Vaccines and Related Biological Products Advisory Committee Meeting December 17, 2020 FDA Briefing Document Moderna COVID-19 Vaccine Sponsor: ModernaTX, Inc. 2 Moderna COVID-19 Vaccine VRBPAC Briefing Document Table of Contents List of Tables.................................................................................................................................3 List of Figures ...............................................................................................................................4 Glossary........................................................................................................................................4 1. Executive Summary.................................................................................................................5 2. Background..............................................................................................................................6 2.1 SARS-CoV-2 Pandemic ....................................................................................................6 2.2 EUA Request for the Moderna COVID-19 Vaccine mRNA-1273......................................7 2.3 U.S. Requirements to Support Issuance of an EUA for a Biological Product..............................................................................................................................8 2.4 Alternatives for Prevention of COVID-19 ..........................................................................9 2.5 Applicable Guidance for Industry ......................................................................................9 2.6 Safety and Effectiveness Information Needed to Support an EUA...................................9 2.7 Continuation of Clinical Trials Following Issuance of an EUA for a COVID-19 Vaccine..........................................................................................................10 2.8 Previous Meetings of the VRBPAC to Discuss Vaccines to Prevent COVID-19........................................................................................................................10 3. Topics for VRBPAC Discussion.............................................................................................11 4. Moderna COVID-19 Vaccine (mRNA-1273)..........................................................................11 4.1 Vaccine Composition, Dosing Regimen..........................................................................11 4.2 Proposed Use Under EUA ..............................................................................................12 5. FDA Review of Clinical Safety and Effectiveness Data .........................................................12 5.1 Overview of Clinical Studies............................................................................................12 5.2 Study mRNA-1273-P301.................................................................................................12 5.2.1 Design.....................................................................................................................12 5.2.2 FDA Assessment of Phase 3 Follow-Up Duration ..................................................17 5.2.3 Participant Disposition and Inclusion in Analysis Populations ................................17 5.2.4 Demographics and Other Baseline Characteristics ................................................19 5.2.5 Vaccine Efficacy......................................................................................................22 5.2.6 Safety......................................................................................................................31 6. Sponsor’s Plans for Continuing Blinded, Placebo-Controlled Follow-Up...............................46 7. Pharmacovigilance Activities .................................................................................................46 8. Benefit/Risk Assessment in the Context of Proposed Indication and Use Under EUA..................................................................................................................................48 8.1 Known Benefits ...............................................................................................................48 8.2 Unknown Benefits/Data Gaps .........................................................................................48 8.3 Known Risks ...................................................................................................................50 8.4 Unknown Risks/Data Gaps .............................................................................................50 3 Moderna COVID-19 Vaccine VRBPAC Briefing Document 9. References ............................................................................................................................52 10.Appendix A. Phase 1 and 2 Studies ......................................................................................53 List of Tables Table 1. Clinical Trials Submitted in Support of Efficacy and Safety Determinations of the Moderna COVID-19 Vaccine mRNA-1273 .........................................................................12 Table 2. Efficacy Set Definitions .................................................................................................15 Table 3. Safety Set Definitions....................................................................................................16 Table 4. Efficacy Analysis Population Study Dispositiona, mRNA-1273-P301............................18 Table 5. Safety Analysis Population Study Dispositiona, mRNA-1273-P301 ..............................19 Table 6. Demographic Characteristics, Per-Protocol Set ...........................................................20 Table 7. Demographic Characteristics, Safety Set .....................................................................21 Table 8. Protocol-Defined Risk for Severe COVID-19 Disease, Safety Seta .............................22 Table 9. Interim Analysis for Primary Efficacy Endpoint, COVID-19 Starting 14 Days After the 2nd Dose, Per-Protocol Set................................................................................................23 Table 10. Subgroup Analyses of Vaccine Efficacy, COVID-19 14 Days After Dose 2 Per Adjudication Committee Assessments, Per-Protocol Set ...................................................24 Table 11. Demographic Characteristics, Participants With COVID-19 Starting 14 Days After Dose 2, Per Adjudication Committee Assessments, Per-Protocol Set ...............................25 Table 12. Vaccine Efficacy by Baseline SARS-CoV-2 Status: First COVID-19 From 14 Days After Dose 2 Per Adjudication Committee Assessment, Full Analysis Set .........................26 Table 13. Vaccine Efficacy by Risk Factor: First COVID-19 Occurrence From 14 Days After Dose 2, Per Adjudication Committee Assessment, Per-Protocol Set.................................26 Table 14. Severe COVID-19 Cases Starting 14 Days After Second Dose Based on Adjudication Committee Assessment, Per-Protocol Set .........................................................................27 Table 15. Vaccine Efficacy of mRNA-1273 to Prevent COVID-19 From Dose 1 by Time Period in Participants Who Only Received One Dose, mITT Set ......................................................28 Table 16. Vaccine Efficacya of mRNA-1273 to Prevent Severe COVID-19 After Dose 1 in Participants Who Only Received One Dose in mITT Set ...................................................29 Table 17. Final Scheduled Efficacy Analysis, Primary Endpoint, COVID-19 Starting 14 Days After the Second Dose per Adjudication Committee Assessments, Per-Protocol Set........29 Table 18. Secondary Efficacy Analysis, Severe COVID-19 Starting 14 Days After the Second Dose per Adjudication Committee Assessments, Per-Protocol Set ...................................30 Table 19. Participants Reporting at Least One Adverse Event, Among All Participants and by Baseline SARS-COV2 Status (Safety Set).........................................................................32 Table 20. Adverse Events Among Adults ≥65 Years of Age (Safety Set)...................................33 Table 21. Frequency of Solicited Local Adverse Reactions Within 7 Days Following Either the First or Second Dose of Vaccine, Participants Age 18 to <64 years, Solicited Safety Set.34 Table 22. Frequency of Solicited Local Adverse Reactions Within 7 Days Following Either the First or Second Dose of Vaccine, Participants Age ≥65 years, Solicited Safety Set ..........35 Table 23. Frequency of Solicited Systemic Adverse Reactions Within 7 Days Following Either the First or Second Dose of Vaccine, Participants Age 18-64 years, Solicited Safety Set.37 4 Moderna COVID-19 Vaccine VRBPAC Briefing Document Table 24. Frequency of Solicited Systemic Adverse Reactions Within 7 Days Following Either the First or Second Dose of Vaccine, Participants Age ≥65 Years, Solicited Safety Set ...38 Table 25. Summary of Unsolicited AEs Regardless of Relationship to the Investigational Vaccine, Through 28 Days After Any Vaccination, Study 301, Safety Set .........................40 Table 26. Unsolicited Adverse Events Occurring in ≥1% of Vaccine Group Participants, by MedDRA Primary System Organ Class and Preferred Term (Safety Analysis Set) ...........40 Table 27. SAEs Considered Related by Investigator..................................................................43 List of Figures Figure 1. Safety Monitoring Plan, Study 301 ..............................................................................15 Figure 2. Cumulative Incidence Curves for the First COVID-19 Occurrence After Randomization, mITT Set.............................................................................................................................28 Glossary AE adverse event AESI adverse event of special interest AIDS acquired immunodeficiency syndrome ARDS acute respiratory distress syndrome CBRN chemical, biological, radiological, or nuclear CDC Centers for Disease Control and Prevention EUA Emergency Use Authorization FDA Food and Drug Administration hACE2 human angiotensin converting enzyme 2 HHS Health and Human Services HIV human immunodeficiency virus IM intramuscular LNP lipid nanoparticle MERS-CoV Middle Eastern respiratory syndrome mRNA messenger RNA NAAT nucleic acid amplification-based test RT-PCR reverse transcription-polymerase chain reaction SAE serious adverse event SARS-CoV-2 severe acute respiratory syndrome coronavirus 2 VE vaccine efficacy VRBPAC Vaccines and Related Biological Products Advisory Committee 5 Moderna COVID-19 Vaccine VRBPAC Briefing Document 1. Executive Summary On November 30, 2020, ModernaTX (the Sponsor) submitted an Emergency Use Authorization (EUA) request to FDA for an investigational COVID-19 vaccine (mRNA-1273) intended to prevent COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The vaccine is based on the SARS-CoV-2 spike glycoprotein (S) antigen encoded by RNA and formulated in lipid nanoparticles (LNPs). The proposed use under an EUA is for active immunization for the prevention of COVID-19 caused by SARS-CoV-2 in individuals 18 years of age and older. The proposed dosing regimen is 2 doses, 100 μg each, administered 1 month apart. The EUA request includes safety and efficacy data from an ongoing Phase 3 randomized, double-blinded and placebo-controlled trial of mRNA-1273 in approximately 30,400 participants. The primary efficacy endpoint is the reduction of incidence of COVID-19 among participants without evidence of SARS-CoV-2 infection before the first dose of vaccine in the period after 14 days post-dose 2. In an interim analysis conducted using a data cutoff of November 7, 2020, a total of 27,817 participants randomized 1:1 to vaccine or placebo with a median 7 weeks of follow-up post-dose 2 were included in the per-protocol efficacy analysis population of participants without evidence of SARS-CoV-2 infection prior to vaccination. Efficacy in preventing confirmed COVID-19 occurring at least 14 days after the second dose of vaccine was 94.5.0% (95% CI 86.5%, 97.8%) with 5 COVID-19 cases in the vaccine group and 90 COVID-19 cases in the placebo group. Subgroup analyses of the primary efficacy endpoint showed similar efficacy point estimates across age groups, genders, racial and ethnic groups, and participants with medical comorbidities associated with high risk of severe COVID-19. Secondary efficacy analyses suggested benefit of the vaccine in preventing severe COVID-19 (11 protocol-defined severe COVID-19 cases in the placebo group vs. 0 cases in the vaccine group), in preventing COVID-19 following the first dose, and in preventing COVID-19 in individuals with prior SARS-CoV-2 infection, although available data for some of these outcomes did not allow for firm conclusions. Efficacy data from the final scheduled analysis of the primary efficacy endpoint (data cutoff of November 21, 2020, with a median follow-up of >2 months post-dose 2) demonstrated a VE of 94.1% (95% CI 89.3%, 96.8%), with 11 COVID-19 cases in the vaccine group and 185 COVID-19 cases in the placebo group and was consistent with results obtained from the interim analysis. The VE in this analysis when stratified by age group was 95.6% (95% CI: 90.6%, 97.9%) for participants 18 to <65 years of age and 86.4% (95% CI: 61.4%, 95.5%) for participants ≥65 years of age. A final secondary efficacy analysis also supported efficacy against protocol-defined severe COVID-19, with 30 cases in the placebo group vs. 0 cases in the vaccine group. Safety data from a November 11, 2020 interim analysis of approximately 30,350 participants ≥18 years of age randomized 1:1 to vaccine or placebo with a median of 7 weeks of follow-up after the second dose supported a favorable safety profile, with no specific safety concerns identified that would preclude issuance of an EUA. These safety data are the primary basis of FDA’s safety review. On December 7, 2020, the Sponsor submitted additional follow-up data from these participants with a cutoff of November 25, 2020, which represents a median of 9 weeks (>2 months) of follow-up post-dose 2. Key safety data from this later submission, including death, other serious adverse events, and unsolicited adverse events of interest were independently verified and confirmed not to change the safety conclusions from the interim safety analysis. The most common solicited adverse reactions associated with mRNA-1273 were injection site pain (91.6%), fatigue (68.5%), headache (63.0%), muscle pain (59.6%), joint pain (44.8%), and 6 Moderna COVID-19 Vaccine VRBPAC Briefing Document chills (43.4%); severe adverse reactions occurred in 0.2% to 9.7% of participants, were more frequent after dose 2 than after dose 1, and were generally less frequent in participants ≥65 years of age as compared to younger participants. Among unsolicited adverse events of clinical interest, which could be possibly related to vaccine, using the November 25, 2020 data cutoff, lymphadenopathy was reported as an unsolicited event in 173 participants (1.1%) in the vaccine group and 95 participants (0.63%) in the placebo group. Lymphadenopathy (axillary swelling and tenderness of the vaccination arm) was a solicited adverse reaction observed after any dose in 21.4% of vaccine recipients <65 years of age and in 12.4% of vaccine recipients ≥65 years of age, as compared with 7.5% and 5.8% of placebo recipients in those age groups, respectively. There was a numerical imbalance in hypersensitivity adverse events across study groups, with 1.5% of vaccine recipients and 1.1% of placebo recipients reporting such events in the safety population. There were no anaphylactic or severe hypersensitivity reactions with close temporal relation to the vaccine. Throughout the safety follow-up period to date, there were three reports of facial paralysis (Bell’s palsy) in the vaccine group and one in the placebo group. Currently available information is insufficient to determine a causal relationship with the vaccine. There were no other notable patterns or numerical imbalances between treatment groups for specific categories of adverse events (including other neurologic, neuro￾inflammatory, and thrombotic events) that would suggest a causal relationship to mRNA-1273. The frequency of serious adverse events was low (1.0% in the mRNA-1273 arm and 1.0% in the placebo arm), without meaningful imbalances between study arms. The most common SAEs in the vaccine group which were numerically higher than the placebo group were myocardial infarction (0.03%), cholecystitis (0.02%), and nephrolithiasis (0.02%), although the small numbers of cases of these events do not suggest a causal relationship. The most common SAEs in the placebo arm which were numerically higher than the vaccine arm, aside from COVID-19 (0.1%), were pneumonia (0.05%) and pulmonary embolism (0.03%). With the exception of more frequent, generally mild to moderate reactogenicity in participants <65 years of age, the safety profile of mRNA-1273 was generally similar across age groups, genders, ethnic and racial groups, participants with or without medical comorbidities, and participants with or without evidence of prior SARS-CoV-2 infection at enrollment. This meeting of the Vaccines and Related Biological Products Advisory Committee (VRBPAC) is being convened to discuss and provide recommendations on whether, based on the totality of scientific evidence available, the benefits of the mRNA-1273 COVID-19 Vaccine outweigh its risks for use in individuals 18 years of age and older. The committee will also discuss what additional studies should be conducted by the vaccine manufacturer following issuance of the EUA to gather further data on the safety and effectiveness of this vaccine. 2. Background 2.1 SARS-CoV-2 Pandemic The SARS-CoV-2 pandemic presents an extraordinary challenge to global health and, as of December 11, 2020, has caused more than 71 million cases of COVID-19 and claimed the lives of more than 1.6 million people worldwide. In the United States, more than 16 million cases have been reported to the Centers for Disease Control and Prevention (CDC), with over 296,000 deaths. Confirmed cases and mortality continue to rise globally. On January 31, 2020, the U.S. Secretary of Health and Human Services (HHS) declared a public health emergency related to COVID-19 and mobilized the Operating Divisions of HHS. Following the World Health Organization’s declaration of the novel coronavirus pandemic on March 11, 2020, the U.S. 7 Moderna COVID-19 Vaccine VRBPAC Briefing Document President declared a national emergency in response to COVID-19 on March 13, 2020. Vaccines to protect against COVID-19 are critical to mitigate the current SARS-CoV-2 pandemic and to prevent future disease outbreaks. SARS-CoV-2 is a novel, zoonotic coronavirus that emerged in late 2019 in patients with pneumonia of unknown cause. 1 The virus was named SARS-CoV-2 because of its similarity to the coronavirus responsible for severe acute respiratory syndrome (SARS-CoV, a lineage B betacoronavirus).2 SARS-CoV-2 is an enveloped, positive sense, single stranded RNA virus sharing more than 70% of its sequence with SARS-CoV, and ~50% with the coronavirus responsible for Middle Eastern respiratory syndrome (MERS-CoV).3 The SARS-CoV-2 spike glycoprotein (S), which is the main target for neutralizing antibodies, binds to its receptor human angiotensin converting enzyme 2 (hACE2) to initiate infection.4 SARS-CoV-2 is the cause of COVID-19, an infectious disease with respiratory and systemic manifestations. Disease symptoms vary, with many persons presenting with asymptomatic or mild disease and some progressing to severe respiratory tract disease including pneumonia and acute respiratory distress syndrome (ARDS), leading to multiorgan failure and death. In an attempt to prevent the spread of disease and to control the pandemic, numerous COVID￾19 vaccine candidates are in development. These vaccines are based on different platforms including mRNA and DNA technologies and include viral vectored, subunit, inactivated, and live￾attenuated vaccines. Most COVID-19 candidate vaccines express the spike protein or parts of the spike protein, i.e., the receptor binding domain, as the immunogenic determinant. 2.2 EUA Request for the Moderna COVID-19 Vaccine mRNA-1273 ModernaTX, Inc. (Sponsor) is developing a vaccine to prevent COVID-19 that is based on the pre-fusion stabilized SARS-CoV-2 spike glycoprotein (S) antigen encoded by mRNA and formulated in a lipid nanoparticle (LNP). The Moderna COVID-19 Vaccine (also referred to as mRNA-1273) is a 2-dose series of 100-μg intramuscular injections administered 1 month apart. The vaccine is supplied as a multi-dose vial (10 doses) containing a frozen suspension -25º to - 15ºC) of mRNA-1273 that must be thawed prior to administration. The vaccine does not contain a preservative. A Phase 3 randomized and placebo-controlled trial using mRNA-1273 in approximately 30,000 participants is currently ongoing to evaluate the vaccine’s safety and efficacy. A prespecified interim efficacy analysis from 27,817 participants using a data cutoff date of November 7, 2020, demonstrated vaccine efficacy (VE) of 94.5% (95% CI: 86.5%, 97.8%) for the prevention of symptomatic confirmed COVID-19 occurring at least 14 days after the second dose. At the time of this interim analysis, the median efficacy follow-up was 7 weeks post completion of the 2- dose series. Safety data from a November 11, 2020, interim analysis with a median of 7 weeks follow-up after the second dose of vaccine were reported to demonstrate an acceptable tolerability profile with no significant safety concerns. On November 30, 2020, ModernaTX submitted an EUA request to FDA, based on the interim analyses described above, for use of mRNA-1273 to prevent COVID-19 caused by SARS-CoV-2 in individuals 18 years of age and older. On December 7, 2020, the Sponsor submitted an amendment to the EUA request with additional accrued safety data on all participants with a median of 2 months (9 weeks) follow-up after the second dose, using a data cutoff date of November 25, 2020, and data from the prespecified final efficacy analysis using a data cutoff of November 21, 2020, which met the median follow-up of 2 months after dose 2 and demonstrated vaccine efficacy of 94.1% (95% 8 Moderna COVID-19 Vaccine VRBPAC Briefing Document CI: 89.3%, 96.8%) for the prevention of symptomatic confirmed COVID-19 occurring at least 14 days after the second dose. Although the complete datasets and analyses from the primary efficacy analysis and associated safety analyses submitted on December 7, 2020, have not been independently verified by the FDA to the same extent as the data for the interim efficacy analyses and associated safety analyses submitted on November 30, 2020, based on comprehensive independent review of the data from the interim analysis, and the consistency of findings across the two analysis time points, FDA considers that the totality of available data are sufficient to support an evaluation of this product for EUA. 2.3 U.S. Requirements to Support Issuance of an EUA for a Biological Product Based on the declaration by the Secretary of HHS that the COVID-19 pandemic constitutes a public health emergency with a significant potential to affect national security or the health and security of United States citizens living abroad, FDA may issue an EUA after determining that certain statutory requirements are met (section 564 of the Federal Food, Drug, and Cosmetic Act (FD&C Act) (21 U.S.C. 360bbb-3)).5 • The chemical, biological, radiological, or nuclear (CBRN) agent referred to in the March 27, 2020 EUA declaration by the Secretary of HHS (SARS-CoV-2) can cause a serious or life￾threatening disease or condition. • Based on the totality of scientific evidence available, including data from adequate and well￾controlled trials, if available, it is reasonable to believe that the product may be effective to prevent, diagnose, or treat such serious or life-threatening disease or condition that can be caused by SARS-CoV-2, or to mitigate a serious or life-threatening disease or condition caused by an FDA-regulated product used to diagnose, treat, or prevent a disease or condition caused by SARS-CoV-2. • The known and potential benefits of the product, when used to diagnose, prevent, or treat the identified serious or life-threatening disease or condition, outweigh the known and potential risks of the product. • There is no adequate, approved, and available alternative to the product for diagnosing, preventing, or treating the disease or condition. If these criteria are met, under an EUA, FDA can allow unapproved medical products (or unapproved uses of approved medical products) to be used in an emergency to diagnose, treat, or prevent serious or life-threatening diseases or conditions caused by threat agents. FDA has been providing regulatory advice to COVID-19 vaccine manufacturers regarding the data needed to determine that a vaccine’s benefit outweigh its risks. This includes demonstrating that manufacturing information ensures product quality and consistency along with data from at least one phase 3 clinical trial demonstrating a vaccine’s safety and efficacy in a clear and compelling manner. In the event an EUA is issued for this product, it would still be considered unapproved and would continue under further investigation (under an Investigational New Drug Application). Licensure of a COVID-19 vaccine will be based on review of additional manufacturing, efficacy, and safety data, providing greater assurance of the comparability of licensed product to product tested in the clinical trials, greater assurance of safety based on larger numbers of vaccine recipients who have been followed for a longer period of time, and additional information about efficacy that addresses, among other questions, the potential for waning of protection over time. 9 Moderna COVID-19 Vaccine VRBPAC Briefing Document 2.4 Alternatives for Prevention of COVID-19 No vaccine or other medical product is FDA approved for prevention of COVID-19. On December 11, 2020, FDA issued an EUA for the Pfizer-BioNTech COVID-19 vaccine for active immunization for prevention of COVID-19 due to SARS-CoV-2 in individuals 16 years of age and older. However, the Pfizer-BioNTech COVID-19 vaccine is not an approved product, and furthermore is not available in quantity sufficient to vaccinate all persons in the U.S. for whom the vaccine is authorized for use. On October 22, 2020, FDA approved remdesivir for use in adult and pediatric patients 12 years of age and older and weighing at least 40 kilograms for the treatment of COVID-19 requiring hospitalization. Several other therapies are currently available under emergency use authorization, but not FDA approved, for treatment of COVID-19. Thus, there is currently no adequate, approved, and available alternative for prevention of COVID-19. 2.5 Applicable Guidance for Industry Risk and benefit considerations are unique for COVID-19 vaccines, given that an EUA may be requested to allow for a vaccine’s rapid and widespread deployment for administration to millions of individuals, including healthy people. FDA published in October 2020 guidance for industry entitled “Emergency Use Authorization for Vaccines to Prevent COVID-19” describing FDA’s current recommendations regarding the manufacturing, nonclinical, and clinical data and information needed under section 564 of the FD&C Act to support the issuance of an EUA for an investigational vaccine to prevent COVID-19, including a discussion of FDA’s current thinking regarding the circumstances under which an EUA for a COVID-19 vaccine would be appropriate.6 2.6 Safety and Effectiveness Information Needed to Support an EUA Effectiveness data Issuance of an EUA requires a determination that the known and potential benefits of the vaccine outweigh the known and potential risks. For a preventive COVID-19 vaccine to be potentially administered to millions of individuals, including healthy individuals, data adequate to inform an assessment of the vaccine’s benefits and risks and support issuance of an EUA would include meeting the prespecified success criteria for the study’s primary efficacy endpoint, as described in the guidance for industry entitled “Development and Licensure of Vaccines to Prevent COVID-19” (i.e., a point estimate for a placebo-controlled efficacy trial of at least 50%, with a lower bound of the appropriately alpha-adjusted confidence interval around the primary efficacy endpoint point estimate of >30%).7 Safety data An EUA request for a COVID-19 vaccine should include all safety data accumulated from studies conducted with the vaccine, with data from Phase 1 and 2 focused on serious adverse events, adverse events of special interest, and cases of severe COVID-19 among study participants. Phase 3 safety data should include characterization of reactogenicity (common and expected adverse reactions shortly following vaccination) in a sufficient number of participants from relevant age groups and should include a high proportion of enrolled participants (numbering well over 3,000) followed for serious adverse events and adverse events of special interest for at least one month after completion of the full vaccination regimen. The Phase 1 and 2 safety data likely will be of a longer duration than the available safety data from the Phase 3 trial at the time of submission of an EUA request and thus, are intended to complement the available data from safety follow-up from ongoing Phase 3 studies. 10 Moderna COVID-19 Vaccine VRBPAC Briefing Document Phase 3 Follow-up Data from Phase 3 studies should include a median follow-up duration of at least 2 months after completion of the full vaccination regimen to help provide adequate information to assess a vaccine’s benefit-risk profile. From a safety perspective, a 2-month median follow-up following completion of the full vaccination regimen will allow identification of potential adverse events that were not apparent in the immediate postvaccination period. Adverse events considered plausibly linked to vaccination generally start within 6 weeks of vaccine receipt. 8 Therefore, a 2- month follow-up period may allow for identification of potential immune-mediated adverse events that began within 6 weeks of vaccination. From the perspective of vaccine efficacy, it is important to assess whether protection mediated by early responses has not started to wane. A 2-month median follow-up is the shortest follow-up period to achieve some confidence that any protection against COVID-19 is likely to be more than short-lived. The EUA request should include a plan for active follow-up for safety (including deaths, hospitalizations, and other serious or clinically significant adverse events) among individuals administered the vaccine under an EUA in order to inform ongoing benefit-risk determinations to support continuation of the EUA. 2.7 Continuation of Clinical Trials Following Issuance of an EUA for a COVID-19 Vaccine FDA does not consider availability of a COVID-19 vaccine under EUA, in and of itself, as grounds for immediately stopping blinded follow-up in an ongoing clinical trial or grounds for offering vaccine to all placebo recipients. To minimize the risk that use of an unapproved vaccine under EUA will interfere with long-term assessment of safety and efficacy in ongoing trials, it is critical to continue to gather data about the vaccine even after it is made available under EUA. An EUA request should therefore include strategies that will be implemented to ensure that ongoing clinical trials of the vaccine are able to assess long-term safety and efficacy (including evaluating for vaccine-associated enhanced respiratory disease and decreased effectiveness as immunity wanes over time) in sufficient numbers of participants to support vaccine licensure. These strategies should address how ongoing trial(s) will handle loss of follow-up information for study participants who choose to withdraw from the study in order to receive the vaccine under an EUA. FDA is aware that some COVID-19 vaccine developers may wish to immediately unblind their trials upon issuance of an EUA in order to rapidly provide vaccine to trial participants who received placebo. Regardless of when vaccination of placebo recipient would occur, there may be advantages to maintaining blinding in a crossover design that provides vaccine to previous placebo recipients and placebo to previous vaccine recipients. Such strategies would impact collection of longer-term placebo-controlled safety data and evaluation of the duration of vaccine efficacy. Ethical and scientific issues associated with offering vaccination to placebo recipients have been discussed in recent statements and articles. 9-11 2.8 Previous Meetings of the VRBPAC to Discuss Vaccines to Prevent COVID-19 On October 22, 2020, the VRBPAC met in open session to discuss, in general, the development, authorization, and/or licensure of vaccines to prevent COVID-19. No specific application was discussed at this meeting. Topics discussed at the meeting included: • FDA’s approach to safety and effectiveness, and chemistry, manufacturing and control (CMC) data as outlined in the respective guidance documents • Considerations for continuation of blinded Phase 3 clinical trials if an EUA has been issued for an investigational COVID-19 vaccine 11 Moderna COVID-19 Vaccine VRBPAC Briefing Document • Studies following licensure and/or issuance of an EUA for COVID-19 vaccines to: o Further evaluate safety, effectiveness and immune markers of protection o Evaluate the safety and effectiveness in specific populations. On December 10, 2020, the VRBPAC met in open session to discuss the EUA request of the Pfizer-BioNTech COVID-19 Vaccine for the prevention of COVID-19 in individuals 16 years of age older. Topics discussed at the meeting but not voted upon included Pfizer’s plan for continuation of blinded, placebo-controlled follow-up in ongoing trials in the event that the vaccine is made available under EUA and gaps in plans for further evaluation of vaccine safety and effectiveness in populations that receive the Pfizer-BioNTech Vaccine under an EUA. The committee voted in favor of a determination that, based on the totality of scientific evidence available, the benefits of the proposed vaccine outweigh its risks for use in individuals 16 years of age and older. 3. Topics for VRBPAC Discussion The Vaccines and Related Biological Products Advisory Committee will convene on December 17, 2020, to discuss and provide recommendations on whether based on the totality of scientific evidence available, the benefits of the Moderna COVID-19 Vaccine outweigh its risks for use in individuals 18 years of age and older. The Committee will also discuss what additional studies should be conducted by the vaccine manufacturer following issuance of the EUA to gather further data on the safety and effectiveness of this vaccine. 4. Moderna COVID-19 Vaccine (mRNA-1273) 4.1 Vaccine Composition, Dosing Regimen The Moderna COVID-19 Vaccine is a white to off-white, sterile, preservative-free frozen suspension for intramuscular injection. The vaccine contains a synthetic messenger ribonucleic acid (mRNA) encoding the pre-fusion stabilized spike glycoprotein (S) of SARS-CoV-2 virus. The vaccine also contains the following ingredients: lipids (SM-102, 1,2-dimyristoyl-rac-glycero￾3-methoxypolyethylene glycol-2000 [PEG2000-DMG], cholesterol, and 1,2-distearoyl-sn￾glycero-3-phosphocholine [DSPC]), tromethamine, tromethamine hydrochloride, acetic acid, sodium acetate, and sucrose. The Moderna COVID-19 Vaccine is provided as a frozen suspension [stored between -25º to -15ºC (-13º to 5ºF)] multi-dose vial containing 10 doses. The vaccine must be thawed prior to administration. After thawing, a maximum of 10 doses (0.5 mL each) can be withdrawn from each vial. Vials can be stored refrigerated between 2° to 8°C (36° to 46°F) for up to 30 days prior to first use. Unopened vials may be stored between 8° to 25°C (46° to 77°F) for up to 12 hours. After the first dose has been withdrawn, the vial should be held between 2° to 25°C (36° to 77°F) and discarded after 6 hours. The Moderna COVID-19 Vaccine, mRNA-1273 (100 μg) is administered intramuscularly as a series of two doses (0.5 mL each), given 28 days apart. FDA has reviewed the CMC data submitted to date for this vaccine and has determined that the CMC information is consistent with the recommendations set forth in FDA’s Guidance on Emergency Use Authorization for Vaccines to Prevent COVID-19. FDA has determined that the Sponsor has provided adequate information to ensure the vaccine’s quality and consistency for authorization of the product under an EUA. 12 Moderna COVID-19 Vaccine VRBPAC Briefing Document 4.2 Proposed Use Under EUA The proposed use of the vaccine under an EUA is for the prevention of COVID-19 in adults 18 years of age and older. 5. FDA Review of Clinical Safety and Effectiveness Data 5.1 Overview of Clinical Studies Data from three ongoing clinical studies were included in the EUA request, which are summarized in Table 1 below. Study mRNA-1273-P301 is a multi-center, Phase 3 randomized, blinded, placebo-controlled safety, immunogenicity, and efficacy study that is the focus of the EUA review. Study mRNA1273-P201 is a Phase 2 dose-confirmation study that explored 2 dose levels of mRNA-1273 and will not be discussed in detail. Study 20-0003 is a Phase 1 open label, dose-ranging, first-in-human study of mRNA-1273 and will also not be discussed in detail. A brief summary of the P201 and 20-0003 study designs and results to date is found in Appendix A, page 53. Table 1. Clinical Trials Submitted in Support of Efficacy and Safety Determinations of the Moderna COVID-19 Vaccine mRNA-1273 Study Number Type of Study (Efficacy, Safety, Nonclinical) Participants randomized (N) Study Design & Type of Control Test Product(s); Dosing Regimens Study Status P301 Efficacy, Safety 30418 A Phase 3, randomized, stratified, observer-blind, placebo￾controlled study mRNA-1273 100 μg Ongoing￾vaccine efficacy demonstrated at the 1st interim analysis P201 Safety, Immunogenicity 600 A Phase 2a, randomized, observer-blind, placebo￾controlled, dose￾confirmation study mRNA-1273 50ug,100μg Ongoing￾Day 57 primary analysis have completed 20-0003* Safety, Immunogenicity 120 A Phase 1 Open-label dose-ranging study mRNA-1273 25ug 50ug,100ug 250ug Ongoing￾Day 119 (25ug, 100ug, 250ug), Day 57 (50ug) *Sponsor: Division of Microbiology and Infectious Diseases (DMID), National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health 5.2 Study mRNA-1273-P301 5.2.1 Design Study mRNA-1273-P301 is an ongoing randomized, stratified, observer-blind, placebo￾controlled study to evaluate the efficacy, safety and immunogenicity of mRNA-1273 administered in 2 doses 28 days apart in adults 18 years of age and older. The study took place in 99 sites in the United States. Participants (N=30,351) were randomized 1:1 to receive intramuscular injections of either 100 µg of mRNA-1273 vaccine (n=15,181) or placebo 13 Moderna COVID-19 Vaccine VRBPAC Briefing Document (n=15,170) on Day 1 and Day 29. Participants were stratified by age and health risk into one of three groups: 18 to <65 years of age and not at risk for progression to severe COVID-19, 18 to <65 years of age and at risk for progression to severe COVID-19, and ≥65 years of age, with the latter two groups consisting of 41.4% of the study population. Participants were considered at risk for progression to severe COVID-19 if they had underlying comorbidities including diabetes, chronic lung disease, severe obesity, significant cardiovascular disease, liver disease, or infection with HIV. The study included 24,907 (82.1%) participants considered at occupational risk for acquiring SARS-CoV-2 infection, of whom 7,613 (25.1%) were healthcare workers. Other essential workers were also represented. The primary efficacy endpoint was efficacy of the vaccine to prevent protocol-defined COVID-19 occurring at least 14 days after the second dose in participants with negative SARS-CoV-2 status at baseline (i.e., negative RT-PCR and negative serology against SARS-CoV-2 nucleocapsid on Day 1). Symptoms of COVID-19 experienced by participants during post-vaccination follow-up prompted an unscheduled illness visit and nasopharyngeal (NP) swab. NP samples were tested for SARS CoV-2 at a central laboratory using a reverse transcription-polymerase chain reaction (RT-PCR) test (Viracor; FDA authorized under EUA), or other sufficiently validated nucleic acid amplification-based test (NAAT). The central laboratory NAAT result is used for the case definition, unless it is not possible to test the sample at the central laboratory. The case-driven study design required 151 COVID-19 cases to trigger the final scheduled efficacy analysis. Two interim analysis timepoints were pre-specified; the first upon accrual of 53 cases and the second upon accrual of 106 cases. The expected duration of study participation is approximately 25 months. Primary Efficacy Endpoint The primary efficacy endpoint was efficacy of the vaccine to prevent protocol-defined COVID-19 occurring at least 14 days after the second dose in participants with negative SARS-CoV-2 status at baseline (i.e., negative RT-PCR and negative serology against SARS-CoV-2 nucleocapsid on Day 1). The primary analysis was based on the Per-Protocol Set, defined as all randomized, baseline SARS-CoV-2 negative participants who received planned doses per schedule and have no major protocol deviations. For the primary efficacy endpoint, the case definition for a confirmed COVID-19 case was defined as: • At least TWO of the following systemic symptoms: Fever (≥38ºC), chills, myalgia, headache, sore throat, new olfactory and taste disorder(s), or • At least ONE of the following respiratory signs/ symptoms: cough, shortness of breath or difficulty breathing, OR clinical or radiographical evidence of pneumonia; and • NP swab, nasal swab, or saliva sample (or respiratory sample, if hospitalized) positive for SARS-CoV-2 by RT-PCR. Vaccine efficacy was defined as the percent reduction (mRNA-1273 vs. placebo) in the hazard of the primary endpoint, i.e. VE =1 – Hazard Ratio (HR). A stratified Cox proportional hazard (PH) model using Efron's method to handle ties and with treatment group as the independent variable was used to estimate the HR, where the same stratification factor used for randomization was applied. The primary objective would be met if the null hypothesis of H0: VE ≤30% is rejected at any of the interim or primary analyses at the respective significance level. The final scheduled efficacy analysis of the primary endpoint was planned when a total of 151 adjudicated cases occurring at least 14 days after the second injection had been accrued. In addition, two interim analyses were planned when 35% (53 cases) and 70% (106 cases) of the 14 Moderna COVID-19 Vaccine VRBPAC Briefing Document total target number of cases had been accrued. The Lan-DeMets spending function was used for approximating O’Brien-Fleming efficacy bounds to preserve the overall Type I error rate at a one-sided α =0.025, yielding nominal one-sided α of 0.0002, 0.0073, and 0.0227 at the first and second interim and the primary analyses, respectively. As conducted, the first and only interim analysis in the study occurred at 95 adjudicated cases of the primary endpoint, where the null hypothesis of H0: VE ≤30% was evaluated at a one-sided alpha of 0.0047. Secondary Efficacy Endpoints Secondary endpoints based on the Per-Protocol Set included the VE of mRNA-1273 to prevent the following: • Severe COVID-19 (as defined below) • COVID-19 based on a less restrictive definition of disease (defined below) occurring at least 14 days after the second dose of vaccine • Death due to COVID-19 • COVID-19 occurring at least 14 days after the first dose of vaccine (including cases that occurred after the second dose) One additional secondary endpoint was based on the Full Analysis Set (FAS): VE of mRNA￾1273 to prevent COVID-19 occurring at least 14 days after the second dose, regardless of prior SARS-CoV-2 infection. One of the secondary efficacy endpoints assessed COVID-19 as defined by a less restrictive definition: a positive NP swab, nasal swab, or saliva sample (or respiratory sample, if hospitalized) for SARS-CoV-2 by RT-PCR and one of the following systemic symptoms: • fever (temperature ≥38ºC), or • chills, • cough, • shortness of breath or difficulty breathing, • fatigue, • muscle aches or body aches, • headache, • new loss of taste or smell, • sore throat, • nasal congestion or rhinorrhea, • nausea or vomiting, or diarrhea Another secondary endpoint assessed cases of severe COVID-19, defined as a case of confirmed COVID-19 plus at least one of the following: • Clinical signs at rest indicative of severe systemic illness (RR ≥30 breaths per minute, HR ≥125 beats per minute, SpO2≤93% on room air at sea level, or PaO2/FiO2<300 mm Hg); • Respiratory failure or Acute Respiratory Distress Syndrome, (defined as needing high-flow oxygen, noninvasive ventilation, mechanical ventilation, or ECMO); • Evidence of shock (SBP <90 mm Hg, DBP <60 mm Hg, or requiring vasopressors) • Significant acute renal, hepatic, or neurologic dysfunction; • Admission to an ICU; • Death 15 Moderna COVID-19 Vaccine VRBPAC Briefing Document Vaccine efficacy of secondary endpoints was estimated from the Cox proportional-hazards model when the primary endpoint reached statistical significance. Estimates based on the Per￾Protocol Set were presented with nominal two-sided 95% confidence intervals. Analysis Populations For the purposes of analysis, the following populations are defined: Table 2. Efficacy Set Definitions Population Description Randomized All participants who are randomized, regardless of the participants’ treatment status in the study. Full Analysis Set All randomized participants who received at least one dose of Investigational Product (IP). mITT Set All participants in the FAS who had no immunologic or virologic evidence of prior COVID-19 (i.e., negative NP swab test at Day 1 and/or bAb against SARS-CoV-2 nucleocapsid below limit of detection [LOD] or lower limit of quantification [LLOQ]) at Day 1 before the first dose of IP. Per Protocol Set All participants in the mITT Set who received planned doses of IP per schedule and have no major protocol deviations, as determined and documented by Sponsor prior to DBL and unblinding, that impact critical or key study data. Safety Set All randomized participants who received at least one dose of IP. Solicited Safety Set All randomized participants who received at least one dose of IP and contributed any solicited adverse reaction data. Evaluation of Safety The primary safety objective for all phases was to describe the safety of mRNA-1273 after 1 or 2 doses. In all studies, participants recorded local reactions, systemic events, and antipyretic/pain medication usage from Day 1 through Day 7 after each dose. Unsolicited adverse events (AEs) are collected from dose 1 to 28 after the last dose and medically attended adverse events (MAAEs) and serious AEs (SAEs) from dose 1 to the end of the study. Figure 1 below shows the study safety monitoring plan. Figure 1. Safety Monitoring Plan, Study 301 16 Moderna COVID-19 Vaccine VRBPAC Briefing Document Safety assessments included the following: • Solicited local and systemic adverse reactions (AR) that occurred during the 7 days following each dose (i.e., the day of vaccination and 6 subsequent days). Solicited ARs were recorded daily using eDiaries. • Unsolicited AEs observed or reported during the 28 days following each dose (i.e., the day of vaccination and 27 subsequent days). Unsolicited AEs are those not included in the protocol-defined solicited AR. • AEs leading to discontinuation from vaccination and/or study participation from Day 1 through Day 759 or withdrawal from the study. • Medically Attended Adverse Events (MAAE) from Day 1 through Day 759 or withdrawal from the study. • Serious Adverse Events (SAEs) from Day 1 through Day 759 or withdrawal from the study. • Abnormal vital sign measurements. • Physical examination findings. • Pregnancy and accompanying outcomes. Safety laboratory evaluations were not assessed in Study P301 but were collected in the phase 2 Study P201. See Appendix A on page 53. Potential COVID-19 illnesses and their sequelae were not to be reported as AEs, with the exception of illnesses that met regulatory criteria for seriousness and were not confirmed to be COVID-19. Such illnesses were evaluated and reported as SAEs. Monitoring for risk of vaccine-enhanced disease was performed by an unblinded team supporting the Data Monitoring Committee that reviewed cases of severe COVID-19 as they were received and reviewed AEs at least weekly for additional potential cases of severe COVID￾19. The stopping rule was triggered when the 1-sided probability of observing the same or a more extreme case split was 5% or less when the true incidence of severe disease was the same for vaccine and placebo participants. The table below shows the Phase 3 safety analyses populations that were used to determine the proportions of study participants who experienced adverse events, including solicited adverse reactions after each dose, unsolicited adverse events, medically attended adverse events, and serious adverse events. Table 3. Safety Set Definitions Population Description Randomized Set All participants who are randomized, regardless of the participants treatment status in the study. Safety Set All randomized participants who received at least one dose of investigational product. The safety set was used for all analyses of safety except solicited adverse reactions. Participants were included in the treatment group corresponding to the investigational product they received. Solicited Safety Set All randomized participants who received at least one dose of investigational product and contributed any solicited adverse reaction data. The solicited safety set was used for the analyses of solicited adverse reactions. Participants were included in the treatment group corresponding to the investigational product they received. Solicited Safety Set-1st Injection All randomized participants who received the 1st dose and provided any solicited reaction data. 17 Moderna COVID-19 Vaccine VRBPAC Briefing Document Population Description Solicited Safety Set-2nd Injection All randomized participants who received the 2nd dose and provided any solicited reaction data. 5.2.2 FDA Assessment of Phase 3 Follow-Up Duration As of the interim analysis cutoff (November 7, 2020, for efficacy, November 11, 2020, for safety), the proportion of participants across groups who received one dose of vaccine or placebo was 100%, and the proportion of participants who received two doses was 91.9% (92.1% vaccine, 91.7% placebo). The median follow-up after dose 2 was 7 weeks across groups. (For participants who did not receive a second dose of vaccine or placebo, follow-up after dose 2 was zero. Among participants who received dose 2, the median follow-up after the second dose was 50.0 days.) The proportion of participants with at least 1 month of follow-up after dose 2 was 76.7% (77.2% vaccine, 76.2% placebo) and with at least 2 months follow-up after dose 2 was 25.3% (25.7% vaccine, 24.9% placebo). FDA has completed its independent validation and evaluation of the datasets from which the Sponsor’s interim safety and efficacy analyses were derived. A second safety data cutoff was performed on November 25, 2020, and final efficacy analysis performed with a data cutoff of November 21, 2020, when 196 primary endpoint cases accrued. These data include a median follow-up of 2 months (9 weeks) for both efficacy and safety. The proportion of participants with at least 1 month of follow-up after dose 2 was 87.9% (88.2% vaccine, 87.7% placebo) and with at least 2 months follow-up after dose 2 was 53.6% (53.8% vaccine, 53.5% placebo). The Sponsor submitted analyses from the final efficacy analysis (Tables, Figures and Listings) on December 4, 2020, and safety analyses (Tables, Figures and Listings) on December 7, 2020, for FDA review under the EUA. Datasets were also submitted on December 7, 2020 and validated by FDA by December 8, 2020. The review of the second dataset submission for the final scheduled efficacy analysis and safety data through November 25, 2020, was not as comprehensive as that of the interim efficacy data and safety data first submitted in support of the EUA. However, preliminary assessments of safety and efficacy data and analyses from second data cutoff do not demonstrate any notable differences compared with the efficacy and safety analyses from November 7, 2020, and November 11, 2020, respectively, and key safety and efficacy data (e.g., the primary analysis, cases of severe COVID-19, and serious adverse events) from the December 7, 2020, submission were verified. FDA therefore considers the totality of submitted data to satisfy the expectation of a median of 2 months follow-up after completion of the full vaccination regimen. 5.2.3 Participant Disposition and Inclusion in Analysis Populations Disposition tables are presented below in Table 4 (Per-Protocol Set) and Table 5 (Safety Set). The proportion of participants excluded from the Per-Protocol Set was balanced between treatment groups, with the majority of those excluded due to positive or unknown baseline SARS-CoV-2 status. Overall, few participants were discontinued or lost to follow-up, and these and other analysis population exclusions were generally balanced between treatment groups. In the per protocol population, 26.3% of vaccine recipients and 25.7% of placebo recipients completed at least 2 months follow-up after dose 2. 18 Moderna COVID-19 Vaccine VRBPAC Briefing Document Table 4. Efficacy Analysis Population Study Dispositiona, mRNA-1273-P301 Disposition Vaccine Group (N=15208) n (%) Placebo Group (N=15210) n (%) Total (N=30418) n (%) Randomized 15208 15210 30418 Full Analysis Set 15180 (99.8) 15170 (99.7) 30350 (99.8) Modified Intent-to-Treat Set 14312 (94.1%) 14370 (94.5%) 28682 (94.3) Participants excluded from PP set 1274 (8.4%) 1327 (8.7%) 2601 (8.6%) Randomized but received no Investigational Product (IP) 28 (0.2%) 40 (0.3%) 68 (0.2%) Baseline SARS-CoV-2 status was positive or not known 868 (5.7%) 800 (5.3%) 1668 (5.5) Received IP other than what the participant was randomized to 5 (<0.1) 7 (<0.1) 12 (<0.1) Discontinued study or study vaccine without receiving the second dose 136 (0.9) 203 (1.3) 339 (1.1) Did not receive second dose of IP 144 (0.9) 155 (1.0) 299 (1.0) Received vaccine out of window 81 (0.5) 98 (0.6) 179 (0.6) Major protocol deviation 12 (<0.1) 24 (0.2) 36 (0.1) Per Protocol Set 13934 (91.6) 13883 (91.3) 27817 (91.4) Completed 1 dose** 13934 (100) 13883 (100) 27817 (100) Completed 2 doses** 13218 (94.9) 13164 (94.8) 26382 (94.8) Completed at least 7 weeks follow-up after dose 2** 7293 (52.3) 7304 (52.6) 14597 (52.5) Completed at least 2 months follow-up after dose 2** 3669 (26.3) 3568 (25.7) 7237 (26.0) Discontinued from Study** 24 (0.2) 34 (0.2) 58 (0.2) Reason for Discontinuation** Adverse Event 0 0 0 Death 0 1 (<0.1) 1 (<0.1) Withdrawal by Participant 18 (0.1) 22 (0.2) 40 (0.1) Lost to Follow-up 2 (<0.1) 9 (<0.1) 11 (<0.1) Protocol Deviation 0 0 0 Physician Decision 2 (<0.1) 0 2 (<0.1) Other 2 (<0.1) 2 (<0.1) 4 (<0.1) Source: Sponsor’s Table 14.1.1.1.1.1, Table 4.1.2.1, Table 14.1.1.1.3.2, Table 14.1.6.2 a EUA request (interim analysis): November 11, 2020 cutoff *Percentage based on number of participants in the Safety Set **Percentage based on number of participants in the Per-Protocol Set Based on the November 11, 2020 safety data cutoff, an overview of participant disposition is presented in the table below. The proportion of randomized participants who discontinued from the study was 0.9% (288 participants) across study groups, with a greater number in the placebo group (168) compared with the vaccine group (120). The most frequently reported reason was withdrawal of consent (67 participants in the vaccine group, 120 in the placebo group). In addition, 51 participants were lost to follow-up (20 in the vaccine group, 31 in the placebo group). In the vaccine group, 3 participants withdrew due to an adverse event (<0.1%, including 1 participant who withdrew due to a SAE) and 3 participants died during the study. In the placebo group, no participants withdrew due to an adverse event, and 4 participants died during the study. 19 Moderna COVID-19 Vaccine VRBPAC Briefing Document Table 5. Safety Analysis Population Study Dispositiona, mRNA-1273-P301 Disposition Vaccine Group (N=15208) n (%) Placebo Group (N=15210) n (%) Total (N=30418) n (%) Randomized 15208 15210 30418 Completed 1 dose 15180 (99.8) 15170 (99.7) 30350 (99.8) Completed 2 doses 13982 (91.9) 13916 (91.5) 27898 (91.7) Exposed (Safety Set) 15184 15166 30350 (99.8) Discontinued from Study 120 (0.8) 168 (1.1) 288 (0.9) Reason for Discontinuation Adverse Event 3 (<0.1) 0 3 (<0.1) Death 3 (<0.1) 4 (<0.1) 7 (<0.1) Withdrawal by Participant 67 (0.4) 120 (0.8) 187 (0.6) Lost to Follow-up 20 (0.1) 31 (0.2) 51 (0.2) Protocol Deviation 1 (<0.1) 1 (<0.1) 2 (<0.1) Physician Decision 17 (0.1) 2 (<0.1) 19 (<0.1) Other 9 (<0.1) 10(<0.1) 19 (<0.1) Completed ≥1 month f/up* 14354 (94.5) 14345 (94.6) 28700 (94.6) Completed ≥2 months f/up* 12021 (79.2) 11974 (79.0) 23995 (79.1) Completed ≥1 month f/up after dose 2* 11717 (77.2) 11559 (76.2) 23276 (76.7) Completed ≥2 months f/up after dose 2* 3894 (25.7) 3773 (24.9) 7667 (25.3) Source: Sponsor’s Table 14.1.1.1.1.1, Table 4.1.2.1, Table 14.1.1.1.3.2, Table 14.1.6.2. a EUA request (interim analysis): November 11, 2020 cutoff 5.2.4 Demographics and Other Baseline Characteristics The Per-Protocol Set included 47.4% females and 25.3% of individuals ≥65 years of age. There were 36.5% of participants considered as representing communities of color with 9.7% African American, 4.7% Asian, and <3% from other racial groups; 20% of participants were Hispanic/Latino. A majority of the participants (82%) were considered at occupational risk for SARS-CoV-2 exposure, with 25.4% of participants being healthcare workers. At least one protocol-defined high-risk condition for severe COVID-19 was present in 22.3% of participants, and 4% of participants had two or more high risk conditions. The protocol-specified risk factors were those conditions that placed an individual at increased risk for severe complications of COVID-19 and were selected based on CDC recommendations12 from March 2020. These conditions included the following: • Chronic lung disease (e.g., emphysema and chronic bronchitis), idiopathic pulmonary fibrosis and cystic fibrosis) or moderate to severe asthma • Significant cardiac disease (e.g., heart failure, coronary artery disease, congenital heart disease, cardiomyopathies, and pulmonary hypertension) • Severe obesity (body mass index ≥40 kg/m2) • Diabetes (Type 1, Type 2 or gestational) • Liver disease • HIV infection There was a similar distribution of demographic characteristics between the treatment groups as well as between the all randomized population, Full Analysis Set, and the Per-Protocol Set. 20 Moderna COVID-19 Vaccine VRBPAC Briefing Document Table 6. Demographic Characteristicsa, Per-Protocol Set Characteristic Vaccine Group (N=13934) n (%) Placebo Group (N=13883) n (%) Total (N=27817) n (%) Sex Female 6661 (47.8) 6514 (46.9) 13175 (47.4) Male 7273 (52.2) 7369 (53.1) 14642 (52.6) Age (years) Mean (SD) 51.6 (15.45) 51.5 (15.55) 51.6 (15.50) Median 53.0 52.0 53.0 Min, max 18, 95 18, 95 18, 95 Age- subgroups (years) 18 to <65 10407 (74.7) 10384 (74.8) 20791 (74.7) 65 and older 3527 (25.3) 3499 (25.2) 7026 (25.3) Race American Indian or Alaska Native 107 (0.8) 110 (0.8) 217 (0.8) Asian 616 (4.4) 684 (4.9) 1300 (4.7) Black or African American 1369 (9.8) 1338 (9.6) 2707 (9.7) Native Hawaiian or Other Pacific Islander 33 (0.2) 30 (0.2) 63 (0.2) White 11078 (79.5) 11005 (79.3) 22083 (79.4) Other 298 (2.1) 293 (2.1) 591 (2.1) Ethnicity Hispanic or Latino 2783 (20.0) 2769 (19.9) 5552 (20.0) Not Hispanic or Latino 11019 (79.1) 10987 (79.1) 22006 (79.1) Race and Ethnicity Non-Hispanic white 8858 (63.6) 8755 (63.1) 17613 (63.3) Communities of color 5054 (36.3) 5102 (36.7) 10156 (36.5) Occupational Risk* 11397 (81.8) 11408 (82.2) 22805 (82.0) Healthcare worker 3541 (25.4) 3531 (25.4) 7072 (25.4) High Risk Condition** No high risk condition 11820 (77.9) 11788 (77.7) 23608 (77.8) One high risk condition present 3116 (22.4) 3075 (22.1) 6191 (22.3) Two or more high risk conditions present 561 (4.0) 554 (4.0) 1115 (4.0) Age and Health Risk for Severe COVID-19*** 18 to <65 years and not at risk 8309 (59.6) 8323 (60.0) 16632 (59.8) 18 to <65 years and at risk 2098 (15.1) 2061 (14.8) 4159 (15.0) ≥65 years 3527 (25.3) 3499 (25.2) 7026 (25.3) Source: Sponsor’s Table14.1.3.4.2. a EUA request (interim analysis): November 11, 2020 data cutoff. Occupational risk includes: Healthcare Workers, Emergency Response, Retail/Restaurant Operations, Manufacturing and Production Operations, Warehouse Shipping and Fulfillment centers, Transportation and Delivery Services, Border Protection and Military Personnel, and Personal care and in-home services, Hospitality and Tourism Workers, Pastoral, Social or Public Health Workers, Educators and Students. ** High risk is defined as patients who meet at least one of the following criteria (protocol-defined): Chronic lung disease (eg, emphysema and chronic bronchitis, idiopathic pulmonary fibrosis, and cystic fibrosis) or moderate to severe asthma; Significant cardiac disease (eg, heart failure, coronary artery disease, congenital heart disease, cardiomyopathies, and pulmonary hypertension); Severe obesity (body mass index ≥40 kg/m2); Diabetes (Type 1, Type 2 or gestational); Liver disease; Human Immunodeficiency Virus (HIV) infection *** Age and health risk for severe COVID-19 is used as stratification factor for randomization. 21 Moderna COVID-19 Vaccine VRBPAC Briefing Document The demographic characteristics among vaccine and placebo participants in the safety population were similar. There were no significant imbalances in demographic and other baseline characteristics between the per-protocol population and the safety population, with median 7-week follow-up. Table 7. Demographic Characteristicsa, Safety Set Characteristic Vaccine Group (N=15184) n (%) Placebo Group (N=15165) n (%) Total (N=30350) n (%) Sex Female 7255 (47.8) 7100 (46.8) 14355 (47.3 Male 7929 (52.2) 8065 (53.2) 15995 (52.7) Age (years) Mean (SD) 51.4 (15.50) 51.3 (15.60) 51.4 (15.55) Median 53.0 52.0 52.0 Min, max 18, 95 18, 95 18, 95 Age – Subgroups (years) ≥18 to <65 11414 (75.2) 11415 (75.3) 22830 (75.2) 65 and older 3770 (24.8) 3750 (24.7) 7520 (24.8) Race American Indian or Alaska Native 110 (0.7) 120 (0.8) 230 (0.8) Asian 653 (4.3) 732 (4.8) 1385 (4.6) Black or African American 1562 (10.3) 1528 (10.1) 3090 (10.2) Native Hawaiian or other Pacific islander 34 (0.2) 32 (0.2) 66 (0.2) White 12032 (79.2) 11990 (79.1) 24023 (79.2) Other 321 (2.1) 315 (2.1) 636 (2.1) Multiracial 315 (2.1) 319 (2.1) 634 (2.1) Ethnicity Hispanic or Latino 3121 (20.6) 3112 (20.5) 6234 (20.5) Not Hispanic or Latino 11920 (78.5) 11914 (78.6) 23834 (78.5) Race and Ethnicity Non-Hispanic White 9534 (62.8) 9458 (62.4) 18992 (62.6) Communities of color 5624 (37.0) 5680 (37.5) 11305 (37.2) Occupational Risk* 12420 (81.8) 12487 (82.3) 24907 (82.1) Healthcare worker 3787 (24.9) 3826 (25.2) 7613 (25.1) High Risk Condition** One high risk condition present 3360 (22.1) 3382 (22.3) 6742 (22.2) No high risk condition 11824 (77.9) 11783 (77.7) 23608 (77.8) Age and Health Risk for Severe COVID-19*** ≥18 to <65 years and not at risk 8889 (58.5) 8884 (58.6) 17773 (58.6) ≥18 to <65 years and at risk 2530 (16.7) 2534 (16.7) 5065 (16.7) ≥65 years 3765 (24.8) 3747 (24.7) 7512 (24.8) 22 Moderna COVID-19 Vaccine VRBPAC Briefing Document Characteristic Vaccine Group (N=15184) n (%) Placebo Group (N=15165) n (%) Total (N=30350) n (%) Baseline SARS CoV-2 status**** Negative 14316 (94.3%) 14366 (94.7) 26862 (94.5%) Positive 341 (2.2%) 334 (2.2%) 675 (2.2%) Missing 527 (3.5%) 465 (3.5%) 993 (3.3%) Source: Sponsor’s Table 14.1.3.2.2 a EUA request (interim analysis): November 11 2020 cutoff. * Occupational risk includes: Healthcare Workers, Emergency Response, Retail/Restaurant Operations, Manufacturing and Production Operations, Warehouse Shipping and Fulfillment centers, Transportation and Delivery Services, Border Protection and Military Personnel, and Personal care and in-home services, Hospitality and Tourism Workers, Pastoral, Social or Public Health Workers, Educators and Students.** **High risk is defined as patients who meet at least one of the following criteria (protocol-defined): Chronic lung disease (eg, emphysema and chronic bronchitis, idiopathic pulmonary fibrosis, and cystic fibrosis) or moderate to severe asthma; Significant cardiac disease (eg, heart failure, coronary artery disease, congenital heart disease, cardiomyopathies, and pulmonary hypertension); Severe obesity (body mass index ≥40 kg/m2); Diabetes (Type 1, Type 2 or gestational); Liver disease; Human immunodeficiency virus (HIV) infection The following table provides the proportions of participants randomized to each of the protocol￾specified strata based on presence or absence of protocol-defined risk factors for severe COVID-19 disease, including age ≥65 years. The presence of these risk factors was assessed at screening via review of the participants medical history. The protocol specified that at least 25% (and up to 50%) of enrolled participants were to be either ≥65 years of age or 18 through <65 years of age with a protocol-defined risk factor. As of the November 11, 2020 cutoff, ~25% of participants were age ≥65 years, and 16.7% of participants were age 18 to <65 years with a protocol-defined risk factor. The remainder of participants (58.6%) were age 18 to <65 years without risks. The proportions of participants in each of these three strata randomized to vaccine or placebo are shown in the table below. Table 8. Protocol-Defined Risk for Severe COVID-19 Disease, Safety Seta Participants Risk Categories Vaccine Group (N=15184) n (%) Placebo Group (N=15165) n (%) Total (N=30350) n (%) Without Any Protocol Risk for Severe COVID-19 11824 (77.9) 11783 (77.7) 23608 (77.8) With Any Protocol Risk for Severe COVID-19 3360 (22.1) 3382 (22.3) 6742 (22.2) Chronic Lung Disease 707 (4.7) 741 (4.9) 1448 (4.8) Significant Cardiac Disease 742 (4.9) 741 (4.9) 1483 (4.9) Severe Obesity 986 (6.5) 978 (6.4) 1964 (6.5) Diabetes 1427 (9.4) 1431 (9.4) 2858 (9.4) Liver Disease 100 (0.7) 96 (0.6) 196 (0.6) HIV Infection 90 (0.6) 86 (0.6) 176 (0.6) Source: Sponsor’s Table 14.1.3.2.2. a EUA request (interim analysis): November 11, 2020 cutoff 5.2.5 Vaccine Efficacy Interim Primary Efficacy Analysis The interim primary efficacy analysis was based on the Per-Protocol Set, which consisted of all participants with negative baseline SARS-CoV-2 status (i.e., negative RT-PCR for SARS-CoV-2 at Day 1 and/or negative serology against SARS-CoV-2 nucleocapsid) and who received 2 doses of investigational product per schedule with no major protocol deviations. The primary efficacy endpoint was vaccine efficacy (VE) in preventing protocol defined COVID-19 occurring at least 14 days after dose 2. Cases were adjudicated by a blinded committee. The primary 23 Moderna COVID-19 Vaccine VRBPAC Briefing Document efficacy success criterion would be met if the null hypothesis of VE ≤30% was rejected at the O’Brien Fleming boundary at either the interim or primary analysis. The efficacy analysis presented is based on the data at the first pre-specified interim analysis timepoint consisting of 95 adjudicated cases. As shown in Table 9, in participants ≥18 years of age, there were 5 COVID-19 cases in the vaccine group and 90 COVID-19 cases in the placebo group, with a VE of 94.5%, a lower bound of the 95% CI of 86.5%, and a one-sided p-value of <0.0001 for testing H0: VE ≤30%, which met the pre-specified success criterion. In participants ≥65 years of age in the Per-Protocol Set, there were no COVID-19 cases in the vaccine group and 15 COVID-19 cases in the placebo group. Table 9. Interim Analysisa for Primary Efficacy Endpoint, COVID-19 Starting 14 Days After the 2nd Dose, Per-Protocol Set Primary Endpoint: COVID-19 (per adjudication committee assessment) Vaccine Group N=13934 Cases n (%) (Incidence rate per 1,000 person￾years) Placebo Group N=13883 Cases n (%) (Incidence rate per 1,000 person￾years) Vaccine Efficacy (VE) % (95% CI)* Met Predefined Success Criterion** All participants 5 (<0.1) 1.840 90 (0.6) 33.365 94.5% (86.5%, 97.8%) Yes 18 to <65 5 / 10407 (<0.1) 2.504 75 / 10384 (0.7) 37.788 93.4% (83.7%, 97.3%) NA 65 and older 0 / 3527 15 / 3499 (0.4) 21.046 100% NA Source: Sponsor’s Table 14.2.2.1.1.1.1, Table 14.2.2.1.1.6.1.1. COVID-19: symptomatic COVID-19 requiring positive RT-PCR result and at least 2 systemic symptoms or 1 respiratory symptom. Cases starting 14 days after the 2nd dose. All potential COVID-19 cases starting 14 days after the 2nd dose in the clinical database as of 07-Nov-2020 have been sent to adjudication committee, and have been adjudicated for this analysis (07-Nov-2020 is the data cutoff date for efficacy). One case (in the placebo group) was assessed as a case by the adjudication committee but did not meet case definition based on statistical analysis plan (participant had body aches, nasal congestion, rhinorrhea, which were not protocol defined symptoms). * VE is calculated as 1-ratio of incidence rates (mRNA-1273/placebo) and 95% CI from the stratified Cox proportional hazard model. **The one-sided p-value is <0.0001 from the stratified Cox proportional hazard model to test the null hypothesis of VE ≤30%, achieving the pre-specified efficacy boundary: the one-sided nominal alpha of 0.0049 based on 95 cases using the Lan-DeMets O'Brien-Fleming spending function. There were an additional 18 COVID-19 cases which met the protocol-defined primary efficacy endpoint but were not able to be adjudicated in time for the interim analysis. Of these 18 cases, one was in the vaccine group, and 17 were in the placebo group. Vaccine efficacy for the primary efficacy endpoint including these unadjudicated cases was similar to the results presented above. Interim Subgroup Analyses of Vaccine Efficacy Subgroup analyses for the primary efficacy endpoint include VE based on age, sex, race and ethnicity, risk factor, and baseline SARS-CoV-2 status and provide additional information on the applicability of these results across the general population. In general, VE among the subgroups are similar to the VE seen in the overall study population. The small number participants and cases in some subgroups, such as participants >75 years of age and participants in certain racial subgroups, limits the interpretability of the individual VE results, but are displayed for completeness. 24 Moderna COVID-19 Vaccine VRBPAC Briefing Document Table 10. Subgroup Analyses of Vaccine Efficacya, COVID-19 14 Days After Dose 2 Per Adjudication Committee Assessments, Per-Protocol Set Subgroup Vaccine Group Cases / N (%) Incidence rate per 1,000 person-years Placebo Group Cases / N (%) Incidence rate per 1,000 person-years VE % (95% CI)* Age (years) 18 to <65 5 / 10407 (<0.1) 2.504 75 / 10384 (0.7) 37.788 93.4% (83.7%, 97.3%) 65 to <75 0 / 2904 12/ 2823 (0.4) 20.883 100% 75 and older 0 / 623 3 / 676 (0.4) 21.726 100% Age and risk for severe COVID-19** 18 and <65 and not at risk 4 / 8309 (<0.1) 2.524 57 / 8323 (0.7) 36.034 93.0% (80.8%, 97.5%) 18 and <65 and at risk 1 / 2098 (<0.1) 2.428 18 / 2061 (0.9) 44.673 94.6% (59.4%, 99.3%) >65 0 / 3527 15 / 3499 (0.4) 21.046 100% Sex Female 3 / 6661 (<0.1) 2.271 45 / 6514 (0.7) 34.991 93.5% (79.2%, 98.0%) Male 2 / 7273 (<0.1) 1.433 45 / 7369 (0.6) 31.883 95.5% (81.5%, 98.9%) Race and Ethnicity Non-Hispanic white 5 / 8858 (<0.1) 2.657 70 /8755 (0.8) 37.721 93.0% (82.6%, 97.2%) Communities of color 0 / 5054 20 / 5102 (0.4) 23.892 100% Ethnicity Hispanic or Latino 0 / 2783 12 / 2769 (0.4) 26.346 100% Not Hispanic or Latino 5 / 11019 (<0.1) 2.243 77 / 10987 (0.7) 34.729 93.6% (84.1%, 97.4%) Race American Indian or Alaska Native 0 / 107 0 / 110 Asian 0 / 616 3 / 684 (0.4) 26.549 100% Black or African American 0 / 1,369 4 / 1338 (0.3) 18.566 100% Native Hawaiian or Other Pacific Islander 0 / 33 0 / 30 White 5 /11078 (<0.1) 2.215 80 / 11005 (0.7) 35.821 93.8% (84.8%, 97.5%) Multiple 0 / 293 1 / 304 (0.3) 100% 25 Moderna COVID-19 Vaccine VRBPAC Briefing Document Subgroup Vaccine Group Cases / N (%) Incidence rate per 1,000 person-years Placebo Group Cases / N (%) Incidence rate per 1,000 person-years VE % (95% CI)* Other 0 / 298 2 / 293 (0.7) 45.645 100% Source: Sponsor’s Table 14.2.2.1.1.6.1.1, Table 14.2.2.1.1.6.3.1, Table 4.2.2.1.1.6.7.1, Table 14.2.2.1.1.6.10.1, Table 14.2.2.1.1.6.4.1, Table 14.2.2.1.1.6.2.1, Table 14.2.2.1.1.6.5.1, Table 14.2.2.1.1.6.6.1 a EUA request (interim analysis): November 7, 2020 data cutoff. * VE is calculated as 1-ratio of incidence rates (mRNA-1273/Placebo) and 95% CI from the stratified Cox proportional hazard model. The VE 95% confidence interval is not presented for subgroups for which the lower bound was not evaluable by the statistical methods used for the analysis. At risk for severe COVID-19 due to comorbidity, regardless of age. High risk is defined as patients who meet at least one of the following criteria (protocol-defined): Chronic lung disease (eg, emphysema and chronic bronchitis, idiopathic pulmonary fibrosis, and cystic f ibrosis) or moderate to severe asthma; Significant cardiac disease (eg, heart failure, coronary artery disease, congenital heart disease, cardiomyopathies, and pulmonary hypertension); Severe obesity (body mass index ≥40 kg/m2); Diabetes (Type 1, Type 2 or gestational); Liver disease; Human Immunodeficiency Virus (HIV) infection **used as stratification factor for randomization The demographics of the participants with confirmed COVID-19 cases contributing to the primary efficacy analysis are displayed below in Table 11. Table 11. Demographic Characteristicsa, Participants With COVID-19 Starting 14 Days After Dose 2, Per Adjudication Committee Assessments, Per-Protocol Set Characteristic Vaccine (Na =5) Nb (%) Placebo (Na =90) Nb (%) Total (Na =95) Nb (%) Sex Female 3 (60) 45 (50) 48 (50.5) Male 2 (40) 45 (50) 47 (49.5) Age group 18 to <65 years 5 (100) 75 (83.3) 80 (84.2) ≥65 to <75 years 0 12 (13.3) 12 (12.6) ≥75 years 0 3 (3.3) 3 (3.2) Race American Indian or Alaska Native 0 0 0 Asian 0 3 (3.3) 3 (3.2) Black or African American 0 4 (4.4) 4 (4.2) Native Hawaiian or Other Pacific Islander 0 0 0 White 5 (100) 80 (88.9) 80 (84.2) Multiracial 0 1 (1.1) 1 (1.1) Other 0 2 (2.2) 2 (2.1) Ethnicity Hispanic or Latino 0 12 (13.3) 12 (12.6) Not Hispanic or Latino 5 (100) 77 (85.6) 82 (86.3) Not reported 0 1 (1.1) 1 (1.1) At risk for severe COVID-19 Yes 1 (20) 24 (26.7) 25 (26.3) No 4 (80) 66 (73.3) 70 (73.7) a N = number of participants in the specified group, or the total sample. This value is the denominator for the percentage calculations. a EUA request (interim analysis): November 07 2020 efficacy data cutoff. a EUA request (interim analysis): November 07 2020 cutoff. b n = Number of participants with the specified characteristic. Only 2.2% of participants had evidence of prior infection at study enrollment, and there was only one COVID-19 case starting 14 days after dose 2 reported from this subgroup, which was in a participant in the placebo group. There is insufficient data to conclude on the efficacy of the vaccine in previously infected individuals. 26 Moderna COVID-19 Vaccine VRBPAC Briefing Document Table 12. Vaccine Efficacy by Baseline SARS-CoV-2 Statusa: First COVID-19 From 14 Days After Dose 2 Per Adjudication Committee Assessment, Full Analysis Set Subgroup Vaccine Group Cases / N (%) Incidence rate per 1,000 person-years Placebo Group Cases / N (%) Incidence rate per 1,000 person-years VE % (95% CI)* Baseline SARS-CoV-2 Regardless of baseline SARS-CoV-2 status 6/15180 92/15170 93.5% (85.2, 97.2) Positive 0/341 1/334 (0.3) 17.038 100% Negative 6/14312 (<0.1) 2.154 90/14370 (0.6) 32.298 93.4% (84.8%, 97.1%) Unknown or missing 0/527 1/465 (0.2) 100% * VE is calculated as 1-ratio of incidence rates (mRNA-1273/Placebo) and 95% CI from the stratified Cox proportional hazard model. The VE 95% confidence interval is not presented for subgroups for which the lower bound was not evaluable by the statistical methods used for the analysis. Additional subgroup analyses of the interim primary efficacy analysis were conducted to evaluate the vaccine efficacy, by risk factor for severe COVID-19. VE point estimates were consistent with the efficacy observed for the overall study population, though interpretation of the results is limited by small numbers of participants and cases. Table 13. Vaccine Efficacy by Risk Factor: First COVID-19 Occurrence From 14 Days After Dose 2, Per Adjudication Committee Assessment, Per-Protocol Set Subgroup Vaccine Group Cases / N (%) Incidence rate per 1,000 person-years Placebo Group Cases / N (%) Incidence rate per 1,000 person-years VE % (95% CI)* At risk for severe COVID-19 due to comorbidity, regardless of age Yes 1 / 3116 (<0.1) 1.604 24 / 3075 (0.8) 39.177 95.9% (69.7%, 99.4%) Chronic Lung Disease 0/661 6/673 (0.9) 42.950 100% Significant Cardiac Disease 0/686 3/678 (0.4) 21.463 100% Severe Obesity (BMI >40 kg/m2) 1/901 (0.1) 5.524 11/884 (1.2) 62.851 91.2% (32.0%, 98.9%) Diabetes 0/1338 7/1309 (0.5) 27.148 100% Liver Disease 0/93 0/90 HIV infection 0/80 1/76 (1.3) 91.108 100% No 4 / 10818 (<0.1) 1.911 66 / 10808 (0.6) 31.657 94.0% (83.5%, 97.8%) Obesity (BMI >30 kg/m2)** 2/5269 (<0.1%) 46/5207 (0.9) 95.8% (82.6, 99.0) a EUA request (interim analysis): November 7, 2020 efficacy data cutoff * VE is calculated as 1-ratio of incidence rates (mRNA-1273/Placebo) and 95% CI from the stratified Cox proportional hazard model. The VE 95% confidence interval is not presented for subgroups for which the lower bound was not evaluable by the statistical methods used for the analysis. ** Post hoc analysis. 27 Moderna COVID-19 Vaccine VRBPAC Briefing Document Interim Secondary Efficacy Analyses Severe COVID-19 Cases All 11 cases of severe COVID-19 at least 14 days after second dose as assessed by the adjudication committee were in the placebo group. Of these 11 participants, 5 had risk factors for severe COVID-19 and 6 did not. Three severe COVID-19 cases resulted in hospitalization and 8 did not. Nine of these cases met the severe COVID-19 case definition based on low oxygen saturation <93% on room air without any other severe disease criteria. One participant had low oxygen saturation as well as systolic blood pressure <90 mmHg. One participant had low oxygen saturation and missing data on whether other criteria were met. The vaccine efficacy of this secondary efficacy endpoint is shown in Table 14. Table 14. Severe COVID-19 Cases Starting 14 Days After Second Dose Based on Adjudication Committee Assessment, Per-Protocol Set Vaccine Group N=13934 Cases n (%) Placebo Group N=13883 Cases n (%) Incidence rate per 1,000 person-years Vaccine Efficacy (VE) % (95% CI)* Severe COVID-19 0 11 (<0.1); 4.072 100% a EUA request (interim analysis): Novemer 07 2020 efficacy data cutoff. * VE is calculated as 1-ratio of incidence rates (mRNA-1273/Placebo) and 95% CI from the stratified Cox proportional hazard model. The VE 95% confidence interval is not presented when the lower bound was not evaluable by the statistical methods used for the analysis. One participant in the mRNA-1273 group, a participant >65 years of age who had risk factors for severe COVID-19, was hospitalized due to oxygen saturation of 88% on room air 2 months after receiving the second dose of vaccine. There was a verbal report of a positive SARS-CoV-2 RT￾PCR test 3 days prior to hospitalization; however, NP swab collected during hospitalization was negative for SARS-CoV-2. Due to absence of a confirmed RT-PCR result at the time of data snapshot, this case was not referred for adjudication and not captured. The pre-hospitalization RT-PCR result was later reported to be positive from an external CLIA-certified laboratory and may represent a severe COVID-19 case with hospitalization in the vaccine group. There were 4 additional severe COVID-19 cases which met the protocol-defined severe COVID￾19 endpoint but were not able to be adjudicated in time for the interim analysis. All 4 cases were in the placebo group. Other Secondary Efficacy Endpoints The secondary efficacy endpoint of VE of mRNA-1273 for the prevention of COVID-19 disease based on a less restrictive definition of COVID-19 disease from 14 days after dose 2 showed similar case splits and VE to the primary efficacy endpoints described above. Efficacy against COVID-19 occurring at least 14 days after the first dose of vaccine, including cases that occurred after the second dose, was also similar to the primary endpoint. There were no deaths due to COVID-19 at the time of the interim analysis to enable an assessment of vaccine efficacy against death due to COVID-19. Cumulative Incidence Curves – Interim Efficacy Analysis Based on the cumulative incidence curve for cases in the mITT efficacy population after randomization (same as date of dose 1), COVID-19 cases appear to have occurred similarly at low rates for both the mRNA-1273 and placebo groups until around Day 14 after dose 1. The 28 Moderna COVID-19 Vaccine VRBPAC Briefing Document curves then diverge, with more cases accumulating in the placebo group than the mRNA-1273 group. Figure 2. Cumulative Incidence Curves for the First COVID-19 Occurrence After Randomization, mITT Set Additional Interim Efficacy Analyses Additional analyses were done to assess efficacy against COVID-19 after one dose of mRNA￾1273. In participants in the mITT set who only received one dose of the vaccine at the time of the interim analysis, VE after one dose was 80.2% (95% CI 55.2%, 92.5%). These participants had a median follow-up time of 28 days (range: 1 to 108 days). The small, non-random sample and short median follow-up time limits the interpretation of these results. There appears to be some protection against COVID-19 disease following one dose; however, these data do not provide sufficient information about longer term protection beyond 28 days after a single dose. Table 15. Vaccine Efficacya of mRNA-1273 to Prevent COVID-19 From Dose 1 by Time Period in Participants Who Only Received One Dose, mITT Set First COVID-19 Occurrence After Dose 1 Vaccine Group N=996 Case n (%) Placebo Group N=1079 Case n (%) VE (%) (95% CI)* After dose 1 7/996 (87.5) 39/1079 (96.7) 80.2% (55.2%, 92.5%) After dose 1 to 14 days after dose 1 5/996 (38.0) 11/1079 (41.1) 50.8% (-53.6%, 86.6%) >14 days after dose 1** 2/983 (87.2) 28/1059 (96.2) 92.1% (68.8%, 99.1%) Surveillance time in person years for given endpoint across all participants within each group at risk for the endpoint * VE is calculated as 1-ratio of incidence rates (mRNA-1273/Placebo). The 95% CI of VE is calculated using the exact method conditional upon the total number of cases, adjusting for person-years **Participants who were not at risk (cases or censored at prior time period) are excluded from this analysis a Based on interim analysis: Novemer 7, 2020 efficacy data cutoff. 29 Moderna COVID-19 Vaccine VRBPAC Briefing Document A similar analysis was conducted to look at vaccine efficacy against severe COVID-19 after one dose. In participants in the mITT group who received only one vaccine, 2 participants in the mRNA-1273 group and 4 participants in the placebo group developed severe COVID-19. Both participants in the vaccine group met the case definition for severe COVID-19 based on oxygen saturation <93% on room air. These results should be interpreted cautiously given the small sample size and case number and the short follow-up duration. Table 16. Vaccine Efficacya of mRNA-1273 to Prevent Severe COVID-19 After Dose 1 in Participants Who Only Received One Dose in mITT Set Vaccine Group N=996 Case n (%) Control Group N=1079 Case n (%) Vaccine Efficacy (95% CI) Number of participants with severe COVID-19 starting after dose 1 2 (0.2) 4 (0.4) 42.6% (-300.8, 94.8) a Based on interim analysis : EUA request (interim efficacy analysis): November 7, 2020 efficacy data cutoff. Final Scheduled Efficacy Analysis Data from the final scheduled efficacy analysis were submitted as an amendment to the EUA request on December 7, 2020. Analyses of efficacy endpoints beyond those presented below have not been independently verified by the FDA. The median efficacy and safety follow-up for participants in the study at of the time of the final scheduled efficacy analysis (November 21, 2020 efficacy data cutoff) was 9 weeks. Vaccine efficacy against COVID-19 starting 14 days after the second dose was 94.1% (95% CI 89.3%, 96.8%) and was consistent with results obtained from the interim analysis. The VE in participants ≥65 years of age appears to be lower than in younger adults 18 to <65 years (86.4% compared to 95.6%) and lower than observed in the interim analysis (100% based on a total of 15 cases). Table 17. Final Scheduled Efficacy Analysis, Primary Endpoint, COVID-19 Starting 14 Days After the Second Dose per Adjudication Committee Assessments, Per-Protocol Set Primary Endpoint: COVID-19 (per adjudication committee assessment) Vaccine Group N=13934 Cases n (%) (Incidence Rate per 1,000 person￾years)* Placebo Group N=13883 Cases n (%) (Incidence Rate per 1,000 person￾years)* Vaccine Efficacy (VE) % (95% CI)** Met Predefined Success Criterion*** All participants 11 (<0.1) 3.328 185 (1.3) 56.510 94.1% (89.3%, 96.8%) Yes 18 to <65 years1 7/10551 (<0.1) 2.875 156/10521 (1.5) 64.625 95.6%; (90.6%, 97.9%) NA 65 years and older2 4/3583 (0.1); 4.595 29/3552 (0.8); 33.728 86.4%; (61.4%, 95.5%) NA 30 Moderna COVID-19 Vaccine VRBPAC Briefing Document Source: Sponsor’s Table 14.2.2.1.1.1.1, Table 14.2.2.1.1.6.1.1 COVID-19: symptomatic COVID-19 requiring positive RT-PCR result and at least 2 systemic symptoms or 1 respiratory symptom. Cases starting 14 days after the second dose. All potential COVID-19 cases starting 14 days after the second dose in the clinical database as of 21-Nov-2020 have been sent to adjudication committee, and have been adjudicated for this analysis (21-Nov-2020 is the data cutoff date for efficacy). One case (in the vaccine group) was adjudicated as a COVID-19 case by the committee but did was meet the case definition per statistical analysis plan due to documented symptoms and positive PCR being more than 14 days apart. 21-Nov-2020 have been sent to adjudication committee, and have been adjudicated for this analysis (21-Nov-2020 is the data cutoff date for efficacy). * Incidence rate is defined as the number of participants with an event divided by the number of participants at risk and adjusted by person-years (total time at risk) in each treatment group. The 95% CI is calculated using the exact method (Poisson distribution) and adjusted by person-years. **VE and 95% CI from the stratified Cox proportional hazard model ***The one-sided p-value is <0.0001 from the stratified Cox proportional hazard model to test the null hypothesis of VE ≤30%, achieving the pre-specified efficacy boundary. 1 Percentage based on number of participants in the 18 to <65 years of age group. 2 Percentage based on number of participants in the ≥65 years of age group. Severe COVID-19 Cases In the primary efficacy analysis, there were an additional 19 cases of severe COVID-19 (one of which resulted in death from COVID-19), for a total of 30 severe COVID-19 cases starting 14 days after dose 2, per adjudication committee assessment. All 30 cases were in the placebo group. Nine of the total 30 severe COVID-19 cases resulted in hospitalization. Of the 19 additional severe cases since the interim analysis, 12 cases met the severe case definition due to low oxygen saturation ≤93% with no other criteria met. The remaining participants met the definition based on the following reasons: death (1 participant), ARDS requiring ECMO (1 participant), low oxygen saturation and renal and neurologic dysfunction (1 participant), low oxygen saturation and low blood pressure (2 participants), need for high flow oxygen (1 participant), low blood pressure only (1 participants). The COVID-19 case which resulted in death was in a 54-year-old participant with diabetes. The possible severe COVID-19 case in a mRNA-1273 vaccine recipient described with the interim efficacy analysis (negative SARS-CoV￾2 PCR per the study central laboratory but reported positive PCR per a CLIA-certified external lab) is not included in the per-protocol analysis below. Table 18. Secondary Efficacy Analysis, Severe COVID-19 Starting 14 Days After the Second Dose per Adjudication Committee Assessments, Per-Protocol Set Severe Cases 14 Days After Dose 2 Based on Adjudication Committee Assessments Vaccine Group N=13934 Cases n (%) (Incidence rate per 1,000 person-years) Placebo Group N=13883 Cases n (%) (Incidence rate per 1,000 person-years) Vaccine Efficacy (VE) % (95% CI)* All participants 0 30 (0.2) 9.138 100% a EUA request (primary analysis): November 21, 2020 efficacy data cutoff. Efficacy Summary The data from the planned interim efficacy analysis, with a cutoff date of November 7, 2020, and median follow-up for efficacy of 7 weeks post-dose 2, met the prespecified success criteria established in the study protocol. Efficacy of the vaccine to prevent COVID-19 occurring at least 14 days after dose 2 was 94.5%, (95% CI 86.5%; 97.8%) in participants without prior evidence of SARS-CoV-2 infection. VE was >93% in the group of participants with or without prior infection, although interpretation of data in participants with positive SARS-CoV-2 status at baseline is limited by the small sample size and case numbers in this subgroup. Efficacy outcomes across demographic subgroups were consistent with the efficacy seen in the overall study population. All 11 cases of severe COVID-19 occurring 14 days after the second dose 31 Moderna COVID-19 Vaccine VRBPAC Briefing Document were in the placebo group, although one severe COVID-19 may have occurred in the vaccine group but did not meet criteria for the protocol-specified case definition. Among participants in the mITT set who only received one dose of vaccine or placebo at the time of the interim analysis, efficacy against COVID-19 starting after dose 1 was 80.2% (95% CI: 55.2%, 92.5%). The efficacy observed after dose 1 and before dose 2, from a post-hoc analysis, cannot support a conclusion on the efficacy of a single dose of the vaccine, because the numbers of participants and time of observation are limited. The trial did not have a single-dose arm to make an adequate comparison. Data from a final efficacy analysis (data cutoff November 21, 2020) was submitted as an amendment after the initial EUA request. The FDA has not independently verified the complete efficacy data from this dataset, beyond those analyses presented above. The final scheduled efficacy analysis on the primary endpoint, demonstrating a VE point estimate of 94.1% (95% CI: 89.3%, 96.8%), appear to align with the data obtained from the interim analysis, except for a lower efficacy observed in participants ≥65 years of age compared to that in younger adults 18 to <65 years of age and compared to the efficacy estimate from the interim analysis. 5.2.6 Safety The safety analyses presented in this review are largely derived from the November 11, 2020 dataset that was the basis for the November 30, 2020 EUA request. FDA has not independently verified the complete safety dataset and analyses from the cutoff date of November 25, 2020. However, all new deaths, SAEs, unsolicited adverse events of interest, and pregnancies were reviewed using the cutoff date of November 25, 2020. No additional safety concerns were raised based on the additional data reviewed by FDA or analyses presented by the Sponsor. The safety analyses from the November 25, 2020 cutoff date, as presented by the Sponsor, appear to align with results from the interim analysis in terms of overall rates and types of solicited and unsolicited adverse events. Adverse events were reported in a higher proportion of vaccine recipients than placebo recipients, and this imbalance was driven by reactogenicity (solicited AEs) reported in the 7 days following each dose of vaccine. The proportions of participants with SAEs, death, and withdrawals due to adverse events were balanced across the study groups. Overall, rates of AEs were lower in participants with baseline positive SARS-CoV-2 status compared with those with baseline negative SARS-CoV-2 status. The tables below provide an overview of the rates of AEs by treatment groups and baseline SARS-CoV-2 status. 32 Moderna COVID-19 Vaccine VRBPAC Briefing Document Table 19. Participants Reporting at Least One Adverse Event, Among All Participants and by Baseline SARS-COV2 Status (Safety Set)a Adverse Event Type Vaccine Group n/N (%) Placebo Group n/N (%) Solicited Safety Set N=15176 N=15162 Solicited adverse reactions after any injection 14338/15176 (94.5) 9027/15162 (59.5) Baseline SARS-COV-2 negative 13566/14309 (94.8%) 8576/14363 (59.7) Baseline SARS-COV-2 positive 279 /340 (82.1%) 151/334 (45.2) Solicited local adverse reaction 13,962/15176 (92.0) 4,381/15161 (28.9) Baseline SARS-COV-2 negative 13211/14309 (92.3) 4147/14362 (28.9) Baseline SARS-COV-2 positive 268/340 (78.8) 74/334 (22.2) Grade 3 solicited injection site reactiona 1386/15176 (9.1) 143/15161 (0.9) Baseline SARS-COV-2 negative 1307/14309 (9.1) 131/14362 (0.9) Baseline SARS-COV-2 positive 23/340 (6.8) 5/334 (1.5) Solicited systemic adverse reaction 12553/15176 (82.7) 8032/15,162 (53.0) Baseline SARS-COV-2 negative 11893/14309 (83.1) 7628/14363(53.1) Baseline SARS-COV-2 positive 237/340 (69.7) 137/334 (41.0) Grade 3 or 4 solicited systemic adverse reaction 2,501/15,176 (16.5) 560/15,162 (3.7) Baseline SARS-COV-2 negative 2383/14309 (16.7) 529/14363 (3.7) Baseline SARS-COV-2 positive 37/340 (10.9) 13/334 (3.9) Safety Set N=15184 N=15165 Unsolicited adverse event up to 28 days after any injection 3325/15184 (21.9) 2949/15165 (19.4) Baseline SARS-COV-2 negative 3204/14316 (22.4) 2846/14366 (19.8) Baseline SARS-COV-2 positive 49/341 (14.4) 56/334 (16.8) Unsolicited adverse event 3283/15184 (21.6) 2902/15165 (19.1) Grade 3 unsolicited adverse event 187/15184 (1.2) 148/15165 (1.0) Related** unsolicited adverse events 1127/15184 (7.4) 609/15165 (4.0) Baseline SARS-COV-2 negative 1095/14316 (7.6) 585/14366 (4.1) Baseline SARS-COV-2 positive 16/341 (4.7) 14/334 (4.2) Related** Grade 3 unsolicited adverse event 69/15184 (0.5) 28/15165 (0.2) Medically attended adverse Event 1215/15184 (8.0) 1276/15165 (8.4) Baseline SARS-COV-2 negative 1167/14316 (8.2) 1243/14366 (8.7) Baseline SARS-COV-2 positive 19/341 (5.6) 18/334 (5.4) Related** medically attended adverse events 122/15184 (0.8) 73/15165 (0.5) Baseline SARS-COV-2 negative 118/14316 (0.8) 68/14366 (0.5) Baseline SARS-COV-2 positive 0/341 5/334 (1.5) Serious adverse event 82/15184 (0.5) 86/15165 (0.6) Baseline SARS-COV-2 negative 79/14316 (0.6) 82/14366 (0.6) Baseline SARS-COV-2 positive 0/341 3/334 (0.9) Related** serious adverse event 5/15184 (<0.1) 4/15165 (<0.1) Baseline SARS-COV-2 negative 5/14316 (<0.1) 4/14366 (<0.1) Baseline SARS-COV-2 positive 0/341 0/334 Death* 4/15184 (<0.1) 4/15165 (<0.1) Related** deaths 0 0 AE leading to discontinuation of the vaccine 41/15184 (0.3) 71/15165 (0.5) Baseline SARS-COV-2 negative 34/14316 (0.2) 68/14366 (0.5) Baseline SARS-COV-2 positive 4/341 (1.2) 3/334 (0.9) Source: Sponsor’s Table 14.3.1.1.3, Table 14.3.1.7.1, Table 14.3.1.7.3, Table 14.3.1.7.7 a There were no reports of Grade 4 injection site adverse reactions a EUA request (interim analysis)-November 11, 2020 **Related as assessed by investigator 33 Moderna COVID-19 Vaccine VRBPAC Briefing Document In subgroup analyses of adults ≥65 years of age, rates of solicited reactions (any, Grade 3 or higher) and all other unsolicited adverse events (AEs) (all and related) were comparable to those observed in all participants. Table 20 below summarizes AEs in participants ≥65 years of age, irrespective of baseline serostatus (as less than 1% of ≥65-year-olds were seropositive at baseline). Table 20. Adverse Events Among Adults ≥65 Years of Age (Safety Set)a Participants Reporting at Least One Vaccine Group n/N (%) Placebo Group n/N (%) Solicited Safety Set Solicited adverse reactions after any injection 3497/3766 (92.9) 2010/3750 (53.6) Solicited local adverse reaction 3337/3766 (88.6) 859/3750 (22.9) Grade 3 solicited local adverse reaction 279/3766 (7.4) 66/3750 (1.8) Solicited systemic adverse reaction 2922/3766 (77.6) 1754/3750 (46.8) Grade 3 or 4 solicited systemic adverse reaction 444/3766 (11.8) 119/3750 (3.2) Safety Set Unsolicited Adverse Event up to 28 days after any 872/3770 (23.1) 734/3750 (19.6) Related** unsolicited adverse events 261/3770 (6.9) 138/3750 (3.7) Medically Attended Adverse Event 336/3770 (8.9) 376/3750 (10.0) Related** medically attended adverse events 22/3770 (0.6) 13/3750 (0.3) Serious Adverse Event 36/3770 (1.0) 42/3750 (1.1) Related** serious adverse event 2/3770 (<0.1) 1/3750 (<0.1) Death 1/3768 (<0.1) 2/3752 (<0.1) Related** deaths 0 0 AE leading to discontinuation of the vaccine 12/3770 (0.3) 17/3750 (0.5) Related** AE leading to discontinuation of the vaccine 3/3370 (<0.1) 4/3750 (0.1) Source: Sponsor’s Table 14.3.1.1.3, Table 14.3.1.7.1, Table 14.3.1.7.3, Table 14.3.1.7.7. a EUA request (interim analysis)-November 11 2020. Data provided in response to Information Request (IR),- received December 7 2020 **Related as assessed by investigator Solicited Adverse Reactions Solicited local and systemic adverse reactions with onset within 7 days after each dose were assessed across groups and are presented in the tables below stratified by age (18 to 64 years; ≥65 years) for all participants. Solicited adverse reactions (AR) were recorded daily by study participants using eDiaries and included the assessment of local injection site reactions (pain, erythema, swelling, and lymphadenopathy) and systemic reactions (fever, headache, fatigue, myalgia, arthralgia, chills, and nausea/vomiting). Local Adverse Reactions Solicited local AR were reported by the majority of vaccine recipients and at higher rates than placebo recipients. Vaccine recipients reported higher rates of local reactions after dose 1 than dose 2. The proportions of participants reporting any local AR were 84.2% and 88.8% after dose 1 and dose 2 in vaccine recipients, compared to 19.8% and 18.8% after dose 1 and dose 2 in placebo recipients, respectively. The proportions reporting at least one grade 3 local AR were 3.5% and 7.0% after dose 1 and dose 2, respectively in vaccine recipients and 0.5% after any dose in placebo recipients. There were no reports of Grade 4 local reactions after any dose across groups. The majority of vaccine recipients (57.6%) reported onset of local AR on Day 1 while at home, and the median duration was 2 days after dose and 3 days after dose 2. 34 Moderna COVID-19 Vaccine VRBPAC Briefing Document Overall across both age cohorts, the most frequently reported local AR was pain, reported by 83.7% vs 19.8% of vaccine/placebo recipients after the first dose (2.8% vs 0.4% reported as Grade 3) and 88.4% vs 17.0% of vaccine/placebo recipients after dose 2 (4.1% vs 0.3% reported as Grade 3). The median durations for pain were 2 days and 3 days after dose 1 and dose 2, respectively. The highest rates of pain were in participants 18 to <64 years after dose 2, with 90.1% reporting any pain and 4.6% reporting Grade 3 pain. Axillary lymphadenopathy (vaccination arm) was the second most frequently reported local AR overall. It was reported in 10.2% vs 4.8% of vaccine/placebo recipients after dose 1 and 14.0% vs 3.9% of vaccine/placebo recipients after dose 2 respectively. Grade 3 axillary lymphadenopathy was reported in 0.3% vs 0.2% vaccine/placebo recipients after dose 1 and in 0.5% vs 0.1% of vaccine/placebo recipients after dose 2. The median duration after dose 1 was 1 day and after dose 2 was 2 days. The highest rates of axillary lymphadenopathy were reported by participants 18 to 64 years of age after dose 2, with 16.0% reporting any severity lymphadenopathy and 0.4% reporting Grade 3 lymphadenopathy. Local reactions that persisted beyond 7 days after any dose were reported by both vaccine recipients and placebo recipients. Local reactions that persisted were reported by 3.7% of vaccine recipients and 1.3% of placebo recipients across both age cohorts. In the younger age cohort, 4.2% of vaccine recipients and 1.4% of placebo recipients reported a local reaction that persisted beyond 7 days, of which 0.6% of vaccine recipients and <0.1% of placebo recipients reported a Grade 3 reaction that persisted. In the older age cohort, 2.3% of vaccine recipients compared to 1.1% of placebo recipients reported a local reaction that persisted, including 0.5% of vaccine recipients, and <0.1% of placebo recipients reporting Grade 3 local reactions. Frequently reported local reactions persisting beyond 7 days in the younger age cohort in vaccine/placebo recipients were pain (1.5%/0.6%) and axillary lymphadenopathy (2.5%/0.7%), and in the older age cohort pain (1.2%/0.6%) and erythema (0.7%/<0.1%). Table 21. Frequency of Solicited Local Adverse Reactions Within 7 Days Following Either the First or Second Dose of Vaccine, Participants Age 18 to <64 years, Solicited Safety Set*a Adverse Reaction Vaccine Group Dose 1 n/N (%) Placebo Group Dose 1 n/N (%) Vaccine Group Dose 2 n/N (%) Placebo Group Dose 2 n/N (%) Any Local 9960/11401 (87.4) 2432/11404 (21.3) 9371/10357 (90.5) 2134/10317 (20.7) Grade 3 452/11401 (4.0) 39/11404 (0.3) 766/10357 (7.4) 41/10317 (0.4) Paina 9908/11401 (86.9) 2179/11404 (19.1) 9335/10357 (90.1) 1942/10317 (18.8) Grade 3 367/11401 (3.2) 23/11404 (0.2) 479/10357 (4.6) 21/10317 (0.2) Erythemab (Redness) 345/11401 (3.0) 46/11404 (0.4) 928/10357 (9.0) 42/10317 (0.4) Grade 3 34/11401 (0.3) 11/11404 (<0.1) 206/10357 (2.0) 12/10317 (0.1) Swellingb (Hardness) 768/11401 (6.7) 33/11404 (0.3) 1309/10357 (12.6) 35/10317 (0.3) Grade 3 62/11401 (0.5) 3/11404 (<0.1) 176/10357 (1.7) 4/10317 (<0.1) 35 Moderna COVID-19 Vaccine VRBPAC Briefing Document Adverse Reaction Vaccine Group Dose 1 n/N (%) Placebo Group Dose 1 n/N (%) Vaccine Group Dose 2 n/N (%) Placebo Group Dose 2 n/N (%) Lymphadenopathyc 1322/11401 (11.6) 567/11404 (5.0) 1654/10357 (16.0) 444/10317 (4.3) Grade 3 36/11401 (0.3) 13/11404 (0.1) 45/10357 (0.4) 10/10317 (<0.1) Source: Sponsor’s Table 14.3.1.1.4, Table 14.3.1.1.5 *Safety Analyses Set: all randomized participants who received ≥1 vaccine or control dose a EUA request (interim analysis)-November 11 2020 Note: Adverse reaction data were collected on the electronic diary (eDiary) by participants and those collected on the eCRF indicated as solicitated adverse reactions. n= # of participants with specified reaction N = number of exposed participants who submitted any data for the event, percentages are based on n/N. a: Pain- Grade 3: any use of Rx pain reliever/prevents daily activity; Grade 4: requires E.R. visit or hospitalization b: Erythema and Swelling/Induration- Grade 3: >100mm/>10cm; Grade 4: necrosis/exfoliative dermatitis c: Axillary Swelling/Tenderness collected as solicited local adverse reaction (i.e. lymphadenopathy: localized axillary swelling or tenderness ipsilateral to the vaccination arm) - Grade 3: any use of Rx pain reliever/prevents daily activity; Grade 4: requires E.R. visit or hospitalization Note: No grade 4 solicited local adverse reactions were reported. Table 22. Frequency of Solicited Local Adverse Reactions Within 7 Days Following Either the First or Second Dose of Vaccine, Participants Age ≥65 years, Solicited Safety Set*a Adverse Reaction Vaccine Group Dose 1 n/N (%) Placebo Group Dose 1 n/N (%) Vaccine Group Dose 2 n/N (%) Placebo Group Dose 2 n/N (%) Any Local 2805/3762 (74.6) 566/3746 (15.1) 3010/3587 (83.9) 473/3549 (13.3) Grade 3 77/3762 (2.0) 39/3746 (1.0) 212/3587 (5.9) 29/3549 (0.8) Paina 2782/3762 (74.0) 481/3746 (12.8) 2990/3587 (83.4) 421/3549 (11.9) Grade 3 50/3762 (1.3) 32/3746 (0.9) 96/3587 (2.7) 17/3549 (0.5) Erythemab (Redness) 86/3761 (2.3) 19/3746 (0.5) 265/3587 (7.4) 13/3549 (0.4) Grade 3 8/3761 (0.2) 2/3746 (<0.1) 75/3587 (2.1) 3/3549 (<0.1) Swellingb (Hardness) 166/3761 (4.4) 19/3746 (0.5) 386/3587 (10.8) 13/3549 (0.4) Grade 3 20/3761 (0.5) 3/3746 (<0.1) 69/3587 (1.9) 7/3549 (0.2) Lymphadenopathyc 231/3761 (6.1) 155/3746 (4.1) 302/3587 (8.4) 90/3549 (2.5) Grade 3 12/3761 (0.3) 14/3746 (0.4) 21/3587 (0.6) 8/3549 (0.2) Source: Sponsor’s Tables 14.3.1.1.4 and 14.3.1.1.5] *Safety Analyses Set: all randomized participants who received ≥1 vaccine or control dose. a EUA request (interim analysis)-November 11 2020. Note: Adverse reaction data were collected on the electronic diary by participants and those collected on the eCRF indicated as solicitated adverse reactions. n= # of participants with specified reaction N = number of exposed participants who submitted any data for the event, percentages are based on n/N. a: Pain- Grade 3: any use of Rx pain reliever/prevents daily activity; Grade 4: requires E.R. visit or hospitalization b: Erythema and Swelling/Induration- Grade 3: >100mm/>10cm; Grade 4: necrosis/exfoliative dermatitis c: Axillary Swelling/Tenderness collected as solicited local adverse reaction (i.e. lymphadenopathy: localized axillary swelling or tenderness ipsilateral to the vaccination arm) - Grade 3: any use of Rx pain reliever/prevents daily activity; Grade 4: requires E.R. visit or hospitalization Note: No grade 4 solicited local adverse reactions were reported. 36 Moderna COVID-19 Vaccine VRBPAC Briefing Document Systemic Adverse Reactions Solicited systemic AR were reported for the majority of vaccine recipients and at higher rates than for placebo recipients. Vaccine recipients had higher rates of systemic reactions after the second dose than the first dose. The proportions of vaccine and placebo participants reporting systemic AR were as follows: reporting any grade was 54.9% vs 42.2% after dose 1 and 79.3% vs 36.5% after dose 2, and reporting Grade 3 was 2.9% vs. 2.0% after dose 1 and 15.7% vs. 2.0% after dose 2, respectively. Across groups and doses <0.1% reported a Grade 4 systemic reaction (mainly fever > 104 ºF). The majority of vaccine recipients reported onset of systemic AR while at home either on Day 1 (33.7%) or on Day 2 (37.0%), and the median duration after any dose was 2 days. Overall, the most frequently reported systemic AR was fatigue, reported by 68.5% of vaccine recipients and 36.1% of placebo recipients. After any dose, Grade 3 fatigue was reported by 9.6% of vaccine participants and 1.3% of placebo recipients. Grade 4 fatigue was reported by 1 participant in the vaccine group and none in the placebo group. After dose 1, any/Grade 3 fatigue was reported by 37.2%/1.0% of vaccine recipients and after dose 2 any/Grade 3 fatigue was reported by 65.2%/9.7% of vaccine recipients. The median duration for fatigue in vaccine recipients was 2 days after any dose. The highest rates of fatigue were reported by participants 18 to 64 years after the 2nd dose, with 67.6% reporting any fatigue, 10.6% reporting Grade 3, and 1 participant reporting Grade 4 (after Dose 1). Rates of other solicited systemic AR were: headache 63.0% vaccine group vs. 36.5% placebo group; myalgia 59.6% vaccine group vs. 20.1% placebo group; arthralgia 44.8% vaccine group vs. 17.2% placebo group; and chills 43.4% vaccine group vs. 9.5% placebo group. The rates of Grade 3 AR were: headache 5.5% vaccine group vs. 2.2% placebo group; myalgia 8.6% vaccine group vs. 0.6% placebo group; arthralgia 5.1% vaccine group vs. 0.5% placebo group; and chills 1.3% vaccine group vs. 0.2% of placebo group. The median duration was 1 day after dose 1 and 1 to 2 days after dose 2. The highest rates of solicited reactions were observed in participants 18 to 64 years after dose 2 and included the following: headache 62.8% (5.0% reported Grade 3), myalgia 61.3% (10.0% Grade 3), arthralgia 45.2% (5.8% Grade 3), and chills 45.8% (1.5% Grade 3). There was one vaccine recipient in the younger age cohort who also reported Grade 4 arthralgia after dose 1. Fever was reported after any dose by 14.8% of vaccine participant and 0.6% of placebo recipients. Fever was reported after dose 1 in 0.8% of vaccine recipients and 15.6% of vaccine recipients after dose 2. Grade 3 (≥102.1 ºF) was reported by <0.1% (11 participants) of vaccine recipients after Dose 1 and 1.3% (186 participants) of vaccine recipients after dose 2. Grade 4 (≥104.0 ºF) fever were reported by 4 vaccine recipients after dose 1 and 11 vaccine recipients after dose 2. In participants 18 to 64 years after dose 2, any fever, Grade 3 fever, and Grade 4 fever were reported in 1,806 participants (17.4%), 168 participants (1.6%), and 10 participants (<0.1%), respectively. Systemic reactions persisting longer than 7 days were reported in both age cohorts of vaccine and placebo recipients after any dose. In the vaccine group, 11.9% of participants reported a solicited reaction that persisted beyond 7 days compared to 9.5% of placebo participants. In the younger age cohort, 9.8% of vaccine recipients and 8.9% of placebo recipients reported a systemic reaction that persisted beyond 7 days; and 2.0% of vaccine recipients and 1.2% of placebo recipients reported Grade 3 or 4 systemic reaction that persisted beyond 7 days. In the older age cohort, 9.4% of vaccine recipients and 8.1% of placebo recipients reported a systemic reaction that persisted; 1.7% of vaccine recipients (63 participants) and 0.8% of placebo 37 Moderna COVID-19 Vaccine VRBPAC Briefing Document recipients (31 participants) reported a Grade 3 or 4 reaction that persisted. The most frequently reported systemic reactions that persisted beyond 7 days in vaccine recipients/placebo recipients 18 to 64 years were fatigue (5.7%/5.0%), headache (4.8%/4.0%), myalgia (2.7%/2.7%), and arthralgia (2.6%/2.8%); in the older cohort were fatigue (5.8%/4.5%), arthralgia (3.7%/3.8%), myalgia (2.9%/2.7%), and headache (2.8%/2.7%). Fever persisted beyond 7 days in 7 vaccine recipients and 4 placebo recipients, all of whom were in the younger age cohort. There were 2 vaccine recipients who reported grade 3 fever that persisted, and none in the placebo group. Table 23. Frequency of Solicited Systemic Adverse Reactions Within 7 Days Following Either the First or Second Dose of Vaccine, Participants Age 18-64 years, Solicited Safety Set*a Adverse Reaction Vaccine Group Dose 1 n/N (%) Placebo Group Dose 1 n/N (%) Vaccine Group Dose 2 n/N (%) Placebo Group Dose 2 n/N (%) Any Systemic 6503/11405 (57.0) 5063/11406 (44.4) 8484/10358 (81.9) 3967/10320 (38.4) Grade 3 363/11405 (3.2) 248/11406 (2.2) 1801/10358 (17.4) 215/10320 (2.1) Grade 4 5/11405 (<0.1) 4/11406 (<0.1) 10/10358 (<0.1) 2/10320 (<0.1) Fever 105/11403 (0.9) 39/11404 (0.3) 1806/10352 (17.4) 38/10315 (0.4) Grade 3 10/11403 (<0.1) 1/11404 (<0.1) 168/10352 (1.6) 1/10315 (<0.1) Grade 4 4/11403 (<0.1) 4/11404 (<0.1) 10/10352 (<0.1) 2/10315 (<0.1) Headache 4031/11401 (35.4) 3303/11404 (29.0) 6500/10357 (62.8) 2617/10317 (25.4) Grade 3 219/11401 (1.9) 162/11404 (1.4) 515/10357 (5.0) 124/10317 (1.2) Fatigue 4384/11401 (38.5) 3282/11404 (28.8) 7002/10357 (67.6) 2530/10315 (24.5) Grade 3 120/11401 (1.1) 83/11404 (0.7) 1099/10357 (10.6) 81/10315 (0.8) Grade 4 1/11401 (<0.1) 0 0 0 Myalgia 2698/11401 (23.7) 1626/11404 (14.3) 6353/10357 (6.1) 1312/10316 (12.7) Grade 3 73/11401 (0.6) 38/11404 (0.3) 1032/10357 (10.0) 39/10316 (0.4) Arthralgia 1892/11401 (16.6) 1327/11404 (11.6) 4685/10357 (45.2) 1087/10315 (10.5) Grade 3 47/11401 (0.4) 29/11404 (0.3) 603/10357 (5.8) 36/10315 (0.3) Grade 4 1/11401 (<0.1) 0 0 0 Nausea/Vomiting 1069/11401 (9.4) 908/11404 (8.0) 2209/10357 (21.3) 754/10315 (7.3) Grade 3 6/11401 (<0.1) 8/11404 (<0.1) 8/10357 (<0.1) 8/10315 (<0.1) 38 Moderna COVID-19 Vaccine VRBPAC Briefing Document Adverse Reaction Vaccine Group Dose 1 n/N (%) Placebo Group Dose 1 n/N (%) Vaccine Group Dose 2 n/N (%) Placebo Group Dose 2 n/N (%) Chills 1051/11401 (9.2) 730/11404 (6.4) 5001/10357 (48.3) 611/10315 (5.9) Grade 3 17/11401 (0.1) 8/11404 (<0.1) 151/10357 (1.5) 14/10315 (0.1) Source: Sponsor’s Tables 14.3.1.1.4 and 14.3.1.1.5 a EUA request (interim analysis)-November 11 2020 *Safety Analyses Set: all randomized participants who received ≥1 vaccine or control dose. Note: Adverse reaction data were collected on the electronic diary (e-Diary) by participants and those collected on the eCRF indicated as solicitated adverse reactions. n=# of participants with specified reaction N = number of exposed participants who submitted any data for the event, percentages are based on n/N a: Fever - Grade 3: ≥39.0 – ≤40.0°C or ≥102.1 – ≤104.0°F; Grade 4: >40.0°C >104.0°F b: Headache – Grade 3: Significant; any use of Rx pain reliever or prevents daily activity; Grade 4: Requires E.R. visit or hospitalization c: Fatigue, Myalgia, Arthralgia – Grade 3: Significant; prevents daily activity; Grade 4: Requires E.R. visit or hospitalization d: Nausea/Vomiting – Grade 3: Prevents daily activity, requires outpatient intravenous hydration; Grade 4: Requires E.R. visit or hospitalization for hypotensive shock e: Chills – Grade 3: Prevents daily activity and requires medical intervention; Grade 4: Requires E.R. visit or hospitalization Table 24. Frequency of Solicited Systemic Adverse Reactions Within 7 Days Following Either the First or Second Dose of Vaccine, Participants Age ≥65 Years, Solicited Safety Set*a Adverse Reaction Vaccine Group Dose 1 n/N (%) Placebo Group Dose 1 n/N (%) Vaccine Group Dose 2 n/N (%) Placebo Group Dose 2 n/N (%) Any Systemic 1818/3761 (48.3) 1335/3748 (35.6) 2580/3589 (71.9) 1102/3549 (31.1) Grade 3 84/3761 (2.2) 63/3748 (1.7) 387/3589 (10.8) 58/3549 (1.6) Grade 4 0 0 2/3589 (<0.1) 1/3549 (<0.1) Fever 10/3760 (0.3) 7/3748 (0.2) 366/3587 (10.2) 5/3549 (0.1) Grade 3 1/3760 (<0.1) 1/3748 (<0.1) 18/3587 (0.5) 0 Grade 4 0 2/3748 (<0.1) 1/3587 (<0.1) 1/3549 (<0.1) Headache 921/3761 (24.5) 724/3745 (19.3) 1665/3587 (46.4) 635/3549 (17.9) Grade 3 52/3761 (1.4) 34/3745 (0.9) 107/3587 (3.0) 32/3549 (0.9) Fatigue 1251/3761 (33.3) 851/3745 (22.7) 2094/3587 (58.4) 695/3549 (19.6) Grade 3 30/3761 (0.8) 23/3745 (0.6) 248/3587 (6.9) 20/3549 (0.6) Myalgia 743/3761 (19.8) 443/3745 (11.8) 1683/3587 (46.9) 385/3549 (10.8) Grade 3 17/3761 (0.5) 9/3745 (0.2) 201/3587 (5.6) 10/3549 (0.3) Arthralgia 618/3761 (16.4) 456/3745 (12.2) 1252/3587 (34.9) 381/3549 (10.7) Grade 3 13/3761 (0.3) 8/3745 (0.2) 122/3587 (3.4) 7/3549 (0.2) 39 Moderna COVID-19 Vaccine VRBPAC Briefing Document Adverse Reaction Vaccine Group Dose 1 n/N (%) Placebo Group Dose 1 n/N (%) Vaccine Group Dose 2 n/N (%) Placebo Group Dose 2 n/N (%) Nausea/Vomiting 194/3761 (5.2) 166/3745 (4.4) 425/3587 (11.8) 129/3549 (3.6) Grade 3 4/3761 (0.1) 4/3745 (0.1) 10/3587 (0.3) 3/3549 (<0.1) Grade 4 0 0 1/3587 (<0.1) 0 Chills 202/3761 (5.4) 148/3745 (4.0) 1099/3587 (30.6) 144/3549 (4.1) Grade 3 7/3761 (0.2) 6/3745 (0.2) 27/3587 (0.8) 2/3549 (<0.1) Source: Sponsor’s Tables 14.3.1.1.4 and 14.3.1.1.5 a EUA request (interim analysis) November 11 2020 *Safety Analyses Set: all randomized participants who received ≥1 vaccine or control dose. Note: Adverse reaction data were collected on the electronic diary (e-Diary) by participants and those collected on the eCRF indicated as solicitated adverse reactions. n=# of participants with specified reaction N = number of exposed participants who submitted any data for the event, percentages are based on n/N a: Fever - Grade 3: ≥39.0 – ≤40.0°C or ≥102.1 – ≤104.0°F; Grade 4: >40.0°C >104.0°F b: Headache – Grade 3: Significant; any use of Rx pain reliever or prevents daily activity; Grade 4: Requires E.R. visit or hospitalization c: Fatigue, Myalgia, Arthralgia – Grade 3: Significant; prevents daily activity; Grade 4: Requires E.R. visit or hospitalization d: Nausea/Vomiting – Grade 3: Prevents daily activity, requires outpatient intravenous hydration; Grade 4: Requires E.R. visit or hospitalization for hypotensive shock e: Chills – Grade 3: Prevents daily activity and requires medical intervention; Grade 4: Requires E.R. visit or hospitalization Unsolicited AEs Unsolicited AEs from the November 11, 2020 data cutoff include safety data from participants who had at least 1 month of follow-up after dose 2 (76.7% of all participants) those who had at least 2 months of follow-up after dose 2 (25.3% of all participants). The median study duration following dose 2 was 7 weeks across study groups. Table 25 below shows unsolicited AEs reported through the first data cutoff. Treatment emergent adverse events (AEs) were defined as any event that occurred during the study and was not present before exposure (study vaccine or placebo), any event that occurred during the study and was not present before exposure, or any event already present that worsened after exposure. The following unsolicited adverse events were specified in the protocol: • Unsolicited AEs observed or reported during the 28 days following each vaccine or placebo dose • AEs leading to discontinuation from vaccination and/or study participation through Day 759 (study completion) or withdrawal from the study • Serious adverse events and medically attended adverse events through Day 759 (study completion) or withdrawal from study Determination of severity for all unsolicited AE were made by the investigators based on medical judgement and definitions of severity as mild, moderate, or severe. The overall proportions of participants who reported an unsolicited adverse event were generally similar, with numerically slightly higher rates of unsolicited AEs in the vaccine group compared to placebo group for some categories of unsolicited nonserious AEs. 40 Moderna COVID-19 Vaccine VRBPAC Briefing Document Table 25. Summary of Unsolicited AEs Regardless of Relationship to the Investigational Vaccine, Through 28 Days After Any Vaccination, Study 301, Safety Set Event Type Nov 11 Dataseta mRNA-1273 (N=15184) n (%) Nov 11 Dataseta Placebo (N=15165) n (%) Nov 25 Datasetb mRNA-1273 (N=15185) n (%) Nov 25 Datasetb Placebo (N=15166) n (%) All unsolicited AEs 3325 (21.9) 2949 (19.4) 3632 (23.9) 3277 (21.6) Medically-attended 1215 (8.0) 1276 (8.4) 1372 (9.0) 1465 (9.7) Severe unsolicited AEs 216 (1.4) 190 (1.3) 234 (1.5) 202 (1.3) Leading to discontinuation from study vaccine 41 (0.3) 71 (0.5) 50 (0.3) 80 (0.5) Serious 82 (0.5) 86 (0.6) 93 (0.6) 89 (0.6) Death 2 (<0.1) 3 (<0.1) 2 (<0.1) 3 (<0.1) Source: Abbreviation: AE = adverse event. Note: An AE is defined as any event not present before exposure to study vaccination or any event already present that worsens in intensity or frequency after exposure. Percentages were based on the number of safety participants. a EUA request (interim analysis)-November 11 2020 b Primary efficacy analysis-November 25, 2020 Unsolicited Adverse Events The table below shows rates of unsolicited AEs that occurred within 28 days of any vaccination and at rates of ≥1% in the vaccine group through the November 11, 2020 data cutoff. The proportion of vaccine recipients who reported an unsolicited AE was 21.9% (3325 participants) compared to 19.4% of placebo participants. A higher frequency of unsolicited adverse events was reported in the vaccine group compared to placebo group and was primarily attributed to local and systemic reactogenicity following vaccination. Table 26. Unsolicited Adverse Events Occurring in ≥1% of Vaccine Group Participants, by MedDRA Primary System Organ Class and Preferred Term (Safety Analysis Set)a System Organ Class Preferred Term Vaccine N=15184 n (%) Vaccine N=15184 n (%) Placebo N=15165 n (%) Placebo N=15165 n (%) Any Severe Any Severe Infections and infestations 521 (3.4) 13 (<0.1) 621 (4.1) 25 (0.2) Vascular disorders 149 (1.0) 28 (0.2) 138 (0.9) 39 (0.3) Nervous system disorders 624 (4.1) 27 (0.2) 552 (3.6) 21 (0.1) Headache 435 (2.9) 19 (0.1) 409 (2.7) 13 (<0.1) Respiratory, thoracic and mediastinal disorders 480 (3.2) 8 (<0.1) 522 (3.4) 9 (<0.1) Cough 148 (1.0) 1 (<0.1) 143 (0.9) 1 (<0.1) Oropharyngeal pain 137 (0.9) 1 (<0.1) 184 (1.2) 3 (<0.1) Gastrointestinal disorders 426 (2.8) 14 (<0.1) 387 (2.6) 16 (0.1) Diarrhea 178 (1.2) 2 (<0.1) 147 (1.0) 1 (<0.1) Skin and subcutaneous tissue disorders 213 (1.4) 4 (<0.1) 158 (1.0) 2 (<0.1) Musculoskeletal and connective tissue disorders 586 (3.9) 24 (0.2) 521 (3.4) 18 (0.1) Arthralgia 174 (1.1) 10 (<0.1) 152 (1.0) 2 (<0.1) Myalgia 172 (1.1) 11 (<0.1) 138 (0.9) 0 41 Moderna COVID-19 Vaccine VRBPAC Briefing Document System Organ Class Preferred Term Vaccine N=15184 n (%) Vaccine N=15184 n (%) Placebo N=15165 n (%) Placebo N=15165 n (%) General disorders and administration site 894 (5.9) 43 (0.3) 560 (3.7) 13 (<0.1) Fatigue 344 (2.3) 12 (<0.1) 307 (2.0) 7 (<0.1) Injection site pain 147 (1.0) 6 (<0.1) 49 (0.3) 1 (<0.1) Injury, poisoning and procedural complications 238 (1.6) 16 (0.1) 262 (1.7) 13 (<0.1) Source: Sponsor’s Tables 14.3.1.8.1 and 14.3.1.17.1 n (%)=number (percentage) of participants reporting the adverse event at least once a EUA request (interim analysis): Novemer 11, 2020 data cutoff. Unsolicited AEs considered related by the investigator to study vaccination were reported by 7.4% of vaccine recipients and 4.0% of placebo recipients. The proportion of participants who reported severe unsolicited AEs was 1.4% following any vaccine dose (275 participants) and 1.3% following any placebo dose (225 participants). The most frequently reported severe AEs that occurred in greater numbers of vaccine than placebo recipients were headache, myalgia, arthralgia, injection site erythema, and injection site pain (Table 26). Medically attended adverse events (MAAE) from dose 1 through 28 day following any dose were reported for 8.0% of participants in the vaccine group (1,839 events in 1,215 participants) and 8.4% of those in the placebo group (1,837 events in 1,276 participants). The majority of these events were considered not related to study vaccinations and were primarily attributed to local and systemic reactogenicity following vaccinations. FDA conducted standard MedDRA queries (SMQs) using FDA-developed software to evaluate for constellations of unsolicited adverse events with onset following dose 1 through the November 11, 2020 cutoff. The SMQs were conducted on adverse event Preferred Terms that could represent various conditions, including but not limited to allergic, neurologic, inflammatory, and autoimmune disorders. FDA assessment of additional safety data accrued through the November 25, 2020 cutoff is ongoing, though specific SMQ of adverse events of clinical interest were assessed. A SMQ evaluating lymphadenopathy-related events (including injection site lymphadenopathy, lymph node pain, and lymphadenitis) through the November 25, 2020 data cut demonstrated a numerical imbalance across study groups, with 1.1% of vaccine recipients (191 events in 173 vaccine recipients) compared to 0.63% of placebo recipients (109 events in 95 participants) reporting such events in the Safety Set. The rates reported in the older cohort (≥65 years) were 0.74% (28 events in 28 participants) in vaccine recipients compared to 0.35% (16 events in 13 participants) in placebo recipients. The rates reported in the younger cohort (18-64 years) were 1.3% (163 events in 145 participants) in vaccine recipients and 0.72% (93 events in 82 participants) in placebo recipients. These events support a plausible relationship to study vaccination and were also reported in the evaluation of solicited local adverse reactions. Local axillary swelling/tenderness was reported in approximately 19% of participants during the 7 days following any dose in the Solicited Safety Set. The median duration following any dose was 1 to 2 days, and <1% reported Grade 3 axillary swelling/tenderness. A SMQ evaluating hypersensitivity-related adverse events through the November 25, 2020 data cutoff demonstrated a numerical imbalance across study groups, with 1.5% of vaccine recipients (258 events in 233 participants) and 1.1% of placebo recipients (185 events in 166 participants) reporting such events in the Safety Set. In the older cohort (age ≥65 years) which 42 Moderna COVID-19 Vaccine VRBPAC Briefing Document comprised 24.8% of the Safety Set, the rates of hypersensitivity were 1.8% (74 events in 68 participants) in vaccine recipients and 1% (45 events in 38 participants) in placebo recipients. In the younger age cohort (18-64 years), the rates were 1.5% (184 events in 165 participants) in vaccine recipients compared to 1.1% (140 events in 128 participants). Overall, the most frequently reported AEs in the hypersensitivity SMQ were injection site rash (0.24% vaccine, 0.01% placebo), injection site urticaria (0.1% vaccine, 0% placebo), and rash maculo-papular (0.07% vaccine, 0.01% placebo). There were no anaphylactic or severe hypersensitivity reactions with close temporal relation to the vaccine. A query of specific adverse events of clinical interest in the Safety Set through November 25, 2020 demonstrated a small imbalance in the number of participants reporting Bell’s palsy (facial paralysis), with 3 vaccine recipients and 1 placebo recipient reporting this MAAE. One case of Bell’s palsy in the vaccine group was considered a SAE; a 67-year-old female with diabetes was hospitalized for stroke due to new facial paralysis 32 days after vaccination. This case was reported as resolving. Another Bell’s palsy case in the vaccine group occurred 28 days after vaccination in a 30-year-old female who reported an upper respiratory infection 27 days prior to onset of her facial paralysis. This case was reported as resolved. An additional case of Bell’s palsy in the vaccine group was reported with the primary analysis safety data (November 25, 2020 data cutoff) and occurred 22 days after vaccination in a 72-year-old female; this event was still ongoing at the time of safety report. The case in the placebo group, reported as resolving, occurred 17 days post injection in a 52-year-old-male. Causality assessment is confounded by predisposing factors in these participants. However, considering the temporal association and biological plausibility, a potential contribution of the vaccine to the manifestations of these events of facial palsy cannot be ruled out. FDA will recommend surveillance for cases of Bell’s palsy with deployment of the vaccine into larger populations. There were no other notable patterns or numerical imbalances between treatment groups for specific categories of adverse events, including other neurologic, neuro-inflammatory, and thrombotic events, that would suggest a causal relationship to the Moderna COVID-19 vaccine. Immediate Adverse Events Immediate solicited reactions occurring within 30 minutes of vaccination were infrequent and there does not appear to be an imbalance between the treatment groups. Review of unsolicited AEs that occurred within 30 minutes of vaccination demonstrated comparable rates across study groups (0.6% vaccine, 0.6% placebo), and none of the events reported in the vaccine group were considered serious. Study Withdrawals due to an Adverse Event (Safety Set) Adverse events that led to discontinuation of vaccination were reported in 0.3% in the vaccine group and 0.5% in the placebo group. Following the November 25, 2020 cutoff, 4 participants were withdrawn from the study due to an adverse event (2 vaccine recipients and 2 placebo recipients). The two AEs reported in the vaccine group were acute pancreatitis and road traffic accident, and the two AEs reported in the placebo group were incarcerated hernia and duodenal ulcer hemorrhage. FDA’s review of data through this latter time point is ongoing. Serious Adverse Events Deaths As of December 3, 2020, 13 deaths were reported (6 vaccine, 7 placebo). Two deaths in the vaccine group were in participants >75 years of age with pre-existing cardiac disease; one 43 Moderna COVID-19 Vaccine VRBPAC Briefing Document participant died of cardiopulmonary arrest 21 days after dose 1, and one participant died of myocardial infarction 45 days after dose 2. Another two vaccine recipients were found deceased at home, and the cause of these deaths is uncertain: a 70-year-old participant with cardiac disease was found deceased 57 days after dose 2, and a 56-year-old participant with hypertension, chronic back pain being treated with opioid medication died 37 days after dose 1 (The official cause of death was listed as head trauma). One case was a 72-year-old vaccine recipient with Crohn’s disease and short bowel syndrome who was hospitalized for thrombocytopenia and acute kidney failure due to obstructive nephrolithiasis 40 days after dose 2 and developed complications resulting in multiorgan failure and death. One vaccine recipient died of suicide 21 days after dose 1. The placebo recipients died from myocardial infarction (n=3), intra-abdominal perforation (n=1), systemic inflammatory response syndrome in the setting of known malignancy (n=1), COVID-19 (n=1), and unknown cause (n=1). These deaths represent events and rates that occur in the general population of individuals in these age groups. Non-fatal Serious Adverse Events Among participants who received at least one dose of vaccine or placebo (N=30,351), the proportion of participants who reported at least one SAE from dose 1 to the primary analysis cutoff date (November 25, 2020) was 1% in the mRNA-1273 group and 1% in the placebo group. The most common SAEs occurring at higher rates in the vaccine group than the placebo group were myocardial infarction (0.03% in vaccine group, 5 cases vs. 3 cases in placebo group), cholecystitis (0.02% in vaccine group, 3 cases vs. 0 cases in placebo group), and nephrolithiasis (0.02% in vaccine group, 3 cases vs. 0 cases in placebo group). The small numbers of cases of these events do not suggest a causal relationship. The most common SAEs occurring at higher rates in the placebo arm than the vaccine arm, aside from COVID-19 (0.1% in placebo group), were pneumonia (0.05% in placebo group) and pulmonary embolism (0.03% in placebo group). Occurrence of other SAEs, including cardiovascular SAEs, were otherwise balanced between treatment groups. As of November 25, 2020, 7 SAEs (4.8%) in the mRNA-1273 group and 5 (3.3%) in the placebo group were assessed by the investigator as related to study vaccination (Table 27). Of the 7 SAEs in the mRNA-1273 group, the Sponsor assessed 4 as related and 3 as unrelated to the vaccine. Table 27. SAEs Considered Related by Investigator Investigational Product SAE Onset (days after last dose) Demographics/ Risk factors Resolution Related per Investigator/ Moderna mRNA-1273 Intractable nausea and vomiting 1 65 F; history of headaches and severe nausea requiring hospitalization Resolved Yes/Yes mRNA-1273 Facial swelling 1 46 F; dermal filler cosmetic injection 6 months prior Resolved Yes/Yes mRNA-1273 Facial swelling 2 51 F; dermal filler cosmetic injection 2 weeks prior Resolved Yes/Yes mRNA-1273 Rheumatoid arthritis 14 57 M; hypothyroid Unresolved Yes/Yes mRNA-1273 Dyspnea with exertion, peripheral edema 8 66 F; diabetes, hypertension Resolving Yes/No 44 Moderna COVID-19 Vaccine VRBPAC Briefing Document Investigational Product SAE Onset (days after last dose) Demographics/ Risk factors Resolution Related per Investigator/ Moderna mRNA-1273 Autonomic dysfunction 24 46 F; hypothyroid; possible sinus infection Unresolved Yes/No mRNA-1273 B-cell lymphocytic lymphoma 31 75 F; history of metastatic lung cancer, breast cancer Unresolved Yes/No Placebo Polymyalgia rheumatica 15 83 M; chronic low back pain Resolving Yes/Yes Placebo Facial swelling, paresthesia, anxiety 7 41 F; dental procedure 2 weeks prior Resolved Yes/No Placebo Procedural hemorrhage 16 52 M; aortic stenosis, hyperlipidemia; aspirin intake Resolved Yes/No Placebo Pulmonary embolism 24 59 M; smoking Unresolved Yes/No Placebo Pneumonia and myocardial infarction 29 70 M; coronary artery disease, chronic kidney disease, diabetes Resolved Yes/No There was one event of lip angioedema 2 days after vaccination in a 29-year-old female participant in the vaccine group which was classified as medically significant but not considered an SAE. The participant has a history of dermal filler injection in the lips (unknown how long prior to vaccination). She reported having a similar reaction after receipt of an influenza vaccine in the past. Taken in context with the SAEs of facial swelling which occurred in 2 participants who had previous history of cosmetic filler injections, it is possible the localized swelling in these cases is due to an inflammatory reaction from interaction between the immune response after vaccination and the dermal filler. This phenomenon has been reported after natural infection (e.g., after an influenza-like illness). In FDA’s opinion following review of the narratives, 3 SAEs are considered likely related, including the one report of intractable nausea/vomiting and 2 reports of facial swelling. The possibility that the vaccine contributed to the SAE reports of rheumatoid arthritis, peripheral edema/dyspnea with exertion, and autonomic dysfunction cannot be excluded. The vaccine was unlikely to have contributed to the other SAEs assessed by the investigator as related. As described in detail in a previous section, there was one report of Bell’s palsy in the vaccine arm which occurred 32 days after vaccination; both the investigator and the Sponsor assessed this event as unrelated to the study vaccine, but in FDA’s assessment a causal relationship cannot be definitively excluded. Subgroup Analyses There were no specific safety concerns identified in subgroup analyses by age, race, ethnicity, medical comorbidities, or prior SARS-CoV-2 infection, and occurrence of solicited, unsolicited, and serious adverse events in these subgroups were generally consistent with the overall study population. Pregnancies Study participants of childbearing potential were screened for pregnancy prior to each vaccination, with a positive test resulting in exclusion or discontinuation from study vaccination. The study is collecting outcomes for all reported pregnancies that occur after vaccination, or 45 Moderna COVID-19 Vaccine VRBPAC Briefing Document before vaccination and not detected by pre-vaccination screening tests. Thirteen pregnancies were reported through December 2, 2020 (6 vaccine, 7 placebo). Study vaccination occurred prior to the last menstrual period (LMP) in 5 participants (2 vaccine, 3 placebo), within 30 days after LMP in 5 participants (2 vaccine, 3 placebo), >30 days after LMP in 2 participants (1 vaccine, 1 placebo), and date of LMP not known in 1 participant (1 vaccine, 0 placebo). Unsolicited AEs related to pregnancy include a case of spontaneous abortion and a case of elective abortion, both in the placebo group. One participant in the placebo group is lost to follow-up. Pregnancy outcomes are otherwise unknown at this time. A combined developmental and perinatal/postnatal reproductive toxicity study of mRNA-1273 in rats was submitted to FDA on December 4, 2020. FDA review of this study concluded that mRNA1273 given prior to mating and during gestation periods at dose of 100 µg did not have any adverse effects on female reproduction, fetal/embryonal development, or postnatal developmental except for skeletal variations which are common and typically resolve postnatally without intervention. Safety Summary The information provided by the Sponsor was adequate for review and to make conclusions about the safety of the mRNA-1273 vaccine in the context of the proposed indication and population for intended use under EUA. The number of participants in the Phase 3 safety population (N=30,350; 15,184 vaccine,15,165 placebo) meets the expectations described in FDA’s Guidance on Development and Licensure of Vaccines to Prevent COVID-19 for efficacy. The initial EUA request was based on data from the pre-specified interim analysis (November 11, 2020 data cutoff) with a median follow-up duration of 7 weeks after dose 2; this interim analysis data is the primary basis of this EUA review and conclusions. Data and analyses from a November 25, 2020 data cut with a median duration of at least 2 months follow-up after completion of the 2-dose primary vaccination series was submitted as an amendment to the EUA request on December 7, 2020. The FDA has not independently verified the complete safety data from the primary analysis, aside from all new deaths (including those reported through December 3, 2020) and SAEs. No new safety concerns have been identified. The rates and types of solicited adverse reactions and unsolicited adverse events are unlikely to change significantly with an additional 2 weeks of follow-up. The totality of the data package submitted in the EUA request meets the Agency’s expectations on the minimum duration of follow-up. Local site reactions and systemic solicited events after vaccination were frequent and mostly mild to moderate. The most common solicited adverse reactions were injection site pain (91.6%), fatigue (68.5%), headache (63.0%), muscle pain (59.6%), joint pain (44.8%), and chills (43.4%); 0.2% to 9.7% were reported as severe, with severe solicited adverse reactions being more frequent after dose 2 than after dose 1 and generally less frequent in adults ≥65 years of age as compared to younger participants. Among adverse events of clinical interest, lymphadenopathy was reported in 173 participants (1.14%) in the vaccine group and 95 participants (0.63%) in the placebo group. There was a numerical imbalance in hypersensitivity adverse events across study groups, with 1.5% of vaccine recipients and 1.1% of placebo recipients reporting such events in the Safety Set. There were no anaphylactic or severe hypersensitivity reactions with close temporal relation to the vaccine. Throughout the safety follow-up period to date, there has been three reports of Bell’s palsy in the vaccine group and one in the placebo group. Currently available information is insufficient to determine a causal relationship with the vaccine. There were no other notable patterns or numerical imbalances between treatment groups for specific categories of adverse events (including other neurologic, neuro-inflammatory, and thrombotic events) that would suggest a causal relationship to mRNA- 46 Moderna COVID-19 Vaccine VRBPAC Briefing Document 1273. As of December 3, 2020, there were a total of 13 deaths reported in the study (6 vaccine, 7 placebo). These deaths represent events and rates that occur in the general population of individuals in these age groups. The frequency of non-fatal serious adverse events was low and without meaningful imbalances between study arms (1% in the mRNA-1273 group and 1% in the placebo group). The most common SAEs in the vaccine group which were numerically higher than the placebo group were myocardial infarction (0.03%), cholecystitis (0.02%), and nephrolithiasis (0.02%), although the small numbers of cases of these events do not suggest a causal relationship. The most common SAEs in the placebo arm which were numerically higher than the vaccine arm, aside from COVID-19 (0.1%), were pneumonia (0.05%) and pulmonary embolism (0.03%). 6. Sponsor’s Plans for Continuing Blinded, Placebo-Controlled Follow-Up ModernaTX expects that participants, including approximately 25% who are healthcare workers, may request unblinding to receive mRNA-1273 or another vaccine potentially available under EUA external to the trial. More extensive participant-driven crossover would be expected to alter the composition of the trial population, with greatly increased participant dropout due to a large proportion of participants belonging to priority vaccination groups desiring to be vaccinated with vaccine made available under EUA. ModernaTX is evaluating the opportunity to amend the protocol to proactively reconsent participants who received placebo to be offered mRNA-1273 vaccination and to remain in the trial, enabling ModernaTX to continue to collect the relevant safety and effectiveness data over the entire two years of follow-up while increasing the likelihood of retaining participants on trial. Adverse events among those vaccinated within the trial will be captured, regardless of the treatment group to which the participants were originally allocated, over the entire follow-up period of 24 months. 7. Pharmacovigilance Activities The Sponsor submitted a Pharmacovigilance Plan to monitor safety concerns that could be associated with the Moderna COVID-19 Vaccine. The Sponsor identified vaccine-associated enhanced disease (which includes but is not limited to vaccine-associated enhanced respiratory disease) and anaphylactic reactions (including anaphylaxis) as important potential risks. Use in the pediatric population, use in pregnant and breast-feeding women, immunogenicity in participants with immunosuppression, concomitant administration with non-COVID vaccines, long-term safety and long-term effectiveness are areas the Sponsor identified as missing information. The Sponsor will conduct both passive and active surveillance activities for continued vaccine safety monitoring. Passive surveillance activities will include submitting spontaneous reports of the following events to the Vaccine Adverse Event Reporting System (VAERS) within 15 days: • Vaccine administration errors whether or not associated with an adverse event • Serious adverse events (irrespective of attribution to vaccination) • Cases of Multisystem Inflammatory Syndrome in adults • Cases of COVID-19 that result in hospitalization or death The Sponsor will also conduct periodic aggregate review of safety data and proposed to submit periodic safety reports at quarterly intervals, or at another interval specified by FDA. FDA has 47 Moderna COVID-19 Vaccine VRBPAC Briefing Document requested that periodic reports be submitted monthly. Each periodic safety report is required to contain descriptive information which includes: • A narrative summary and analysis of adverse events submitted during the reporting interval, including interval and cumulative counts by age groups, special populations (e.g., pregnant women), and adverse events of special interest • Newly identified safety concerns in the interval • Actions taken since the last report because of adverse experiences (e.g., changes made to Vaccination Provider fact sheets, changes made to studies or studies initiated) Sponsor studies will include completion of long-term follow-up from ongoing clinical trials as well as the following three planned surveillance studies. • Pregnancy Cohort: The Sponsor plans to establish a passive pregnancy registry to monitor vaccination during pregnancy within populations expected to receive the vaccine under EUA, and to submit a protocol for FDA review and approval. • Active Follow-up for Safety: This study is an active safety surveillance activity conducting retrospective analyses of medical and pharmacy claims data to address three objectives; estimation of background rates of 23 prespecified adverse events of special interest (AESI), descriptive analyses of observed versus expected rates, and self-controlled risk interval analyses that will be conducted if certain criteria are met from the descriptive analyses. The planned study duration is through December 2022. • Real World Effectiveness Study: This study is a prospective cohort study to be conducted at Kaiser Permanente Southern California to evaluate vaccine effectiveness in preventing the following outcomes: laboratory confirmed and clinical COVID-19 infection, hospitalization, and mortality for COVID-19. Vaccinated participants will receive Moderna COVID-19 Vaccine between January 1, 2021 and December 31, 2021, and the comparator group will be age matched, unvaccinated KPSC members. The planned study duration is through December 31, 2023. FDA will provide feedback on these studies after further review of protocols once submitted by the Sponsor. Reporting to VAERS and ModernaTX, Inc. Providers administering the Moderna COVID-19 Vaccine must report to VAERS (as required by the National Childhood Vaccine Injury Act) and to ModernaTX the following information associated with the vaccine of which they become aware: • Vaccine administration errors whether or not associated with an adverse event • Serious adverse events (irrespective of attribution to vaccination) • Cases of Multisystem Inflammatory Syndrome in adults • Cases of COVID-19 that result in hospitalization or death Additional VAERS Reporting An additional source of VAERS reports will be through a program administered by the CDC known as v-safe. V-safe is a smartphone-based opt-in program that uses text messaging and web surveys from CDC to check in with vaccine recipients for health problems following COVID￾19 vaccination. The system also will provide telephone follow-up to anyone who reports 48 Moderna COVID-19 Vaccine VRBPAC Briefing Document medically significant (important) adverse events. Responses indicating missed work, inability to do normal daily activities, or that the recipient received care from a doctor or other healthcare professional will trigger the VAERS Call Center to reach out to the participant and collect information for a VAERS report, if appropriate. 8. Benefit/Risk Assessment in the Context of Proposed Indication and Use Under EUA 8.1 Known Benefits The known benefits among recipients of the proposed vaccine relative to placebo are: • Reduction in the risk of confirmed COVID-19 occurring at least 14 days after the second dose of vaccine • Reduction in the risk of confirmed severe COVID-19 occurring at least 14 days after the second dose of vaccine The 2-dose vaccination regimen was highly effective in preventing PCR-confirmed COVID-19 occurring at least 14 days after receipt of the second dose. Secondary efficacy analyses showed consistency with outcomes in the primary efficacy analysis; the vaccine was effective in preventing COVID-19 using a less restrictive definition of the disease and considering all cases starting 14 days after the first injection. Efficacy findings in the interim analysis were also consistent across various subgroups, including racial and ethnic minorities, participants ages 65 years and older, and those at risk for severe COVID-19 disease due to obesity, diabetes, cardiac disease, liver disease, chronic lung disease, mild to severe asthma, and infection with HIV, although the efficacy estimate in participants ages 65 years and older was slightly lower in the primary efficacy analysis. 8.2 Unknown Benefits/Data Gaps Duration of protection As the interim and final analyses have a limited length of follow-up, it is not possible to assess sustained efficacy over a period longer than 2 months. Effectiveness in certain populations at high-risk of severe COVID-19 Although the proportion of participants at high risk of severe COVID-19 is adequate for the overall evaluation of safety in the available follow-up period, the subsets of certain groups such as immunocompromised individuals (e.g., those with HIV/AIDS) are too small to evaluate efficacy outcomes. Effectiveness in individuals previously infected with SARS-CoV-2 Limited data suggest that individuals with prior SARS-CoV-2 infection can be at risk of COVID￾19 (i.e., re-infection) and may benefit from vaccination. Regarding the benefit of the mRNA-1273 for individuals with prior infection with SARS-CoV2, participants with a known history of SARS￾CoV-2 infection were excluded from the Phase 3 study, and there was only one case of COVID￾19 among study participants with positive SARS-COV-2 infection status at baseline. Thus, the study was not designed to assess the benefit in individuals with prior SARS-CoV-2 infection. Effectiveness in pediatric populations No efficacy data are available from participants ages 17 years and younger. 49 Moderna COVID-19 Vaccine VRBPAC Briefing Document Future vaccine effectiveness as influenced by characteristics of the pandemic, changes in the virus, and/or potential effects of co-infections The study enrollment and follow-up occurred during the period of July 27, 2020 to November 21, 2020, in sites across the United States. The evolution of the pandemic characteristics, such as increased attack rates, increased exposure of subpopulations, as well as potential changes in the virus infectivity, antigenically significant mutations to the S protein, and/or the effect of co￾infections may potentially limit the generalizability of the efficacy conclusions over time. Continued evaluation of vaccine effectiveness following issuance of an EUA and/or licensure will be critical to address these uncertainties. Vaccine effectiveness against asymptomatic infection Data are limited to assess the effect of the vaccine in preventing asymptomatic infection as measured by detection of the virus and/or detection of antibodies against non-vaccine antigens that would indicate infection rather than an immune response induced by the vaccine. Additional evaluations will be needed to assess the effect of the vaccine in preventing asymptomatic infection, including data from clinical trials and from the vaccine’s use post-authorization. Vaccine effectiveness against long-term effects of COVID-19 disease COVID-19 disease may have long-term effects on certain organs, and at present it is not possible to assess whether the vaccine will have an impact on specific long-term sequelae of COVID-19 disease in individuals who are infected despite vaccination. Demonstrated high efficacy against symptomatic COVID-19 should translate to overall prevention of COVID-19- related sequelae in vaccinated populations, though it is possible that asymptomatic infections may not be prevented as effectively as symptomatic infections and may be associated with sequelae that are either late-onset or undetected at the time of infection (e.g., myocarditis). Additional evaluations will be needed to assess the effect of the vaccine in preventing long-term effects of COVID-19, including data from clinical trials and from the vaccine’s use post￾authorization. Vaccine effectiveness against mortality A larger number of individuals at high risk of COVID-19 and higher attack rates would be needed to confirm efficacy of the vaccine against mortality. However, non-COVID vaccines (e.g., influenza) that are efficacious against disease have also been shown to prevent disease￾associated death.13-16 Benefits in preventing death should be evaluated in large observational studies following authorization. Vaccine effectiveness against transmission of SARS-CoV-2 Data are limited to assess the effect of the vaccine against transmission of SARS-CoV-2 from individuals who are infected despite vaccination. Demonstrated high efficacy against symptomatic COVID-19 may translate to overall prevention of transmission in populations with high enough vaccine uptake, though it is possible that if efficacy against asymptomatic infection were lower than efficacy against symptomatic infection, asymptomatic cases in combination with reduced mask-wearing and social distancing could result in significant continued transmission. Additional evaluations including data from clinical trials and from vaccine use post-authorization will be needed to assess the effect of the vaccine in preventing virus shedding and transmission, in particular in individuals with asymptomatic infection. 50 Moderna COVID-19 Vaccine VRBPAC Briefing Document 8.3 Known Risks The vaccine elicited increased local and systemic adverse reactions as compared to those in the placebo arm, usually lasting a few days. The most common solicited adverse reactions were pain at injection site (91.6%), fatigue (68.5%), headache (63.0%), muscle pain (59.6%), joint pain (44.8%), and chills (43.4%). Adverse reactions characterized as reactogenicity were generally mild to moderate; 0.2% to 9.7% of these events were reported as severe, with severe solicited adverse reactions being more frequent after dose 2 than after dose 1 and generally less frequent in older adults (≥65 years of age) as compared to younger participants. Among reported unsolicited adverse events, lymphadenopathy occurred much more frequently in the vaccine group than the placebo group and is plausibly related to vaccination. The number of participants reporting hypersensitivity-related adverse events was numerically higher in the vaccine group compared with the placebo group (258 events in 233 participants [1.5%] vs. 185 events in 166 participants [1.1%]). There were no anaphylactic or severe hypersensitivity reactions with close temporal relation to the vaccine. Serious adverse events, while uncommon (1.0% in both treatment groups), represented medical events that occur in the general population at similar frequency as observed in the study. Of the 7 SAEs in the mRNA-1273 group that were considered as related by the investigator, FDA considered 3 as related: intractable nausea and vomiting (n=1), facial swelling (n=2). For the serious adverse events of rheumatoid arthritis, peripheral edema/dyspnea with exertion, and autonomic dysfunction, a possibility of vaccine contribution cannot be excluded. For the event of B-cell lymphoma, an alternative etiology is more likely. An SAE of Bell’s palsy occurred in a vaccine recipient, for which a causal relationship to vaccination cannot be concluded at this time. No specific safety concerns were identified in subgroup analyses by age, race, ethnicity, medical comorbidities, or prior SARS-CoV-2 infection. 8.4 Unknown Risks/Data Gaps Safety in certain subpopulations There are currently insufficient data to make conclusions about the safety of the vaccine in subpopulations such as children less than 18 years of age, pregnant and lactating individuals, and immunocompromised individuals. FDA review of a combined developmental and perinatal/postnatal reproductive toxicity study of mRNA-1273 in female rats concluded that mRNA1273 given prior to mating and during gestation periods at dose of 100 µg did not have any effects on female reproduction, fetal/embryonal development, or postnatal developmental except for skeletal variations which are common and typically resolve postnatally without intervention Adverse reactions that are very uncommon or that require longer follow-up to be detected Following authorization of the vaccine, use in large numbers of individuals may reveal additional, potentially less frequent and/or more serious adverse events not detected in the trial safety population of approximately 30,000 participants over the period of follow-up at this time. Active and passive safety surveillance will continue during the post-authorization period to detect new safety signals. 51 Moderna COVID-19 Vaccine VRBPAC Briefing Document Although the safety database revealed an imbalance of cases of Bell’s palsy (3 in the vaccine group and 1 in the placebo group), causal relationship is less certain because the number of cases was small and not more frequent than expected in the general population. Further signal detection efforts for these adverse events will be informative with more widespread use of the vaccine. Vaccine-enhanced disease Available data do not indicate a risk of vaccine-enhanced disease, and conversely suggest effectiveness against severe disease within the available follow-up period. However, risk of vaccine-enhanced disease over time, potentially associated with waning immunity, remains unknown and needs to be evaluated further in ongoing clinical trials and in observational studies that could be conducted following authorization and/or licensure. 52 Moderna COVID-19 Vaccine VRBPAC Briefing Document 9. References 1. Zhu N, Zhang D, Wang W, et al. A Novel Coronavirus from Patients with Pneumonia in China, 2019. The New England journal of medicine. 2020;382(8):727-733. 2. Coronaviridae Study Group of the International Committee on Taxonomy of V. The species Severe acute respiratory syndrome-related coronavirus: classifying 2019-nCoV and naming it SARS-CoV-2. 3. Lu R, Zhao X, Li J, et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet (London, England). 2020;395(10224):565-574. 4. Hoffmann M, Kleine-Weber H, Schroeder S, et al. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell. 2020;181(2):271-280.e278. 5. Federal Food, Drug and Cosmetic Act, 21 U.S.C. § 360bbb–3 and 360bbb-3b. (2011). 6. FDA. Guidance for Industry: Emergency Use Authorization for Vaccines to Prevent COVID-19. October 2020. https://www.fda.gov/regulatory-information/search-fda￾guidance-documents/emergency-use-authorization-vaccines-prevent-covid-19. 7. FDA. Guidance for Industry: Development and Licensure of Vaccines to Prevent COVID￾19. June 2020. https://www.fda.gov/regulatory-information/search-fda-guidance￾documents/development-and-licensure-vaccines-prevent-covid-19. 8. National Vaccine Injury Compensation Program. Vaccine Injury Table, Revised and Effective March 21, 2017. https://www.hrsa.gov/sites/default/files/hrsa/vaccine￾compensation/vaccine-injury-table.pdf. 9. International Coalition of Medicines Regulatory Authorities. Statement on continuation of vaccine trials. http://www.icmra.info/drupal/en/covid￾19/statement_on_continuation_of_vaccine_trials. 2020. 10. Krause PR, Fleming TR, Longini IM, et al. Placebo-Controlled Trials of Covid-19 Vaccines - Why We Still Need Them. The New England journal of medicine. 2020. 11. Wendler D, Ochoa J, Millum J, Grady C, Taylor HA. COVID-19 vaccine trial ethics once we have efficacious vaccines. Science. 2020:eabf5084. 12. Centers for Disease Control and Prevention. Evidence used to update the list of underlying medical conditions that increase a person’s risk of severe illness from COVID-19. 2020. https://www.cdc.gov/coronavirus/2019-ncov/need-extra￾precautions/evidence-table.html. 13. Roush SW, Murphy TV, Vaccine-Preventable Disease Table Working Group, the. Historical Comparisons of Morbidity and Mortality for Vaccine-Preventable Diseases in the United States. JAMA. 2007;298(18):2155-2163. 14. Verhees RAF, Dondorp W, Thijs C, Dinant GJ, Knottnerus JA. Influenza vaccination in the elderly: Is a trial on mortality ethically acceptable? Vaccine. 2018;36(21):2991-2997. 15. Flannery B, Reynolds SB, Blanton L, et al. Influenza Vaccine Effectiveness Against Pediatric Deaths: 2010–2014. 2017;139(5):e20164244. 16. Rolfes MA, Flannery B, Chung JR, et al. Effects of Influenza Vaccination in the United States During the 2017-2018 Influenza Season. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2019;69(11):1845-1853. 53 Moderna COVID-19 Vaccine VRBPAC Briefing Document 10. Appendix A. Phase 1 and 2 Studies Study DMID Protocol 20-0003 Study Design DMID Protocol 20-0003 is an ongoing Phase 1, open-label, first-in-human, dose-ranging study to evaluate the safety and immunogenicity of mRNA-1273 in healthy adults 18 years of age and older. A total of 120 participants without risk factors for progression to severe COVID-19 were enrolled into one of 10 age and dose cohorts to receive 2 injections of 25 µg, 50 µg, 100 µg, or 250 µg of mRNA-1273 given 28 days apart. The study included 60 participants 18 through 55 years of age, 30 participants 56 through 70 years of age, and 30 participants 71 years and older. Participants will be followed safety and immunogenicity for 12 months after last vaccination. Study Objectives/Endpoints Relevant to the EUA The immunogenicity objectives are to evaluate the binding antibody (bAb) concentrations for spike IgG as measured by ELISA and neutralizing antibody (nAb) titers as measured by PsVNA for all dose levels at baseline and at various time points after vaccination. The study also evaluated T-cell responses elicited by the mRNA-1273 vaccine as assessed by an intracellular cytokine stimulation assay. All participants are followed for solicited adverse reactions through 7 days post each vaccination. Unsolicited AEs are collected through 28 days after each vaccination. All SAEs and medically attended adverse events are collected through the end of the study. Statistical Analysis No formal statistical hypothesis was tested in this study, and all results were descriptive. Study Results The study showed a dose response in participants across all age groups as measured by both binding and neutralizing antibodies after 2 doses. There was a comparable response between the 100-µg and 250-µg dose groups, and both were greater compared to the 25-µg group. The bAb and nAb levels seen after 2 doses of 100 µg or 250 µg of mRNA-1273 were similar in magnitude compared to those seen in pooled convalescent sera from patients recovered from COVID-19. All dose levels elicited CD4+ T-cell responses that were strongly biased toward expression of Th1 cytokines, with minimal Th2 cytokine expression. This Th1-dominant profile was clinically reassuring in terms of risk of developing vaccine-induced disease. These results, along with the interim safety data showing a lower incidence of reactogenicity in the 100ug group compared to the 250ug group, led to the selection of the 100ug dose to advance to Phase 2 and 3. Preliminary safety data from this Phase 1 study show a similar profile to that observed in the Phase 3 study. No SAEs or severe COVID-19 cases have been reported from this study as of November 16, 2020. Study mRNA-1273-P201 Study Design Study mRNA-1273-P201 is an ongoing phase 2a, randomized, observer-blind, placebo￾controlled, dose-confirmation study to evaluate the safety, reactogenicity, and immunogenicity of mRNA-1273 in healthy adults 18 years and older. The study enrolled 600 participants, consisting of 300 participants 18 to <55 years old and 300 participants 55 years and older, who 54 Moderna COVID-19 Vaccine VRBPAC Briefing Document were randomized equally to receive either 2 doses of 50ug of mRNA-1273, 100ug of mRNA￾1273, or saline placebo given 28 days apart. Participants will be followed for safety and immunogenicity for 12 months post last vaccination. Study Objectives/Endpoints Relevant to the EUA The immunogenicity objectives are to evaluate the immunogenicity of 2 doses of mRNA-1273 at the 2 dose levels (50 µg and 100 µg) administered 28 days apart as assessed by level of bAb and by nAb titers at baseline and at various time points after vaccination. All participants are followed for solicited adverse reactions through 7 days post each vaccination. Unsolicited AEs are collected through 28 days after each vaccination. All SAEs and medically attended adverse events are collected through the end of the study. Statistical Analysis No formal statistical hypothesis was tested in this study and all results were descriptive. Study Results The immune response as assessed by bAb and nAb after 2 doses were comparable in the 50-µg and 100-µg dose groups, with an overall geometric mean fold rise (GMFR)>20-fold in bAB as measured by ELISA and >50-fold in nAb as measured by microneutralization assay at 28 days post-dose 2. In the 100-µg dose group, the older age cohort (≥55 years) had slightly lower bAb response when compared to the younger age cohort (18 to <55 years) at 28 days post-dose 2, but the nAb response was similar between both age groups Safety profile was similar to that reported in the Phase 3 study. Laboratory evaluations (including complete blood count, liver function tests, kidney functions tests, and coagulation studies) were conducted for participants ≥55 years of age (N=100) at baseline and at 1 month after the second dose (Day 29, Day 57). According to narratives that the Sponsor provided to FDA on December 6, 2020, there were 2 participants in the 100-µg group who experienced Grade 3 decreases in hemoglobin (Grade 0 reported at baseline), but both Grade 3 values were within normal range and not clinically significant. The overall event rates were not provided. As of December 6, 2020, there were 3 SAEs reported in the vaccine group: a 65-year-old participant with community acquired pneumonia 25 days after vaccination, a 72-year-old participant with arrhythmia after being struck by lightning 28 days after vaccination, and an 87- year-old participant with worsening of chronic bradycardia 45 days after vaccination. On FDA review of the narratives, none of these SAEs are assessed as related. There were no cases of severe COVID-19 reported in the study.