Summary Basis of Decision for Nuvaxovid

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

Recent Activity for Nuvaxovid

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 Nuvaxovid

Updated: 2024-03-22

The following table describes post-authorization activity for Nuvaxovid, a product which contains the medicinal ingredient severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) recombinant spike protein. 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.

Drug Identification Number (DIN):

  • DIN 02525364 - 5 mcg/0.5 mL SARS-CoV-2 recombinant spike protein, suspension, intramuscular administration

Post-Authorization Activity Table (PAAT)

Activity/submission type, control number Date submitted Decision and date Summary of activities
DIN 02525364 cancelled (post market) Not applicable Discontinuation date: 2024-02-29 The manufacturer notified Health Canada that sale of the drug has been discontinued post market. Health Canada cancelled the DIN(s) pursuant to section C.01.014.6(1)(a) of the Food and Drug Regulations.
PBRER # 278381 2023-08-18 Review completed 20234-01-22 Submission filed in response to commitments made as per the terms and conditions imposed on the authorization issued under the Food and Drug Regulations. PBRER #3 for the period 2022-12-20 to 2023-06-19. The sponsor was asked to provide updated assessments for the ongoing monitoring of safety events in the next PBRER for Nuvaxovid XBB.1.5.
SNDS # 275840 2023-05-31 Issued NOC 2023-11-16 Submission filed as a Level I – Supplement for a new 2-vial pack presentation for the 5-dose vial drug product. The submission was reviewed and considered acceptable, and an NOC was issued.
SNDS # 273313 2023-03-15 Cancellation Letter Received 2023-11-09 Submission filed as a Level I – Supplement for the expansion of the indication to include a booster dose regimen for adolescent patients 12-17 years of age and to update the PM with new safety and efficacy data. In addition, data were provided from studies 2019nCoV-301, -302, and -501 as per the terms and conditions imposed on the authorization issued under the Food and Drug Regulations. A Summary of Cancellation was published. 
SNDS # 273625 2023-03-24 Issued NOC 2023-07-18 Submission filed as a Level I – Supplement for a new 5-dose vial presentation. The data were reviewed and considered acceptable, and an NOC was issued.
PBRER # 272740 2023-02-27 Review completed 2023-07-24 Submission filed in response to commitments made as per the terms and conditions imposed on the authorization issued under the Food and Drug Regulations. PBRER #2 for the period 2022-06-20 to 2022-12-19. The information was reviewed and found acceptable. No further action was required.

Amended Terms and Conditions
Control # 255370

Not applicable

Terms and conditions amended post authorization
2023-07-10

Health Canada updated the Risk Management Plan Terms and Conditions for Nuvaxovid to reflect the accumulation of safety data and information gained in the post-market setting for this vaccine.
SNDS # 270288 2022-12-01 Issued NOC 2023-04-12 Submission filed as per the terms and conditions imposed on the authorization issued under the Food and Drug Regulations as a Level I – Supplement to add French language text to the inner and outer labels. The submission was reviewed and considered acceptable, and an NOC was issued.
NC # 272826 2023-03-01 Issued NOL 2023-04-03 Submission filed as a Level II (90 day) Notifiable Change (Moderate Quality Changes) to extend the shelf life of the drug substance. The submission was reviewed and considered acceptable, and an NOL was issued.
NC # 273204 2023-03-09 Issued NOL 2023-03-28 Submission filed as a Level II (90 day) Notifiable Change (Moderate Quality Changes) to change the specifications used to release the drug product and to extend the shelf life of the drug product from 9 to 12 months. The submission was reviewed and considered acceptable, and an NOL was issued.
NC # 272045 2023-02-06 Issued NOL 2023-03-24 Submission filed as a Level II (90 day) Notifiable Change (Moderate Quality Changes) to extend the shelf-life of a drug substance intermediate. The submission was reviewed and considered acceptable, and an NOL was issued.
NC # 272247 2023-02-09 Issued NOL 2023-03-16 Submission filed as a Level II (90 day) Notifiable Change (Moderate Quality Changes) to extend the in-use shelf life of the drug product from 6 to 12 hours when stored at 2oC to 8oC. The submission was reviewed and considered acceptable, and an NOL was issued.
NC # 272286 2023-02-09 Issued NOL 2023-03-13 Submission filed as a Level II (90 day) Notifiable Change (Moderate Quality Changes) to change the qualification protocol for the interim reference standard. The submission was reviewed and considered acceptable, and an NOL was issued.
SNDS # 268946 2022-10-21 Issued NOC 2023-03-09 Submission filed as a Level I – Supplement for a scale-up of the drug substance manufacturing process. The data were reviewed and considered acceptable, and an NOC was issued.
SNDS # 271559 2023-01-20 Cancellation Letter Received
2023-02-01

Submission filed as a Level I – Supplement to change the stability protocol of the drug substance. The changes were not in scope of an SNDS but were considered to be Level II and III changes. The sponsor cancelled the submission administratively.

Bi-monthly safety report Control # 270486 2022-12-09 Review completed
2023-02-17

Information filed as per the terms and conditions imposed on the authorization issued under the Food and Drug Regulations. Bi-monthly safety report for the period 2022-09-01 to 2022-11-15. The sponsor was asked to continue with the standard monitoring.

SNDS # 268596 2022-10-12 Issued NOC
2023-02-02

Submission filed as a Level II – Supplement (Safety) to update the PM with safety information related to anaphylaxis, myocarditis, pericarditis, and hypoesthesia/paraesthesia. The submission was reviewed and considered acceptable. As a result of the SNDS, modifications were made to the Warnings and Precautions, and Adverse Reactions sections of the PM, and corresponding changes were made to Part III: Patient Medication Information. An NOC was issued.

PBRER # 267302 2022-08-25 Review completed
2023-01-19

Submission filed in response to commitments made as per the terms and conditions imposed on the authorization issued under the Food and Drug Regulations. PBRER #1 for the period 2021-12-20 to 2022-06-19. The information was reviewed and found acceptable. No further action was required.

Monthly safety report Control # 268712 2022-10-14 Review completed
2023-01-03

Information filed as per the terms and conditions imposed on the authorization issued under the Food and Drug Regulations. Monthly safety report #8 for the period 2022-09-01 to 2022-09-30. The sponsor was asked to continue with the standard monitoring.

SNDS # 265342 2022-06-21 Issued NOC
2022-12-06

Submission filed as a Level I – Supplement to seek authorization for the use of Nuvaxovid in adolescents 12 to 17 years of age. The submission was reviewed and considered acceptable, and an NOC was issued. A Regulatory Decision Summary was published.

Monthly safety report Control # 267901 2022-09-15 Review completed
2022-12-05

Information filed as per the terms and conditions imposed on the authorization issued under the Food and Drug Regulations. Monthly safety report #7 for the period 2022-08-01 to 2022-08-31. The sponsor was asked to continue with the standard monitoring.

SNDS # 266285 2022-08-05 Issued NOC (subject to terms and conditions)
2022-11-17

Submission filed as a Level I – Supplement to seek authorization for a booster dose of Nuvaxovid. The submission was reviewed and considered acceptable, and an NOC was issued. Terms and conditions were imposed on the authorization. A Regulatory Decision Summary was published.

Monthly safety report Control # 266986 2022-08-15 Review completed
2022-11-15

Information filed as per the terms and conditions imposed on the authorization issued under the Food and Drug Regulations. Monthly safety report #6 for the period 2022-07-01 to 2022-07-31. The sponsor was asked to continue with the standard monitoring.

Post-Market Information Request Review Control # 255370 2022-10-07 Review completed
2022-11-09

Request filed regarding transitioning monthly summary safety reports to bi-monthly summary safety reports. Health Canada agreed that the sponsor will submit a bi-monthly summary safety report for one cycle, then submit 6-month PBRERs as described in the terms and conditions imposed on the authorization issued under the Food and Drug Regulations.

NC # 267838 2022-09-13 Issued NOL
2022-10-24

Submission filed as a Level II (90 day) Notifiable Change (Moderate Quality Changes) regarding a change in the reference standard for the drug substance. The submission was reviewed and considered acceptable, and an NOL was issued.

SNDS # 268098 2022-09-23 Cancellation Letter Received
2022-10-06

Submission filed as a Level II – Supplement (Safety) to update the PM. The proposed revisions exceeded the scope of a Labelling SNDS. The submission was therefore cancelled administratively by the sponsor so as to be filed as a Clinical SNDS.

NC # 265872 2022-07-11 Issued NOL
2022-09-14

Submission filed as a Level II (90 day) Notifiable Change (Moderate Quality Changes) to change the primary container closure system for the drug substance, to extend the shelf life of the drug substance, and to change the supplier of an adjuvant. The submission was reviewed and considered acceptable, and an NOL was issued.

Amended Terms and Conditions Control # 255370 Not applicable Terms and conditions amended post authorization
2022-09-14

Health Canada imposed a new Term and Condition to the authorization of Nuvaxovid. Monthly safety reports are to continue to be submitted.

Monthly safety report
Control # 266288
2022-07-22 Review completed
2022-08-12

Information filed as per the terms and conditions imposed on the authorization issued under the Food and Drug Regulations. Monthly safety report #5 for the period 2022-06-01 to 2022-06-30. The sponsor was asked to provide updated assessments for the ongoing monitoring of safety events.

SNDS # 265228 2022-06-16 Issued NOC
2022-08-12

Submission filed as a Level I – Supplement to make updates to the manufacturing process of the drug substance. The information was reviewed and considered acceptable. An NOC was issued.

Monthly safety report
Control # 265205
2022-06-15 Review completed
2022-07-14

Information filed as per the terms and conditions imposed on the authorization issued under the Food and Drug Regulations. Monthly safety report #4 for the period 2022-05-01 to 2022-05-31. The sponsor was asked to provide updated assessments for the ongoing monitoring of safety events.

NC # 264027 2022-05-11 Issued NOL
2022-07-04

Submission filed as a Level II (90 day) Notifiable Change (Moderate Quality Changes) to extend the shelf lives of the drug substance and the adjuvant. The submission was reviewed and considered acceptable, and an NOL was issue.

Monthly safety report
Control # 264258
2022-05-13 Review completed
2022-06-15

Information filed as per the terms and conditions imposed on the authorization issued under the Food and Drug Regulations. Monthly safety report #3 for the period 2022-04-01 to 2022-04-30. The sponsor was asked to provide updated assessments for the ongoing monitoring of safety events.

Monthly safety report
Control # 263401
2022-04-14 Review completed
2022-05-26

Information filed as per the terms and conditions imposed on the authorization issued under the Food and Drug Regulations. Monthly safety report #2 for the period 2022-03-01 to 2022-03-31. The sponsor was asked to continue with the standard monitoring.

Monthly safety report
Control # 262392
2022-03-15 Review completed
2022-04-01
Information filed as per the terms and conditions imposed on the authorization issued under the Food and Drug Regulations. Monthly safety report #1 for the period 2021-12-20 to 2022-02-28. The sponsor was asked to continue with the standard monitoring.
Drug product (DIN 02525364) market notification Not applicable Date of first sale:
2022-03-31

The manufacturer notified Health Canada of the date of first sale pursuant to C.01.014.3 of the Food and Drug Regulations.

Health Product Risk Communication Not applicable Posted
2022-02-23
Health Product Risk Communication posted (Distribution of Nuvaxovid with English-only Vial and Carton Labels), containing information about labelling, product safety and supply for healthcare professionals.
NDS # 255370 2021-08-27 Issued NOC (subject to terms and conditions)
2022-02-17
NOC issued for New Drug Submission. Terms and conditions were imposed on the authorization.
Application # 248761 2021-01-29 Closed
2021-09-16
Application closed with expiry of Interim Order Respecting the Importation, Sale and Advertising of Drugs for Use in Relation to COVID-19.
Summary Basis of Decision (SBD) for Nuvaxovid

Date SBD issued: 2022-04-08

The following information relates to the New Drug Submission for Nuvaxovid.

SARS-CoV-2 recombinant spike protein

Drug Identification Number (DIN):

  • DIN 02525364 - 5 mcg/0.5 mL SARS-CoV-2 recombinant spike protein, suspension, intramuscular administration

Novavax Inc.

New Drug Submission Control Number: 255370

 

On February 17, 2022, Health Canada issued a Notice of Compliance (NOC), subject to terms and conditions, to Novavax Inc. for the vaccine Nuvaxovid.

The Food and Drug Regulations were amended on March 18, 2021 to incorporate flexibilities into the existing new drug submission (NDS) regulatory pathway, thereby facilitating the regulatory process for authorization of new drugs that treat or prevent coronavirus disease 2019 (COVID‑19). The modified requirements allow an NDS for a designated COVID‑19 drug to be filed through a rolling submission process, i.e., as the information becomes available. Sponsors are responsible for completing the required documentation and providing the necessary evidence to Health Canada. Health Canada will issue an NOC for a COVID‑19 drug if it is determined that the benefits and risks of the product are supported by evidence of the safety, efficacy, and consistent quality of the drug.

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 Nuvaxovid is favourable for active immunization to prevent COVID-19 caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in individuals 18 years of age and older.

Nuvaxovid is authorized in accordance with the Food and Drug Regulations, subject to terms and conditions that need to be met by the sponsor. Terms and conditions may be imposed or amended at any time. Additionally, failure to comply with the terms and conditions may result in compliance and enforcement actions being taken by Health Canada.

For further information on the amended regulatory pathway, refer to the Guidance on Amendments to the Food and Drug Regulations for Drugs for Use in Relation to COVID‑19.

1 What was approved?

Nuvaxovid was authorized for active immunization to prevent coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in individuals 18 years of age and older.

Nuvaxovid is not authorized for use in pediatric patients (less than 18 years of age), as its safety and effectiveness have not been established in this population.

Clinical studies of Nuvaxovid include participants 65 years of age and older and their data contribute to the overall assessment of safety and efficacy.

Nuvaxovid (5 mcg/0.5 mL, SARS-CoV-2 recombinant spike protein) is presented as a suspension. In addition to the medicinal ingredient, the suspension contains 50 mcg of Matrix‑M adjuvant in each 0.5 mL dose and the following non-medicinal ingredients: disodium hydrogen phosphate heptahydrate, sodium dihydrogen phosphate monohydrate, sodium chloride, polysorbate 80, sodium hydroxide, hydrochloric acid, and water for injection. The Matrix-M adjuvant contains Quillaja saponaria saponins fraction A and fraction C and the following non-medicinal ingredients: cholesterol, phosphatidylcholine, potassium chloride, potassium dihydrogen phosphate, disodium hydrogen phosphate dihydrate, and sodium chloride.

The use of Nuvaxovid is contraindicated in individuals who are hypersensitive to the active ingredient or to any ingredients in the formulation, including any non-medicinal ingredient, or component of the container.

Nuvaxovid was approved for use under the conditions stated in its Product Monograph taking into consideration the potential risks associated with its administration. The Nuvaxovid Product Monograph is available through the Drug Product Database and on the Health Canada COVID-19 vaccines and treatments portal.

For more information about the rationale for Health Canada's decision, refer to the Clinical, Non-clinical, and Quality (Chemistry and Manufacturing) Basis for Decision sections.

2 Why was Nuvaxovid approved?

Health Canada considers that the benefit-risk profile of Nuvaxovid is favourable for active immunization to prevent coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in individuals 18 years of age and older.

Nuvaxovid is authorized in accordance with the Food and Drug Regulations, subject to terms and conditions that need to be met by the sponsor.

Coronavirus disease 2019 is the infectious disease caused by the novel coronavirus, SARS-CoV-2, which has spread rapidly and globally since its emergence in late 2019. On March 11, 2020, the World Health Organization declared COVID-19 a pandemic. In Canada, there have been 3,217,184 confirmed cases of COVID-19 and approximately 35,787 deaths as of February 17, 2022, the date of authorization of Nuvaxovid.

Severe acute respiratory syndrome coronavirus 2 is a highly transmissible and pathogenic coronavirus. The majority of SARS-CoV-2-infected patients experience mild respiratory disease. However, following infection, some patients develop severe disease that requires oxygen support or critical disease with complications such as respiratory failure, sepsis and septic shock, thromboembolism, and/or multiorgan failure, including acute kidney injury and cardiac injury. Medical conditions or other factors that place patients at high risk for progression to severe COVID-19 include older age, obesity, current smoker, chronic kidney disease, diabetes, immunosuppressive disease or immunosuppressive treatment, cardiovascular disease, chronic lung disease, sickle cell disease, neurodevelopmental disorders, active cancer, and medically related technological dependence. Other medical conditions or factors (e.g., race or ethnicity) may also place individual patients at high risk for progression to severe COVID-19.

Health Canada has previously authorized the following vaccines for protection against COVID-19: Comirnaty (previously Pfizer‑BioNTech COVID‑19 Vaccine), Spikevax (previously COVID‑19 Vaccine Moderna), Vaxzevria (previously AstraZeneca COVID‑19 Vaccine), Covishield, and Janssen COVID-19 Vaccine.

Two of the currently authorized vaccines are messenger ribonucleic acid (mRNA) vaccines (Comirnaty and Spikevax) while the other two are viral-vectored vaccines (Vaxzevria and Janssen COVID-19 Vaccine). Covishield was authorized under the Interim Order Respecting the Importation, Sale and Advertising of Drugs for Use in Relation to COVID-19 and the authorization has since expired. While care for individuals who have COVID-19 has improved with clinical experience, having multiple vaccine options is desirable.

Nuvaxovid is a sterile, preservative-free, aqueous buffered suspension of the SARS-CoV-2 recombinant spike (rS) protein that is co-formulated with Matrix-M adjuvant and a formulation buffer. The SARS-CoV-2 rS protein in Nuvaxovid is produced in the Spodoptera frugiperda insect cell line infected with a baculovirus that encodes full-length, SARS-CoV-2 spike gene-producing trimeric spike proteins from the original (Wuhan) strain. Matrix-M adjuvant contains Quillaja saponaria saponins fraction A and fraction C. The vaccine elicits both humoral and cellular immune responses directed against the spike protein which may contribute to protection against COVID-19.

Clinical data supporting the efficacy, immunogenicity, and safety of Nuvaxovid were primarily evaluated in six ongoing clinical studies conducted in Australia, South Africa, the United Kingdom, the United States, Mexico, and India. Of these studies, two (Studies 2019nCoV-301 and 2019nCoV-302) were considered pivotal in the establishment of efficacy for Nuvaxovid. Both pivotal studies were Phase III, multicentre, randomized, observer-blinded, and placebo-controlled. Study 2019nCoV-301 was conducted in the United States and Mexico. In total, 29,949 participants 18 years of age and older were randomized in a 2:1 ratio to receive two intramuscular injections of either Nuvaxovid or placebo, 21 days apart. Study 2019nCoV-302, conducted in the United Kingdom, included 15,187 participants 18 to 84 years of age randomized in a 1:1 ratio to receive two intramuscular injections of either Nuvaxovid or placebo, 21 days apart.

Both studies included participants with a clinically stable underlying co-morbidity, as well as participants with well-controlled human immunodeficiency virus (HIV) infection. Both studies excluded participants who were significantly immunocompromised, pregnant, or who had a history of laboratory-confirmed COVID-19. The demographic and baseline characteristics were balanced amongst recipients of Nuvaxovid or placebo in both studies. It is noteworthy that the data provided from both the pivotal studies were collected at a time when the Alpha variant was the primary circulating strain of SARS-CoV-2. This was prior to the emergence of the Delta and Omicron variants. Novavax Inc. is required to provide data regarding protection against current and emerging variants of concern as one of the terms and conditions associated with the authorization of Nuvaxovid.

Study 2019nCoV-301, Phase III; Participants in final efficacy analysis

 

Vaccine group

Placebo group

Total

Male

9,050 (52.3%)

4,131 (50.7%)

13,181 (51.8%)

Female

8,262 (47.7%)

4,009(49.3%)

12,271 (48.2%)

Total number of participants

(18 years of age and older)

17,312

8,140

25,452

Study 2019nCoV-302, Phase III; Participants in final efficacy analysis

Male

3,609 (51.4%)

3,629 (51.7%)

7,238 (51.6%)

Female

3,411 (48.6%)

3,390 (48.3%)

6,801 (48.4%)

Total number of participants

(18 years of age and older)

7,020

7,019

14,039

Participants in safety analysis

Total number of participants

(18 years of age and older) who received at least one dose

29,297

19,401

48,698

The primary efficacy endpoint for both pivotal studies was the vaccine efficacy of Nuvaxovid against the first episode of laboratory-confirmed (polymerase chain reaction [PCR]-positive) symptomatic mild, moderate, or severe COVID-19 with an onset at least 7 days following the administration of Dose 2 in participants who were seronegative to SARS-CoV-2 at baseline. The vaccine efficacy success criteria for the primary endpoint as predefined by the sponsor was a vaccine efficacy rate with a lower bound of the 95% confidence interval (LBCI) above 30%. Health Canada's vaccine efficacy success criteria as defined in the Guidance for Market Authorization Requirements for COVID-19 Vaccines is a target threshold of at least 50% vaccine efficacy, with an LBCI above 30%.

As of the data cut-off date of May 31, 2021, the primary efficacy analysis set for Study 2019nCoV-301 included 25,452 participants who received either Nuvaxovid (total number [n] = 17,312) or placebo (n = 8,140). For Study 2019nCoV-302, as of the cut-off date of January 29, 2021, the final primary efficacy analysis set included 14,039 participants who received either Nuvaxovid (n = 7,020) or placebo (n = 7,019).

As demonstrated through the results of the final primary efficacy analysis, Nuvaxovid met the primary endpoints for both pivotal studies and exceeded the success criterion of a vaccine efficacy greater than 50% and with the LBCI above 30% set out by Health Canada's Guidance for Market Authorization Requirements for COVID-19 Vaccines.

Vaccine efficacy success criteria

As defined in Health Canada's Guidance for Market Authorization Requirements for COVID-19 Vaccines

Target threshold of at least 50% vaccine efficacy, with a lower bound of the 95% confidence interval (CI) above 30%

Predefined by sponsor for Studies 2019nCoV-301 and 2019nCoV-302

A vaccine efficacy success criteria for the primary endpoint of a lower bound of the 95% confidence interval (CI) above 30%

Efficacy rates observed in final efficacy analysis (Study 2019nCoV-301)

 

Vaccine efficacy rate

95% Confidence Interval

Overall

Participants 18 years of age and older

90.4%

82.9% to 94.6%

Participants 18 years to 64 years of age

91.5%

84.2% to 95.4%

Participants 65 years of age or older

57.5%

-486.91% to 96.9%

Efficacy rates observed in final efficacy analysis (Study 2019nCoV-302)

 

Vaccine efficacy rate

95% Confidence Interval

Overall
Participants 18 years of age and older

89.7%

80.2% to 94.6%

Participants 18 years to 64 years of age

89.8%

79.7% to 94.9%

Participants 65 years of age or older

88.9%

20.2% to 99.7%

For Study 2019nCoV-301, the overall vaccine efficacy of Nuvaxovid was 90.4% (95% CI: 82.9, 94.6) for prevention of the onset of COVID-19 from 7 days after Dose 2 in participants 18 years of age and older. Thus, Nuvaxovid met the study's primary efficacy endpoint criterion for success with an LBCI greater than 30%. In addition, Nuvaxovid met Health Canada's required threshold of at least 50% vaccine efficacy, with an LBCI greater than 30%. No cases of moderate or severe COVID-19 were reported in the 17,312 Nuvaxovid recipients compared with 10 cases of moderate COVID-19 and 4 cases of severe COVID-19 reported in the 8,140 placebo recipients.

For Study 2019nCoV-302, the overall vaccine efficacy of Nuvaxovid was 89.7% (95% CI: 80.2, 94.6) for prevention of the onset of COVID-19 from 7 days after Dose 2 in participants 18 years of age and older. Nuvaxovid met the study's primary efficacy endpoint criterion for success, with an LBCI greater than 30%. In addition, Nuvaxovid met Health Canada's required threshold of at least 50% vaccine efficacy, with an LBCI greater than 30%. There were 9 cases of moderate COVID‑19 and no cases of severe COVID-19 reported in the 7,020 Nuvaxovid recipients compared with 63 cases of moderate and 5 cases of severe COVID-19 reported in the 7,019 placebo recipients.

For Study 2019nCoV-301, vaccine efficacy data for participants 65 years of age and older were limited as the event rates in this subgroup were too low to allow for meaningful interpretation.

The immunogenicity of Nuvaxovid was primarily evaluated in the two pivotal studies (2019nCoV-301 and Study 2019nCoV-302), the Phase IIa/b Study 2019nCoV-501, and Study ICMR/SII-COVOVAX. Results from these studies indicate that Nuvaxovid elicited SARS-CoV-2 immune responses.

Clinical safety data for Nuvaxovid was pooled from the two pivotal studies and the supportive Study 2019nCoV-501. At the time of the analysis, a total of 48,698 participants 18 years of age and older received at least one dose of Nuvaxovid (n = 29,297) or placebo (n = 19,401). The median duration of follow-up was 70 days post-Dose 2, with 32,993 (66%) participants completing more than 2 months of follow-up. At the time of vaccination, the median age of participants who received Nuvaxovid was 48 years (range 18 to 95 years): 84.1% of participants were between 18 and 64 years of age and 15.9% of participants were 65 years of age or older.

The frequency and severity of solicited local and systemic reactions were collected within 7 days following each dose of Nuvaxovid or placebo. Of the pooled reactogenicity data, which included participants 18 years of age and older who received at least one dose of Nuvaxovid (n = 21,395) or placebo (n = 12,197), the most frequent adverse reactions in Nuvaxovid recipients were injection site tenderness (68%), injection site pain (56%), fatigue (45%), myalgia (44%), headache (41%), malaise (35%), arthralgia (20%), and nausea or vomiting (11%). The corresponding rates in placebo recipients were injection site tenderness (14%), injection site pain (12%), fatigue (18%), myalgia (11%), headache (18%), arthralgia (6%), and nausea or vomiting (5%). Adverse reactions were usually mild to moderate in severity with a median duration of 2 days or less for local events and 1 day or less for systemic events following vaccination. When compared with Dose 1, local and systemic adverse reactions were more frequently reported after Dose 2.

Across the pooled studies, participants were monitored for unsolicited adverse events after receipt of Dose 1 through 28 days after Dose 2 (at 49 days). The overall frequency of unsolicited adverse events was calculated as events per 100 person‑years (e/100 PY) for participants who received at least one dose of Nuvaxovid or placebo. For those who received Nuvaxovid, there were 157 e/100 PY for participants 18 to 64 years of age (younger cohort) and 153 e/100 PY for participants 65 years of age and older (older cohort). In comparison, for those who received the placebo, there were 133 e/100 PY for the younger cohort and 124 e/100 PY for the older cohort.

Serious adverse events (SAEs) were uncommon across both treatment groups, with a higher incidence rate in participants who received placebo (4.09 e/100 PY) than in participants who received Nuvaxovid (3.82 e/100 PY). Incidence rates for SAEs in the younger cohort were 3.31 e/100 PY in Nuvaxovid recipients and 3.59 e/100 PY in placebo recipients. There was a slightly higher incidence rate among participants in the older cohort, with incidence rates of 6.69 e/100 PY in Nuvaxovid recipients and 6.65 e/100 PY in placebo recipients.

There were no other notable patterns of imbalance between treatment groups for specific categories of serious adverse events or adverse events of interest. No deaths related to the vaccine were reported in the main and supportive clinical studies.

Myocarditis was identified in two male teenagers shortly after receiving a second dose of the vaccine resulting in a mild clinical course with complete resolution and no sequelae. The information available at the time of authorization was insufficient to determine a causal relationship with the vaccine.

The overall frequency of non-serious unsolicited adverse events was higher in the Nuvaxovid group than in the placebo group, with events of fatigue, injection site pain, pyrexia, and myalgia occurring beyond the 7-day post-injection period largely accounting for the differences between the treatment groups. In addition, an imbalance of chills and pain in an extremity were reported. Chills occurred in 0.55% (n = 166) of participants who received Nuvaxovid and 0.11% (n = 21) of participants who received placebo. Pain in an extremity occurred in 1.36% (n = 410) of participants who received Nuvaxovid and 0.36% (n = 72) of participants who received placebo. There were no other notable imbalances between treatment groups for unsolicited non-serious adverse events that would suggest a causal relationship to Nuvaxovid.

The safety and efficacy of Nuvaxovid in pregnant women have not yet been established. Administration of Nuvaxovid in pregnancy should only be considered when the potential benefits outweigh any potential risks for the mother and fetus.

It is unknown if Nuvaxovid is excreted in human milk. A risk to the newborns/infants cannot be excluded. The developmental and health benefits of breastfeeding should be considered along with the mother’s clinical need for immunization against COVID-19.

A Core (European Union) Risk Management Plan (RMP) and a Canadian RMP Addendum for Nuvaxovid were submitted by Novavax Inc. to Health Canada. The RMP is designed to describe known and potential safety issues, to present the monitoring scheme, and, when needed, to describe measures that will be put in place to minimize risks associated with the product. The RMP for Nuvaxovid includes information about the important potential risks of vaccine associated enhanced disease (VAED) including vaccine associated enhanced respiratory disease (VAERD), anaphylaxis, and myocarditis and pericarditis. The RMP also identified eight areas of missing (limited/no clinical data) information: "use during pregnancy and while breastfeeding", "use in immunocompromised patients", "use in frail patients with comorbidities", "use in patients with autoimmune or inflammatory disorders", "interaction with other vaccines", "long-term safety", "use in pediatric subjects", and "long-term effectiveness".

Overall, the RMP was considered to be acceptable and identified appropriate monitoring (pharmacovigilance) activities and risk minimization measures (i.e., Product Monograph and labelling) based on the known safety profile of the product. The identified limitations and areas of missing information are managed through labelling and the RMP, and will continue to be investigated through ongoing and planned studies. The Phase III studies are ongoing and will continue to collect information on the long-term safety and efficacy of Nuvaxovid. In addition, there are post-authorization commitments for monitoring the long-term safety and effectiveness of Nuvaxovid. As outlined in the terms and conditions, the RMP will be updated to reflect additional safety information, including that which is relevant to the Canadian-specific context, as it becomes available. In addition to regulatory requirements for post-market monitoring and prioritized reporting of adverse events following immunization, monthly safety summary reports will be provided to Health Canada and will include information related to special populations (e.g., pregnant women). Results related to safety and effectiveness from ongoing and planned studies will be submitted as they become available. For more information, refer to the terms and conditions available on the Health Canada COVID‑19 vaccines and treatments portal.

The submitted inner and outer labels, package insert and Patient Medication Information section of the Nuvaxovid Product Monograph meet the necessary regulatory labelling, plain language and design element requirements.

The sponsor submitted a brand name assessment that included testing for look‑alike sound‑alike attributes. Upon review, the proposed name Nuvaxovid was accepted.

Nuvaxovid has been shown to have a favourable benefit-risk profile and an acceptable safety profile based on non-clinical and clinical studies. Collectively, the results of the clinical efficacy and safety evaluation demonstrate that Nuvaxovid met the safety requirements as specified in Health Canada's Guidance for Market Authorization Requirements for COVID‑19 Vaccines. The vaccine was found to be safe and well tolerated in participants 18 years of age and older when administered according to the recommended dosage regimen. Participants from the pivotal studies 2019nCoV-301 and 2019nCoV-302 will be followed for up to 24 and 12 months, respectively, for assessments of both safety and efficacy against COVID-19. These data will be provided to Health Canada when available along with safety data from 6 months of follow-up. At the time of authorization, important limitations of the data included the lack of information on the long-term safety and efficacy of the vaccine, the duration of protection, protection against current and emerging variants, and the lack of or limited data for special populations (e.g., individuals who are significantly immunocompromised, pregnant, have a history of COVID-19, or have severe comorbidities). These limitations are considered adequately managed through labelling, terms and conditions associated with the authorization of Nuvaxovid, the RMP, and adequate monitoring. Appropriate warnings and precautions are in place in the Nuvaxovid Product Monograph to address the identified risks.

This New Drug Submission complies with the requirements of sections C.08.002 and C.08.005.1 and therefore Health Canada has issued a Notice of Compliance (NOC) pursuant to section C.08.004 of the Food and Drug Regulations. The NOC in respect of Nuvaxovid is accompanied by terms and conditions imposed in accordance with section C.01.014.21 of the Food and Drug Regulations. Of note, terms and conditions may be imposed or amended at any time. All terms and conditions are enforceable under section 21.7 of the Food and Drugs Act. Failure to comply with the terms and conditions may result in compliance and enforcement actions being taken by Health Canada.

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 Nuvaxovid?

The application for authorization of Nuvaxovid was filed on January 29, 2021, in accordance with section 3 of the Interim Order Respecting the Importation, Sale and Advertising of Drugs for Use in Relation to COVID-19 (Interim Order). The intent of the Interim Order was to expedite the authorization of COVID-19 drugs; it was signed by the Minister of Health on September 16, 2020 and was in place for one year. The Interim Order allowed the Minister to account for the urgent public health needs relating to COVID-19 in deciding whether to authorize a COVID-19 drug based on the provided evidence of safety, efficacy, and quality. Before the Interim Order expired, Novavax Inc. filed a New Drug Submission (NDS) for Nuvaxovid on August 27, 2021.

The Food and Drug Regulations were amended on March 18, 2021 to incorporate flexibilities into the existing new drug submission (NDS) regulatory pathway, thereby facilitating the regulatory process for authorization of new drugs that treat or prevent coronavirus disease 2019 (COVID‑19). For example, to expedite the review process, the modified requirements allow an NDS for a designated COVID‑19 drug to be filed through a rolling submission process, i.e., as the information becomes available. This in turn allows Health Canada to commence a rolling review process of the information submitted. As outlined in the Guidance on amendments to the Food and Drug Regulations for drugs for use in relation to COVID‑19, Health Canada will begin its review using the information submitted by the sponsor and accept new evidence as it becomes available until the submission is deemed complete. The rolling process can reduce the time it takes to authorize these critical new drugs while maintaining appropriate standards of safety, efficacy, and quality. Sponsors are responsible for completing the required documentation and providing the necessary evidence to Health Canada. Health Canada will issue a Notice of Compliance (NOC) for a COVID‑19 drug if it is determined that the benefits and risks of the product are supported by evidence of the safety, efficacy, and consistent quality of the drug. Importantly, the amended regulations also allow the use of terms and conditions in order to ensure appropriate oversight, manage uncertainties or mitigate risks related to the drug in the context of the public health need due to COVID‑19.

The information for this NDS was provided on a rolling basis. Following an expedited review of the data submitted, Health Canada determined that sufficient evidence was provided to support the conclusion that the benefits of Nuvaxovid outweigh the risks under the conditions of use recommended, with consideration given to the uncertainties relating to those benefits and risks as well as the public health need related to COVID‑19. Health Canada issued an NOC for Nuvaxovid, with imposed terms and conditions, on February 17, 2022.

For further information on the amended NDS regulatory pathway, refer to the Guidance on Amendments to the Food and Drug Regulations for Drugs for Use in Relation to COVID‑19.

The review of the quality, non-clinical, and clinical components of the New Drug Submission (NDS) for Nuvaxovid was based on a critical assessment of the data package submitted to Health Canada. The review completed by the European Medicines Agency (EMA) was used as an added reference, as per Method 3 described in the Draft Guidance Document: The Use of Foreign Reviews by Health Canada. The Canadian regulatory decision on the Nuvaxovid NDS was made independently based on the Canadian review.

 

Submission Milestones: Nuvaxovid

Submission Milestone Date
Application Milestone: Control # 248761  
Pre-application meeting 2021-01-20
Initial application filed by sponsor under the Interim Order 2021-01-29
Initial non-clinical data submitted by sponsor 2021-01-29
Initial clinical data submitted by sponsor 2021-03-24
Application closed with expiry of Interim Order 2021-09-16
Submission Milestone: Control # 255370  
Initial New Drug Submission filed by sponsor 2021-08-27
Initial quality data submitted by sponsor 2021-09-20
Screening Acceptance Letter issued 2021-10-12
Risk Management Plan submitted by sponsor 2021-10-21
Health Canada Risk Management Plan evaluation completed 2022-01-31
Health Canada non-clinical evaluation completed 2022-02-16
Health Canada clinical/medical evaluation completed 2022-02-16
Health Canada quality evaluation completed 2022-02-16
Health Canada labelling evaluation completed 2022-02-16
Terms and conditions finalized by Health Canada 2022-02-16
Final Product Monograph (English) submitted by sponsor 2022-02-16
Final Product Monograph (French) submitted by sponsor 2022-02-16
Notice of Compliance issued by Director General, Biologic and Radiopharmaceutical Drugs Directorate 2022-02-17
4 What follow-up measures will the company take?

The Notice of Compliance issued in respect of Nuvaxovid is accompanied by terms and conditions imposed on the drug identification number assigned to Nuvaxovid in accordance with section C.01.014.21 of the Food and Drug Regulations. Of note, terms and conditions may be imposed or amended at any time. Failure to comply with the terms and conditions may result in compliance and enforcement actions being taken by Health Canada.

These terms and conditions set out requirements relating to clinical, non-clinical, quality (chemistry and manufacturing), labelling, and pharmacovigilance information. They were put in place to ensure appropriate oversight, manage uncertainties or mitigate risks, and ascertain the continued quality, safety, and efficacy of the product.

The terms and conditions include (but are not limited to) the requirements listed below.

With respect to information on clinical and non-clinical studies, the sponsor is required to submit the following when the data become available:

  • Safety data from Studies 2019nCoV-301 and 2019nCoV-302 for 6 months post initial vaccination.
  • Results from the blinded crossover period of Study 2019nCoV-301, including safety and efficacy data for 24 months after the second dose.
  • Results from the blinded crossover period of Study 2019nCoV-302, including safety and efficacy data for 12 months after the second dose.
  • Safety, efficacy, and immunogenicity results from the adolescent participants of Study 2019nCoV-301.
  • Data regarding protection against current and emerging variants of concern.

Additionally, the sponsor is required to:

  • Update the Product Monograph with information on the pregnancy registry.
  • Submit prompt reporting of adverse reactions to the Canada Vigilance Program.
  • Submit Monthly Safety Summary Reports for the first 6 months of marketing of Nuvaxovid in Canada.
  • Submit Periodic Safety Update Reports/Periodic Benefit Risk Evaluation Reports every 6 months.
  • Submit an updated Core Risk Management Plan with the Canadian Addendum in a timely manner if a safety issue is identified that requires immediate regulatory action or as requested by Health Canada.
  • Submit final snapshots of all components of the electronic platform containing the approved Canadian-specific information for Nuvaxovid in French and English for Health Canada's review and records, prior to launch of the electronic platform.
  • Develop Canadian-specific bilingual labelling for Nuvaxovid and implement such labelling once supplies are transitioned to Canadian-dedicated supplies. Health Canada should be kept informed of estimated timelines and proposed strategies concerning the development and implementation of Canadian-specific bilingual labels.
  • Provide data to support the approval of the new master reference standard and the first working reference standard for the relative potency assay.
  • Provide data to support the approval of the use of Matrix-C batches manufactured at AGC-SEA for drug product formulation.
  • Provide updates from on-going stability studies for drug substance, drug product and adjuvant lots manufactured using commercial processes at the approved facilities.
  • Provide final reports/summaries of shipping qualification studies to support transportation of drug product lots and of adjuvant to the drug product manufacturing site.
  • Provide results from ongoing extractables and leachables studies for the container closure systems of drug product and adjuvant.
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

The New Drug Submission for Nuvaxovid (severe acute respiratory syndrome coronavirus 2 [SARS‑CoV‑2] recombinant spike protein) was submitted and reviewed in accordance with the Food and Drug Regulations, which permitted a rolling submission and review process. Following review of the provided information, a Notice of Compliance was issued in relation to Nuvaxovid, with accompanying terms and conditions to manage any uncertainties or mitigate risks related to the drug.

As described above, the clinical review of the New Drug Submission for Nuvaxovid was conducted as per Method 3 described in the Draft Guidance Document: The Use of Foreign Reviews by Health Canada.

The clinical efficacy, immunogenicity, and safety of Nuvaxovid were primarily evaluated using the available data from two ongoing pivotal, placebo-controlled, Phase III studies: Study 2019nCoV-301 conducted in the United States and Mexico, and Study 2019nCoV-302 conducted in the United Kingdom. In addition, supportive data were also evaluated from four ongoing clinical studies: Study 2019nCoV-101 (Part 1), Study 2019nCoV-101 (Part 2), Study 2019nCoV-501, and Study ICMR/SII-COVOVAX. These studies were conducted in Australia, South Africa, the United States, and India. The Nuvaxovid vaccine used in clinical studies contained the recombinant SARS-CoV-2 spike protein derived from the original (Wuhan) strain of SARS-CoV-2.

Clinical Pharmacology

Nuvaxovid is a sterile, preservative-free, aqueous buffered suspension of the SARS-CoV-2 recombinant spike (rS) protein. It is co-formulated with Matrix-M adjuvant and a formulation buffer. The SARS-CoV-2 rS protein in Nuvaxovid is produced in the Spodoptera frugiperda insect cell line infected with a baculovirus that encodes full-length, SARS-CoV-2 spike gene-producing trimeric spike proteins from the original (Wuhan) strain. The Matrix-M adjuvant contains Quillaja saponaria saponins fraction A and fraction C. The addition of the saponin-based Matrix-M adjuvant facilitates activation of the cells of the innate immune system, which enhances the magnitude of the spike protein-specific immune response. The vaccine elicits both humoral and cellular immune responses via B- and T‑cell immune responses to the spike protein, including neutralizing antibodies, which may contribute to protection against coronavirus disease 2019 (COVID-19).

Pharmacokinetic studies to demonstrate absorption, distribution, metabolism, and excretion of SARS-CoV-2 recombinant spike protein were not conducted and are typically not required for vaccines.

Immunogenicity was assessed as part of the clinical efficacy evaluation of Nuvaxovid.

For further details, please refer to the Nuvaxovid Product Monograph, approved by Health Canada and available through the Drug Product Database and on the Health Canada COVID‑19 vaccines and treatments portal.

Clinical Efficacy

The efficacy of Nuvaxovid was evaluated in three clinical studies. The Phase IIa/b Study 2019nCoV-501 served as a proof of concept study for efficacy. Vaccine efficacy was subsequently evaluated in the two pivotal, placebo-controlled, Phase III studies: Study 2019nCoV-301 and Study 2019nCoV-302. All three studies were ongoing at the time of Nuvaxovid authorization.

The primary efficacy endpoint for all studies was the vaccine efficacy of Nuvaxovid against the first episode of laboratory‑confirmed (polymerase‑chain reaction [PCR]-positive) symptomatic mild, moderate, or severe COVID-19 (with minor differences in the definitions across studies) with an onset at least 7 days following the administration of Dose 2 in participants seronegative for SARS-CoV-2 infection at baseline. For the pivotal studies, the vaccine efficacy success criteria for the primary endpoint as predefined by the sponsor, was a vaccine efficacy rate with a lower bound of the 95% confidence interval (LBCI) greater than 30%. Health Canada's vaccine efficacy success criteria as defined in the Guidance for Market Authorization Requirements for COVID-19 Vaccines is a target threshold of at least 50% vaccine efficacy, with an LBCI above 30%. Although there were some differences in study design, the efficacy analysis population (referred to as the Per-Protocol Efficacy [PP-EFF] analysis set) for all studies included baseline seronegative participants who received both doses of either Nuvaxovid or placebo (Dose 1 on Day 0 and Dose 2 between Days 21 and 28) who did not experience an exclusionary protocol deviation, and did not have evidence of SARS-CoV-2 infection through 6 days after Dose 2.

Both pivotal studies excluded participants who were significantly immunocompromised, pregnant, or who had a history of laboratory-confirmed COVID-19. Participants with clinically stable underlying comorbidity were included in both studies, as were participants with well-controlled human immunodeficiency virus (HIV) infection. It is noteworthy that the data provided from both of the pivotal studies were collected prior to the emergence of the Delta and Omicron variants.

Pivotal Efficacy Studies

Study 2019nCoV-301

Study 2019nCoV-301 was a Phase III, multicentre, randomized, observer-blinded, placebo-controlled study conducted in participants 18 years of age and older in the United States and Mexico. Upon enrolment, participants were stratified by age (18 to 64 years or 65 years and older) and were randomized in a 2:1 ratio to receive two intramuscular injections of Nuvaxovid or placebo, 21 days apart.

The enrolment of adult participants for Study 2019nCoV-301 was completed on February 18, 2021. Following the collection of sufficient efficacy and safety data to support the application, participants were permitted to be re-vaccinated with two injections of the alternative study product to what they initially received (i.e. those who received Nuvaxovid could receive placebo and those who received placebo could receive Nuvaxovid) 21 days apart. This period, which commenced on April 20, 2021, was referred to as the blinded crossover period. Safety and efficacy events were evaluated until each participant’s first blinded crossover vaccination or as of the data cut-off date of May 31, 2021. Participants are to be followed for assessments of safety and efficacy against COVID-19 for up to 24 months after the second dose.

The demographic and baseline characteristics were balanced amongst participants who received Nuvaxovid or placebo. Of the 29,949 participants randomized, 15.1% of participants in the Nuvaxovid group and 23.3% of participants in the placebo group requested unblinding to receive an authorized COVID-19 vaccine. In the PP-EFF analysis set, Nuvaxovid recipients (total number [n] = 17,312) had a median age of 47 years (range: 18 to 95 years), 88% were 18 to 64 years of age, 12% were aged 65 and older, and 48% were female. In the Nuvaxovid group, 94% of recipients were from the United States and 6% were from Mexico. Most of the Nuvaxovid recipients (76%) were White, 11% were Black or African American, 6% were American Indian (including Native Americans) or Alaskan Native, and 4% were Asian. Of all Nuvaxovid recipients, 22% were Hispanic or Latino. In 16,493 (95%) of the Nuvaxovid recipients, at least one pre-existing comorbidity or lifestyle characteristic associated with an increased risk of severe COVID-19 was present. Comorbidities included: obesity (body mass index [BMI] ≥30 kg/m2); chronic lung disease; diabetes mellitus type II, cardiovascular disease; chronic kidney disease; or HIV. Other high-risk characteristics included an age of 65 years or older (with or without comorbidities) or an age of less than 65 years with comorbidities and/or living or working conditions involving known frequent exposure to SARS-CoV-2 or to densely populated circumstances.

As of the data cut-off date of May 31, 2021, the primary efficacy analysis population (referred to as the PP-EFF analysis set) included 25,452 participants who received two doses of either Nuvaxovid (n = 17,312) or placebo (n = 8,140) as two doses, 21 to 28 days apart. Participants in this analysis set did not experience an exclusionary protocol deviation, and did not have evidence of SARS‑CoV‑2 infection through 6 days after the second dose. Cases of COVID-19 were confirmed by PCR through a central laboratory.

As of the data cut-off point, 14 cases of COVID-19 were reported in Nuvaxovid recipients and 63 cases were reported in the placebo recipients from 7 days onward following the administration of Dose 2. The vaccine efficacy of Nuvaxovid to prevent the onset of COVID-19 from 7 days following Dose 2 was estimated to be 90.4% (95% Confidence Interval [CI]: 82.9, 94.6). Based on this outcome, Nuvaxovid met the study's primary efficacy endpoint criterion for success with an LBCI greater than 30%. In addition, Nuvaxovid met Health Canada's required threshold of at least 50% vaccine efficacy, with an LBCI greater than 30%.

In the PP-EFF analysis set, there were no cases of moderate or severe COVID-19 reported in the 17,312 Nuvaxovid participants compared with 10 cases of moderate COVID-19 and 4 cases of severe COVID-19 reported in the 8,140 placebo recipients.

The efficacy results for Study 2019nCoV-301 reflect enrolment that occurred during a time period when strains classified as variants of concern or variants of interest were circulating in the Unites States and Mexico. Based on exploratory sequencing conducted in the study, the Alpha variant made up the majority of the cases (31 of the 61 cases for which these data were available).

Study 2019nCoV-302

Study 2019nCoV-302 was a Phase III, multicentre, randomized, observer-blinded, placebo-controlled study conducted in participants 18 to 84 years of age in the United Kingdom. Upon enrolment, participants were stratified by age (18 to 64 years or 65 to 84 years) and were randomized in a 1:1 ratio to receive two intramuscular injections of Nuvaxovid or placebo, 21 days apart.

Enrolment for Study 2019nCoV-302 was completed on November 28, 2020. The data cut-off dates for efficacy and safety were January 29, 2021 and February 23, 2021, respectively. Participants are to be followed for assessments of safety and efficacy against COVID-19 for up to 12 months after the last vaccination.

The demographic and baseline characteristics were balanced amongst participants who received Nuvaxovid or placebo. Of the 15,187 participants randomized, 33.8% of participants in the Nuvaxovid group and 35.4% of participants in the placebo group requested to receive an authorized COVID-19 vaccine. In the PP-EFF analysis set, Nuvaxovid recipients had a median age of 56 years (range: 18 to 84 years), 72% were 18 to 64 years old, 28% were aged 65 to 84, and 49% were female. The majority (95%) of Nuvaxovid recipients were White, 3% were Asian, 1.0% were multiple races, and 0.4% were Black or African American. Of all Nuvaxovid recipients, 0.9% were Hispanic or Latino. Forty-five percent (45%) of Nuvaxovid recipients had at least one comorbid condition.

As of the data cut-off date of January 29, 2021, the PP-EFF analysis set included 14,039 participants who received two doses of either Nuvaxovid (n = 7,020) or placebo (n = 7,019), 21 to 28 days apart. None of the participants experienced an exclusionary protocol deviation and none had evidence of SARS‑CoV‑2 infection through 6 days after the second dose. Cases of COVID-19 were confirmed by PCR through a central laboratory.

As of the data cut-off point, 10 cases of COVID-19 were reported in Nuvaxovid recipients and 96 cases in placebo recipients from 7 days onward following the administration of Dose 2. The vaccine efficacy of Nuvaxovid was estimated to be 89.7% (95% CI: 80.2, 94.6) for prevention of the onset of COVID-19 from 7 days following Dose 2. Based on this outcome, Nuvaxovid met the study's primary efficacy endpoint criterion for success with an LBCI greater than 30%. In addition, Nuvaxovid met Health Canada's required threshold of at least 50% vaccine efficacy, with an LBCI greater than 30%.

Nine cases of moderate COVID‑19 and no cases of severe COVID-19 were reported in the 7,020 Nuvaxovid recipients compared with 63 cases of moderate COVID‑19 and 5 cases of severe COVID-19 reported in the 7,019 placebo recipients in the PP-EFF analysis set. These efficacy results reflect enrolment that occurred during a time period when strains classified as variants of concern or variants of interest were circulating in the United Kingdom. Based on exploratory sequencing conducted in the study, the Alpha variant made up the majority of the cases (66 out of the 95 cases for which these data were available).

Supportive Efficacy

Study 2019nCoV‑501

Study 2019nCoV-501 was a Phase IIa/b, multicentre, randomized, observer-blinded, placebo-controlled study conducted in South Africa. The study enrolled HIV-negative participants who were 18 to 84 years of age as well as medically stable HIV-positive participants 18 to 64 years of age. As of the data cut-off date of February 23, 2021, the PP-EFF analysis set included 2,770 participants who had received two doses of either Nuvaxovid (n = 1,408) or placebo (n = 1,362), 21 days apart.

Demographic and baseline characteristics were balanced amongst participants who received Nuvaxovid or placebo. In the PP-EFF analysis set, for participants who received Nuvaxovid, the median age was 28 years (range: 18 to 84 years); 40% were female; 91% were Black or African American, 2% were White, 3% were multiple races, and 1% were Asian; 2% were Hispanic or Latino; and 5.5% were HIV-positive.

As of the data cut-off point, there were 51 cases of COVID-19 reported in Nuvaxovid recipients and 96 cases in placebo recipients from 7 days onward following the administration of Dose 2. The vaccine efficacy of Nuvaxovid was 48.6% (95% CI: 28.4, 63.1) for prevention of the onset of COVID-19 from 7 days after Dose 2. Nuvaxovid met the study's primary efficacy endpoint criterion for success with an LBCI >0%, thereby demonstrating proof of concept of efficacy.

These efficacy results reflect enrolment that occurred during a time period when strains classified as variants of concern or variants of interest were circulating in South Africa. Based on exploratory DNA sequence analysis conducted in the study, the Beta variant made up the majority of the cases (38 out of the 41 cases for which these data were available).

Immunogenicity

The immunogenicity of Nuvaxovid was primarily evaluated in the pivotal studies 2019nCoV-301 and Study 2019nCoV-302, along with Study 2019nCoV-501, and Study ICMR/SII-COVOVAX. In all four studies, blood samples for serologic assessments were collected from all enrolled participants before vaccination, during the immediate period following the first set of vaccinations (Day 0, 21 and 35), and as planned long-term follow-up (Months 12, 18 and 24). Based on the specifics of each study, amounts of anti-spike protein immunoglobulin G (IgG) were measured and compared to amounts on Day 0 using geometric mean titers (GMTs) reported as geometric mean enzyme‑linked immunosorbent assay (ELISA) units (GMEUs), geometric mean fold rises (GMFRs), and seroconversion rates (SCRs), or the percentage of subjects with a postvaccination titer ≥4-fold over baseline and ≥2-fold over a pre-existing titer. Similarly, neutralization antibody levels were measured from baseline at Day 0 comparing the GMFRs of active vaccine to placebo, as well as corresponding SCRs.

Study 2019nCoV-301

In the pivotal Study 2019nCoV-301, the immunogenicity analysis was conducted in a subset of 1,200 randomly selected participants following the initial set of vaccinations. This subset was intentionally over-selected for seniors by selecting near equally from the two main age categories (49.4% being 18 to <65 years of age, and 50.6% being ≥65 years of age). Therefore, this subset was not representative of the study population analyzed for efficacy in the PP-EFF analysis, but was useful for exploring the effect of age on the immune response of Nuvaxovid.

In participants who were SARS-CoV-2 seronegative at baseline, serum IgG antibody GMEUs in the Nuvaxovid group were approximately 1.7-fold higher in the younger cohort than in the older cohort at Day 35 (14 days after Dose 2). However, SCRs in the Nuvaxovid groups were markedly increased relative to placebo across all age groups (97.3% vs. 4.5% for participants ≥18 years of age; 98.0% vs 3.7% for 18 to <65 years of age; and 96.6% vs 5.3% for ≥65 years of age). Similar results were also seen for human angiotensin-converting enzyme 2 (hACE2) receptor binding inhibition antibodies and neutralizing antibodies.

Study 2019nCoV-302

In the pivotal Study 2019nCoV-302, the per-protocol immunogenicity (PP-IMM) anti-spike protein serology subset included 831 participants (5.5%), with 414 in the Nuvaxovid group and 417 in the placebo group. The PP-IMM neutralization assay subset included 761 (5.0%) participants, with 381 in the Nuvaxovid group and 380 in the placebo group.

Two weeks following Dose 2 (Day 35), serum GMTs of the anti-spike protein IgG in the Nuvaxovid group were increased approximately 300- to 400-fold relative to the placebo group across all age groups (46,679.3 endotoxin units [EU]/mL vs. 129.5 EU/mL for participants 18 to 84 years of age). Serum GMTs of the anti-spike protein IgG in the Nuvaxovid group were approximately 1.4-fold higher in the younger cohort (18 to 64 years) than in the older cohort (65 to 84 years). At Day 35, neutralizing antibody levels were also increased in Nuvaxovid recipients relative to placebo recipients, regardless of baseline serostatus, across all age groups: 18 to 84 years, 18 to 64 years, and 65 to 84 years. In most participants, neutralizing antibody GMTs in the Nuvaxovid group were markedly increased approximately 80- to 120-fold relative to the placebo group across all age groups (1214.6 vs 11.3 for participants 18 to 84 years of age) at 2 weeks following Dose 2 (Day 35). Neutralizing antibody GMTs in the Nuvaxovid group were approximately 1.4-fold higher in the younger cohort (18 to 64 years) than in the older cohort (65 to 84 years). This finding is consistent with findings from Study 2019nCoV-301.

Study 2019nCoV-501

In the supportive Study 2019nCoV-501, immune responses following Dose 2 of Nuvaxovid were markedly higher compared with those following Dose 1, regardless of HIV status or SARS-CoV-2 serostatus at baseline. In HIV-negative and HIV-positive serologically naïve participants, GMFRs for anti-spike protein IgG antibodies following Dose 2 were 283.7 at Day 35 relative to 123.0 at Day 0. Following Dose 1, these values were 10.7 at Day 21 relative to 4.4 at Day 0. Among participants who were SARS-CoV-2 seronegative at baseline, anti-spike protein IgG antibody responses at Day 35 were approximately 2-fold higher for HIV-negative participants than those for HIV-positive participants. However, for participants who were seropositive at baseline, anti-spike protein IgG antibody responses at Day 35 were comparable between HIV-negative and HIV-positive participants. Regardless of HIV status, anti-spike protein IgG antibody responses at Day 35 were markedly higher in SARS-CoV-2 seropositive participants compared with seronegative participants, which is indicative of cell-mediated immune memory.

Neutralizing antibody responses against wild-type virus showed a similarly robust pattern of induction at Day 35 (14 days after second vaccination) to those seen for anti-spike protein IgG antibodies. At Day 35, the GMFR in neutralizing antibodies were measured for all participants (stratified by serostatus: SARS‑CoV‑2 seronegative or seropositive at baseline). In HIV‑negative participants, 53.4‑ to 70.4‑fold increases in the GMFR were measured in those who received Nuvaxovid, and 1.1‑ to 1.2‑fold increases were measured in those who received the placebo. In HIV‑positive participants, 30.6‑ to 36.9‑fold increases in the GMFR were measured in those who received Nuvaxovid, and 0.9‑ to 1.2‑fold increases were measured in those who received the placebo.

In HIV-negative participants, seroconversion rates were 97.1% and 97.4% for baseline SARS-CoV-2 seronegative and seropositive individuals, respectively. In stable HIV-positive participants, seroconversion rates were 98.4% and 92.3% for baseline SARS-CoV-2 seronegative and seropositive individuals, respectively. Among baseline SARS-CoV-2 seronegative participants, neutralizing antibody responses were two-fold higher in HIV-negative participants compared with stable HIV-positive participants. However, responses were comparable among baseline SARS-CoV-2 seropositive participants, regardless of HIV status. Neutralizing antibody responses at Day 35 were markedly higher in SARS-CoV-2 seropositive compared to seronegative participants, regardless of HIV status. Taken together, prior exposure to COVID-19 infection or another COVID-19 vaccine may improve immune response to this vaccine regardless of HIV status.

ICMR/SII-COVOVAX Study

The ICMR/SII-COVOVAX study was primarily conducted as an immunobridging Phase II/III noninferiority study comparing the Novavax vaccine produced in the United States with a future commercialized vaccine to be mass produced in India under the domestic brand of Covovax (a hybrid United States drug substance/India drug product).

The Phase III portion of the study randomized 1,400 participants. A total of 1,396 (99.7%) participants received Dose 1 of a study vaccine (Covovax or Novavax vaccine), and 1,375 (98.2%) received Dose 2 with 1,361 (97.2%) completing Day 36 follow-up. Of these participants, 458 (32.7%) were enrolled in the Immunogenicity and Reactogenicity Cohort and were randomized 3:1 to receive Dose 1 of Covovax or Novavax vaccine. A total of 452 of 458 (98.7%) participants received Dose 2 with 449 of 458 (98%) participants providing blood samples for immunogenicity assessments on Day 36 (Visit 3). Although the median age was 33 to 34 years of age within the immunogenicity cohort, demographics and baseline characteristics were well-balanced relative to the general population in India with baseline SARS-CoV-2 seropositivity being about 34% in both groups.

Baseline GMEUs of anti-spike protein IgG antibodies were comparable between the groups. For both groups, the rise in GMEUs (GMFR) of anti-spike protein IgG antibodies was higher than 17-fold (17.7 for Covovax and 20.3 for Novovax) at Day 22 after Dose 1, and higher than 65-fold (65.6 for Covovax, 88.6 for Novovax) at Day 36 (14 days after Dose 2). The immunogenicity of Covovax was assessed based on the primary endpoint of the ratio of GMEUs of anti‑spike protein IgG antibodies, with a prespecified requirement of a noninferiority margin of 0.67 for the lower bound of the 95% CI for Day 36. The primary immunogenicity endpoint was met as the GMEU ratio between the Covovax and Novavax vaccine products at Day 36 was 0.92 (95% CI [0.79, 1.06]) with the lower bound of the 95% CI (0.79) exceeding the noninferiority margin of 0.67. Similar GMFR and GMEU ratio findings were noted for neutralizing antibodies measured by a microneutralization assay using wild type SARS CoV-2.

Taken together, Nuvaxovid (and the Covovax version) appear to provide a robust humoral immune response (both binding and neutralizing antibodies against SARS-CoV-2 spike protein) following Dose 2. These immune responses were observed regardless of SARS-CoV-2 serostatus, age, or HIV status, though the response may be mildly attenuated in older age groups or in stable-HIV-positive patients. However, this may not be a clinically meaningful difference. Duration of immunity in the post-marketing period will require additional larger scale immunogenicity studies to determine the need for, and spacing of, future booster doses of Nuvaxovid.

Indication

The New Drug Submission for Nuvaxovid was filed by the sponsor with the following indication, which Health Canada subsequently approved:

  • Nuvaxovid (COVID-19 Vaccine [recombinant protein, adjuvanted]) is indicated for active immunization to prevent coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in individuals 18 years of age and older.

For more information, refer to the Nuvaxovid Product Monograph, approved by Health Canada and available through the Drug Product Database and on the Health Canada COVID‑19 vaccines and treatments portal.

Clinical Safety

The evidence of safety for Nuvaxovid was primarily based on pooled data from the two pivotal Studies 2019nCoV-301 and 2019nCoV-302, and the supportive Study 2019nCoV-501, described in the Clinical Efficacy section above.

At the time of the analysis, a total of 48,698 participants 18 years of age and older received at least one dose of Nuvaxovid (n = 29,297) or placebo (n = 19,401). The median duration of follow-up was 70 days after Dose 2, with 32,993 (66%) participants completing more than 2 months of follow-up. At the time of vaccination, the median age of participants who received Nuvaxovid was 48 years (range 18 to 95 years): 84.1% of participants were between 18 and 64 years of age and 15.9% of participants were 65 years of age or older.

Reactogenicity

Solicited adverse events (AEs) were consistently collected and pooled across the three studies but excluded data from the 2019nCoV-302 substudy. Adverse events that occurred at the injection site were recorded as local reactogenicity. These AEs included pain, tenderness, erythema/redness, and swelling/induration. Adverse events that were more generalized and occurred at other sites throughout the body were recorded as systemic reactogenicity. These AEs included fever, nausea/vomiting, headache, fatigue/malaise, myalgia/muscle pain, and arthralgia/joint pain. Any reactions that persisted for more than 7 days after vaccination were also recorded as a treatment‑emergent adverse event (TEAE) starting on the same day as the reactogenicity event. There was a general trend of increased local and general reactogenicity within the Nuvaxovid versus (vs.) placebo groups, especially after Dose 2. However, these differences were attenuated in the stable HIV-positive subset for tenderness, erythema, and for swelling in the HIV-positive subset in Study 2019nCoV-501.

The pooled reactogenicity data included participants 18 years of age and older who received at least one dose of Nuvaxovid (n = 21,395) or placebo (n = 12,197). Overall, 21,395 pooled participants were assessed for local reactogenicity after Dose 1 of Nuvaxovid and 20,259 after Dose 2. From the placebo group, there were 12,197 pooled participants studied after Dose 1 and 11,434 after Dose 2. The rates for any local AE of any grade temporally associated with Nuvaxovid administration were 57.4% after Dose 1 and 75.4% after Dose 2 in participants 18 to 64 years of age (younger cohort). Corresponding rates for placebo recipients in the younger cohort were 20.3% after Dose 1 and 19.8% after Dose 2. The rates for any local AE of any grade temporally associated with Nuvaxovid administration were 37.3% after Dose 1 and 60.0% after Dose 2 in participants 65 years of age and older (older cohort). Corresponding rates for placebo recipients in the older cohort were 14.4% after Dose 1 and 14.4% after Dose 2. Following Dose 2, the most frequently reported solicited local AEs (in Nuvaxovid and placebo recipients, respectively) were: tenderness at the injection site (69.3% vs. 14.4% in the younger cohort and 54.9% vs. 9.6% in the older cohort), followed by pain (57.7.0% vs. 12.7% in the younger cohort and 40.5% vs. 9.5% in the older cohort), erythema (6.3% vs. 0.3% in the younger cohort and 5.3% vs. 0.3% in the older cohort) and swelling (5.8% vs. 0.3% in the younger cohort and 5.7% vs. 0.6% in the older cohort).

Overall, reports of solicited systemic adverse reactions increased with successive doses of Nuvaxovid, but not with placebo. Similar to local reactogenicity, there was an increase of Grade 3 or higher AEs in the Nuvaxovid group but not the placebo group. In the younger cohort, the rate for any systemic AE of any grade temporally associated with Nuvaxovid administration was 47.5% after Dose 1 and 66.5% after Dose 2. Corresponding rates for placebo recipients for the younger cohort were 37.9% and 32.7%, respectively. In the older cohort, the rates for any systemic AE of any grade temporally associated with Nuvaxovid administration were 31.7% after Dose 1 and 47.0% after Dose 2. Corresponding rates for placebo recipients in the older cohort were 29.4% and 25.4%, respectively. The most frequently reported solicited systemic AEs after Dose 2 of Nuvaxovid (vs. placebo) were: fatigue (46.8% [vs. 19.0%] in the younger cohort and 28.6% [vs 13.9%] in the older cohort), muscle pain/myalgia (46.0% [vs. 10.7%] in the younger cohort and 26.4% [vs. 9.4%] in the older cohort), headache (43.3% [vs. 18.5%] in the younger cohort and 23.6% [vs. 12.8%] in the older cohort), malaise (36.9% [vs. 10.7%] in the younger cohort and 21.0% [vs. 8.3%] in the older cohort), joint pain/arthralgia (21.5% [vs. 6.5%] in the younger cohort and 12.5% [vs. 5.6%] in the older cohort), followed by nausea/vomiting (11.3% [vs. 5.2%] in the younger cohort and 5.1% [vs. 3.3%] in the older cohort) and fever (5.7% [vs. 0.5%] in the younger cohort and 1.9% [vs. 0.9%] in the older cohort). Similar to local reactogenicity, the younger cohort had higher relative rates of systemic AEs compared with the older cohort.

Unsolicited Adverse Events

Data regarding unsolicited AEs were collected from Day 0 to 49 (28 days following Dose 2 of the vaccine) across the three studies and were presented as incidence rates (IR) of events per 100 person-years (e/100 PY). The data strongly reflected the above solicited reactogenicity but extended beyond the first 7 days. This was illustrated in the System Organ Class (SOC) of General Disorders and Administrative Site Conditions, where higher incidence rates were observed for Nuvaxovid recipients in subcategories such as injection site pain followed by fatigue, pyrexia, and chills. Similarly, higher incident rates were observed in Nuvaxovid recipients in the SOCs of Musculoskeletal and Connective Tissue Disorders (e.g., myalgia and pain in an extremity), and Nervous System Disorder (e.g., headaches). Unsolicited AEs occurring in other SOCs and their corresponding subcategories were substantially less common, and interpretation was indeterminate.

The same pattern was seen for unsolicited severe AEs (Grade 3 or higher) unsolicited AEs during the period from Day 0 to 28 days following Dose 2 of Nuvaxovid, controlling for background placebo events. The reported unsolicited severe AEs include: fatigue (8% and 8% in the older and younger group, respectively); injection site pain (3% and 4%); malaise (4% and 2%); pyrexia (2% and 4%); headaches (4% and 3%); myalgia (4% and 4%); and hypertension (2% and 9%).

Unsolicited serious adverse events (SAEs) were reported from Day 0 to the end of follow-up for all events. These included events clearly unrelated to vaccination. In the pooled analysis, the SOC of Infections and Infestations had the highest event counts and incident rates in participants receiving Nuvaxovid and placebo (46 events and an incident rate of 0.63 e/100 PY for Nuvaxovid; 55 events and an incident rate of 1.16 e/100 PY for placebo). The majority of the events in both groups were associated with COVID-19, COVID-19 pneumonia, pneumonia, and appendicitis, with all incidence rates higher in the placebo group.

In the younger cohort, there were no SAEs with an incidence rate greater than 0.10 e/100 PY in the Nuvaxovid group while three events had incidence rates greater than 0.10 e/100 PY in the placebo group: COVID-19 pneumonia (0.25 e/100 PY), COVID-19 (0.23 e/100 PY), and appendicitis (0.15 e/100 PY). In the older cohort, SAEs that occurred at an incidence rate greater than 0.20 e/100 PY were COVID-19 (0.37 e/100 PY) and prostate cancer (0.28 e/100 PY) in participants who received Nuvaxovid, and pneumonia (0.51 e/100 PY), COVID-19 (0.26 e/100 PY), COVID 19 pneumonia (0.26 e/100 PY), and atrial fibrillation (0.26 e/100 PY) in participants who received the placebo.

Overall, there were more potential immune-mediated medical conditions in the Nuvaxovid group (27 events with an incident rate of 0.37 e/100 PY) than placebo (8 events with an incident rate of 0.04 e/100 PY), as well as across the two age groups. The main SOCs of concern where the incident rates for the Nuvaxovid group were higher than placebo included Musculoskeletal and Connective Tissue Disorders (0.08 vs 0.04 e/100 PY) including the cluster of arthritis and polymyalgia rheumatica, Nervous System Disorders (0.08 vs. 0.04 e/100 PY) but with no clear clustering of conditions, Endocrine Disorders (0.06 vs. 0.00 e/100 PY) including a clustering around autoimmune thyroiditis (Grave’s disease), and Eye Disorders (0.06 vs. 0.00 e/100 PY) clustering around uveitis and iridocyclitis. Health Canada recommended these disease groupings be monitored in the post-marketing period.

Based on compelling case reports, autoimmune conditions that may warrant further review in the post-marketing period include: immune thrombocytopenic purpura (ITP), Bell’s palsy, Grave’s disease, uveitis, iridocyclitis, rheumatoid arthritis, psoriasis and psoriatic arthritis, polymyalgia rheumatica, and chilblains. Within this group, seven cases of myocarditis and/or pericarditis were also identified and carefully reviewed. Of these, two cases among male teenagers (19 and 16 years of age) were pathognomonic for vaccine-associated myocarditis already described with approved messenger ribonucleic acid (mRNA) vaccines. A statement is included within the Product Monograph for Nuvaxovid to better inform providers and patients of the potential risk of myocarditis pending additional post-marketing data.

Special Populations

Clinical studies of Nuvaxovid include participants 65 years of age and older and their data contribute to the overall assessment of safety and efficacy. The safety and efficacy of Nuvaxovid in individuals under 18 years of age have not yet been established. Use in pediatric subjects is included in the Risk Management Plan (RMP) as missing information.

The safety and efficacy of Nuvaxovid in pregnant women have not yet been established. Women who were pregnant or breastfeeding were excluded from the clinical studies. As of October 26, 2021, a total of 137 pregnancies were reported in the two pivotal studies (2019nCoV-301 and 2019nCoV-302): 95 in the vaccine group and 42 in the placebo group. Of the pregnancies in Nuvaxovid recipients, 8 resulted in live births, 11 were voluntarily terminated, 16 resulted in spontaneous abortion (miscarriage) and 60 were ongoing at the time of authorization. Due to the limited duration of follow-up, the outcome of the remaining pregnancies is pending.

It is unknown if Nuvaxovid is excreted in human milk. Data are not available to assess the effects of the vaccine on the breastfed infant or on milk production/excretion. Use in pregnancy and while breastfeeding is included in the RMP as missing information.

No data are currently available on significantly immunocompromised persons, including individuals taking immunosuppressant therapy. Immunocompromised persons may have a diminished immune response to the vaccine and use in immunocompromised patients is included in the RMP as missing (limited/ no clinical data) information.

Risk Management Plan

A Core (European Union) Risk Management Plan (RMP) and a Canadian RMP Addendum for Nuvaxovid were submitted by Novavax Inc. to Health Canada as part of the submission for authorization. The RMP is designed to describe known and potential safety issues, to present the monitoring plan, and when needed, to describe measures that will be put in place to minimize risks associated with the product.

The following information relates to the RMP submitted by Novavax Inc., as part of the New Drug Submission for Nuvaxovid. It is the sponsor’s responsibility to monitor the safety profile of this vaccine and to submit an update to the RMP if there is a significant change to the benefits, harms or uncertainties associated with this vaccine. Updates to the RMP will be reflected in the Post-Authorization Activity Table for Nuvaxovid.

Based on results from non-clinical and clinical studies, the RMP included no important identified risks and three important potential risks: vaccine‑associated enhanced disease, including vaccine‑associated enhanced respiratory disease; anaphylaxis; and myocarditis and pericarditis. There were eight areas of missing (limited/no clinical data) information identified: "use during pregnancy and while breastfeeding", "use in immunocompromised patients", "use in frail patients with comorbidities", "use in patients with autoimmune or inflammatory disorders", "interaction with other vaccines", "long-term safety", "use in pediatric subjects", and "long-term effectiveness". The proposed routine and additional pharmacovigilance activities for the above risks are considered to be acceptable and the sponsor confirmed that they would be applied in the Canadian context. Additional pharmacovigilance activities include continued safety surveillance of ongoing clinical studies as well as five planned post-authorization studies: two non-interventional safety studies, two non-interventional effectiveness studies, and one pregnancy exposure registry. Routine risk minimization measures (i.e., Product Monograph and labelling) are also considered to be appropriate. The sponsor is expected to provide an updated Core (European Union) RMP and Canadian Addendum in a timely manner if a signal of safety issue is identified in ongoing post-authorization surveillance.

In addition, in the post-market period the sponsor will provide information with respect to the following:

  • Older adults: Older adults were included in the clinical development program. The sponsor will submit monthly safety reports to Health Canada, which will include analyses of subpopulations by age and gender.
  • Children: Children were not included in the clinical development program. This subpopulation was identified as missing information in the RMP. The sponsor will submit monthly safety reports to Health Canada. Studies of Nuvaxovid in children are planned.
  • Pregnant women: More information is needed about the use of the vaccine for pregnant women. This subpopulation was identified as missing information in the RMP. The sponsor will monitor and submit monthly safety reports to Health Canada. The sponsor will assess outcomes in pregnancy and lactation as part of ongoing and planned studies, including a pregnancy exposure registry.
  • Breastfeeding women: This subpopulation was identified as missing information in the RMP. The sponsor will submit monthly safety reports to Health Canada. The sponsor will assess outcomes in pregnancy and lactation as part of ongoing and planned studies, including a pregnancy exposure registry.
  • Immunocompromised individuals and patients with chronic or debilitating conditions: These individuals were not included in the clinical development program. This subpopulation was identified as missing information in the RMP. The sponsor will submit monthly safety reports to Health Canada.
  • Anaphylaxis: The risk of anaphylaxis was identified as an important potential risk in the RMP. This risk will be assessed via routine pharmacovigilance activities, and reported in monthly safety reports.
  • Myocarditis and Pericarditis: The risk of myocarditis and pericarditis was identified as an important potential risk in the RMP. This risk will be assessed via routine pharmacovigilance activities, and reported in monthly safety reports.
  • Indigenous populations in Canada: Adverse drug reactions (ADRs) from all subpopulations, including Indigenous, will be reported as expeditiously as applicable, and will be assessed in the monthly safety reports.

The sponsor is required to promptly report adverse reactions and provide Monthly Safety Summary Reports. These reports will be reflected in the Post-Authorization Activity Table for Nuvaxovid.

Results related to safety and effectiveness from ongoing and planned studies will be submitted for review as they become available. Together, the results from these studies and the monthly safety summary reports are expected to address data gaps related to the long-term safety and effectiveness of the vaccine, interactions with other vaccines, and specific subpopulations (e.g., pregnant and breastfeeding women, pediatric populations younger than 18 years of age, patients with autoimmune or inflammatory disorders, immunocompromised patients and frail patients with comorbidities).

Collectively, the results of the clinical safety evaluation demonstrated that Nuvaxovid met the vaccine safety requirements as specified in Health Canada’s Guidance for Market Authorization Requirements for COVID-19 Vaccines. Overall, Nuvaxovid is a safe vaccine with some local and systemic reactogenicity associated with the immune response after the second dose. The authorization is accompanied by terms and conditions to manage uncertainties and mitigate risks related to the drug. Appropriate warnings and precautions are in place in the approved Nuvaxovid Product Monograph to address the identified risks.

For more information, refer to the Nuvaxovid Product Monograph, approved by Health Canada and available through the Drug Product Database and on the Health Canada COVID‑19 vaccines and treatments portal.

7.2 Non-Clinical Basis for Decision

The New Drug Submission for Nuvaxovid (SARS-CoV-2 recombinant spike [rS] protein) was submitted and reviewed in accordance with the Food and Drug Regulations, which permitted a rolling submission and review process. Following review of the provided information, a Notice of Compliance was issued in relation to Nuvaxovid, with accompanying terms and conditions to manage any uncertainties or mitigate risks related to the drug.

As described above, the review of the non-clinical component of the New Drug Submission for Nuvaxovid was conducted as per Method 3 described in the Draft Guidance Document: The Use of Foreign Reviews by Health Canada.

The non-clinical data submitted in the application for authorization of Nuvaxovid, known as SARS-CoV-2 rS with Matrix-M adjuvant, included a comprehensive pharmacology and toxicology program.

Non‑Clinical Pharmacology

The non-clinical pharmacology program in support of SARS‑CoV‑2 rS with Matrix‑M adjuvant included multiple immunogenicity studies that evaluated both humoral and/or cell-mediated immune responses in rodents and nonhuman primates (NHP). Several live virus challenge studies were also performed in mice, hamsters, and two NHP models (cynomolgus and rhesus macaques) to further evaluate the protective efficacy of the vaccine. Histopathological assessments of the lungs were also included as part of these viral challenge studies to evaluate the potential risk of vaccine‑associated enhanced disease (VAED).

The results from pharmacology studies showed that SARS‑CoV‑2 rS with Matrix‑M adjuvant induced high titer anti‑spike protein immunoglobulin G antibodies, as well as functional antibodies, including human angiotensin converting enzyme 2 (hACE2) binding inhibition and wild‑type virus neutralizing antibody responses, in multiple species (mice, hamsters, and NHP). These studies also showed that immune responses increased with the addition of the Matrix‑M adjuvant versus the SARS‑CoV‑2 rS antigen alone, and following a second vaccine administration versus a single dose. In NHP models, two doses corresponding to the recommended dose in humans of 5 or 25 mcg SARS‑CoV‑2 rS with 50 mcg Matrix‑M adjuvant resulted in maximal immune responses with no notable differences between the two antigen doses. In mice, administration of the SARS‑CoV‑2 rS with Matrix‑M adjuvant led to a T helper 1 (Th1)‑dominant response, as demonstrated by the induction of strong Th1‑type CD4+ T‑cell responses. These included multifunctional effector phenotypes (producing interferon gamma, tumor necrosis factor α, and interleukin‑2) that were generally induced at much higher levels than interleukin‑4‑producing (T helper 2 [Th2]) cells.

In baboons, SARS-CoV-2 rS with Matrix‑M adjuvant induced strong Th1‑dominant CD4+ T-cell responses, which included polyfunctional effector phenotypes, when administered at the recommended human dose level. Further, in rhesus macaques, immunization with SARS‑CoV‑2 rS with Matrix-M adjuvant elicited high levels of both spike protein‑specific and receptor‑binding domain‑specific antibodies. Changes in circulating immune cell abundance after vaccination were consistent with typical responses to a potent adjuvant, as well as recruitment of lymphocytes to lymphoid organs. When baboons received a second vaccine, B‑cell responses following the second dose were consistent with a rapid recall of memory B cells, T‑cell responses again indicating a Th1‑skewed response, and the presence of circulating T follicular helper (Tfh) cells, which suggests favourable conditions for a diverse, high‑affinity, and durable antibody response.

Live virus challenge studies were also performed in mice, hamsters, and two NHP models (cynomolgus and rhesus macaques) following either a single‑ or two‑dose vaccine schedule. Histopathological evaluations of the lungs were also conducted as part of these studies to assess the potential risk for VAED. In a murine challenge model in mice with respiratory tract hACE2 expression transduced via an adenovirus vector, SARS-CoV-2 rS with Matrix-M adjuvant provided protection against SARS-CoV-2 virus challenge with reduced SARS-CoV-2 virus titers in the lungs (2 to 4 log10 reduction), reduced lung inflammation, and reduced systemic morbidity. In addition, based on histopathological evaluation of the lungs following a live virus challenge, there was no evidence of vaccine‑associated enhanced respiratory disease (VAERD).

In the hamster, cynomolgus macaque, and rhesus macaque challenge models, SARS‑CoV‑2 rS with Matrix-M was immunogenic in all species by inducing high titers of anti‑spike protein immunoglobulin G antibodies, as well as functional hACE2 binding inhibiting and virus neutralizing antibodies. In the hamster challenge model, SARS‑CoV‑2 rS with Matrix‑M adjuvant conferred protection against the SARS‑CoV‑2 virus as evidenced by amelioration of body weight loss, protection from activity reduction, decreased evidence of viral replication, accelerated viral clearance, and protection against severe SARS‑CoV‑2‑induced lung histopathological changes compared with placebo.

In the cynomolgus and rhesus macaque challenge models, all animals vaccinated with SARS‑CoV‑2 rS with Matrix-M adjuvant (administered as either a single‑ or two‑dose schedule) were protected against the SARS‑CoV‑2 virus as evidenced by decreased or absent subgenomic ribonucleic acid in bronchoalveolar lavages and nasal swabs up to 7 days post‑challenge. While subgenomic ribonucleic acid may not be as evanescent as originally believed, its absence nonetheless clearly indicates suppression (or elimination) of new virus replication. These results indicate that animals vaccinated with Matrix-M adjuvanted SARS‑CoV‑2 rS, including the dose and regimen currently proposed for use in humans (i.e., two doses of 5 mcg SARS‑CoV‑2 rS + 50 mcg Matrix-M adjuvant administered 21 days apart), were protected from viral replication in the upper and lower respiratory tract following challenge with live SARS‑CoV‑2. Also, there was no evidence for the potential risk of VAED following exposure to SARS‑CoV‑2 in any of the studies.

Non‑Clinical Toxicology

The toxicology program in support of the SARS‑CoV‑2 rS with Matrix-M adjuvant included repeat‑dose toxicity, biodistribution, and developmental toxicity studies.

The toxicological potential of SARS‑CoV‑2 rS, with or without Matrix-M adjuvant, was evaluated in a repeat-dose toxicology study conducted in New Zealand White (NZW) rabbits. Intramuscular injections 50 mcg SARS‑CoV‑2 rS, with or without 50 µg Matrix-M adjuvant, were administered to the NZW rabbits up to 4 times (on Day 1, Day 8, Day 15, and Day 36). The results from the study showed that SARS-CoV-2 rS with Matrix‑M adjuvant was well‑tolerated with no adverse findings. Further, the effects on clinical pathology parameters (fibrinogen, C‑reactive protein, and/or globulin) and histopathology (subacute inflammation at injection sites and adjacent tissue) were consistent with immune stimulation following administration of a vaccine and decreased or resolved completely during the recovery interval.

In support of the toxicological safety of the Matrix-M adjuvant, the sponsor submitted seven repeat‑dose toxicity studies conducted in rats and rabbits. These studies were conducted with either the Matrix-M adjuvant alone or in combination with other nanoparticle vaccine antigens. The results from these studies showed that the Matrix-M adjuvant was well tolerated up to dose levels equal to the dose used in the repeat‑dose toxicity study (50 mcg in NZW rabbits) when used in combination with one or more antigens. However, these studies did not provide specific toxicological information on the Matrix‑M adjuvant with the SARS‑CoV‑2 rS antigen. The studies are therefore considered as supportive, since the formulation proposed for clinical use should also be the one used to demonstrate the safety of the adjuvant. However, the abovementioned repeat-dose toxicology study conducted in NZW rabbits addressed the toxicological impacts of the SARS‑CoV‑2 rS antigen in combination with the Matrix-M adjuvant.

A study evaluating the potential biodistribution of the Matrix-M adjuvant in male and female mice showed that the saponin‑labelled adjuvant was transiently observed mostly at the site of injection, draining lymph nodes, and in the urine. Very low levels were also observed in the spleen, kidney, liver, and plasma. The cholesterol‑labelled adjuvant was found in higher concentrations in most of the tissues, at later timepoints, which is consistent with the normal presence of cholesterol and its metabolites in all mammalian tissues. These observations are considered acceptable given there were no corresponding findings in the repeat‑dose studies in two species.

A developmental and reproductive toxicology study was conducted in rats. The SARS‑CoV‑2 rS, with or without Matrix‑M adjuvant, was not associated with adverse effects on mating or fertility performance, or neurodevelopmental or physical effects in offspring, when administered as four intramuscular vaccine doses spanning the mating period and gestation until gestational Day 15. Administration of SARS‑CoV‑2 rS with Matrix‑M adjuvant induced immunoglobulin G antibodies in all treated females and in pooled blood samples from fetuses and pups. In these animals, antibody titers increased to approximately 12,000 to 19,000 by study day 15, increased by ten-fold following the second dose on study day 28 to between 192,000 and 203,000, and persisted through to gestational day or post natal day 21. Mean immunoglobulin G titers on the order of 104 in fetuses terminated on gestational Day 21 and 105 in the first filial generation offspring of pups suggests that vaccine-induced immunoglobulin G antibodies were transferred to offspring.

In vitro genotoxicity studies were conducted with the Matrix‑M adjuvant. The adjuvant was shown to be non‑mutagenic in both the bacterial reverse mutation assay and mammalian cell micronucleus assay.

Overall, the non‑clinical pharmacology and toxicology profile of SARS‑CoV‑2 rS with Matrix‑M adjuvant supports the proposed clinical use of Nuvaxovid. The results of the non‑clinical studies as well as the potential risks to humans have been included in the Nuvaxovid Product Monograph. Considering the intended use of Nuvaxovid, there are no pharmacological or toxicological issues within this submission to preclude authorization of the product.

For more information, refer to the Nuvaxovid Product Monograph, approved by Health Canada and available through the Drug Product Database and on the Health Canada COVID‑19 vaccines and treatments portal.

7.3 Quality Basis for Decision

The New Drug Submission for Nuvaxovid (SARS-CoV-2 recombinant spike protein) was submitted and reviewed in accordance with the Food and Drug Regulations, which permitted a rolling submission and review process. Following review of the provided information, a Notice of Compliance was issued in relation to Nuvaxovid, with accompanying terms and conditions to manage any uncertainties or mitigate risks related to the drug.

As described above, the review of the quality component of the New Drug Submission for Nuvaxovid was conducted as per Method 3 described in the Draft Guidance Document: The Use of Foreign Reviews by Health Canada.

Nuvaxovid (also known as NVX‑CoV2373) is an adjuvanted, recombinant spike protein nanoparticle vaccine developed to prevent COVID‑19 caused by the severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2). The vaccine is composed of a purified, full-length SARS-CoV-2 recombinant spike (rS) protein derived from the original (Wuhan) strain which is co‑formulated with a saponin‑based Matrix‑M adjuvant. The Matrix‑M adjuvant is added to enhance the magnitude of the spike protein‑specific immune response. The two vaccine components, SARS‑CoV‑2 rS and the Matrix‑M adjuvant, elicit B‑ and T‑cell immune responses to the spike protein, including neutralizing antibodies, which may contribute to protection against COVID-19.

Characterization of the Drug Substance

The drug substance, SARS‑CoV‑2 rS, is a protein product of a recombinant SARS‑CoV‑2 spike gene encoding 1,260 amino acids of the spike protein (the full‑length 1,273 amino acid protein minus the signal peptide). The spike gene was optimized for expression in Spodoptera frugiperda (Sf9) insect cells from a full-length, prefusion, stabilised SARS‑CoV‑2 spike genetic sequence derived from the original (Wuhan) strain. A total of five amino acid changes were introduced in the spike gene, three in the S1/S2 furin cleavage site and two in the HR1 domain where two proline substitutions were inserted to stabilize the protein. Several detailed characterization studies were performed to ensure the drug substance consistently maintained the desired structure, stability, purity, and protein content.

The drug substance, SARS‑CoV‑2 rS, is co‑formulated with a novel saponin‑based adjuvant, Matrix‑M. The formulation of Matrix‑M consists of two individually formed nanoparticles (Matrix‑A and Matrix‑C), each with a different and well‑characterized saponin fraction (fraction A and fraction C) with complementary properties.

Manufacturing Process of the Drug Substance and Drug Product and Process Controls

The drug substance, SARS‑CoV‑2 rS, is a protein produced by using a recombinant baculovirus system. The SARS‑CoV‑2 rS protein is created in a Sf9 insect cell line, which is infected with a baculovirus encoding a modified SARS‑CoV‑2 spike protein derived from the original (Wuhan) strain. The manufacturing process begins with thawing, revival, and expansion of the Sf9 cells in shake flasks, followed by expansion in a fermentor/bioreactor using serum‑free medium. The Sf9 cells are infected with recombinant baculovirus expressing the modified SARS CoV 2 spike protein gene. Following infection, spike proteins are produced and expressed on the surface of the cells. Once mature, the spike proteins are harvested, purified and concentrated to produce the SARS‑CoV‑2 rS drug substance.

The novel Matrix‑M adjuvant is produced by mixing two components, Matrix-A and Matrix-C. Both components (Matrix‑A and Matrix‑C) are produced from purified saponin fractions (A and C) derived from saponin raw material that is extracted from the bark of Quillaja saponaria Molina trees. Cholesterol and phosphatidylcholine are present as excipients in the matrix solutions.

The drug product manufacturing process includes preparation of the formulation buffer, processing of the drug substance (SARS‑CoV‑2 rS) and adjuvant Matrix‑M components (Matrix‑A and Matrix‑C), combining the drug substance with the Matrix‑M adjuvant and adding the formulation buffer, blending, and final filling steps. The classification of process parameters and the predetermined acceptance criteria for process controls of the drug product are based on an understanding of the process, manufacturing experience and data obtained during product development, including the quality characteristics of clinical lots (Phase II/III).

As part of the overall process control strategy, the sponsor has determined critical quality attributes, material attributes, and process parameters. In‑process controls for the drug substance and drug product are based on scientifically relevant assays and appropriate specifications to monitor key quality attributes. The sponsor provided enough information to support the consistency of production. This information, together with additional characterization studies and the experience of the sponsor with other vaccines based on the same nanoparticle technology platform, is considered sufficient to support authorization with accompanying terms and conditions.

Nuvaxovid is a Schedule D (biologic) drug and is, therefore, subject to Health Canada's Lot Release Program before sale as per the Guidance for Sponsors: Lot Release Program for Schedule D (Biologic) Drugs.

Control of the Drug Substance and Drug Product

The drug substance and drug product are tested against suitable reference standards to verify that they meet approved specifications.

Analytical tests and process validation studies were conducted on the drug substance. Two different cell culture media were used during process validation. Process parameters and the results of routine quality control and non‑routine testing were provided for three process performance qualification lots each, manufactured using the two cell culture media, and the consistency of the manufacturing process is considered acceptable.

Process parameters and the results of routine quality control and non-routine testing (filling homogeneity, purity, potency, and SARS‑CoV‑2 rS content) for three process performance qualification lots were provided for the drug product. The consistency of the manufacturing process is considered acceptable. 

Analytical comparability studies conducted by the sponsor, along with immunogenicity data from a Phase II study in humans support the comparability of the commercial drug substance and drug product lots manufactured in India to clinical trial lots manufactured in the United States. Based on the totality of data provided to Health Canada, it was concluded that the clinical performance of the commercial lots is expected to be similar to that of the clinical trial lots.

In light of the favourable benefit-risk profile based on the clinical information for Nuvaxovid, and taking into account the public health need related to the COVID‑19 pandemic, Health Canada considered it acceptable to address the quality uncertainties as part of the terms and conditions associated with the authorization. The sponsor is required to address the quality‑specific comments and provide additional data as outlined in the terms and conditions.

Stability of the Drug Substance and Drug Product

Based on the stability data submitted, the proposed shelf life and storage conditions for the drug substance and drug product were adequately supported and are considered to be satisfactory. The stability data provided for the drug product support the proposed 9 month shelf life when stored at 2 °C to 8 °C and protected from light.

In‑use storage conditions are outlined in the Nuvaxovid Product Monograph. After the first dose has been withdrawn, the vial/filled syringe may be held at 2 °C to 25 °C for up to 6 hours after first puncture. If the vaccine is not used within this time, it must be discarded.

The stability profiles were based on the evaluation and monitoring of stability-indicating attributes. The analytical procedures used in the stability programs assess the composition, strength, purity, safety and general quality attributes of the drug product.

Further ongoing stability studies for the drug substance, drug product, and adjuvant lots manufactured at commercial scale will be submitted to Health Canada for review post‑authorization (see terms and conditions).

The proposed packaging and components are considered acceptable.

Facilities and Equipment

Travel restrictions due to the COVID-19 pandemic prevented Health Canada from conducting on‑site evaluations of the drug substance and drug product manufacturing facilities. The facilities involved in production are compliant with Good Manufacturing Practices.

Based on the information provided in the submission, the design, operations, and controls of the facilities and equipment involved in production are considered suitable for the activities and products manufactured.

Adventitious Agents Safety Evaluation

The manufacturing process of Nuvaxovid incorporates adequate control measures to prevent contamination and maintain microbial control. Controlled selection and appropriate specifications for raw materials and excipients, specifications, in-process controls and release testing for starting materials, the drug substance and drug product, assure adventitious agents safety for Nuvaxovid. Therefore, the overall control strategy for adventitious agents is considered acceptable.