EMD Serono, a Division of EMD Inc. Canada. Upon review, the RMP was considered acceptable. The RMP is designed to describe known and potential safety issues, to present the monitoring scheme and when needed, to describe measures that will be put in place to minimize risks associated with the product.
For more information, refer to the Tepmetko Product Monograph, approved by Health Canada and available through the Drug Product Database.
7.2 Non-Clinical Basis for Decision
The medicinal ingredient in Tepmetko is tepotinib, supplied as tepotinib hydrochloride. In a non-clinical biochemical kinase assay, tepotinib inhibited the kinase activity of the mesenchymal-epithelial transition (MET) receptor. Tepotinib showed specificity for the MET receptor in a series of four kinase panel screens. No off-target kinase inhibition was observed at therapeutically meaningful concentrations. However, inhibitory action at clinically achievable concentrations were noted for the imidazoline 1 receptor and the melatonin 2 receptor with an inhibition of 106% and 102%, respectively.
Tepotinib and tepotinib hydrochloride inhibited cancer cell proliferation, migration, and anchorage-independent growth in functional cellular assays and inhibited MET phosphorylation in tumour tissue in mice. A major circulating metabolite of tepotinib, MSC2571109A, inhibited hepatocyte growth factor (HGF)-dependent MET phosphorylation with half-maximal inhibitory concentration values two- to five-fold higher than tepotinib hydrochloride. Tepotinib and tepotinib hydrochloride demonstrated potent anti-tumour efficacy in cancer models carrying oncogenic alterations to the MET gene, including MET tyrosine kinase receptor exon 14 (METex14) skipping alterations, as well as cancer models with MET overexpression and high HGF/MET expression. Tepotinib hydrochloride had no effect on wound healing in mice at single doses of up to 50 mg/kg. Cardiovascular safety analysis showed that neither tepotinib nor MSC2571109A exhibited the potential to induce QT prolongation in vitro and in vivo in rats and dogs. Central nervous system and respiratory functions were not altered with single doses of tepotinib up to 200 mg/kg in rats.
The metabolite MSC2571109A represented more than 25% of the plasma exposure of tepotinib in humans. In vitro data identifies both tepotinib and MSC2571109A as inhibitors and substrates of multiple enzymes and transporters. Tepotinib showed a rapid absorption in mice, but a slow absorption in rats, dogs, and monkeys following oral administration. In vivo studies in various animal models showed a moderate oral bioavailability (21.4% to 55.3%) of tepotinib in all of the species tested. In vitro studies showed that tepotinib is a substrate of permeability glycoprotein (P-gp). Thus, at low doses, tepotinib is susceptible to efflux leading to low drug absorption through intestinal membranes. However, at high doses, tepotinib is capable of saturating its own efflux, thus leading to increased transmembrane drug absorption. An in vitro study showed that tepotinib has a high ability for passive permeability. Some in vivo studies showed that the radiolabeled tepotinib accumulated in the intestine or gastrointestinal tract of rats following intravenous administration and it was also secreted in bile. This is indicative of biliary or fecal excretion of the drug. Tepotinib showed a moderate-to-high plasma protein binding affinity across species. In human plasma, the in vitro protein binding affinity for tepotinib was 96.6% to 98.4%. Tepotinib binds primarily to human serum albumin and acid alpha-1 glycoprotein. Tepotinib and/or its metabolite bind to melanin. In a study conducted using a xenograft mouse model to which human tumour cells were transplanted, tepotinib showed a high tumour-to-plasma ratio of 11.6 at 6 hours post dosing. Only the parent drug molecule was found to accumulate in tumour tissues, whereas the metabolites were found to accumulate in the plasma. Thus, only the parent molecule is expected be responsible for the anti-tumour efficacy of this drug in humans.
In vitro studies showed that the two-phase sequential biotransformation of tepotinib is predominantly accomplished by cytochrome P450 (CYP) 3A4 and CYP2C8. Other non-CYP metabolizing enzymes, such as flavin-containing mono-oxygenase, also contribute to the phase 1 (oxidative) metabolism of tepotinib, therefore, it appears unlikely that concomitantly administered effectors of CYP3A4 and CYP2C8 could affect the pharmacokinetics of tepotinib. Pharmacokinetic/pharmacodynamic modeling and simulation in xenograft mouse models allowed predicting that a greater than 95% inhibition of MET phosphorylation is required for observing significant tumour regression and that the minimum target efficacious plasma concentration of tepotinib in humans should be 146 ng/mL for observing an optimal therapeutic response.
Single-dose toxicity studies in rats and mice did not reveal any evidence for toxicity at dose levels of up to 2,000 mg/kg tepotinib. Information on acute toxicity in dogs was obtained from a short-term dose escalation study indicating that an oral dose of 45 mg/kg/day was clinically well tolerated, while higher doses of 90 mg/kg/day and 180 mg/kg/day led to reduced food consumption and decreases in body weight. In rats, histopathology revealed mild liver cell hypertrophy and minimal-to-mild activation of the thyroid follicular epithelium and macrophage aggregates in the mesenteric lymph nodes. Additionally, in four-week studies at high doses, treatment-related incidences of alveolar macrophage aggregates (foam cells) were seen in the lungs in some female rats. The macrophage aggregates in the lungs and lymph nodes were reversible and not associated with any inflammatory reaction at doses ≤450 mg/kg/day. In all studies, slight dose-dependent increases of liver enzyme values (e.g., alanine aminotransferase, aspartate aminotransferase, alkaline phosphatase, and glutamate dehydrogenase) were observed. All changes proved to be reversible or showed indications for reversibility.
Thyroid activation was also noted; however, as described by the sponsor, this is a known secondary compensatory phenomenon in situations of enhanced liver cell metabolism due to xenobiotics. The no-observed-adverse-effect levels in rats were set up at 90 mg/kg/day in the 4-week studies and 45 mg/kg/day in the 26-week study. In dogs, symptoms included a dose-dependent incidence and severity of vomiting and soft and/or mucous stool with bloody admixture/diarrhea, and were accompanied by body weight decreases and reduced food consumption. Increased hepatobiliary liver enzymes and pronounced cholangitis and pericholangitis associated with inflammatory infiltrates in the liver were the main findings in the 4-week and 39-week dog studies. All findings showed a trend toward normalization or complete reversal to normal after an adequate recovery period of up to 12 weeks. Additionally, cardiovascular investigations performed during the repeated dose toxicity study in dogs showed no relevant effects on heart rate, arterial blood pressure, and electrocardiogram parameters including the heart rate-corrected QT intervals. The no-observed-adverse-effect levels were set up in dogs at 10 mg/kg/day in the 4-week and 39-week studies, and 30 mg/kg/day in the 13-week study.
Tepotinib and its metabolites did not show any evidence of genotoxic activity in vitro and in vivo. Carcinogenicity studies were not conducted based on the current development in patients suffering from advanced cancer and in line with the International Council for Harmonisation guidelines. Two preliminary embryo-fetal development studies in rabbits revealed maternotoxic effects (starting at a dose level of 50 mg/kg/day) and a dose-dependent increase in the number of skeletal malformations (teratogenicity) (starting at a dose level of 5 mg/kg/day). In general toxicology studies with tepotinib, histopathology of reproductive organs did not reveal adverse effects. In addition, sperm analysis (i.e., morphology and motility) conducted at the end of the dosing period in the 26-week repeat-dose toxicity study in rats did not show any treatment-related change. Overall, these data indicate no risk for impairment of fertility. No dedicated local tolerance studies have been conducted since the compound is developed as a tablet that is administered via the oral route. No toxicologically relevant findings were observed at histological evaluations of the gastric and intestinal tract included in repeat-dose studies. Non-clinical investigations indicate no risk for phototoxicity. Toxicologically relevant findings identified within the non-clinical safety program are all considered clinically monitorable and manageable with appropriate risk mitigation measures in place. Overall, the toxicology data for Tepmetko supports the proposed indication for the treatment of adult patients with advanced non-small cell lung cancer whose tumours have a mutation that leads to METex14 skipping. Although the systemic exposure in the toxicology studies was comparatively lower than the systemic exposure measured in humans, the animal studies allowed for the identification of potential target organs of toxicity (liver/hepatobiliary system). In addition, embryo-fetal development studies in rabbits showed that tepotinib may induce skeletal malformations (teratogenicity).
In view of the intended use of Tepmetko, there are no pharmacological/toxicological issues within this submission that preclude authorization of the product. Appropriate warnings and precautionary measures are in place in the Tepmetko Product Monograph to address the identified safety concerns, including a Serious Warnings and Precautions box describing the risk of hepatotoxicity, interstitial lung disease/pneumonitis, and embryo-fetal toxicity.
For more information, refer to the Tepmetko 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 Tepmetko 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 30 months is acceptable when the drug product is stored at room temperature (15 ºC to 30 ºC) and in the original package to protect from moisture.
Proposed limits of drug-related impurities are considered adequately qualified (i.e., within International Council for Harmonisation limits and/or qualified from toxicological studies).
All sites involved in production are compliant with Good Manufacturing Practices.
All non-medicinal ingredients (described earlier) found in the drug product are acceptable for use in drugs according to the Food and Drug Regulations.
One excipient, lactose monohydrate, in the ready-to-use film-coating mixture (Opadry II pink) is of animal origin. Lactose monohydrate is produced from milk obtained from healthy animals (bovine) in the same condition as those used to collect milk for human consumption. A letter of attestation confirming the material is compliant with the Note for Guidance on Minimising the Risk of Transmitting Animal Spongiform Encephalopathy Agents via Human and Veterinary Medicinal Products (EMEA/410/01, Revision 3) was provided, indicating that it is considered to be safe for human use.
Related Drug Products
Product name | DIN | Company name | Active ingredient(s) & strength |
---|---|---|---|
TEPMETKO | 02516322 | EMD SERONO, A DIVISION OF EMD INC., CANADA | TEPOTINIB (TEPOTINIB HYDROCHLORIDE) 225 MG |