COVID-19 vaccine clinical research

COVID-19 vaccine clinical research uses clinical research to establish the characteristics of COVID-19 vaccines. These characteristics include efficacy, effectiveness, and safety. As of November 2022, 40 vaccines are authorized by at least one national regulatory authority for public use:[1][2]

As of June 2022, 353 vaccine candidates are in various stages of development, with 135 in clinical research, including 38 in phase I trials, 32 in phase I–II trials, 39 in phase III trials, and 9 in phase IV development.[1]

Formulation

A wide variety of technologies are being used to formulate vaccines against COVID-19. The development and deployment of mRNA vaccines and viral vector vaccines has been outstandingly rapid and can be described as revolutionary. However, global vaccine equity against COVID-19 has not been achieved. Conventional vaccine manufacturing approaches using whole inactivated virus (WIV), protein-based subunit vaccines, and virus-like particles (VLPs) may offer advantages in the development of vaccines for use in low- and middle-income countries (LMICs) and in addressing vaccine access gaps.[6]

Many vaccine candidates use adjuvants to enhance immunogenicity, as part of the delivery system or as an accompanying immune stimulant.[8][9] Vaccine adjuvant formulations using aluminum salts or "alum" may be particularly effective for technologies using inactivated COVID-19 virus and for recombinant protein-based or vector-based vaccines.[6][10]

Status

Clinical trials

The clinical trial process typically consists of three phases, each following the success of the prior phase. Trials are doubly blind in that neither the researcher nor the subject know whether they receive the vaccine or a placebo. Each phase involves randomly-selected subjects who are randomly assigned to serve either as recipients are controls:

  • Phase I trials test primarily for safety and preliminary dosing in healthy subjects. Dozens of subjects.
  • Phase II trials evaluate immunogenicity, dose levels (efficacy based on biomarkers) and adverse effects.[11][12] Hundreds of subjects. Sometimes Phase I and II trials are combined.[12]
  • Phase III trials typically involve more participants at multiple sites, include a control group, and test effectiveness of the vaccine to prevent the disease (an "interventional" or "pivotal" trial), while monitoring for adverse effects at the selected dose.[11][12] Safety, efficacy, and clinical endpoints may vary, including the definition of side effects, infection or amount of transmission, and whether the vaccine prevents moderate or severe infection.[13][14][15]

A clinical trial design in progress may adopt an "adaptive design". If accumulating data provide insights about the treatment, the endpoints or other aspects or the trial can be adjusted.[16][17] Adaptive designs may shorten trial durations and use fewer subjects, possibly expediting decisions, avoiding duplication of research efforts, and enhancing coordination of design changes.[16][18]

Vaccine candidates in human trials

The table below shows various vaccine candidates and the phases which they had completed per the references. Current phases are also shown along with other details.

COVID-19 candidate vaccines in Phase I–III trials
COVID‑19 vaccine candidates in Phase I–III trials[19][20][21]
()
Vaccine candidates,
developers, and sponsors
Country of originType (technology)Current phase (participants)
design
Completed phase[a] (participants)
Immune response
Pending authorization
Sanofi–GSK COVID-19 vaccine (VAT00008, Vidprevtyn)
Sanofi Pasteur, GSK
France, United KingdomSubunit (SARS-CoV-2 S adjuvanted recombinant protein)Phase III (37,430)[22][23]
A Parallel-group, Phase III, Multi-stage, Modified Double-blind, Multi-armed Study to Assess the Efficacy, Safety, and Immunogenicity of Two SARS-CoV-2 Adjuvanted Recombinant Protein Vaccines (Monovalent and Bivalent) for Prevention Against COVID-19 in Adults 18 Years of Age and Older.
May 2021 – Mar 2023, Colombia, Dominican Republic, Ghana, Honduras, India (3,000), Japan, Kenya,[24] Mexico,[25] Nigeria, Pakistan, Sri Lanka, Uganda, United States
Phase I–II (1,160)
Phase I-IIa (440): Immunogenicity and Safety of SARS-CoV-2 Recombinant Protein Vaccine Formulations (With or Without Adjuvant) in Healthy Adults 18 Years of Age and Older.[26]
Phase IIb (720): Immunogenicity and Safety of SARS-CoV-2 Recombinant Protein Vaccine With AS03 Adjuvant in Adults 18 Years of Age and Older.[27]
Sep 2020 – Apr 2022, United States
Emergency (5)
Nanocovax[34]
Nanogen Pharmaceutical Biotechnology JSC
VietnamSubunit (SARS‑CoV‑2 recombinant spike protein with aluminum adjuvant)[35][36]Phase III (13,000)[37][38]
Adaptive, multicenter, randomized, double-blind, placebo-controlled
Jun 2021 – Jul 2022, Vietnam
Phase I–II (620)[39]
Phase I (60): Open label, dose escalation.
Phase II (560): Randomization, double-blind, multicenter, placebo-controlled.
Dec 2020 – Jun 2021, Vietnam
Emergency (1)
UB-612
United Biomedical,Inc, Vaxxinity, DASA
Brazil, Taiwan, United StatesSubunit (Multitope peptide based S1-RBD-protein based vaccine)Phase III (18,320)[41][42]
Phase IIb/III (7,320): Randomized, Multicenter, Double-Blind, Placebo Controlled, Dose-Response.
Phase III (11,000)
Jan 2021 – Mar 2023, Taiwan (phase 2b/3), India (phase 3)[43]
Phase I–II (3,910)[44]
Phase 1 (60): Open-label study
Phase IIa (3,850): Placebo-controlled, Randomized, Observer-blind Study.
Sep 2020 – Jan 2021, Taiwan
Emergency (1)
SCB-2019[46][47]
Clover Biopharmaceuticals,[48][49] Dynavax Technologies,[50] CEPI
ChinaSubunit (spike protein trimeric subunit with combined CpG 1018 and aluminium adjuvant)Phase III (30,300)
Phase II/III (30,000): Randomized, double-blind, controlled.
Phase III (300): Double-blind, randomized, controlled.[51]
Mar 2021 – Oct 2022, Belgium, Brazil, Colombia, Dominican Republic, Germany, Nepal, Panama, the Philippines, Poland, South Africa, Ukraine
Phase I–II (950)
Phase I (150): Randomized, Double-blind, Placebo-controlled, First-in-human.
Phase II (800): Multi-center, Double-blind, Randomized, Controlled.[52]
Jun 2020 – Oct 2021, Australia (phase 1), China (phase 2)
Emergency (1)
S-268019
Shionogi
JapanSubunitPhase III (54,915)[53][54]
Phase II/III: Open-label.
Phase III: Randomized, observer-blind, placebo-controlled cross-over.
Oct 2021 – Dec 2022, Japan (3,100), Vietnam
Phase I–II (300)[55]
Randomized, double-blind, placebo-controlled, parallel-group.
Dec 2020 – Aug 2021, Japan
West China Hospital COVID-19 vaccine
Jiangsu Province Centers for Disease Control and Prevention, West China Hospital (WestVac Biopharma), Sichuan University
ChinaSubunit (recombinant with Sf9 cell)Phase III (40,000)[56]
Multicenter, randomized, double-blind, placebo-controlled.
Jun 2021 – Feb 2022, Indonesia, Kenya, Malaysia,[57] Mexico, Nepal, the Philippines (5,000)[58]
Phase I–II (5,128)[59][60][61]
Phase I (168): Single-center, Randomized, Placebo-controlled, Double-blind.
Phase IIa (960):Single-center, Randomized, Double-Blinded, Placebo-Controlled.
Phase IIb (4,000):Single-center, Randomized, Double-Blinded, Placebo-Controlled.
Aug 2020 – May 2021, China
DelNS1-2019-nCoV-RBD-OPT (DelNS1-nCoV-RBD LAIV)
Beijing Wantai Biological Pharmacy, University of Hong Kong, Xiamen University
China, Hong KongReplicating viral vector (flu-based-RBD[clarification needed])Phase III (40,000)[62]
Multi-center, Randomized, Double-blind, Placebo controlled.
Oct 2021 – Apr 2022, the Philippines
Phase I–II (895)[63][64]
Phase I (60+115=175)
Phase II (720)
Sep 2020 – Sep 2022, China (60), Hong Kong (115)
Versamune-CoV-2FC [pt]
Farmacore Biotechnology, PDS Biotechnology Corporation, Faculty of Medicine of Ribeirão Preto
Brazil, United StatesSubunitPhase III (30,000)[65]
Double-blind, randomized controlled.
Aug–Dec 2021, Brazil
Phase I–II (360)[66][67][68]
Double-blind, randomized controlled.
Mar–Aug 2021, Brazil
Walvax COVID-19 vaccine (ARCoV)[69]
PLA Academy of Military Science, Walvax Biotech,[70] Suzhou Abogen Biosciences
ChinaRNAPhase III (28,000)[71]
Multi-center, Randomized, Double-blind, Placebo-controlled
May–Nov 2021, China,[72] Colombia, Indonesia, Malaysia, Mexico, Nepal, Pakistan, the Philippines, Turkey
Phase I–II (908)
Phase I (168)
Phase II (420)
Phase I/II (320)[73]
Jun 2020 – Oct 2021, China[74]
V-01
Livzon Mabpharm, Inc.
ChinaSubunit (SARS-CoV-2 recombinant fusion protein)Phase III (22,500)[75]
Global, multi-center, randomized, double-blind, placebo-controlled.
Aug 2021–Mar 2023, the Philippines
Phase I (1,060)[76][77]
Phase I (180): Single-center, randomized, double-blind and placebo-controlled.
Phase II (880): Randomized, double-blind, and placebo-controlled.
Feb–May 2021, China
ARCT-154 (VBC-COV19-154 in Vietnam)[78][79][80]
Arcturus Therapeutics, Vinbiocare
United States, VietnamRNAPhase III (20,600)
Phase IIIa (600): Randomized, double-blinded, placebo controlled.
Phase IIIb (20,000): Randomized, double-blinded, placebo controlled.[81][82]
Oct-Dec 2021, Vietnam
Phase I–II (400)
Phase I (100): Randomized, double-blinded, placebo controlled.
Phase II (300): Randomized, double-blinded, placebo controlled.
Aug-Oct 2021, Vietnam[83]
ReCOV
Jiangsu Rec-Biotechnology Co Ltd
ChinaSubunit (Recombinant two-component spike and RBD protein (CHO cell))Phase II–III (20,301)[84]
Multi-center, randomized, double-blind, placebo-controlled.
Dec 2021–Dec 2022, China, New Zealand, the Philippines
Phase I (160)[85]
First-in-human, randomized, double-blind, placebo-controlled, dose-finding.
Jun–Dec 2021, New Zealand
BriLife (IIBR-100)[86]
The Israel Institute for Biological Research
IsraelVesicular stomatitis vector (recombinant)Phase III (20,000)[87]
Randomized, multi-center, placebo-controlled.
Sept – Dec 2021, Israel
Phase I–II (1,040)[88]
Randomized, multi-center, placebo-controlled, dose-escalation.
Oct 2020 – May 2021, Israel
Zhongyianke Biotech–Liaoning Maokangyuan Biotech COVID-19 vaccine
Zhongyianke Biotech, Liaoning Maokangyuan Biotech, Academy of Military Medical Sciences
ChinaSubunit (Recombinant)Phase III (14,600)[89]
International multicenter, randomized, double-blind, placebo-controlled.
Sep 2021–?, China
Phase I–II (696)[90]
Phase I (216): Randomized, placebo-controlled, double-blind.
Phase II (480): Single-center, randomized, double blinded, placebo controlled.[91]
Oct 2020 – Jul 2021, China
GX-19 (GX-19N)[92][93][94]
Genexine consortium,[95][96] International Vaccine Institute
South KoreaDNAPhase II–III (14,000)[97]
Randomized, double-blinded, placebo-controlled.
Oct 2021 – Oct 2022, Indonesia, Seoul
Phase I–II (410)
Phase I-II (170+210+30): Multi-center, some open-labeled, some double-blinded, single arm, randomized, placebo-controlled
Jun 2020 – Jul 2021, Seoul
GRAd-COV2[98][99]
ReiThera, Lazzaro Spallanzani National Institute for Infectious Diseases
ItalyAdenovirus vector (modified gorilla adenovirus vector, GRAd)Phase III (10,300)[100][101]
Randomized, stratified, observer-blind, placebo-controlled.
Mar–Oct 2021, Italy
Phase I (90)[102]
Subjects (two groups: 18–55 and 65–85 years old) randomly receiving one of three escalating doses of GRAd-COV2 or a placebo, then monitored over a 24-week period. 93% of subjects who received GRAd-COV2 developed anti-bodies.
Aug–Dec 2020, Rome
Inovio COVID-19 Vaccine (INO-4800)[103][104]
Inovio, CEPI, Korea National Institute of Health, International Vaccine Institute
South Korea, United StatesDNA vaccine (plasmid delivered by electroporation)Phase III (7,517)
Randomized, placebo-controlled, multi-center.[105]
Nov 2020 – Jan 2023, Brazil, Colombia, Mexico, the Philippines, United States[b]
Phase I–II (920)
Phase Ia (120): Open-label trial.
Phase Ib-IIa (160): Dose-Ranging Trial.[106]
Phase II (640): Randomized, double-blinded, placebo-controlled, dose-finding.[107]
April 2020 – Feb 2022, China (phase II), South Korea (phase Ib-IIa), United States
DS-5670[108]
Daiichi Sankyo[109]
JapanRNAPhase II–III (5,028)[110]
Randomized, Active-comparator, Observer-blind.
Dec 2021 – Jul 2023, Japan
Phase I–II (152)[111]
A Phase 1/2 Study to Assess the Safety, Immunogenicity and Recommended Dose of DS-5670a (COVID-19 Vaccine) in Japanese Healthy Adults and Elderly Subjects.
Mar 2021 – Jul 2022, Japan
GBP510
SK Bioscience Co. Ltd., GSK
South Korea, United KingdomSubunit (Recombinant protein nanoparticle with adjuvanted with AS03)Phase III (4,000)[112]
Randomized, active-controlled, observer-blind, parallel-group, multi-center.[113]
Aug 2021-Mar 2022, South Korea
Phase I–II (580)[114][115]
Phase I-II (260-320): Placebo-controlled, randomized, observer-blinded, dose-finding.
Jan–Aug 2021, South Korea
HGC019[116]
Gennova Biopharmaceuticals, HDT Biotech Corporation[117]
India, United StatesRNAPhase II–III (4,400)[118]
A prospective, multicentre, randomized, active-controlled (with COVISHIELD), observer-blind study to evaluate safety, tolerability and immunogenicity in healthy adults.
Phase II (400)
Phase III (4,000)
Sep 2021 – Sep 2022, India
Phase I–II (620)[119][120][121]
Randomized, phase I/II, placebo-controlled, dose-ranging, parallel-group, crossover, multi-centre study to evaluate the safety, tolerability and immunogenicity in healthy adult subjects.
Phase I (120) open-label study in healthy 18-70 year-olds.
Phase II (500) observer-blind study in healthy 18-75 year-olds.
Apr 2021 – Oct 2021, India
KD-414
KM Biologics Co
JapanInactivated SARS‑CoV‑2Phase II–III (2,000)[122]
Multicenter, open-label, non-randomized.
Oct 2021 – Mar 2023, Japan
Phase I–II (210)[123]
Randomized, double blind, placebo control, parallel group.[124]
Mar 2021 – Dec 2022, Japan
LYB001
Yantai Patronus Biotech Co., Ltd[125]
ChinaVirus-like particle[126]Phase II–III (1,900)[127]
Phase II: Randomized, double blinded, placebo-controlled
Phase III: Single-armed, open-label expanded.
Jan 2022 – Mar 2023, Laos
Phase I (100)[128]
Randomized, double blinded, placebo-controlled.
Dec 2021 – Feb 2022, Laos
AKS-452
Akston Biosciences, University Medical Center Groningen
NetherlandsSubunitPhase II–III (1,600)[129]
Randomized, double-blinded, placebo-controlled, parallel-group, multi-centre, adaptive, seamless bridging.
Oct 2021–Dec 2022, India
Phase I–II (112)[130]
Non-randomized, Single-center, open-label, combinatorial.
Apr–Sep 2021, Netherlands
AG0302-COVID‑19[131][132]
AnGes Inc.,[133] AMED
JapanDNA vaccine (plasmid)Phase II–III (500)
Randomized, double-blind, placebo controlled[134]
Nov 2020 – Apr 2021, Japan
Phase I–II (30)
Randomized/non-randomized, single-center, two doses
Jun–Nov 2020, Osaka
202-CoV
Shanghai Zerun Biotechnology Co., Ltd., Walvax Biotech
ChinaSubunit (Spike protein (CHO cell) 202-CoV with CpG / alum adjuvant)Phase II (1,056)[135]
Randomized, Double-blinded, Placebo-controlled.
July–Dec 2021, China
Phase I (144)[136]
Randomized, double-blinded, placebo-controlled.
July–Dec 2021, China
Vaxart COVID-19 vaccine
Vaxart
United StatesViral vectorPhase II (896)[137]
Double-Blind, Multi-Center, Randomized, Placebo-Controlled, Dose-Ranging.
Oct 2021 – Mar 2022, United States
Phase I (83)[138][139]
Phase Ia (35): Double-blind, randomized, placebo-controlled, first-in-Human.
Phase Ib (48): Multicenter, randomized, double-blind, placebo-controlled.
Sep 2020 – Aug 2021, United States
PTX-COVID19-B[140]
Providence Therapeutics
CanadaRNAPhase II (890)[141]
Randomized, double-dummy, observer-blind.
Aug 2021–Feb 2022, Canada
Phase I (60)[140]
First-in-Human, Observer-Blinded, Randomized, Placebo Controlled.[142]
Jan–May 2021, Canada
Unnamed
Ningbo Rong’an Biological Pharmaceutical Co., Ltd.
ChinaInactivated SARS‑CoV‑2Phase II (600)[143]
Randomized, double-blind, placebo-controlled.
Oct 2021 – Mar 2022, China
Phase I (150)[144]
Randomized, double-blind, placebo-controlled.
Aug – Oct 2021, China
Unnamed
Tsinghua University, Tianjin Medical University,[145] Walvax Biotech
ChinaViral vectorPhase II (360)[146]
Jul–Nov 2021, China
Phase I (60)[147]
May–Jun 2021, China
INNA-051
Ena Respiratory
AustraliaViral vectorPhase II (423)[148]
Randomized, double-blind, placebo-controlled.
Mar  – Dec 2022, Australia
Phase I (124)[149]
Randomised, double blind, placebo-controlled.
Jun – Oct 2021, Australia
mRNA-1283
Moderna
United StatesRNAPhase II (420)[150]
Randomized, stratified, observer-blind.
Dec 2021 – Jan 2023, United States
Phase I (106)[151]
Randomized, observer-blind, dose-ranging study.
Mar 2021 – Apr 2022, United States
Unnamed
Ihsan Gursel, Scientific and Technological Research Council of Turkey
TurkeyVirus-like particlePhase II (330)[152]
Randomized, parallel dose assigned, double blind, multi center.
Jun – Sep 2021, Turkey
Phase I (36)[153]
double-blinded, randomised, placebo controlled.
Mar – May 2021, Turkey
COH04S1
GeoVax, City of Hope Medical Center
United StatesViral vectorPhase II (240)[154]
Multi-center, observer-blinded, EUA vaccine-controlled, randomized.
Aug 2021 – Jun 2023, California
Phase I (129)[155]
Dose Escalation Study.
Dec 2020 – Nov 2022, California
ABNCoV2
Bavarian Nordic.[156] Radboud University Nijmegen
Denmark, NetherlandsVirus-like particlePhase II (210)[157][158]
Single center, sequential dose-escalation, open labelled trial.
Aug–Dec 2021, Germany
Phase I (42)[159]
Single center, sequential dose-escalation, open labelled trial.
Mar–Dec 2021, Netherlands
SCB-2020S
Clover Biopharmaceuticals[160]
ChinaSubunitPhase I–II (150)[161]
Randomized, controlled, observer-blind.
Aug 2021 – Apr 2022, Australia
Preclinical
SCTV01C
Sinocelltech
ChinaSubunitPhase I–II (1,712)[162][163][164]
540+420+752=1,712: multicenter, randomized, double-blinded trial.
Aug 2021 – Jun 2023, China
Preclinical
NDV-HXP-S (ButanVac, COVIVAC, HXP-GPOVac, Patria)
Icahn School of Medicine at Mount Sinai, Institute of Vaccines and Medical Biologicals,[165] Butantan Institute, Laboratorio Avimex, National Council of Science and Technology, Mahidol University, University of Texas at Austin
Brazil, Mexico, Thailand, United States, VietnamNewcastle disease virus (NDV) vector (expressing the spike protein of SARS-CoV-2, with or without the adjuvant CpG 1018)/Inactivated SARS‑CoV‑2Phase I–II (12,750)
Randomized, placebo-controlled, observer-blind.
Mar 2021 – May 2022; Brazil (5,394), Mexico (Phase I: 90, Phase II: 396),[166] Thailand (460),[167] United States (Phase I: 35),[168] Vietnam (495)[169][170]
Preclinical
Stemirna COVID-19 vaccine
Stemirna Therapeutics Co. Ltd.
ChinaRNAPhase I–II (880)[171][172]
Phase I (240): Randomized, double-blind, placebo-controlled.
Phase I/II (640): Open-label.
Mar 2021–Feb 2022, China (phase I), Lao (phase I/II)
Preclinical
ARCT-021[173][174]
Arcturus Therapeutics, Duke–NUS Medical School
United States, SingaporeRNAPhase I–II (798)
Phase I/II (92): Randomized, double-blinded, placebo controlled
Phase II (600): Randomized, observer-blind, placebo-controlled, multiregional, multicenter trial in healthy adults to evaluate the safety, reactogenicity, and immunogenicity.[175]
Phase IIa (106): Open label extension trial to assess the safety and long-term immunogenicity by giving single-dose vaccine to the participants from the parent study that received placebo or were seronegative at screening.[176]
Aug 2020 – Apr 2022, Singapore, United States (phase IIa)
Preclinical
Unnamed
PT Bio Farma
IndonesiaSubunitPhase I–II (780)[177]
Observer-Blind, Randomized, Controlled.
Oct 2021 – Jan 2022, Indonesia
Preclinical
VBI-2902[178]
Variation Biotechnologies
United StatesVirus-like particlePhase I (141)[179]
Randomized, observer-blind, dose-escalation, placebo-controlled
Mar 2021 – Nov 2022, Canada
Preclinical
ICC Vaccine[180]
Novavax
United StatesSubunitPhase I–II (640)[181]
Randomized, observer-blinded.
Sep 2021 – Mar 2022, Australia
Preclinical
EuCorVac-19[182]
EuBiologics Co
South KoreaSubunit (spike protein using the recombinant protein technology and with an adjuvant)Phase I–II (280)
Dose-exploration, randomized, observer-blind, placebo-controlled
Feb 2021 – Mar 2022, the Philipppines (phase II), South Korea (phase I/II)
Preclinical
PHH-1V
Hipra[183]
SpainSubunitPhase I–II (286)[184][185]
Phase I/IIa (30): Randomized, controlled, observer-blinded, dose-escalation.
Phase IIb (256): Randomized, controlled, observer-blinded.
Aug–Dec 2021, Spain (phase I/IIa), Vietnam (phase IIb)
Preclinical
RBD SARS-CoV-2 HBsAg VLP
SpyBiotech
United KingdomVirus-like particlePhase I–II (280)[186]
Randomized, placebo-controlled, multi-center.
Aug 2020 – ?, Australia
Preclinical
AV-COVID-19
AIVITA Biomedical, Inc., Ministry of Health (Indonesia)
United States, IndonesiaDendritic cell vaccine (autologous dendritic cells previously loaded ex vivo with SARS-CoV-2 spike protein, with or without GM-CSF)Phase I–II (202)[187][188]
Adaptive.
Dec 2020 – Feb 2022, Indonesia (phase I), United States (phase I/II)
Preclinical
COVID-eVax
Takis Biotech
ItalyDNAPhase I–II (160)[189]
Multicenter, open label.
Phase I: First-in-human, dose escalation.
Phase II: single arm or two arms, randomized, dose expansion.
Feb–Sep 2021, Italy
Preclinical
BBV154[190]
Bharat Biotech[191]
IndiaAdenovirus vector (intranasal)Phase I–II (375)[190][192]
Phase I (175): Randomized, double-blinded, multicenter.
Phase II (200): Randomized, double blind, multicenter.[193]
Mar 2021–?, India
Preclinical
VB10.2129 and VB10.2210
Nykode Therapeutics[194][195]
NorwayDNAPhase I–II (160)[196][197]
Open Label, Dose Escalation.
Oct 2021–Jun 2022, Norway
Preclinical
ChulaCov19
Chulalongkorn University
ThailandRNAPhase I–II (72)[198]
Phase 1 (72): single-center, dose-escalation, first in human study in 2 age groups: 18-55 years-old and 56-75 years-old.
Phase 2: Multi-center, observer-blinded, placebo-controlled study to assess the safety, reactogenicity, and immunogenicity in healthy adults between 18-75 years old.
May-September 2021, Thailand
Preclinical
COVID‑19/aAPC[199]
Shenzhen Genoimmune Medical Institute[200]
ChinaLentiviral vector (with minigene modifying aAPCs)Phase I (100)[199]
Single group, open-label study to evaluate safety and immunity.
Feb 2020 – Jul 2023, Shenzhen
Preclinical
LV-SMENP-DC[201]
Shenzhen Genoimmune Medical Institute[200]
ChinaLentiviral vector (with minigene modifying DCs)Phase I–II (100)[201]
Single-group, open label, multi-center study to evaluate safety and efficacy.
Mar 2020 – Jul 2023, Shenzhen
Preclinical
ImmunityBio COVID-19 vaccine (hAd5)
ImmunityBio
United StatesViral vectorPhase I–II (540)[202][203][204][205][206]
Phase 1/2 Study of the Safety, Reactogenicity, and Immunogenicity of a Subcutaneously- and Orally- Administered Supplemental Spike & Nucleocapsid-targeted COVID-19 Vaccine to Enhance T Cell Based Immunogenicity in Participants Who Have Already Received Prime + Boost Vaccines Authorized For Emergency Use.
Oct 2020  – Sep 2021, South Africa, United States
Preclinical
COVAC[207]
VIDO (University of Saskatchewan)
CanadaSubunit (spike protein + SWE adjuvant)Phase I (120)[207]
Randomized, observer-blind, dose-escalation.[208][209]
Feb 2021 – Apr 2023, Brazil Halifax
Preclinical
COVI-VAC (CDX-005)[210]
Codagenix Inc.
United StatesAttenuatedPhase I (48)[211]
First-in-human, randomised, double-blind, placebo-controlled, dose-escalation
Dec 2020 – Jun 2021, United Kingdom
Preclinical
CoV2 SAM (LNP)
GSK
United KingdomRNAPhase I (40)[212]
Open-label, dose escalation, non-randomized
Feb–Jun 2021, United States
Preclinical
COVIGEN[213]
Bionet Asia, Technovalia, University of Sydney
Australia, ThailandDNAPhase I (150)[214]
Double-blind, dose-ranging, randomised, placebo-controlled.
Feb 2021 – Jun 2022, Australia, Thailand
Preclinical
MV-014-212[215]
Meissa Vaccine Inc.
United StatesAttenuatedPhase I (130)[216]
Randomized, double-blinded, multicenter.
Mar 2021 – Oct 2022, United States
Preclinical
KBP-201
Kentucky Bioprocessing
United StatesSubunitPhase I–II (180)[217]
First-in-human, observer-blinded, randomized, placebo-controlled, parallel group
Dec 2020 – May 2022, United States
Preclinical
AdCLD-CoV19
Cellid Co
South KoreaViral vectorPhase I–II (150)[218]
Phase I: Dose Escalation, Single Center, Open.
Phase IIa: Multicenter, Randomized, Open.
Dec 2020 – Jul 2021, South Korea
Preclinical
AdimrSC-2f
Adimmune Corporation
TaiwanSubunit (Recombinant RBD +/− Aluminium)Phase I–II (310)[219][220]
Phase I (70): Randomized, single center, open-label, dose-finding.
Phase I/II (240): Placebo-controlled, randomized, double-blind, dose-finding.
Aug 2020–Sep 2022, Indonesia (phase I/II), Taiwan (phase I)
Preclinical
GLS-5310
GeneOne Life Science Inc.
South KoreaDNAPhase I–II (345)[221]
Multicenter, Randomized, Combined Phase I Dose-escalation and Phase IIa Double-blind.
Dec 2020 – Jul 2022, South Korea
Preclinical
Covigenix VAX-001
Entos Pharmaceuticals Inc.
CanadaDNAPhase I–II (72)[222]
Placebo-controlled, randomized, observer-blind, dose ranging adaptive.
Mar–Aug 2021, Canada
Preclinical
NBP2001
SK Bioscience Co. Ltd.
South KoreaSubunit (Recombinant protein with adjuvanted with alum)Phase I (50)[223]
Placebo-controlled, Randomized, Observer-blinded, Dose-escalation.
Dec 2020 – Apr 2021, South Korea
Preclinical
CoVAC-1
University of Tübingen
GermanySubunit (Peptide)Phase I–II (104)[224][225]
Phase I (36): Placebo-controlled, Randomized, Observer-blinded, Dose-escalation.
Phase I/II (68): B-pVAC-SARS-CoV-2: Phase I/II Multicenter Safety and Immunogenicity Trial of Multi-peptide Vaccination to Prevent COVID-19 Infection in Adults With Bcell/ Antibody Deficiency.
Nov 2020 – Feb 2022, Germany
Preclinical
bacTRL-Spike
Symvivo
CanadaDNAPhase I (24)[226]
Randomized, observer-blind, placebo-controlled.
Nov 2020 – Feb 2022, Australia
Preclinical
ChAdV68-S (SAM-LNP-S)
NIAID, Gritstone Oncology
United StatesViral vectorPhase I (150)[227]
Open-label, dose and age escalation, parallel design.
Mar 2021 – Sep 2022, United States
Preclinical
SpFN COVID-19 vaccine
WRAIR's Emerging Infectious Diseases Branch (EIDB)
United StatesSubunitPhase I (72)[228]
Randomized, double-blind, placebo-controlled.
Apr 2021 – Oct 2022, United States
Preclinical
MVA-SARS-2-S (MVA-SARS-2-ST)
University Medical Center Hamburg-Eppendorf
GermanyViral vectorPhase I–II (270)[229][230]
Phase I (30): Open, Single-center.
Phase Ib/IIa (240): Multi-center, Randomized Controlled.
Oct 2020 – Mar 2022, Germany
Preclinical
Koçak-19 Inaktif Adjuvanlı COVID-19 vaccine
Kocak Farma
TurkeyInactivated SARS‑CoV‑2Phase I (38)[231]
Phase 1 Study for the Determination of Safety and Immunogenicity of Different Strengths of Koçak-19 Inaktif Adjuvanlı COVID-19 Vaccine, Given Twice Intramuscularly to Healthy Volunteers, in a Placebo Controlled Study Design.
Mar–Jun 2021, Turkey
Preclinical
CoV2-OGEN1
Syneos Health, US Specialty Formulations
United StatesSubunitPhase I (45)[232]
First-In-Human
Jun–Dec 2021, New Zealand
Preclinical
CoVepiT
OSE Immunotherapeutics
FranceSubunitPhase I (48)[233][234]
Randomized, open label.
Apr–Sept 2021, France
Preclinical
HDT-301[235] (QTP104)
HDT Biotech Corporation, Senai Cimatec, Quartis[236]
Brazil, South Korea, United StatesRNAPhase I (189)[237][238]
Phase I (90+63+36): Randomized, open-label, dose-escalation.
Aug 2021–Jul 2023, Brazil, South Korea, United States
Preclinical
SC-Ad6-1
Tetherex Pharmaceuticals
United StatesViral vectorPhase I (40)[239]
First-In-Human, Open-label, Single Ascending Dose and Multidose.
Jun–Dec 2021, Australia
Preclinical
Unnamed
Osman ERGANIS, Scientific and Technological Research Council of Turkey
TurkeyInactivated SARS‑CoV‑2Phase I (50)[240]
Phase I Study Evaluating the Safety and Efficacy of the Protective Adjuvanted Inactivated Vaccine Developed Against SARS-CoV-2 in Healthy Participants, Administered as Two Injections Subcutaneously in Two Different Dosages.
Apr–Oct 2021, Turkey
Preclinical
EXG-5003
Elixirgen Therapeutics, Fujita Health University
Japan, United StatesRNAPhase I–II (60)[241]
First in Human, randomized, placebo-controlled.
Apr 2021 – Jan 2023, Japan
Preclinical
IVX-411
Icosavax, Seqirus Inc.
United StatesVirus-like particlePhase I–II (168)[242][243]
Phase I/II (84): Randomized, observer-blinded, placebo-controlled.
Jun 2021–2022, Australia
Preclinical
QazCoVac-P[244]
Research Institute for Biological Safety Problems
KazakhstanSubunitPhase I–II (244)[245]
Phase I: Randomized, blind, placebo-controlled.
Phase II: Randomized, open phase.
Jun – Dec 2021, Kazakhstan
Preclinical
LNP-nCOV saRNA-02
MRC/UVRI & LSHTM Uganda Research Unit
UgandaRNAPhase I (42)[246]
A Clinical Trial to Assess the Safety and Immunogenicity of LNP-nCOV saRNA-02, a Self-amplifying Ribonucleic Acid (saRNA) Vaccine, in SARS-CoV-2 Seronegative and Seropositive Uganda Population.
Sep 2021 – Jun 2022, Uganda
Preclinical
Baiya SARS-CoV-2 Vax 1[247]
Baiya Phytopharm Co Ltd.
ThailandPlant-based Subunit (RBD-Fc + adjuvant)Phase I (96)[248]
Randomized, open-label, dose-finding.
Sep–Dec 2021, Thailand
Preclinical
CVXGA1
CyanVac LLC
United StatesViral vectorPhase I (80)[249]
Open-label
July–Dec 2021, United States
Preclinical
Unnamed
St. Petersburg Scientific Research Institute of Vaccines and Sera of Russia at the Federal Medical Biological Agency
RussiaSubunit (Recombinant)Phase I–II (200)[250][251]
Jul–Aug 2021, Russia
Preclinical
LVRNA009
Liverna Therapeutics Inc.
ChinaRNAPhase I (24)[252]
July–Nov 2021, China
Preclinical
ARCT-165
Arcturus Therapeutics
United StatesRNAPhase I–II (72)[253]
Randomized, observer-blind.
Aug 2021–Mar 2023, Singapore, United States
Preclinical
BCD-250
Biocad
RussiaViral vectorPhase I–II (160)[254]
Randomized, double-blind, placebo-controlled, adaptive, seamless phase I/II.
Aug 2021–Aug 2022, Russia
Preclinical
COVID-19-EDV
EnGeneIC
AustraliaViral vectorPhase I (18)[255]
Open label, non-randomised, dose escalation.
Aug 2021–Jan 2022, Australia
Preclinical
COVIDITY
Scancell
United KingdomDNA[256]Phase I (40)[257]
Open-label, two-arm.
Sep 2021–Apr 2022, South Africa
Preclinical
SII Vaccine
Novavax
United StatesSubunitPhase I–II (240)[258]
randomized, observer-blinded, open-label.
Oct–Nov 2021, Australia
Preclinical
EG-COVID
Eyegene
South KoreamRNAPhase I–II (120)[259][260][261]
Phase I/IIa: Multi-center, Open-label.
Feb 2022–May 2023, South Korea
Preclinical
PIKA COVID-19 vaccine
Yisheng Biopharma
ChinaSubunitPhase I (45)[262]
Open-label, dose-escalation.
Sep–Nov 2021, New Zealand
Preclinical
Ad5-triCoV/Mac
McMaster University, Canadian Institutes of Health Research (CIHR)
CanadaViral vectorPhase I (30)[263]
Open-label.
Nov 2021–Jun 2023, Canada
Preclinical
Unnamed
University of Hong Kong, Immuno Cure 3 Limited
Hong KongDNAPhase I (30)[264]
Randomized, double-blinded, placebo-controlled, dose-escalation.
Nov 2021–Jun 2022, Hong Kong
Preclinical
MigVax-101
Oravax Medical[265][266][267]
IsraelVirus-like particlePhase I
Oct 2021–?, South Africa
Preclinical
IN-B009
HK inno.N[268]
South KoreaSubunit (Recombinant protein)Phase I (40)[269]
Open-label, dose-escalation.
Sep 2021–Feb 2023, South Korea
Preclinical
naNO-COVID
Emergex Vaccines
United KingdomSubunitPhase I (26)[270]
Double-blind, randomized, vehicle-controlled, dose-finding.
Nov 2021–Sep 2022, Switzerland
Preclinical
Betuvax-CoV-2
Human Stem Cells Institute
RussiaSubunitPhase I–II (170)[271][272]
Sep 2021–?, Russia
Preclinical
Covi Vax[273]
National Research Centre
EgyptInactivated SARS‑CoV‑2Phase I (72)[274]
Randomized, open-labeled
Nov 2021–Feb 2023, Egypt
Preclinical
VLPCOV-01
VLP Therapeutics
United StatesmRNAPhase I (45)[275]
Randomized, placebo-controlled, parallel group, first-in-human.
Aug 2021–Jan 2023, Japan
Preclinical
GRT-R910
Gritstone Oncology
United StatesmRNAPhase I (120)[276]
A Phase 1 Trial to Evaluate the Safety, Immunogenicity, and Reactogenicity of a Self-Amplifying mRNA Prophylactic Vaccine Boost Against SARS-CoV-2 in Previously Vaccinated Healthy Elderly Adults.
Sep 2021–Nov 2022, United Kingdom
Preclinical
Unnamed
DreamTec Limited
Hong KongSubunitPhase I (30)[277]
Development of a COVID19 Oral Vaccine Consisting of Bacillus Subtilis Spores Expressing and Displaying the Receptor Binding Domain of Spike Protein of SARS-COV2.
Apr–Dec 2021, Hong Kong
Preclinical
Almansour-001
Imam Abdulrahman Bin Faisal University, ICON plc
Ireland, Saudi ArabiaDNAPhase I (30)[278]
Single center, randomized, observer blind, dosage finding.
Feb–Jul 2022, Ireland, Saudi Arabia
Preclinical
Unnamed
North's Academy of Medical Science Medical biology institute
North KoreaSubunit (spike protein with Angiotensin-converting enzyme 2)Phase I–II (?)[279]
Jul 2020, North Korea
Preclinical
Vabiotech COVID-19 vaccine
Vaccine and Biological Production Company No. 1 (Vabiotech)
VietnamSubunitPreclinical
Awaited for the conduct on Phase I trial.[280]
?
INO-4802
Inovio
United StatesDNAPreclinical
Awaited for the conduct on Phase I/II trials.[281]
?
Bangavax (Bancovid)[282][283]
Globe Biotech Ltd. of Bangladesh
BangladeshRNAPreclinical
Awaiting for approval from Bangladesh government to conduct the first clinical trial.[284]
?
Unnamed
Indian Immunologicals, Griffith University[285]
Australia, IndiaAttenuatedPreclinical?
EPV-CoV-19[286]
EpiVax
United StatesSubunit (T cell epitope-based protein)Preclinical?
Unnamed
Intravacc[287]
NetherlandsSubunitPreclinical?
CureVac–GSK COVID-19 vaccine[288]
CureVac, GSK
Germany, United KingdomRNAPreclinical?
DYAI-100[289] Sorrento Therapeutics, Dyadic International, Inc.[290]United StatesSubunitPreclinical?
Unnamed[291]
Ministry of Health (Malaysia), Malaysia Institute of Medical Research Malaysia, Universiti Putra Malaysia
MalaysiaRNAPreclinical?
CureVac COVID-19 vaccine (CVnCoV)
CureVac, CEPI
GermanyRNA (unmodified RNA)[292]Terminated (44,433)[293][294][295][296][297]
Phase 2b/3 (39,693): Multicenter efficacy and safety trial in adults.
Phase 3 (2,360+180+1,200+1,000=4,740): Randomized, placebo-controlled, multicenter, some observer-blinded, some open-labeled.
Nov 2020 – Jun 2022, Argentina, Belgium, Colombia, Dominican Republic, France, Germany, Mexico, Netherlands, Panama, Peru, Spain[298]
Phase I–II (944)[299][300]
Phase I (284): Partially blind, controlled, dose-escalation to evaluate safety, reactogenicity and immunogenicity.
Phase IIa (660):Partially observer-blind, multicenter, controlled, dose-confirmation.
Jun 2020 – Oct 2021, Belgium (phase I), Germany (phase I), Panama (phase IIa), Peru (phase IIa)
Emergency (2)
CORVax12
OncoSec Medical, Providence Health & Services
United StatesDNATerminated (36)[303]
Open-label, non-randomized, parallel assignment study to evaluate the safety of prime & boost doses with/without the combination of electroporated IL-12p70 plasmid in 2 age groups: 18-50 years-old and > 50 years-old.
Dec 2020 – Jul 2021, United States
Preclinical
Sanofi–Translate Bio COVID-19 vaccine (MRT5500)[304]
Sanofi Pasteur and Translate Bio
France, United StatesRNATerminated (415)[305]
Interventional, randomized, parallel-group, sequential study consisting of a sentinel cohort followed by the full enrollment cohort. The sentinel cohort is an open-label, step-wise, dose-ranging study to evaluate the safety of 3 dose levels with 2 vaccinations. The full enrollment cohort is a quadruple-blinded study of safety and immunogenicity in 2 age groups, with half receiving a single injection, and the other half receiving 2 injections.
Mar 2021 – Sep 2021, Honduras, United States, Australia
Preclinical
AdCOVID
Altimmune Inc.
United StatesViral vectorTerminated (180)[306][307]
Double-blind, randomized, placebo-controlled, first-in-Human.
Feb 2021 – Feb 2022, United States
Preclinical
LNP-nCoVsaRNA[308]
MRC clinical trials unit at Imperial College London
United KingdomRNATerminated (105)
Randomized trial, with dose escalation study (15) and expanded safety study (at least 200)
Jun 2020 – Jul 2021, United Kingdom
?
TMV-083
Institut Pasteur
FranceViral vectorTerminated (90)[309]
Randomized, Placebo-controlled.
Aug 2020 – Jun 2021, Belgium, France
?
SARS-CoV-2 Sclamp/V451[310][311][unreliable source?]
UQ, Syneos Health, CEPI, Seqirus
AustraliaSubunit (molecular clamp stabilized spike protein with MF59)Terminated (120)
Randomised, double-blind, placebo-controlled, dose-ranging.
False positive HIV test found among participants.
Jul–Oct 2020, Brisbane
?
V590[312] and V591/MV-SARS-CoV-2[313] Merck & Co. (Themis BIOscience), Institut Pasteur, University of Pittsburgh's Center for Vaccine Research (CVR), CEPIUnited States, FranceVesicular stomatitis virus vector[314] / Measles virus vector[315][unreliable source?]Terminated
In phase I, immune responses were inferior to those seen following natural infection and those reported for other SARS-CoV-2/COVID-19 vaccines.[316]


Homologous prime-boost vaccination

In July 2021, the U.S. Food and Drug Administration (FDA) and the Centers for Disease Control and Prevention (CDC) issued a joint statement reporting that a booster dose is not necessary for those who have been fully vaccinated.[317]

In August 2021, the FDA and the CDC authorized the use of an additional mRNA vaccine dose for immunocompromised individuals.[318][319] The authorization was extended to cover other specific groups in September 2021.[320][321][322]

In October 2021, the FDA and the CDC authorized the use of either homologous or heterologous vaccine booster doses.[323][324]

Heterologous prime-boost vaccination

The World Health Organization (WHO) defines heterologous prime-boost immunization as the "administration of two different vectors or delivery systems expressing the same or overlapping antigenic inserts."[325] A heterologous scheme can sometimes be more immunogenic than some homologous schemes.[326]

In October 2021, the FDA and the CDC authorized the use of either homologous or heterologous vaccine booster doses.[323][324]

Some experts believe that heterologous prime-boost vaccination courses can boost immunity, and several studies have begun to examine this effect.[327] Despite the absence of clinical data on the efficacy and safety of such heterologous combinations, Canada and several European countries have recommended a heterologous second dose for people who have received the first dose of the Oxford–AstraZeneca vaccine.[328]

In February 2021, the Oxford Vaccine Group launched the Com-COV vaccine trial to investigate heterologous prime-boost courses of different COVID-19 vaccines.[329] As of June 2021, the group is conducting two phase II studies: Com-COV and Com-COV2.[330]

In Com-COV, the two heterologous combinations of the Oxford–AstraZeneca and Pfizer–BioNTech vaccines were compared with the two homologous combinations of the same vaccines, with an interval of 28 or 84 days between doses.[331][332][unreliable medical source?]

In Com-COV2, the first dose is the Oxford–AstraZeneca vaccine or the Pfizer vaccine, and the second dose is the Moderna vaccine, the Novavax vaccine, or a homologous vaccine equal to the first dose, with an interval of 56 or 84 days between doses.[333]

A study in the UK is evaluating annual heterologous boosters by randomly combining the following vaccines: Oxford–AstraZeneca, Pfizer–BioNTech, Moderna, Novavax, VLA2001, CureVac, and Janssen.[334]

On 16 December, WHO recommendations on heterologous vaccinations suggested a general trend of increased immunogenicity when one of the doses is of an mRNA vaccine, particularly as the last dose. The immunogenicity of a homologous mRNA course is roughly equivalent to a heterologous scheme involving a vector vaccine and an mRNA vaccine. However, the WHO has emphasized the need to address many evidence gaps in heterologous regimens, including duration of protection, optimal interval between doses, influence of fractional dosing, effectiveness against variants and long-term safety.[335]

Heterologous second dose regimes in clinical trial
First doseSecond doseSchedulesCurrent phase (participants), periods and locations
Oxford–AstraZeneca
Pfizer–BioNTech
Oxford–AstraZeneca
Pfizer–BioNTech
Days 0 and 28
Days 0 and 84
Phase II (820)[331]
Feb–Aug 2021, United Kingdom
Sputnik LightOxford–AstraZeneca
Moderna
Sinopharm BIBP
Phase II (121)[336]
Feb–Aug 2021, Argentina
Oxford–AstraZeneca
Pfizer–BioNTech
Oxford–AstraZeneca
Pfizer–BioNTech
Moderna
Novavax
Days 0 and 56–84Phase II (1,050)[333]
Mar 2021 – Sep 2022, United Kingdom
ConvideciaZF2001Days 0 and 28
Days 0 and 56
Phase IV (120)[337]
Apr–Dec 2021, China
Oxford–AstraZenecaPfizer–BioNTechDays 0 and 28Phase II (676)[338]
Apr 2021 – Apr 2022, Spain
Oxford–AstraZeneca
Pfizer–BioNTech
Moderna
Pfizer–BioNTech
Moderna
Days 0 and 28
Days 0 and 112
Phase II (1,200)[339]
May 2021 – Mar 2023, Canada
Pfizer–BioNTech
Moderna
Pfizer–BioNTech
Moderna
Days 0 and 42Phase II (400)[340]
May 2021 – Jan 2022, France
Oxford–AstraZenecaPfizer–BioNTechDays 0 and 28
Days 0 and 21–49
Phase II (3,000)[341]
May–Dec 2021, Austria
JanssenPfizer–BioNTech
Janssen
Moderna
Days 0 and 84Phase II (432)[342]
Jun 2021 – Sep 2022, Netherlands
Heterologous booster dose regimes in clinical trial
Initial courseBooster doseIntervalCurrent phase (participants), periods and locations
CoronaVac (2 doses)CoronaVac
Pfizer–BioNTech
Oxford–AstraZeneca
19 weeks or morePhase IV (2,017,878)[343][344]
Aug–Nov 2021, Chile

Efficacy

Cumulative incidence curves for symptomatic COVID-19 infections after the first dose of the Pfizer–BioNTech vaccine (tozinameran) or placebo in a double-blind clinical trial (red: placebo; blue: tozinameran)[345]

Vaccine efficacy is the reduction in risk of getting the disease by vaccinated participants in a controlled trial compared with the risk of getting the disease by unvaccinated participants.[346] An efficacy of 0% means that the vaccine does not work (identical to placebo). An efficacy of 50% means that there are half as many cases of infection as in unvaccinated individuals.[citation needed]

COVID-19 vaccine efficacy may be adversely affected if the arm is held improperly or squeezed so the vaccine is injected subcutaneously instead of into the muscle.[347][348] The CDC guidance is to not repeat doses that are administered subcutaneously.[349]

It is not straightforward to compare the efficacies of the different vaccines because the trials were run with different populations, geographies, and variants of the virus.[350] In the case of COVID-19 prior to the advent of the delta variant, it was thought that a vaccine efficacy of 67% may be enough to slow the pandemic, but the current vaccines do not confer sterilizing immunity,[351] which is necessary to prevent transmission. Vaccine efficacy reflects disease prevention, a poor indicator of transmissibility of SARS‑CoV‑2 since asymptomatic people can be highly infectious.[352] The US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) set a cutoff of 50% as the efficacy required to approve a COVID-19 vaccine, with the lower limit of the 95% confidence interval being greater than 30%.[353][354][355] Aiming for a realistic population vaccination coverage rate of 75%, and depending on the actual basic reproduction number, the necessary effectiveness of a COVID-19 vaccine is expected to need to be at least 70% to prevent an epidemic and at least 80% to extinguish it without further measures, such as social distancing.[356]

The observed substantial efficacy of certain mRNA vaccines even after partial (1-dose) immunization[357][345] indicates a non-linear dose-efficacy relation already seen in the phase I-II study.[358] It suggests that personalization of the vaccine dose (regular dose to the elderly, reduced dose to the healthy young,[359] additional booster dose to the immunosuppressed[360]) might allow accelerating vaccination campaigns in settings of limited supplies, thereby shortening the pandemic, as predicted by pandemic modeling.[361]

Ranges below are 95% confidence intervals unless indicated otherwise, and all values are for all participants regardless of age, according to the references for each of the trials. By definition, the accuracy of the estimates without an associated confidence interval is unknown publicly. Efficacy against severe COVID-19 is the most important, since hospitalizations and deaths are a public health burden whose prevention is a priority.[362] Authorized and approved vaccines have shown the following efficacies:

COVID-19 vaccine efficacy
()
VaccineEfficacy by severity of COVID-19Trial locationRefs
Mild or moderate[A]Severe without hospitalization or death[B]Severe with hospitalization or death[C]
Oxford–AstraZeneca81% (6091%)[D]100% (97.5% CI, 72100%)100%[E]Multinational[363]
74% (6882%)[F]100%[E]100%[E]United States[364]
Pfizer–BioNTech95% (9098%)[G]66% (−125 to 96%)[H][G]Multinational[365]
95% (9098%)[G]Not reportedNot reportedUnited States[366]
Janssen66% (5575%)[I][J]85% (5497%)[J]100%[E][J][K]Multinational[367]
72% (5882%)[I][J]86% (−9 to 100%)[H][J]100%[E][J][K]United States
68% (4981%)[I][J]88% (8100%)[H][J]100%[E][J][K]Brazil
64% (4179%)[I][J]82% (4695%)[J]100%[E][J][K]South Africa
Moderna94% (8997%)[L]100%[E][M]100%[E][M]United States[369]
Sinopharm BIBP78% (6586%)100%[E]100%[E]Multinational[370]
79% (6688%)Not reported79% (2694%)[H]Multinational[371]
Sputnik V92% (8695%)100% (94100%)100%[E]Russia[372]
CoronaVac51% (3662%)[N]84% (5894%)[N]100% (56100%)[N]Brazil[373][374][375]
84% (6592%)100%[E]100% (20100%)[H]Turkey[376]
Covaxin78% (6586%)93% (57100%)India[377][378]
Sputnik Light79%[E]Not reportedNot reportedRussia[379]
Convidecia66%[E][N]91%[E][N]Not reportedMultinational[380][unreliable medical source?]
Sinopharm WIBP73% (5882%)100%[E][O]100%[E][O]Multinational[381]
Abdala92% (8696%)Not reportedNot reportedCuba[382][383][unreliable medical source?]
Soberana 0271% (5979%)63%[E]Not reportedCuba[384]
67% (5979%)97%[E]95%[E]Iran[385][386]
Soberana 02 and Soberana Plus92% (8796%)100%[E]Not reportedCuba[384]
Novavax90% (7595%)100%[E][O]100%[E][O]United Kingdom[387][388][389]
60% (2080%)[H]100%[E][O]100%[E][O]South Africa
90% (8395%)Not reportedNot reportedUnited States
Not reportedNot reportedMexico
CureVac48%[E]Not reportedNot reportedMultinational[390]
ZyCoV-D67%[E]Not reportedNot reportedIndia[391][unreliable medical source?]
EpiVacCorona79%[E]Not reportedNot reportedRussia[392][unreliable medical source?]
ZF200182%[E]Not reportedNot reportedMultinational[393][unreliable medical source?]
SCB-201967% (5477%)Not reportedNot reportedMultinational[394]
CoVLP71% (5980%)Not reportedNot reportedMultinational[395]
Sanofi–GSK COVID-19 vaccine58% (2777%)Not reportedNot reportedMultinational[396]


Effectiveness

Death rates for unvaccinated Americans substantially exceed those who were vaccinated, with bivalent boosters further reducing the death rate.[397]

Evidence from vaccine use during the pandemic shows vaccination can reduce infection and is most effective at preventing severe COVID-19 symptoms and death, but is less good at preventing mild COVID-19. Efficacy wanes over time but can be maintained with boosters.[398] In 2021, the CDC reported that unvaccinated people were 10 times more likely to be hospitalized and 11 times more likely to die than fully vaccinated people.[399][400]

The CDC reported that vaccine effectiveness fell from 91% against Alpha to 66% against Delta.[401] One expert stated that "those who are infected following vaccination are still not getting sick and not dying like was happening before vaccination."[402] By late August 2021, the Delta variant accounted for 99 percent of U.S. cases and was found to double the risk of severe illness and hospitalization for those not yet vaccinated.[403]

In November 2021, a study by the ECDC estimated that 470,000 lives over the age of 60 had been saved since the start of the vaccination roll-out in the European region.[404]

On 10 December 2021, the UK Health Security Agency reported that early data indicated a 20- to 40-fold reduction in neutralizing activity for Omicron by sera from Pfizer 2-dose vaccinees relative to earlier strains. After a booster dose (usually with an mRNA vaccine),[405] vaccine effectiveness against symptomatic disease was at 70%–75%, and the effectiveness against severe disease was expected to be higher.[406]

According to early December 2021 CDC data, "unvaccinated adults were about 97 times more likely to die from COVID-19 than fully vaccinated people who had received boosters".[407]

A meta-analysis looking into COVID-19 vaccine differences in immunosuppressed individuals found that people with a weakened immune system are less able to produce neutralizing antibodies. For example, organ transplant recipients need three vaccines to achieve seroconversion.[408] A study on the serologic response to mRNA vaccines among patients with lymphoma, leukemia, and myeloma found that one-quarter of patients did not produce measurable antibodies, varying by cancer type.[409]

In February 2023, a systematic review in The Lancet said that the protection afforded by infection was comparable to that from vaccination, albeit with an increased risk of severe illness and death from the disease of an initial infection.[410]

A January 2024 study by the CDC found that staying up to date on the vaccines could reduce the risk of strokes, blood clots and heart attacks related to COVID-19 in people aged 65 years or older or with a condition that makes them more vulnerable to said conditions.[411][412]

Studies

Real-world studies of vaccine effectiveness measure the extent to which a certain vaccine prevents infection, symptoms, hospitalization and death for the vaccinated individuals in a large population under routine conditions.[413]

  • In Israel, among the 715,425 individuals vaccinated by the mRNA vaccines from 20 December 2020, to 28 January 2021, starting seven days after the second shot, only 317 people (0.04%) displayed mild/moderate COVID-19 symptoms and only 16 people (0.002%) were hospitalized.[414]
  • CDC reported that under real-world conditions, mRNA vaccine effectiveness was 90% against infections regardless of symptom status; while effectiveness of partial immunization was 80%.[415]
  • In the UK, 15,121 health care workers from 104 hospitals who had tested negative for antibodies prior to the study, were followed by RT-PCR tests twice a week from 7 December 2020 to 5 February 2021, a study compared the positive results for the 90.7% vaccinated share of their cohort with the 9.3% unvaccinated share, and found that the Pfizer-BioNTech vaccine reduced all infections (including asymptomatic), by 72% (58–86%) three weeks after the first dose and 86% (76–97%) one week after the second dose, while Alpha was dominant.[416][needs update]
  • In Israel a study conducted from 17 January to 6 March 2021, found that Pfizer/BioNTech reduced asymptomatic Alpha infections by 94% and symptomatic COVID-19 infections by 97%.[417]
  • A study on the Queensland Population having only ever been exposed to the Omicron strain of COVID-19 found vaccine effectiveness against symptomatic hospitalizations to be 70% in the general population and similar for First Nations peoples.[418]
  • A study among pre-surgical patients across the Mayo Clinic system in the United States, showed that mRNA vaccines were 80% protective against asymptomatic infections.[419]
  • A UK study found that a single dose of the Oxford–AstraZeneca COVID-19 vaccine is about 73% (2790%) effective in people aged 70 and older.[420]
  • A study finds that nearly all teenagers admitted to intensive care units because of COVID-19 were unvaccinated.[421]

Pregnancy and fertility

Studies have not observed a correlation between COVID vaccination and fertility.[422][423]

A UK study found COVID vaccination is safe for pregnant women and is associated with a 15% decrease in the odds of stillbirth. Vaccination is recommended for pregnant women because pregnancy increases the risk of severe COVID. Researchers at St George's, University of London, and the Royal College of Obstetricians and Gynaecologists investigated 23 published studies and trials involving 117,552 vaccinated pregnant women. There was no increased risk of complications during pregnancy. Almost all pregnant women admitted to UK hospitals with COVID were unvaccinated.[424][425]

A US study of 46,079 pregnancies concluded that COVID vaccination is safe and does not raise the risk of preterm birth or small size babies.[426]

()
VaccineInitial effectiveness by severity of COVID-19Study locationRefs
AsymptomaticSymptomaticHospitalizationDeath
Oxford–AstraZeneca70% (6971%)Not reported87% (8588%)90% (8892%)Brazil[427]
Not reported89% (7894%)[i]Not reportedNot reportedEngland[429]
Not reportedNot reportedNot reported89%[ii]Argentina[430]
72% (6974%)Not reportedNot reported88% (7994%)Hungary[431]
Pfizer–BioNTech92% (9192%)97% (9797%)98% (9798%)97% (9697%)Israel[432]
92% (8895%)94% (8798%)87% (55100%)97%[ii]Israel[433][434]
83% (8384%)Not reportedNot reported91% (8992%)Hungary[431]
Not reported78% (7779%)98% (9699%)96% (9597%)Uruguay[435]
85% (7496%)Not reportedNot reportedEngland[436]
90% (6897%)Not reported100%[ii][iii]United States[437]
Moderna89% (8790%)Not reportedNot reported94% (9196%)Hungary[431]
90% (6897%)Not reported100%[ii][iii]United States[437]
Sinopharm BIBPNot reportedNot reportedNot reported84%[ii]Argentina[430]
69% (6770%)Not reportedNot reported88% (8689%)Hungary[431]
50% (4952%)Not reportedNot reported94% (9196%)Peru[438]
Sputnik VNot reported98%[ii]Not reportedNot reportedRussia[439][440]
Not reported98%[ii]100%[ii][iii]100%[ii][iii]United Arab Emirates[441]
Not reportedNot reportedNot reported93%[ii]Argentina[430]
86% (8487%)Not reportedNot reported98% (9699%)Hungary[431]
CoronaVac54% (5355%)Not reported73% (7274%)74% (7375%)Brazil[427]
Not reported66% (6567%)88% (8788%)86% (8588%)Chile[442][443]
Not reported60% (5961%)91% (8993%)95% (9396%)Uruguay[435]
Not reported94%[ii]96%[ii]98%[ii]Indonesia[444][445]
Not reported80%[ii]86%[ii]95%[ii]Brazil[446][447]
Sputnik Light79% (7582%)[ii][iv]Not reported88% (8092%)[ii][iv]85% (7591%)[ii][iv]Argentina[448]
()
Initial courseBooster doseInitial effectiveness by severity of COVID-19Study locationRefs
AsymptomaticSymptomaticHospitalizationDeath
CoronaVacCoronaVacNot reported80%[I]88%[I]Not reportedChile[449]
Pfizer–BioNTechNot reported90%[I]87%[I]Not reportedChile[449]
Oxford–AstraZenecaNot reported93%[I]96%[I]Not reportedChile[449]


Variants

World Health Organization video describing how variants proliferate in unvaccinated areas

The interplay between the SARS-CoV-2 virus and its human hosts was initially natural but is now being altered by the prompt availability of vaccines.[450] The potential emergence of a SARS-CoV-2 variant that is moderately or fully resistant to the antibody response elicited by the COVID-19 vaccines may necessitate modification of the vaccines.[451] The emergence of vaccine-resistant variants is more likely in a highly vaccinated population with uncontrolled transmission.[452] Trials indicate many vaccines developed for the initial strain have lower efficacy for some variants against symptomatic COVID-19.[453] As of February 2021, the US Food and Drug Administration believed that all FDA authorized vaccines remained effective in protecting against circulating strains of SARS-CoV-2.[451]

Alpha (lineage B.1.1.7)

Limited evidence from various preliminary studies reviewed by the WHO indicated retained efficacy/effectiveness against disease from Alpha with the Oxford–AstraZeneca vaccine, Pfizer–BioNTech and Novavax, with no data for other vaccines yet. Relevant to how vaccines can end the pandemic by preventing asymptomatic infection, they have also indicated retained antibody neutralization against Alpha with most of the widely distributed vaccines (Sputnik V, Pfizer–BioNTech, Moderna, CoronaVac, Sinopharm BIBP, Covaxin), minimal to moderate reduction with the Oxford–AstraZeneca and no data for other vaccines yet.[454]

In December 2020, a new SARS‑CoV‑2 variant, the Alpha variant or lineage B.1.1.7, was identified in the UK.[455]

Early results suggest protection to the variant from the Pfizer-BioNTech and Moderna vaccines.[456][457]

One study indicated that the Oxford–AstraZeneca COVID-19 vaccine had an efficacy of 42–89% against Alpha, versus 71–91% against other variants.[458][unreliable medical source?]

Preliminary data from a clinical trial indicates that the Novavax vaccine is ~96% effective for symptoms against the original variant and ~86% against Alpha.[459]

Beta (lineage B.1.351)

Limited evidence from various preliminary studies reviewed by the WHO have indicated reduced efficacy/effectiveness against disease from Beta with the Oxford–AstraZeneca vaccine (possibly substantial), Novavax (moderate), Pfizer–BioNTech and Janssen (minimal), with no data for other vaccines yet. Relevant to how vaccines can end the pandemic by preventing asymptomatic infection, they have also indicated possibly reduced antibody neutralization against Beta with most of the widely distributed vaccines (Oxford–AstraZeneca, Sputnik V, Janssen, Pfizer–BioNTech, Moderna, Novavax; minimal to substantial reduction) except CoronaVac and Sinopharm BIBP (minimal to modest reduction), with no data for other vaccines yet.[454]

Moderna has launched a trial of a vaccine to tackle the Beta variant or lineage B.1.351.[460] On 17 February 2021, Pfizer announced neutralization activity was reduced by two-thirds for this variant, while stating that no claims about the efficacy of the vaccine in preventing illness for this variant could yet be made.[461] Decreased neutralizing activity of sera from patients vaccinated with the Moderna and Pfizer-BioNTech vaccines against Beta was later confirmed by several studies.[457][462] On 1 April 2021, an update on a Pfizer/BioNTech South African vaccine trial stated that the vaccine was 100% effective so far (i.e., vaccinated participants saw no cases), with six of nine infections in the placebo control group being the Beta variant.[463]

In January 2021, Johnson & Johnson, which held trials for its Janssen vaccine in South Africa, reported the level of protection against moderate to severe COVID-19 infection was 72% in the United States and 57% in South Africa.[464][465][466]

On 6 February 2021, the Financial Times reported that provisional trial data from a study undertaken by South Africa's University of the Witwatersrand in conjunction with Oxford University demonstrated reduced efficacy of the Oxford–AstraZeneca COVID-19 vaccine against the variant.[467] The study found that in a sample size of 2,000 the AZD1222 vaccine afforded only "minimal protection" in all but the most severe cases of COVID-19.[468] On 7 February 2021, the Minister for Health for South Africa suspended the planned deployment of about a million doses of the vaccine whilst they examine the data and await advice on how to proceed.[468][469]

In a study reported in March[470] and May 2021,[471] the efficacy of the Novavax vaccine (NVX-CoV2373) was tested in a preliminary randomized, placebo-controlled study involving 2684 participants who were negative for COVID at baseline testing. Beta was the predominant variant to occur, with post-hoc analysis indicating a vaccine efficacy of Novavax against Beta of 51.0% for HIV-negative participants.[6][471]

Gamma (lineage P.1)

Limited evidence from various preliminary studies[when?] reviewed by the WHO have indicated likely retained efficacy/effectiveness against disease from Gamma with CoronaVac and Sinopharm BIBP, with no data for other vaccines yet. Relevant to how vaccines can end the pandemic by preventing asymptomatic infection, they have also indicated retained antibody neutralization against Gamma with Oxford–AstraZeneca and CoronaVac (no to minimal reduction) and slightly reduced neutralization with Pfizer–BioNTech and Moderna (minimal to moderate reduction), with no data for other vaccines yet.[454]

The Gamma variant or lineage P.1 variant (also known as 20J/501Y.V3), initially identified in Brazil, seems to partially escape vaccination with the Pfizer-BioNTech vaccine.[462]

Delta (lineage B.1.617.2)

Limited evidence from various preliminary studies[when?] reviewed by the WHO have indicated likely retained efficacy/effectiveness against disease from Delta with the Oxford–AstraZeneca vaccine and Pfizer–BioNTech, with no data for other vaccines yet. Relevant to how vaccines can end the pandemic by preventing asymptomatic infection, they have also indicated reduced antibody neutralization against Delta with single-dose Oxford–AstraZeneca (substantial reduction), Pfizer–BioNTech and Covaxin (modest to moderate reduction), with no data for other vaccines yet.[454]

In October 2020, a new variant was discovered in India, which was named lineage B.1.617. There were very few detections until January 2021, but by April it had spread to at least 20 countries in all continents except Antarctica and South America.[472][473][474] Mutations present in the spike protein in the B.1.617 lineage are associated with reduced antibody neutralization in laboratory experiments.[475][476] The variant has frequently been referred to as a 'Double mutant', even though in this respect it is not unusual.[477] the latter two of which may cause it to easily avoid antibodies.[478] In an update on 15 April 2021, PHE designated lineage B.1.617 as a 'Variant under investigation', VUI-21APR-01.[479] On 6 May 2021, Public Health England escalated lineage B.1.617.2 from a Variant Under Investigation to a Variant of Concern based on an assessment of transmissibility being at least equivalent to the Alpha variant.[480]

Omicron (lineage BA.2 and BA.2.12.2)

COVID-19 vaccine effectiveness was studied in adults without immunocompromising conditions in 10 US states between December 18, 2021 – June 10, 2022, when Omicron was prevalent. 3 doses of mRNA COVID-19 vaccines was 69% against COVID-19–associated hospitalization 7–119 days after the third vaccine dose and 52% against COVID-19–associated hospitalization more than 4 months after the 3rd dose. Among adults aged ≥50 years, COVID-19 vaccine effectiveness against COVID-19–associated hospitalization ≥120 days after receipt of dose 3 was only 32%, increasing to 66% ≥7 days after the fourth dose.[481]

Effect of neutralizing antibodies

One study found that the in vitro concentration (titer) of neutralizing antibodies elicited by a COVID-19 vaccine is a strong correlate of immune protection. The relationship between protection and neutralizing activity is nonlinear. A neutralization as low as 3% (95% CI, 113%) of the level of convalescence results in 50% efficacy against severe disease, with 20% (1428%) resulting in 50% efficacy against detectable infection. Protection against infection quickly decays, leaving individuals susceptible to mild infections, while protection against severe disease is largely retained and much more durable. The observed half-life of neutralizing titers was 65 days for mRNA vaccines (Pfizer–BioNTech, Moderna) during the first 4 months, increasing to 108 days over 8 months. Greater initial efficacy against infection likely results in a higher level of protection against serious disease in the long term (beyond 10 years, as seen in other vaccines such as smallpox, measles, mumps, and rubella), although the authors acknowledge that their simulations consider only protection from neutralizing antibodies and ignore other immune protection mechanisms, such as cell-mediated immunity, which may be more durable. This observation also applies to efficacy against variants and is particularly significant for vaccines with a lower initial efficacy; for example, a 5-fold reduction in neutralization would indicate a reduction in initial efficacy from 95% to 77% against a specific variant, and from a lower efficacy of 70% to 32% against that variant. For the Oxford–AstraZeneca vaccine, the observed efficacy is below the predicted 95% confidence interval. It is higher for Sputnik V and the convalescent response, and is within the predicted interval for the other vaccines evaluated (Pfizer–BioNTech, Moderna, Janssen, CoronaVac, Covaxin, Novavax).[482]

Side effects

All vaccines, including COVID-19 ones, can have minor side effects related to the mild trauma associated with the introduction of a foreign substance into the body.[483][484] These include soreness, redness, rash, and inflammation at the injection site. Other common side effects include fatigue, headache, myalgia (muscle pain), and arthralgia (joint pain) which generally resolve within a few days.[485]

Serious adverse events that follow the administration of vaccines are of high interest to the public.[486] It is important to recognize that the occurrence of an adverse event following vaccination does not necessarily mean that the vaccine caused the adverse event; the health problem may have been unrelated. Reporting of all adverse events, careful followup and statistical analysis of occurrences are required to determine whether or not a specific health problem is more likely to occur after a vaccine is administered.[487][488][489]

More serious side effects are very rare. Before COVID-19 vaccines such as Moderna and Pfizer/BioNTech were authorized for use in the general population, they had to pass phase III studies involving tens of thousands of people. Any serious side effects that did not appear during that testing are likely to occur less often than ~1 in 10,000 cases. It is important that Phase III trials be diverse, to ensure that safety results apply broadly. It is possible that side effects may affect a population that was not adequately represented during the initial testing. Pregnant women, immunocompromised people, and children are usually excluded from initial studies because they may be at higher risk. Further studies may be done to ensure their safety before vaccines are authorized for use in such populations. Subsequent examinations of the use of COVID vaccines in pregnant people and in children have shown similar outcomes to the general population and do not suggest greater risk for these groups.[489]

References