Summary Basis of Decision for Ervebo

Review decision

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


Product type:

Drug

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

Recent Activity for Ervebo

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 Ervebo

Updated:

2023-03-24

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

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

Drug Identification Number (DIN):

DIN 02532603 – 72 million plaque forming units (pfu)/mL, Ebola Zaire Vaccine (rVSVΔG-ZEBOV-GP, live), solution, intramuscular injection

Post-Authorization Activity Table (PAAT)

Activity/submission type, control numberDate submittedDecision and dateSummary of activities
NC # 2700122022-11-23Issued NOL
2023-02-14

Submission filed as a Level II (90 day) Notifiable Change (Moderate Quality Changes) for changes to the drug product manufacturing process. The submission was reviewed and considered acceptable, and an NOL was issued.

NC # 2698772022-11-22Issued NOL
2023-02-14

Submission filed as a Level II (90 day) Notifiable Change (Moderate Quality Changes) to transfer drug product release testing to a new facility. The submission was reviewed and considered acceptable, and an NOL was issued.

NC # 2698692022-11-22Issued NOL
2022-12-08

Submission filed as a Level II (90 day) Notifiable Change (Moderate Quality Changes) to change the specifications used to release the drug substance. The submission was reviewed and considered acceptable, and an NOL was issued.

NDS # 2565682021-09-10Issued NOC
2022-11-09

NOC issued for New Drug Submission.

Summary Basis of Decision (SBD) for Ervebo

Date SBD issued: 2023-03-24

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

Ebola Zaire Vaccine (rVSVΔG-ZEBOV-GP, live)

Drug Identification Number (DIN):

  • DIN 02532603 - 72 million plaque forming units (pfu)/mL, Ebola Zaire Vaccine (rVSVΔG-ZEBOV-GP, live), solution, intramuscular injection

Merck Canada Inc.

New Drug Submission Control Number: 256568

On November 9, 2022, Health Canada issued a Notice of Compliance to Merck Canada Inc. for the vaccine Ervebo.

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 Ervebo is favourable for active immunization of individuals 18 years of age or older to protect against Ebola Virus Disease (EVD) caused by Zaire Ebola virus.

1 What was approved?

Ervebo, an active immunizing agent, was authorized for active immunization of individuals 18 years of age or older to protect against Ebola Virus Disease (EVD) caused by Zaire Ebola virus.

Based on the data submitted and reviewed by Health Canada, the safety, immunogenicity, and efficacy of Ervebo in children and adolescents (less than 18 years of age) have not yet been established. Therefore, Health Canada has not authorized an indication for pediatric use.

Across the clinical development program, 552 subjects 65 years of age or older received Ervebo. Clinical studies of Ervebo did not include sufficient numbers of subjects in this age group to determine whether geriatric subjects respond differently from younger adult subjects regarding safety and immunogenicity.

Ervebo (72 million plaque forming units/mL, Ebola Zaire Vaccine [rVSVΔG-ZEBOV-GP, live]) is presented as a solution. In addition to the medicinal ingredient, the solution contains hydrochloric acid and sodium hydroxide for pH adjustments, recombinant human serum albumin, trometamol buffer, and water for injection.

The use of Ervebo is contraindicated in patients who are hypersensitive to this vaccine or to any ingredient in the formulation, including any non-medicinal ingredient, rice protein, or component of the container.

The drug product was approved for use under the conditions stated in its Product Monograph taking into consideration the potential risks associated with its administration. The Ervebo Product Monograph is available through the Drug Product Database.

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 Ervebo approved?

Health Canada considers that the benefit-risk profile of Ervebo is favourable for active immunization of individuals 18 years of age or older to protect against Ebola Virus Disease (EVD) caused by Zaire Ebola virus (ZEBOV).

Ebola Virus Disease is a rare, severe, acute illness caused by viruses from the genus Ebolavirus. The Zaire Ebola virus is considered the most virulent strain of Ebolavirus, having the highest case fatality rate and being responsible for the majority of outbreaks to date. With an incubation period of 2 to 21 days, the symptom onset of EVD is around 4 to 10 days after exposure. Fatal cases will experience severe symptoms and succumb to the disease within 10 days of symptom onset. Symptoms include a sudden onset of flu-like symptoms, such as fever, myalgia, severe headache, and malaise, followed by gastrointestinal symptoms and fluid loss and haemorrhage as a late manifestation. Human case fatality ranges from 25% to 90%. Non-fatal cases typically begin recovery 6 to 11 days after symptom onset, with full recovery occurring over a long period of time and often associated with long-term sequelae. During recovery, Ebola virus can persist in some body fluids, such as semen, urine, and breast milk.

Prevention of EVD is currently accomplished through education on avoidance of risk factors and quarantine of infected individuals. On October 14, 2020, the United States Food and Drug Administration (FDA) approved Inmazeb (atoltivimab, maftivimab, and odesivimab-ebgn), a mixture of three monoclonal antibodies, as treatment for ZEBOV infection in adult and pediatric patients. On December 21, 2020, the United States FDA also approved Ebanga (Ansuvimab-zykl), a human monoclonal antibody, for the treatment for ZEBOV infection in adults and children. Treatment also includes supportive therapy, which is less than ideally effective with case fatality rates remaining high, even with intensive supportive care.

Ervebo is a genetically engineered, replication-competent, attenuated, live vaccine that induces immune responses following a single intramuscular administration of 72 million or more plaque forming units per dose. The vaccine is a recombinant vesicular stomatitis virus (VSV) which has the gene encoding for the VSV envelope glycoprotein G deleted from its ribonucleic acid and replaced with the gene encoding for the ZEBOV envelope glycoprotein (rVSVΔG-ZEBOV-GP).

To support authorization of Ervebo, efficacy, immunogenicity, and safety data from thirteen clinical trials were provided which encompassed eight Phase I trials, one Phase II trial, one Phase II/III trial, and three Phase III trials that were designed and conducted independently by a number of different sponsors. Of the thirteen trials, V920-010 was considered the pivotal trial for the demonstration of efficacy.

V920-010 was a field-based, Phase III, open-label, cluster-randomized, controlled ring vaccination trial conducted in Guinea during the 2014-2016 EVD outbreak. The trial was designed to evaluate the efficacy, effectiveness, and safety of one dose of Ervebo in the prevention of EVD when implemented as ring vaccination. Ring vaccination is a strategy to vaccinate individuals at highest risk of infection due to their social network connection to a patient with laboratory-confirmed EVD. Individuals who met the criteria for a defined ring were randomized (1:1) at the ring level into either the immediate or delayed (21 days) vaccination arms, then vaccinated with Ervebo.

The primary objective of V920-010 was to assess vaccine efficacy against laboratory-confirmed EVD between 10 and 31 days by performing a clinical trial comparing immediate versus (vs.) delayed ring vaccination. The final primary efficacy analysis compared all vaccinated subjects in the immediate arm (2,108 subjects in 51 rings) to all eligible subjects who consented on Day 0 in the delayed arm (1,429 subjects in 46 rings). Ten cases (in 4 rings) of confirmed EVD were observed in eligible subjects in the delayed vaccination arm compared to 0 cases in the immediate vaccination arm. Vaccine efficacy was estimated to be 100% (95% confidence interval [CI]: 63.5% to 100%). Given the data evaluated, the vaccine is expected to be highly efficacious and is expected to confer protection for a disease that is highly contagious and with a high mortality rate. Questions remain regarding the long-term durability of the vaccine efficacy, the need for a booster dose, and the potential that the vaccine may not protect all individuals. Given the high mortality rate of the disease, individuals at risk should continue to protect themselves from exposure to ZEBOV following vaccination.

Analyses for an integrated summary of immunogenicity (ISI) were performed to provide a broad evaluation of the immunogenicity data collected in the Ervebo Phase II/III clinical program. The ISI analyses include the calculation of the geometric mean titers (GMTs) at baseline, Month 1, Month 6, and Month 12, and the percentages of subjects with a seroresponse defined as a ≥2‑fold increase from baseline and ≥200 enzyme-linked immunosorbent assay [ELISA] units [EU]/mL for the glycoprotein-ELISA (GP-ELISA) and a ≥4‑fold increase from baseline for both the GP-ELISA and plaque reduction neutralization test (PRNT). Seroconversion was defined as a post-vaccination ELISA titre of ≥200 EU/mL that was also a ≥4-fold increase in ZEBOV immunoglobulin G (IgG) over baseline. The values for the GP-ELISA and PRNT GMTs were higher than baseline at the Month 1, Month 6, and Month 12 post-vaccination time points for the vaccination groups. For the GP-ELISA, 95.2% and 98.1% of subjects had a seroresponse in the African and ex-African subpopulations at any time post vaccination, respectively. For the PRNT, 57.4% and 83.3% of subjects had a seroresponse in the African and ex-African subpopulations at any time post vaccination, respectively. The immunogenicity data are considered informative but in the absence of an immunocorrelate of protection, the clinical relevance to efficacy is unknown.

A Phase III blinded V920-012 trial, mostly conducted in the United States with some subjects from Canada and Spain, was identified as the main safety study by Health Canada. V920-012 was supported by two earlier trials; a Phase I/II trial conducted in Switzerland (V920-005) and a Phase I trial conducted in the United States (V920-004).

Like many vaccines that induce a robust immune response, the majority of Ervebo recipients, compared with placebo recipients (71.8% vs. 14.3%), experienced at least one local injection-site reaction in V920-012, such as pain, swelling, or redness. First identified in earlier studies, 3.5% of Ervebo recipients (vs. 0% of placebo recipients) in V920-012 reported vaccine-related arthritis, with a median onset of 10 to 11 days post vaccination and a median duration of 5 to 6 days with nearly all cases resolving by end of study. Also, vaccine-related rashes of different types were noted more commonly in vaccine recipients compared with placebo recipients (3.8% vs. 1.5%), as were skin blisters (1.5% vs. 0%). One of 3 subjects who had synovial fluid tested by reverse transcription polymerase chain reaction (RT-PCR) for vaccine virus had positive results (2,301 copies/mL), and 1 of 6 subjects who had skin biopsies tested by RT-PCR for vaccine virus had a positive result (60 copies/mL), which could suggest a vaccine aetiology.

In V920-005, systemic adverse events (AEs) from Day 1 to Day 14 post vaccination were reported in 91.2% of Ervebo recipients vs. 69.2% of placebo recipients, and in 59.6% vs. 41.5% of recipients, respectively, in V920-004. Common solicited systemic AEs included headache, fatigue, myalgia, chills, arthralgia, fever (subjective and objective), nausea, loss of appetite, and diarrhea. In V920-012, systemic AEs were not solicited; 82.8% of Ervebo recipients vs. 44.4% of placebo recipients experienced one or more common adverse events from Day 1 to Day 42 post vaccination. Adverse events, which occurred in 1% or more of subjects who received study vaccination included: diarrhea, nausea, vomiting, asthenia, chills, influenza-like illness, peripheral swelling, malaise, pyrexia, nasopharyngitis, anorexia, arthritis, muscle spasms, stiffness, lethargy, paraesthesia, rhinorrhea, blister, and rash.

In all three trials, no serious adverse events or deaths were considered to be related to Ervebo. Viremia (i.e., vaccine virus in the blood) and viral shedding in saliva and/or urine were tested in V920-004 and V920-005 but were found in few subjects and were short-lived (i.e., less than 28 days post vaccination) suggesting a low risk of transmission to others or the environment following immunization.

Based on the totality of the information, the benefit-risk profile for Ervebo for active immunization of individuals 18 years of age or older to protect against EVD caused by ZEBOV is considered favourable.

The identified safety concerns have been addressed through appropriate labelling in the Product Monograph.

A Risk Management Plan (RMP) for Ervebo was submitted by Merck Canada 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. Upon review, the RMP was considered to be acceptable.

The submitted inner and outer labels, package insert, and Patient Medication Information section of the Ervebo 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 Ervebo was accepted.

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

3 What steps led to the approval of Ervebo?

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

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

Submission Milestones: Ervebo

Submission MilestoneDate
Pre-submission meeting2015-11-18
Pre-submission meeting2021-04-14
New Drug Submission filed2021-09-10
Screening
Screening Acceptance Letter issued2021-10-29
Review
Request granted to pause review clock for 71 days (extension to respond to clarification request)2022-07-11
Review of Risk Management Plan completed2022-10-17
Request granted to pause review clock for 5 days (extension to respond to clarification request)2022-10-18
Quality evaluation completed2022-10-28
Non-clinical evaluation completed2022-11-02
Clinical/medical evaluation completed2022-11-04
Labelling review completed2022-11-04
Biostatistics evaluation completed2022-11-09
Notice of Compliance issued by Director General, Biologic and Radiopharmaceutical Drugs Directorate2022-11-09

4 What follow-up measures will the company take?

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

6 What other information is available about drugs?

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

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

Clinical Pharmacology

Ervebo consists of a live, attenuated recombinant vesicular stomatitis virus-based vector expressing the envelope glycoprotein gene of Zaire Ebola virus (ZEBOV). Immunization of subjects with the vaccine results in an immune response and protection from Zaire Ebola Virus Disease (EVD). The relative contributions of innate, humoral, and cell-mediated immunity to protection from ZEBOV are unknown.

Clinical pharmacology studies with Ervebo were not conducted and are typically not required for vaccines.

Immunogenicity (see Immunogenicity section) was assessed as part of the clinical efficacy evaluation of Ervebo.

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

Clinical Efficacy

The authorization of Ervebo was supported by efficacy, immunogenicity, and safety data from thirteen clinical trials which encompassed eight Phase I trials, one Phase II trial, one Phase II/III trial, and three Phase III trials that were designed and conducted independently by a number of different sponsors. Of the thirteen trials, V920-010 was considered the pivotal trial for the demonstration of the clinical efficacy of Ervebo.

Clinical Efficacy

V920-010, conducted in Guinea during the 2014-2016 EVD outbreak, was a field-based, Phase III, open-label, cluster-randomized, controlled, ring vaccination trial designed to evaluate the efficacy, effectiveness, and safety of one dose of Ervebo in the prevention of EVD when implemented as ring vaccination. Ring vaccination is a strategy to vaccinate individuals at highest risk of infection due to their social network connection to a patient with laboratory-confirmed EVD. Individuals who met the criteria for a defined ring were randomized (1:1) at the ring level into either the immediate or delayed (21 days) vaccination arms, then vaccinated with Ervebo. Subjects were followed up to 84 days post vaccination for EVD.

The primary objective of V920-010 was to assess vaccine efficacy against laboratory-confirmed EVD between 10 and 31 days following randomization by performing a clinical trial comparing immediate versus (vs.) delayed ring vaccination. The final primary efficacy analysis compared all vaccinated subjects in the immediate arm (2,108 subjects in 51 rings) to all eligible subjects who consented on Day 0 in the delayed arm (1,429 subjects in 46 rings). The vaccine efficacy with Ervebo was 100% (95% confidence interval: 63.5% to 100%), with 10 cases (4 rings) of confirmed EVD observed in subjects in the delayed vaccination arm vs. 0 cases in the immediate vaccination arm. Cases of EVD occurring less than 10 days following randomization were excluded from the final primary efficacy analysis to maintain the comparability of the populations with respect to exposure to the index case. Cases of EVD occurring more than 31 days following randomization were also excluded from the final primary efficacy analysis to account for vaccination in the delayed arm on Day 21. The true vaccine efficacy and the duration of protection are unknown given the inherent biases with such studies as well as the challenges of conducting a ring vaccination study in the setting of an outbreak.

Given the data evaluated, the vaccine is expected to be efficacious to a great degree and is expected to confer protection for a disease that is highly contagious with a high mortality rate. Taking into account the high mortality rate of the disease, individuals at risk should continue to protect themselves from exposure to ZEBOV following vaccination.

Immunogenicity

The clinical development program consisted of twelve clinical trials that provided immunogenicity data. Eight Phase I trials (V920-001 through V920-008) were conducted in North America (United States and Canada), Europe (Switzerland and Germany), and non-epidemic regions of Africa (Gabon and Kenya). These trials evaluated dose ranges from 3 x 103 to 1 x 108 plaque forming units (pfu). Antibody titers, measurable by glycoprotein-enzyme-linked immunosorbent assay (GP-ELISA) at Day 14 in most recipients of Ervebo, had the greatest fold rise at Day 28 post vaccination for most of the dosing regimens and declined slightly but plateaued thereafter through Day 180 or through Day 360/365 (depending on the study). Plaque reduction neutralization test (PRNT) antibody titers were elevated as early as Day 14 and peaked at Day 28. The results revealed that the greatest fold rise occurred 1 to 2 months post vaccination and were sustained through Day 360. Immunogenicity data from these studies supported the selection of the 2 x 107 pfu dose that was used in the pivotal trials.

Additionally, four Phase II/III trials (V920-009, V920-011, V920-012, and V920-018) provided immunogenicity data from subjects who received Ervebo as a single nominal dose of 2 x 107 pfu (all four trials) or 1 x 108 pfu (V920-012 only). Three of the trials were conducted in African countries (Liberia, Sierra Leone, and Guinea) and the other was conducted in Canada, the United States, and Spain (V920-012).

Analyses for an integrated summary of immunogenicity (ISI) were performed to provide a broad evaluation of the immunogenicity data collected in the Ervebo Phase II/III clinical program. The ISI analyses include the calculation of geometric mean titers (GMTs) at baseline, Month 1, Month 6, and Month 12, and the percentages of subjects with a seroresponse defined as a ≥2–fold increase from baseline and ≥200 ELISA units [EU]/mL for the GP-ELISA and a ≥4–fold increase from baseline for both the GP–ELISA and PRNT. Seroconversion was defined as a post-vaccination ELISA titre of at least 200 EU/mL that was also a ≥4–fold increase over baseline in ZEBOV immunoglobulin G (IgG). The values for the GP–ELISA and PRNT GMTs were higher than baseline at the Month 1, Month 6, and Month 12 post-vaccination time points for the vaccination groups.

In V920-012, the durability of immunogenicity was evaluated through Month 24 post vaccination. The values for GP–ELISA and PRNT GMTs for the Ervebo groups were higher than baseline through Month 24 post vaccination. Whereas GP–ELISA

GMTs peaked at Day 28 post vaccination with an approximate 25% decline observed at Month 24 post vaccination, PRNT GMTs peaked at Month 18 post vaccination and no appreciable decline was observed thereafter. The GMTs for the placebo group were less than the lower limit of quantification of the GP–ELISA and the PRNT at all time points.

The GP–ELISA geometric mean fold rises (GMFRs) demonstrated that GP–ELISA titers for the Ervebo groups increased from baseline by approximately 64–fold at Day 28 post vaccination and remained elevated approximately 48–fold at Month 24 post vaccination. The PRNT GMFRs demonstrated that PRNT titers for the Ervebo groups increased from baseline by approximately 11–fold at Day 28 post vaccination, peaked by approximately 16–fold at Month 18 post vaccination, and remained elevated approximately 15–fold at Month 24 post vaccination.

Immunogenicity specimens from subjects in the African trials underwent gamma irradiation to allow for processing in a biosafety level 2 facility. A study conducted by the sponsor showed that gamma irradiation results in an approximate 20% elevation in measured antibody response for negative clinical samples and an approximate 20% reduction in post-vaccination antibody response in the GP–ELISA. The effect of gamma irradiation was not consistently observed for the PRNT. The sponsor reasons that this is why GP–ELISA and PRNT GMTs were generally lower for subjects in the African trials compared with those in V920-012. For the GP–ELISA, 95.2% and 98.1% of subjects had a seroresponse in the African and ex-African subpopulations, respectively, at any time post vaccination. For the PRNT, 57.4% and 83.3% of subjects had a seroresponse in the African and ex-African subpopulations, respectively, at any time post vaccination. From a proof-of-principle perspective (as the data were not correlated to individual efficacy data), the robust increases of the GP–ELISA binding and PRNT neutralizing antibodies across the main and supportive studies suggest that the efficacy findings are explained by the immunogenicity responses. The sponsor concluded that although the GP–ELISA may be a correlate of protection as a result of vaccination, it is not prudent to define a threshold value for protection at this time given a number of considerations and limitations associated with the analyses conducted. The immunogenicity data are considered informative but in the absence of an immunocorrelate of protection, the clinical relevance to efficacy is unknown.

Key efficacy issues and uncertainties

Important limitations of the data at this time include the lack of information on the long-term efficacy and safety of the vaccine. Namely, the duration of protection, the level of protection, and the protection against other Ebola virus strains or viral variants of the Zaire strain are unknown. There are also limited data on efficacy in special populations (e.g., pediatrics and individuals who are significantly immunocompromised, pregnant/lactating, or with severe comorbidities). Additionally, the need for and timing of a booster dose is unknown, and antibodies against the viral vector were not measured, leaving a lack of understanding of the kinetics of antibodies against the viral vector and their potential impact on the immune response to repeated vaccinations. Furthermore, effectiveness of the vaccine when administered concurrently with antiviral medication, immunoglobulin, and/or blood or plasma transfusions is unknown. Given the high mortality rate of the disease, individuals at risk should continue to protect themselves from exposure to ZEBOV following vaccination. These limitations are considered adequately managed through labelling associated with authorization, and the Risk Management Plan for Ervebo.

Indication

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

Ervebo (Ebola Zaire Vaccine [rVSVΔG-ZEBOV-GP, live]) is indicated for active immunization of individuals 18 years of age or older to protect against Ebola Virus Disease (EVD) caused by Zaire Ebola virus.

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

Clinical Safety

The clinical safety evaluation of Ervebo focused on potential reactogenicity as assessed by solicited and unsolicited injection site and systemic adverse events (AEs) after vaccination with Ervebo. The review of vaccine safety data provided in the submission was complex due to a heterogeneous group of thirteen blinded and open-label trials (V920-001 to V920-012, and V920-018) conducted in Africa, North America, and Europe. These trials were conducted among a diverse set of study populations using different methods of measuring solicited and unsolicited AEs, and were initiated by different organizations, including the World Health Organization and the sponsor. As such, the sponsor was unable to pool safety data from across the different trials (16,765 adult subjects) but used a study-by-study strategy for evaluation of safety data.

The Phase III blinded V920-012 trial, mostly conducted in the United States with some subjects from Canada and Spain, was identified as the main safety study by Health Canada. V920-012 was supported by two earlier trials; the Phase I/II blinded V920-005 trial (Switzerland) and the Phase I blinded V920-004 trial (United States). V920-005 and V920-004 were considered key safety studies that confirmed AEs of special interest (AESI) clustering around delayed post-vaccination arthritis syndrome and delayed post-vaccination rash syndrome. The main efficacy study, V920-010 (Guinea), was excluded from the safety analysis due to a high baseline morbidity that would not be expected in a Canadian population (e.g., malaria, EVD, etc.), as well as being an open-label design without a placebo control group for comparison.

Solicited local (injection-site) AEs were collected in V920-012, V920-005, and V920-004, with the majority of Ervebo recipients experiencing local reactions. In V920-012, 71.8% of 1,051 Ervebo recipients vs. 14.3% of 133 placebo recipients experienced at least one injection site AE during the first 5 days post vaccination, with the most frequently reported being injection site pain (69.5% vs. 12.8%), injection site swelling (16.5% vs. 3.0%), and injection site erythema (11.9% vs. 1.5%), all with a median duration of 4 days. In the first 14 days post vaccination, solicited local treatment-emergent adverse reactions (TEAEs) were experienced by 52.0% of 102 Ervebo recipients vs. 23.1% of 13 placebo recipients in V920-005 and by 43.1% of 418 Ervebo recipients vs. 10.6% of 94 placebo recipients in V920-004.

Solicited systemic AEs were not specifically collected in V920-012 but were collected in V920-005 and V920-004 within 14 days post vaccination. In V920-012, systemic reactogenicity was captured in the data for unsolicited AEs within 42 days post vaccination. Common solicited systemic AEs included headache, fatigue, myalgia, chills, arthralgia, fever (subjective and objective), nausea, loss of appetite, and diarrhea. Overall, V920-005 reported more systemic reactogenicity, occurring in 91.2% of Ervebo recipients vs. 69.2% of placebo recipients compared with 59.6% vs. 41.5% of recipients, respectively, in V920-004.

In V920-012, the majority (82.8%) of 1,051 Ervebo recipients compared with 44.4% of 133 placebo recipients experienced one or more common AE from Day 1 to Day 42 post vaccination. The most common System Organ Classes with unsolicited AEs were (Ervebo vs. placebo) “general disorders and administrative site conditions” (78% vs. 18.8%), “musculoskeletal and connective tissue disorder” (29.9% vs. 6.8%), “nervous system disorders” (26.1% vs. 14.3%), “gastrointestinal disorders” (12.0% vs. 6.0%), and “infections and infestations” (7.5% vs. 9.0%). Adverse events, which occurred in 1% or more of subjects, included: diarrhea, nausea, vomiting, asthenia, chills, influenza-like illness, peripheral swelling, malaise, pyrexia, nasopharyngitis, anorexia, arthritis, muscle spasms, stiffness, lethargy, paraesthesia, rhinorrhea, blister, and rash.

During the implementation of the Phase I/II V920-005 blinded trial, investigators initiated a safety hold in response to the appearance of delayed-onset arthritis and rash syndromes (or both) among vaccine recipients during the first month of enrollment. Overall, 23.5% of 102 Ervebo recipients experienced arthritis TEAEs vs. 0% of placebo recipients, with a median time of onset of 10.5 days post vaccination and a median duration of 34.5 days. All but 2 subjects had no prior history of arthritis. Arthritis resolved in 50% of subjects by 1 year post vaccination, and in another 25% of subjects by 2 years post vaccination.

In V920-004, 4.5% of 418 Ervebo recipients experienced “temporally associated post-vaccination arthritis” compared with 3.2% of 94 placebo recipients. All vaccine-related cases resolved by the end of the study, with the exception for 1 subject with recurrent arthritis. The median time to onset was 12 days in the Ervebo group vs. 15 days in the placebo group, with a median duration of 8 days vs. 47 days, respectively. In the follow-up Phase III V920-012 trial, 3.5% of 1,051 Ervebo recipients and 0% of 133 placebo recipients experienced an “arthritis” AE (composite term for arthritis, monoarthritis, polyarthritis, osteoarthritis, joint swelling, or joint effusion). The median onset was 10 to 11 days post vaccination with a median duration of 5 to 6 days and no dose relationship (regular vs. high dose). In V920-005, the joint fluid samples of 3 subjects were positive for vaccine virus as determined by reverse transcription-polymerase chain reaction (RT-PCR) on Day 14 or Day 28, whereas 1 of 3 subjects tested positive in V920-012, which could suggest a vaccine aetiology.

Similarly, in V920-005, “skin- and mucosal-related treatment-emergent adverse events”, or skin- and mucosal-related TEAEs, were noted in 29.4% of 102 Ervebo recipients vs. 31% of 13 placebo recipients, with a median onset of 8 days post vaccination for Ervebo recipients vs. 8.5 days post vaccination for placebo recipients, and lasting 10.5 days vs. 11 days, respectively. However, skin- and mucosal-related TEAEs in the Ervebo group were different, and included the following conditions not experienced in the placebo group: maculopapular rash, skin lesions, cutaneous vasculitis, as well as papule, generalized rash, macular rash, popular rash, and vesicular rash. Five subjects who were vaccinated had a total of eight skin samples that were positive for virus vector, suggesting a vaccine aetiology. Overall, 10.8% of Ervebo recipients had concurrent arthritis and skin-related TEAEs, compared with 0% of placebo recipients.

In V920-004, 5.7% of 418 Ervebo recipients vs. 3.2% of 94 placebo recipients experienced “temporally associated post-vaccination dermatitis” (e.g., dermatitis, petechiae, macular rash, papular rash, vesicular rash, skin lesion, skin mass, or skin ulcer) with an onset within 56 days of vaccination. The overall median time to onset was 9 days for Ervebo recipients and 5 days for placebo recipients, with a median duration of 7 days and 54 days, respectively. A total of 1.2% of Ervebo recipients experienced concurrent post-vaccination arthritis and dermatitis with no similar cases within the placebo group.

In the main safety study, V920-012, 3.8% of 1,051 Ervebo recipients and 1.5% of 133 placebo recipients experienced “rash” AEs (composite term for petechiae, purpura, rash, rash generalized, rash macular, rash popular, and rash vesicular but not dermatitis) from Day 1 to Day 42 post vaccination. The median time to onset was 7.5 to 10.5 days for Ervebo recipients and 3.5 days for placebo recipients, with a median duration 6 to 18 days and 14 days, respectively. Additionally, a total of 1.5% of 1,051 Ervebo recipients experienced vesicular (blistering) lesion from Day 1 to Day 42 post vaccination vs. 0% of placebo recipients. Vaccine virus was detected in skin biopsies by RT-PCR in 1 of 6 subjects, suggesting a vaccine aetiology.

In V920-012, V920-005, and V920-004, no serious adverse events or deaths were considered to be vaccine related. Of the 408 Ervebo recipients with PCR data in V920-004, 20% had detectable vesicular stomatitis virus viremia at one or more time points post vaccination (vs. 0% in placebo). Viremia was most prevalent at Day 1 and Day 2 post vaccination, with 13% of Ervebo recipients testing positive for vaccine virus at each of these time points. The number of Ervebo recipients who were viremic dropped to 2% by Day 3 and to 0.5% by Day 4, with viremia resolving in all subjects by Day 28. Higher doses of Ervebo were associated with higher percentages of subjects experiencing vaccine viremia. In V920-005, 44% of Ervebo recipients had detectable vaccine viremia at Day 1 post vaccination (vs. 0% of placebo recipients). At Day 7, only 2% of the 92 subjects with available PCR data had detectable vaccine viremia. No vaccine viremia was detectable on Day 14 or Day 28. This suggests that the detection of administered vaccine product in the blood shortly after administration was short-lived. In V920-004, vaccine virus shedding/excretion in vaccine viremic subjects was infrequent as only 1 Ervebo recipient had detectable vaccine virus in saliva and urine within the first week. Similarly, in V920-005, it was planned that all subject samples were to be tested for vaccine virus ribonucleic acid shedding in saliva and urine following vaccination, but testing was stopped after the first safety analysis (i.e., after the first 10 subjects were tested) when it was noted that few samples from subjects were yielding positive results. Vaccine viral shedding was not assessed in the Phase III V920-012 trial. Overall, this suggests that the risk of transmission of vaccine virus by blood, saliva, or urine is low and short-lived. To avoid the theoretical risk of vaccine virus transmission, the Ervebo product monograph includes a precaution for vaccine recipients to avoid close contact with, and exposure of, high-risk individuals to blood and bodily fluids for at least 6 weeks following vaccination.

Key safety issues and uncertainties

Important limitations of the data at this time include the lack of information on the long-term efficacy and safety of Ervebo. Ervebo has been extensively used in field trials and during ZEBOV outbreaks subsequent to the trial data submitted within this submission. Fewer than the 3,000 vaccine exposures from the Phase III V920-012 blinded trial and the supportive Phase I blinded trials (V920-004 and V920-005) were used as the appropriate safety analysis dataset for the Canadian Product Monograph. However, this was considered adequate for the following reasons: participants were representative of the Canadian population, rates of AEs in vaccine recipients were properly controlled by a concurrent placebo group, and AESIs of arthritis and rash were common enough to sufficiently characterize the potential risk. Whether the post-marketing rates of AESIs will be higher, lower, or similar to trial rates is unknown. Similarly, solicited information from the smaller Phase I studies suggests that the risk of transmission of vaccine virus in blood, saliva, and urine appears to be low and short-lived. Thus, AESIs and transmission risk should continue to be monitored.

Appropriate warnings and precautions are in place in the approved Ervebo Product Monograph to address the identified safety concerns.

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

7.2 Non-Clinical Basis for Decision

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

The non-clinical package consisted of pharmacological studies and a toxicological testing program which covered local tolerance and general toxicity in repeat-dose toxicity studies and a developmental and reproductive toxicology study (DART) study. Additional studies were also conducted, namely, an in vivo biodistribution and persistence study and an exploratory neurovirulence study. The pharmacodynamic properties of Ervebo were evaluated in multiple previously published non-Good Laboratory Practices (GLP) immunogenicity and efficacy studies in mice, guinea pigs, and in non-human primates (NHPs), and subsequently confirmed in additional non-GLP NHP studies conducted by the sponsor. Pivotal toxicology studies were GLP-compliant, including repeat-dose toxicity studies in mice and NHPs, a DART study in rats, and a biodistribution study in NHPs. Primary pharmacodynamics studies, an exploratory neurovirulence test, and an exploratory immunogenicity and viremia study were non-GLP.

When administered intramuscularly to NHPs, Ervebo was highly immunogenic across a wide dose range (3 x 101 to 1 x 108 plaque forming units [pfu]). Additionally, these studies support the immunogenicity of the vaccine, with ZEBOV-GP-specific immunoglobulin G (IgG) detectable in most animals 14 days after a single immunization with all animals seroconverting within 28 days. Moreover, protective efficacy against Ebola Virus Disease (EVD) was demonstrated, with 95% to 100% protection against mortality following a lethal intramuscular Zaire Ebola virus (ZEBOV) injection challenge 42 days after immunization. The studies indicated that no sterile immunity was obtained by vaccination since a number of vaccinated animals that survived had detectable viremia after challenge, and some also had clinical signs indicative of infection. The exact duration of protection following a single dose is not fully understood. No major objections were identified, and no additional studies were required. It is considered that the provided non-clinical pharmacology data are sufficient to support the use of Ervebo in individuals at high risk of exposure in the short-term, e.g., in an outbreak setting.

The pivotal toxicity studies were adequately designed and conducted, and the results did not raise any safety signal of concern. The repeat-dose toxicity studies in mice and NHPs showed no evidence of systemic toxicity, and local tolerance was considered to be consistent with what would be expected for an intramuscular vaccine injection. The exploratory neurovirulence study revealed no tropism for the central nervous system, and even when the vaccine virus was introduced intrathalamically in the central nervous system of NHPs, no neurological symptoms were observed. Additionally, the biodistribution study in NHPs showed no evidence of Ervebo distribution to the brain. In rats, DART study data demonstrated no effect of Ervebo (5.28 x 107 pfu/animal) on mating, fertility, or fetal development following either single or multiple doses.

Wild-type vesicular stomatitis virus is a pathogen in livestock and can occasionally be transmitted to humans in close contact with infected animals. Replication of Ervebo was investigated in rodents, pigs, arthropods, and NHPs. Based on limited shedding in adults, the results of a toxicity study in NHPs, and the lack of horizontal transmission in pigs, it was inferred that the overall risk of Ervebo to human health and the environment is considered negligible. However, it is reasoned that as a precaution, vaccine recipients should attempt to avoid exposure of livestock to blood and bodily fluids for at least 6 weeks following vaccination to avoid the theoretical risk of spread of the Ervebo vaccine virus.

Overall, the non-clinical pharmacology and toxicology profile of Ervebo sufficiently establishes proof of concept and exhibits an appropriate safety profile as a vaccine that could be used for active immunization of at-risk individuals to protect against EVD caused by ZEBOV.

The results of the non-clinical studies as well as the potential risks to humans have been included in the Ervebo Product Monograph.

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

7.3 Quality Basis for Decision

Characterization of the Drug Substance

The Ervebo drug substance (Ebola Zaire Vaccine [rVSVΔG-ZEBOV-GP, live]) is a live, attenuated recombinant virus consisting of a vesicular stomatitis virus (VSV)-based vector. The gene for the VSV envelope glycoprotein (VSV-GP) is deleted and replaced by the gene for the Zaire Ebola virus (Kikwit serotype) envelope glycoprotein (ZEBOV-GP). The binding of virus particles to host cells, and hence the development of Ebola virus infection, is mediated by ZEBOV-GP.

Detailed characterization studies were performed to provide assurance that the identity, purity, and genetic stability of the rVSVΔG-ZEBOV-GP master virus seed met acceptance criteria. Genetic stability of the master virus seed was assessed by next generation sequencing and was found to be comparable through two consecutive viral passages.

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

The Ervebo drug substance, a recombinant virus, is propagated in Vero cell culture and the supernatant is harvested, clarified, enzyme treated, purified, stabilized to obtain bulk drug substance, and then frozen.

The Ervebo drug product is supplied as a solution for intramuscular injection. Each 1 mL dose is formulated to contain 72 million or more plaque forming units of vaccine virus in a stabilizer solution consisting of recombinant human serum albumin and tromethamine buffer.

The manufacture of the drug product involves thawing of the bulk drug substance which is then aseptically formulated with sterile filtered stabilizer solution to achieve a target concentration prior to being aseptically filled into single-dose vials for injection. Final steps in the manufacture of the drug product include visual inspection, labelling, packaging in 10‑vial cartons, and freezing.

Changes to the bulk drug substance manufacturing process made throughout the pharmaceutical development are considered acceptable upon review. The drug product manufacturing process did not significantly change throughout development.

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

None of the non-medicinal ingredients (excipients, described earlier) found in the drug product are prohibited by the Food and Drug Regulations. The compatibility of the medicinal ingredient with the excipients is supported by the stability data provided.

Control of the Drug Substance and Drug Product

The drug substance and drug product are tested using appropriate controls to verify that they meet approved specifications, and analytical procedures are validated and in compliance with International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) guidelines.

Each lot of Ervebo is tested for appearance, content, identity, potency, purity, and impurities. Established test specifications and validated analytical test methods are considered acceptable.

Ervebo 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.

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 proposed 36-month shelf life at ‑70 ± 10 °C for Ervebo is considered acceptable, with time allowances of 2 weeks at 5 ± 3 °C and 4 hours at 25 ± 2 °C after the product is thawed.

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

The proposed packaging and components are considered acceptable.

Facilities and Equipment

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

Based on a risk assessment score determined by Health Canada, an on-site evaluation of the manufacturing facility was not deemed necessary.

The facility involved in production is compliant with good manufacturing practices.

Adventitious Agents Safety Evaluation

The drug substance and drug product manufacturing process incorporates adequate control measures to prevent contamination and maintain microbial control. Pre-harvest culture fluids, as well as cell banks and virus seed, are rigorously tested to ensure freedom from adventitious microorganisms (bioburden, mycoplasma, and viruses), and appropriate limits are set. Purification process steps designed to remove potential adventitious viruses are adequately validated.

A raw material of biological origin (fetal bovine serum) used in the manufacture of the master cell bank was sourced from a spongiform encephalopathy/transmissible spongiform encephalopathy-negligible region and is in compliance with the Note for Guidance on Minimising the Risk of Transmitting Animal Spongiform Encephalopathy Agents via Human and Veterinary Medicinal Products (EMEA/410/01, Revision 3).

No primary raw materials used in the manufacture of working cell bank, master virus seed, bulk drug substance, or drug product are of animal origin.