Abstract
The number of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected patients keeps rising in most of the European countries despite the pandemic precaution measures. The current antiviral and anti-inflammatory therapeutic approaches are only supportive, have limited efficacy, and the prevention in reducing the transmission of SARS-CoV-2 virus is the best hope for public health. It is presumed that an effective vaccination against SARS-CoV-2 infection could mobilize the innate and adaptive immune responses and provide a protection against severe forms of coronavirus disease 2019 (COVID-19) disease. As the race for the effective and safe vaccine has begun, different strategies were introduced. To date, viral vector-based vaccines, genetic vaccines, attenuated vaccines, and protein-based vaccines are the major vaccine types tested in the clinical trials. Over 80 clinical trials have been initiated; however, only 18 vaccines have reached the clinical phase II/III or III, and 4 vaccine candidates are under consideration or have been approved for the use so far. In addition, the protective effect of the off-target vaccines, such as Bacillus Calmette-Guérin and measles vaccine, is being explored in randomized prospective clinical trials with SARS-CoV-2-infected patients. In this review, we discuss the most promising anti-COVID-19 vaccine clinical trials and different vaccination strategies in order to provide more clarity into the ongoing clinical trials.
Keywords: Severe acute respiratory syndrome coronavirus 2, Coronavirus disease 2019, Vaccine, Clinical trials, Bacillus Calmette-Guérin, Measles
Introduction
The rapid spread of coronavirus disease 2019 (COVID-19) has globally become a serious issue [1]. The disease, caused by a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has been first reported in December 2019 (Wuhan, China) and declared by the World Health Organization (WHO) as a pandemic on March 11, 2020 [2]. To date, the COVID-19-associated deaths keep rising in most of the European countries. This phenomenon can be influenced by the pandemic precaution measures, such as mandatory face masks wearing; however, the end of the pandemic may not be seen until an effective anti-COVID-19 vaccine is developed [3, 4]. SARS-CoV-2 belongs to the single-stranded RNA viruses which originated in bats [5]. SARS-CoV-2 exhibits the same structural and molecular patterns as other coronaviruses, such as structural proteins S (spike), E (envelope), M (membrane), and N (nucleocapsid) (Fig. 1) [6, 7]. The binding of the virus and its further entry into the host cell through angiotensin-converting enzyme 2 is mediated by the viral S protein. As the S protein is processed by a protease transmembrane protease serine 2, viral fusion with the host cell occurs. The first genetic analyses of the virus revealed an 89% nucleotide identity with bat virus SARS-like-CoVZXC21 [6]. Further investigation, however, confirmed a high similarity (96%) between the SARS-CoV-2 and the betaCoV RaTG13 of bats [1, 8, 9]. In COVID-19, it has been attempted by multiple studies to identify the intermediate host and on the basis of the available data, pangolins are most likely the mammals serving as a SARS-CoV-2 intermediate host [10]. The understanding and identification of an intermediate host is of major importance. The reason stems from the fact that blocking the human-host contact may restrain further spread of the novel disease variants, such as the Cluster 5 identified originally in minks. This cluster includes Y453F mutation in the spike protein and was reported among people in Denmark. Additionally, the virus was shown to infect multiple animal species under experimental conditions, and also few cases of household cats and dogs were reported to be positive for SARS-CoV-2 RNA. Therefore, concerns regarding the new sources of infection, novel potential viral strains, and uncontrolled outbreaks rise [11, 12, 13]. Moreover, the identification of an intermediate host may allow novel vaccine testing [1]. As a matter of fact, in Middle East respiratory syndrome disease, the first vaccine candidates were tested in dromedary camels [14]. The total number of deaths in COVID-19 is affected by the high transmissibility (assessed by the basic reproduction number, R0) [15]. COVID-19 is mainly transmitted through respiratory droplets from sneezing and coughing; however, few other transmission routes have been described, such as alimentary transmission or through conjunctival mucosa [1, 4]. To date, severe and deadly forms of COVID-19 have been reported. Underlying health conditions were seen in cases with life-threatening course of COVID-19, and thus, the COVID-19 pandemic represents an enormous threat for the elderly [16] or chronically ill patients, particularly for those suffering from severe obesity, CKD, diabetes, arterial hypertension, or asthma [17]. On the other hand, the viral load is presumably one of the factors that dictate the clinical course of the disease [18]. This may also be affected by the transmission route, and therefore, asymptomatic COVID-19 patients were reported throughout multiple studies [19, 20]. Currently, multiple therapeutic approaches are being applied to deal with the infection. However, these approaches are rather supportive, and the prevention in reducing the transmission is the best hope for public health [1, 5, 21]. We have reviewed the current status of all anti-COVID-19 vaccines that have reached the clinical trials in humans. As the race for the effective and safe vaccine has begun, different strategies were introduced. In this review, we discuss the most promising anti-COVID-19 vaccine clinical trials and discuss different vaccination strategies in order to provide more clarity into the ongoing clinical trials.
Fig. 1.
Structure of SARS-CoV-2 virus: spike (S) glycoprotein, membrane (M) glycoprotein, envelope (E) glycoprotein, hemagglutinin-esterase (HA) glycoprotein, and nucleocapsid (N) phosphoprotein [97, 98].
Methods
We conducted a comprehensive review of the literature on the progress of anti-SARS-CoV-2 vaccine development preventing the rapid spread of COVID-19 disease. The vaccines that were registered until December 2020 in the Clinical Trials database by the National Library of Medicine at the US National Institutes of Health were reviewed [22]. The authors followed the proposed guidelines for biomedical narrative review preparation [23].
Immune Response to SARS-CoV-2 Virus
The immune system affects the severity of the COVID-19 disease [7]. SARS-CoV-2 infection has an impact on both innate and adaptive immune responses. It has been described that SARS-CoV-2 enters the human body through physical barriers, such as respiratory tract, oral mucosa, and conjunctival epithelium [24, 25]. The dendritic cells, macrophages, and neutrophils represent the first line of defense, and their functions may be promoted by the production of type I and III interferons by SARS-CoV-2-infected epithelial cells [26]. The adaptive T-cell- and B-cell-mediated immune responses are also presented in COVID-19 disease and, however, can be suppressed by SARS-CoV-2 [7]. In some cases, the innate immune cells may contribute to the excessive inflammation and, therefore, to the disease progression. The inability to reach control over the infection may result in dysregulated inflammatory responses that are potentially lethal. The IgM and IgG antibodies to SARS-CoV-2 are detectable within 1–2 weeks and began to decrease by 8 weeks [27]. Several studies also reported that IgA response peaks earlier than IgM [28]. The antibody response particularly leads to production of neutralizing antibodies to the S protein and to the nucleoprotein. S protein is also the main target of the majority of newly designed vaccines [29]. The magnitude of neutralizing antibodies positively correlates with the disease severity and the robustness of T-cell response [30]. T-cell responses were detectable in individuals recovering from mild COVID-19 who did not have detectable antibody responses to SARS-CoV-2 [31, 32]. The effective vaccination may not eradicate the SARS-CoV-2 virus but may at least protect from severe and deadly forms of the COVID-19 disease [7]. Current knowledge regarding the diverse aspects of SARS-CoV-2-immune system interplay shall be reflected in the vaccine design, including the selection of antigens, the vaccine platforms and adjuvants, the vaccination routes, and the dosage regimens [33, 34]. The key points of the SARS-CoV-2 vaccination strategies are discussed below. To date, over 80 clinical trials have been registered in the Clinical Trials database by the National Library of Medicine at the US National Institutes of Health; however, only 34 of them are active and recruiting (11 of phase I, 8 of phase I/II, 3 of phase II, 1 of phase II/III, and 11 of phase III) [22]. Moreover, 2 vaccine candidates have been approved for use by the US Food and Drug Administration (FDA) − BNT162/Comirnaty and mRNA-1273). BNT162/Comirnaty has been also permitted by the European Medicines Agency (EMA). The vaccination program with BNT162/Comirnaty has been recently initiated in many European countries. The main features of the registered and ongoing anti-SARS-CoV-2 vaccine clinical trial are summarized in Table 1 (Phase I and I/II) and Table 2 (Phase II, II/III, and III).
Table 1.
Summary of active and recruiting Phase I and I/II trials registered in the Clinical Trials database (National Library of Medicine at the US National Institutes of Health)
| Vaccine name | Mechanism of action | Trial design | Outcome | Dosage regimen | Participants, n | Estimated study completion | Sponsor | Identifier |
|---|---|---|---|---|---|---|---|---|
|
Phase I clinical trials SCB-2019 |
Recombinant protein with adjuvant AS03 or CpG/alum | Randomized, double-blinded, placebo-controlled |
Safety and immunogenicity | 2 i.m. doses | 150 patients (18–75 yr) | March, 2021 | Clover Biopharmaceuticals AUS Pty Ltd | NCT04405908 |
| AdimrSC-2f | Baculovirus vector with/without alum | Open-label | Safety and immunogenicity | 1 dose | 70 patients (20–60 yr) | March, 2021 | Adimmune Corporation | NCT04522089 |
| MVA-SARS-2-S | Modified vaccinia virus ankara vector | Open-label | Safety, tolerability and immunogenicity | 2 doses | 30 patients (18–55 yr) | May, 2021 | NCT04569383 | |
| MVC-COV1901 | Reombinant protein with adjuvant CpG1018 | Open-label | Safety and immunogenicity | 2 i.m. dose | 45 patients (20–50 yr) | June, 2021 | Medigen Vaccine Biologics Corp. | NCT04487210 |
| COVAX-19 | Recombinant protein with adjuvant Advax-SM | Randomized, controlled | Safety and immunogenicity | 1 dose | 40 patients (18–45 yr) | July, 2021 | Vaxine Pty Ltd | NCT04453852 |
| GRAd-COV2 | Gorilla Adenovirus vector | Open-label | Safety and immunogenicity | 1 i.m. dose | 90 patients (18–85 yr) | July, 2021 | ReiThera Srl | NCT04528641 |
| UB-612 | Recombinant protein | Open-label | Safety, tolerability and immunogenicity | 1 dose | 60 patients (20–55 yr) | August, 2021 | United Biomedical Inc., Asia | NCT04545749 |
| SARS-CoV-2 Sclamp | Recombinant protein with adjuvant MF59 | Randomized, double-blinded, placebo-controlled | Safety and immunogenicity | 2 doses | 216 patients (>56 yr) | September, 2021 | The University of Queensland | NCT04495933 |
| TMV-083 | Measles vector | Randomized placebo-controlled and dose escalation | Safety, tolerability and immunogenicity | 1–2 i.m. dose(s) | 90 patients (18–55 yr) | October, 2 021 | Institut Pasteur | NCT04497298 |
| CoVac-1 | Recombinant peptide with adjuvant XS15 | Open label | Safety and immunogenicity | single s.c. dose | 36 patients (15–≥75 yr) | December, 2021 | University Hospital Tuebingen | NCT04546841 |
| bacTRL-Spike | DNA (Bifidobacterium longum vector) | Observer-blinded, randomized, placebo-controlled | Safety, tolerability and immunogenicity | 1 p.o. dose | 12 patients (>18 yr) | February, 2022 | Symvivo Corporation | NCT04334980 |
| Phase I/II clinical trials | ||||||||
| ARCT-021 | mRNA | Randomized, double-blinded, placebo-controlled | Safety, tolerability and immunogenicity | 1 i.m. dose | 92 patients (21–80 yr) | January, 2021 | Arcturus Therapeutics, Inc. | NCT04480957 |
| SARS-CoV-2 | Inactivated virus | Randomized, double-blinded, placebo-controlled | Safety and immunogenicity | 2 doses | 552 patients (3–17 yr) | September, 2021 | Sinovac Research and Development Co., Ltd. | NCT04551547 |
| SARS-CoV-2 | Inactivated virus | Randomized, double-blinded, placebo-controlled | Safety and immunogenicity | 2 doses | 942 patients (18–59 yr) | September, 2021 | Chinese Academy of Medical Sciences | NCT04412538 |
| Recombinant new coronavirus | Recombinant protein | Randomized, double-blined, placebo-controlled | Safety and tolerability | 1 i.m. dose | 50 patients (>60 yr) | December, 2021 | Anhui Zhifei Longcom Biologic Pharmacy Co., Ltd. | NCT04550351 |
| INO-4800 | DNA (plasmid vector pGX9501) | Open label | Tolerability, safety, and immunogenicity | 2 i.d. doses | 160 patients (19–64 yr) | February, 2022 | International Vaccine Institute | NCT04447781 |
| GX-19 | DNA | Randomized, double-blind, placebo-controlled | Safety, tolerability, and immunogenicity | 2 i.m. doses | 210 patients (18–50 yr) | June, 2022 | Genexine, Inc. | NCT04445389 |
| LV-SMENP-DC | Lentivirus modified DC and antigen-specific CTLs | Open-label trial | Safety and efficacy | 1 s.c. dose/1 infusion | 100 patients (6 mo–80 yr) | December, 2024 | Shenzhen Geno-Immune Medical Institute | NCT04276896 |
| COVID-19/aAPC | Inactivated artificial APC upon lentivirus modification | Open label | Safety and immune reactivity | 3 s.c. doses | 100 patients (6 mo–80 yr) | December, 2024 | Shenzhen Geno-Immune Medical Institute | NCT04276896 |
APC, antigen-presenting cell.
Table 2.
Summary of active and recruiting Phase II, II/III, and III trials registered in Clinical Trials database (National Library of Medicine at the US National Institutes of Health)
| Vaccine name | Mechanism of action | Trial design | Outcome | Dosage regimen | Participants, n | Estimated study completion | Sponsor | Identifier |
|---|---|---|---|---|---|---|---|---|
|
Phase II clinical trials CVnCoV |
mRNA | Observer-blinded, multicenter, controlled | Safety, reactogenicity, and immunogenicity | 1–2 i.m. doses | 660 patients (>18 yr) | November, 2021 | Cure Vac AG | NCT04515147 |
| NVX-CoV2373 | Recombinant protein with adjuvant matrix-M1 | Observer-blinded, randomized, placebo-controlled | Efficacy, immunogenicity, and safety | 2 i.m. doses | 4,400 patients (18–64 yr) | November, 2021 | Novavax | NCT04533399 |
| Ad5-nCoV | Adenovirus-5 vector | Randomized, double-blinded, placebo-controlled | Safety and immunogenicity | 2 i.m. doses | 481 patients (>6 yr) | October, 2022 | CanSino Biologics Inc. | NCT04566770 |
| Phase II/III clinical trials | ||||||||
| BNT162 | mRNA | Observer-blinded, randomized, placebo-controlled | Safety, tolerability, immunogenicity and efficacy | 2 i.m. doses | 43,998 patients (>12 yr) | January, 2023 | BioNTech RNA Pharmaceuticals GmbH | NCT04368728 |
| Phase III clinical trials | ||||||||
| CoronaVac | Inactivated virus | Randomized, double-blinded, placebo-controlled | Efficacy, safety and immunogenicity | 2 doses | 13,000 patients (>18 yr) | April, 2021 | Health Institutes of NCT04582344 Turkey | |
| SARS-CoV-2 | Inactivated virus | Randomized, double-blinded | Non-inferiority of the commercial scale | 2 doses | 1,040 patients (>18 yr) | May, 2021 | Sinovac Research and Development Co., Ltd. | NCT04617483 |
| SARS-CoV-2 | Inactivated virus | Randomized, double-blinded, placebo-controlled | Efficacy, safety, and immunogenicity | 2 doses | 6,000 patients (18–60 yr) | September, 2021 | Universidad Perunana Cayetano Heredia | NCT04612972 |
| ChAdOxl nCoV-19 | ChAdOxl vector | Randomized, controlled | Efficacy, safety, and immunogenicity | 1–2 doses | 10,300 patients (>18 yr) | September, 2021 | University of Oxford | NCT04536051 |
| SARS-CoV-2 | Inactivated virus | Randomized, double-blinded, parallel placebo-controlled | Efficacy, safety, and immunogenicity | 2 doses | 45,000 patients (>18 yr) | September, 2021 | China National Biotec Group Company limited | NCT04510207 |
| SARS-CoV-2 | Inactivated virus | Observer-blinded, randomized, placebo-controlled | Efficacy, safety, and immunogenicity | 2 i.m. doses | 1,620 patients (18–59 yr) | September, 2021 | PT Bio Farma | NCT04508075 |
| Adsorbed COVID-19 | Inactivated virus | Randomized, double-blinded, placebo-Controlled | Efficacy and safety in health-care professionals | 2 doses | 13,060 patients (18–59 yr) | October, 2021 | Butan tan Institute | NCT04456595 |
| SARS-CoV-2 rS/matrix-M 1-adjuvant | Recombinant S protein | Randomized, observer-blinded, placebo-controlled | Efficacy and safety | 2 doses | 15,000 patients (18–84 yr) | January, 2022 | Novovax | NCT04583995 |
| Ad5-nCoV | Adenovirus-5 vector | Randomized, double-blinded, placebo-controlled | Efficacy, safety, and immunogenicity | 1 i.m. dose | 40,000 patients(>18 yr) | January, 2022 | CanSino Biologics Inc. | NCT04526990 |
| AZD1222 | ChAdOxl vector | Randomized, double-blinded, placebo-controlled | Efficacy, safety, and immunogenicity | 2 i.m. doses | 40,000 patients (>18 yr) | February, 2023 | AstraZeneca | NCT04516746 |
| Ad26.COV2.S | Adenovirus-26 vector | Randomized, double-blinded, placebo-controlled | Efficacy and safety | 1 i.m. dose | 60,000 patients (>18 yr) | March, 2023 | Janssen Vaccines & NCT04505722 Prevention B.V. | |
Inactivated Vaccines
Inactivated vaccines are based on presenting the form of pathogen with a loss of disease-producing capacity. The virus cultivation occurs in cell lines that represent a substrate for the production of large quantities of antigen. Virus multiplication is often followed by a purification and concentration prior to the vaccine inactivation [35]. Formaldehyde and beta-propiolactone are used in the majority of licensed human antiviral vaccines to inactivate the virus [36]. Multiple doses or adjuvants are required to achieve sufficient efficacy of inactivated vaccines [37]. To date, 4 inactivated vaccines have reached the phase III clinical trials and are currently under evaluation (#NCT04510207, #NCT04508075, and #NCT04456595).
Subunit Vaccines
Subunit vaccines are composed of purified antigens instead of whole microorganisms, and different carriers serve as a transporter for those antigens. In the anti-SARS-CoV-2 subunit vaccines, the antigens are represented by viral proteins, peptides, or nanoparticles. Because of relatively low immunogenicity of the subunit vaccines, adjuvants are required to create a stronger immune response [38]. Currently, aluminum salts, virosomes, AS03 (α-tocopherol, surfactant polysorbate 80, and squalene), AS04 (Monophosphoryl lipid A, MPLA) and MF59 (squalene) are the most widespread licensing adjuvants [39, 40]. These adjuvant systems are also used in a number of anti-SARS-CoV-2 vaccines; however, novel adjuvants are tested as well. Advax-SM (clinical trial #NCT04453852) is an adjuvant composed of polysaccharide delta-inulin and CpG oligodeoxynucleotide (CpG ODN). CpG ODN is a TLR 9 agonist with T-helper 1 skewing properties [41]. Granulocyte macrophage colony-stimulating factor is a proinflammatory cytokine which may also serve as an adjuvant (#NCT03305341 and #NCT04386252).
Nonetheless, subunit vaccines provide a high level of safety. Bacterial expression systems represent the most commonly used technique to produce recombinant proteins with high expression. However, in antigens where posttranslational modification is required, the use of mammalian or insect cells may be considered [42]. Other offered alternatives include transgenic plants [43]. This technology has been also adopted as a source of SARS-CoV-2 virus spike protein for the purpose of vaccine development (phase I/II trial #NCT04473690). To date, there are no SARS-CoV-2-recombinant vaccines tested in phase III. Three vaccines are being evaluated in clinical phase I/II (#NCT04527575 and #NCT04537208) and phase II (#NCT04533399). Recombinant technologies, including bacterial, insect, or mammalian cell-based expression systems can also be used for the generation of virus-like particles (VLPs). VLPs that are formed by a capsid protein do not contain infectious viral RNA or DNA. Moreover, the antisense RNA can inhibit virus expression, and the viral RNA/DNA may activate different pattern recognition receptors and trigger antiviral immune responses. These responses are primarily characterized by a production of type I interferons and proinflammatory cytokines [44]. VLPs-based anti-SARS-CoV-2 vaccine (#NCT04450004) is currently tested in phase I clinical trials.
DNA Vaccines
DNA vaccines deliver coronavirus's genes to the human cells. The vaccination principle depends on the DNA translocation into the cell nucleus where the transcription of the antigen is initiated and followed by a translation. DNA vaccines frequently use plasmids as vectors. Depending on the route of vaccine administration (intramuscular, intradermal, and subcutaneous), either myocytes or keratinocytes are addressed. Nonetheless, antigen-presenting cells residing close to the site of application can be transfected directly by DNA vaccines as well. In such cases, the expressed antigens are loaded onto MHC I and MHC II molecules due to the cross-priming potential [45]. The produced antigens are either released by exosomes or apoptotic bodies which lead to a recognition by antigen-presenting cells and further evolvement of humoral or cytotoxic immune responses. Different delivery devices are used to create a robust immune response [46, 47]. The main safety concerns imply a possible integration of transfected DNA into somatic and/or germ cells of the host. In such cases, a dysregulation of gene expression might occur and lead to various mutations. However, only extrachromosomal plasmids with a very low level of chromosomal integration are usually employed in the development of DNA vaccines. Furthermore, the majority of plasmids remain at the site of administration [48]. Three anti-SARS-CoV-2 DNA vaccines are currently in phase I/II of clinical assessment (#NCT04527081, #NCT04447781, and #NCT04445389).
RNA Vaccines
Messenger RNA (mRNA) vaccines were first tested in early 1990s; however, their use was limited because of their instability [49]. The mRNA encodes the genetic information to produce an antigen, and thus, RNA vaccines also lead to a production of coronavirus's proteins in vivo. The in vitro generation of an RNA vaccine includes a reaction of a DNA plasmid template and a recombinant RNA polymerase. In addition, a synthetic cap analog and a poly(A) tail are added to form a mature RNA sequence. The stabilization is further achieved by various transport systems (such as lipid nanoparticles, nano-emulsions, and cationic peptides) or methods enabling facilitated transfection (gene gun and electroporation). Conventional mRNA vaccines are based on the initiation of the transient antigen expression in the cytoplasm of the host cells. Another platform is represented by self-amplifying mRNA vaccines that contain both the genes coding the targeted antigen as well as the genes required for the self-replication (mostly RNA-dependent RNA polymerase) [50]. The conventional mRNA vaccines induce a prompt antigen expression, and the expressed antigens generate both humoral and cellular immune responses [51, 52, 53, 54]. In self-amplifying mRNA vaccines, a delayed antigen expression may prevail and limit the efficacy of the vaccine. Yet, the self-amplifying mRNA vaccine platform reaches higher yields, and thus, an equivalent protection is conferred at much lower doses [55]. Regarding the safety profiles, the replicons of both abovementioned platforms are not capable of producing viral particles due to the lack of viral structural proteins. Moreover, neither conventional nor self-amplifying mRNA vaccines can integrate into the host genome. The mRNA-based vaccines were able to induce production of functional antibodies with neutralizing properties in rabies, influenza, or Zika virus and represent also a promising vaccination strategy in the prevention against COVID-19 infection [56]. The efficacy and safety are being assessed in the ongoing phase II and phase III clinical trials (#NCT04515147, #NCT04368728, and #NCT04470427).
Viral Vector-Based Vaccines
Viral vector-based vaccines (VBVs) are constructed by engineering a viral vector to carry coronavirus genes and slowly replicate in the host cells. The replication leads to the production of coronavirus proteins and a subsequent immune system activation. Potential viral vectors include a broad spectrum of both DNA and RNA viruses, such as adenoviruses, parvoviruses (e.g., adeno-associated viruses), togaviruses (e.g., Semliki Forest virus), paramyxoviruses (e.g., measles virus, Newcastle disease virus or human parainfluenza virus), rhabdoviruses (e.g., vesicular stomatitis virus), and poxviruses (e.g., Modified Vaccinia Ankara). These viral vectors can be constructed as replicating or nonreplicating vectors [57]. The efficacy of VBV may be significantly affected by the preexisting immunity of the host. This can be avoided by the use of nonhuman or rare serotype vectors [58, 59]. The main safety concerns include the potential of viral genes to integrate into the host genome and uncontrolled replication. On the other hand, the high yield production supports the use of VBV particularly in the time of disease outbreaks [60]. In SARS-CoV-2 vaccine development, the most commonly used vectors are the adenoviral vectors, such as ChAdOx (#NCT04536051 and #NCT04516746), adenovirus type 5 (#NCT04564716, #NCT04540419, and #NCT04526990), and adenovirus type 26 (#NCT04564716 and #NCT04505722). All these vaccines are currently being evaluated in phase III clinical trials. However, lentivirus (#NCT04276896 and #NCT04428073), measles (#NCT04498247 and #NCT04497298), baculovirus (#NCT04522089), or MVA (#NCT04569383) are also being tested.
Routes of Administration
The route of administration is another crucial aspect that significantly affects the vaccine efficacy. Conventional vaccination approaches include mucosal and parenteral administration. Parenteral route generally includes intramuscular (IM), subcutaneous (SC), and intradermal (ID) application [61]. Due to an increased infiltration of dermis with DCs, the ID application initiates greater adaptive immune response than the IM application providing a significant dose sparing effect. However, improved efficacy is associated with a less favorable safety profile [62]. Mucosal vaccines including the intranasal and oral administration routes provide a number of advantages, particularly the avoidance of a needle application and a lower risk of systemic adverse events (AEs). Nevertheless, the systemic responses to mucosal vaccination are weaker as compared to parenterally administrated vaccinations [63]. The majority of vaccinations are administered intramuscularly. However, the intradermal administration (#NCT03305341 and #NCT04447781), oral administration (#NCT04334980), or combined administration (intramuscular and mucosal) of the vaccines (#NCT04552366) is being evaluated.
The Most Promising Anti-COVID-19 Vaccines
The first vaccine with favorable results was the ChAdOx1 nCoV-19 (also known as AZD1222, AstraZeneca/University of Oxford). This vaccine was evaluated in July 2020 in the phase I/II single-blind randomized trial with 1077 participants. The patients were exposed to 2 doses of recombinant adenovirus vaccine ChAdOx1 nCoV-19 in a 28-day interval. Neutralizing antibodies against SARS-CoV-2 spike protein were detected in 91% of patients after the first dose. The production of virus-specific antibodies peaked on day 28, and a robust T-cell response was also observed. Severe AEs were not reported [64]. The efficacy has been recently confirmed in a pooled interim analysis of 4 phase I/–III clinical trials. [65]. The preliminary results of a double-blind, randomized, placebo-controlled phase I/II trial with another vaccine candidate Ad26.COV2 (Janssen-Cilag International N.V.) were published in September 2020 (data published as a preprint). The study included >800 patients, and the seroconversion rate with the production of anti-spike protein-neutralizing antibodies was seen in 83–100% patients across the cohorts. The specific T-helper 1 response was detected in 80–83% of the participants with a robust activation of CD8+ T cells. Local and systemic AEs included fever, headache, myalgia, and injection site pain. In November 2020, the preliminary results of an open-label clinical trial including 45 healthy adults treated with mRNA-1273 vaccine (Moderna Biotech Spain, S.L.) were shown. The vaccine was administered in 2 doses and various concentrations (25, 100, and 250 μg). The seroconversion occurred in all participants, and the response depended on the administered dose. On the other hand, higher doses were associated with increased risk of systemic AEs (reported in 33% participants) [66]. The mRNA BNT162 (Pfizer/BioNTech) vaccine has been proven in a large observer-blinded, randomized, placebo-controlled trial with >43,000 participants to be a safe and potent vaccine. Two doses (30 μg per dose) were administered in a 21-day interval. The overall reported efficacy of 95% was observed across different subgroups defined by age, sex, race, ethnicity, baseline BMI, and the presence of coexisting conditions. A 52% efficacy was observed after the first dose indicating early protection. Injection site reactions, fatigue, headaches, and fevers were the most common AEs (reported in 27% of patients) [67].
Off-Target Vaccinations
It has been shown that various vaccination principles bear a potential to prevent or at least to suppress the detrimental effect of COVID-19. The cross-protection has been discussed particularly in association with the Bacillus Calmette-Guérin (BCG) vaccination. In BCG vaccinated populations, the incidence and the severity of the COVID-19 disease appears to be lower than BCG-non-vaccinated populations [68, 69, 70, 71, 72]. A similar phenomenon of a cross-protection was also described in individuals after the measles infection, or the measles, mumps, rubella vaccination [73, 74]. These findings were supported by previous observations that a nonspecific effect of these vaccines protects against other infections including those of viral origin [75, 76, 77, 78]. Therefore, a number of prospective randomized clinical trials has been initiated to validate the preventive effect of both BCG vaccine (#NCT04379336, #NCT04537663, #NCT04327206, #NCT04328441, #NCT04461379, #NCT04369794, #NCT04414267, #NCT04384549, and other) and the measles vaccine (NCT04357028 and NCT04475081).
Discussion
An effective vaccination against SARS-CoV-2 infection could mobilize the innate and adaptive immune responses and provide a protection against severe forms of COVID-19. Since the SARS-CoV-2 virus may undergo mutational changes and antigenically evolve over time, the vaccine may become, as in influenza, a seasonal protection. On the other hand, coronaviruses have a low mutation rate in comparison to other RNA viruses, particularly Influenza type A [79]. The anti-SARS-CoV-2 vaccination may not lead to the eradication of the disease, however, may most certainly decrease the disease-related mortality and morbidity [75, 80, 81].
In COVID-19, live vaccines have not yet been registered in human clinical trials. Previous studies have shown that booster (secondary) vaccination with life-attenuated viruses generate only limited immune response as compared to the first vaccination dose [82, 83]. Also, the preexisting immunity caused by previous COVID-19 infection may inhibit the efficacy of live attenuated vaccines and the presence of neutralizing antibodies can be associated with the virus neutralization. Moreover, the genome instability may lead to a back mutation recovering their virulence, mainly in viruses with higher mutation rate [84, 85, 86]. Therefore, the live vaccines may not represent the optimal vaccine type in prevention of COVID-19 infection [87].
Other classical vaccination approaches, such as inactivated or recombinant subunit vaccines, are currently being tested against COVID-19 infection in clinical trials. Their efficacy is, however, also limited by relatively low response rates and short-term immune memory. Therefore, both approaches require the use of potent adjuvants such as CpG ODN, ADVAX-SM, or granulocyte macrophage colony-stimulating factor representing novel strategies to enhance immune response. Another obstacle of inactivated vaccines represents the risk of reversed outcome associated with enhanced virus-mediated disease and fatal consequences. In the cases of respiratory syncytial virus vaccination, the vaccine was found to be immunogenic; however, the elicited antibodies were nonprotective and respiratory syncytial virus disease progression in single cases resulted in death in vaccinated infants [88, 89]. Therefore, novel vaccine designs, such as mRNA vaccines and recombinant VBV vaccines are being extensively investigated to avoid these barriers. FDA and EMA have been recently authorized mRNA vaccine candidate BNT162/comirnaty and mRNA-1273 for the use [90]. Furthermore, other vaccine candidates, including Ad26.COV2.S and ChAdOx1-SARS-CoV-2 (AZD1222), are currently under consideration [91]. To note, novel vaccine approaches raise many safety concerns. However, strategies following the good manufacturing practice principles and appropriate preclinical and clinical testing under the surveillance of regulatory authorities should ensure good safety profile [92]. The efficacy and safety also remain an issue in immunocompromised patients with primary or secondary immunodeficiencies. Generally, administration of attenuated life virus vaccines has to be considered with caution and indicated upon careful individual assessment of risk and benefits in these patients. Non-live vaccines such as influenza or pneumococcal vaccine are regarded as safe, even though their efficacy may be reduced in patients with severely impaired antibody response [93, 94]. Similar principles might be applied also in anti-SARS-CoV-2 vaccination; however, neither recommendations nor guidelines are available yet. The level of virus-specific antibodies does not correlate with the acquired immune response mediated by T cells that might be preserved in the majority of the antibody deficient patients [95, 96]. Thus, the examination of T-cell response should also be considered in healthy subjects. Surprisingly, other vaccines such as BCG and measles vaccine have also shown efficacy in the prevention of COVID-19 disease. The leading vaccine candidates are currently being distributed and among selected population with great expectations to reduce the COVID-19 spread.
Statement of Ethics
Ethics approval was not required.
Conflict of Interest Statement
Z.S., J.S., J.B., and T.M. declare that they have no conflict of interest regarding the publication of this article.
Funding Sources
The preparation of the manuscript was supported by internal financial sources of the Department of Immunology, Second Faculty of Medicine, Charles University, and University Hospital Motol. No external funding sources were required.
Author Contributions
Z.S.: substantial contributions to the conception and design of the manuscript, analysis and interpretation of the literature, and drafting. J.S.: substantial contributions to the analysis and interpretation of the literature and drafting. J.B.: revising the manuscript critically for important intellectual content and final approval of the version to be published. T.M.: substantial contributions to the conception and design of the manuscript, analysis and interpretation of the literature, drafting, and revising it critically for important intellectual content.
Acknowledgements
The preparation of this review article was endorsed by the Department of Immunology, Second Faculty of Medicine, Charles University, University Hospital Motol, and Department of Paediatric and Adult Rheumatology, University Hospital Motol, Prague, Czech Republic.
References
- 1.Weston S, Frieman MB. COVID-19: knowns, unknowns, and questions. mSphere. 2020;5((2)):e00203–20. doi: 10.1128/mSphere.00203-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Hiscott J, Alexandridi M, Muscolini M, Tassone E, Palermo E, Soultsioti M, et al. The global impact of the coronavirus pandemic. Cytokine Growth Factor Rev. 2020;53:1–9. doi: 10.1016/j.cytogfr.2020.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Kaur SP, Gupta V. COVID-19 vaccine: a comprehensive status report. Virus Res. 2020;288:198114. doi: 10.1016/j.virusres.2020.198114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Strizova Z, Bartunkova J, Smrz D. Can wearing face masks in public affect transmission route and viral load in COVID-19? Cent Eur J Public Health. 2020;28((2)):161–2. doi: 10.21101/cejph.a6290. [DOI] [PubMed] [Google Scholar]
- 5.Prompetchara E, Ketloy C, Palaga T. Immune responses in COVID-19 and potential vaccines: lessons learned from SARS and MERS epidemic. Asian Pac J Allergy Immunol. 2020;38((1)):1–9. doi: 10.12932/AP-200220-0772. [DOI] [PubMed] [Google Scholar]
- 6.Chan JF, Kok KH, Zhu Z, Chu H, To KK, Yuan S, et al. Genomic characterization of the 2019 novel human-pathogenic coronavirus isolated from a patient with atypical pneumonia after visiting Wuhan. Emerg Microbes Infect. 2020;9((1)):221–36. doi: 10.1080/22221751.2020.1719902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Paces J, Strizova Z, Smrz D, Cerny J. COVID-19 and the immune system. Physiol Res. 2020;69((3)):379–88. doi: 10.33549/physiolres.934492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Zhou H, Chen X, Hu T, Li J, Song H, Liu Y, et al. A novel bat coronavirus closely related to SARS-CoV-2 contains natural insertions at the S1/S2 cleavage site of the spike protein. Curr Biol. 2020;30((11)):2196–203.e3. doi: 10.1016/j.cub.2020.05.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Zheng J. SARS-CoV-2: an emerging coronavirus that causes a global threat. Int J Biol Sci. 2020;16((10)):1678–85. doi: 10.7150/ijbs.45053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Lam TT, Jia N, Zhang YW, Shum MH, Jiang JF, Zhu HC, et al. Identifying SARS-CoV-2-related coronaviruses in Malayan pangolins. Nature. 2020;583((7815)):282–5. doi: 10.1038/s41586-020-2169-0. [DOI] [PubMed] [Google Scholar]
- 11.Johansen MD, Irving A, Montagutelli X, Tate MD, Rudloff I, Nold MF, et al. Animal and translational models of SARS-CoV-2 infection and COVID-19. Mucosal Immunol. 2020;13((6)):877–91. doi: 10.1038/s41385-020-00340-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mallapaty S. COVID mink analysis shows mutations are not dangerous: yet. Nature. 2020;587((7834)):340–1. doi: 10.1038/d41586-020-03218-z. [DOI] [PubMed] [Google Scholar]
- 13.Patterson EI, Elia G, Grassi A, Giordano A, Desario C, Medardo M, et al. Evidence of exposure to SARS-CoV-2 in cats and dogs from households in Italy. Nat Commun. 2020;11((1)):6231. doi: 10.1038/s41467-020-20097-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Haagmans BL, van den Brand JM, Raj VS, Volz A, Wohlsein P, Smits SL, et al. An orthopoxvirus-based vaccine reduces virus excretion after MERS-CoV infection in dromedary camels. Science. 2016;351((6268)):77–81. doi: 10.1126/science.aad1283. [DOI] [PubMed] [Google Scholar]
- 15.Petrosillo N, Viceconte G, Ergonul O, Ippolito G, Petersen E. COVID-19, SARS and MERS: are they closely related? Clin Microbiol Infect. 2020;26((6)):729–34. doi: 10.1016/j.cmi.2020.03.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Daoust JF. Elderly people and responses to COVID-19 in 27 Countries. PLoS One. 2020;15((7)):e0235590. doi: 10.1371/journal.pone.0235590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ko JY, Danielson ML, Town M, Derado G, Greenlund KJ, Daily Kirley P, et al. Risk factors for COVID-19-associated hospitalization: COVID-19-associated hospitalization surveillance network and behavioral risk factor surveillance system. Clin Infect Dis. 2020 Sep 18;:ciaa1419. doi: 10.1093/cid/ciaa1419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Fajnzylber J, Regan J, Coxen K, Corry H, Wong C, Rosenthal A, et al. SARS-CoV-2 viral load is associated with increased disease severity and mortality. Nat Commun. 2020;11((1)):5493. doi: 10.1038/s41467-020-19057-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Gao Z, Xu Y, Sun C, Wang X, Guo Y, Qiu S, et al. A systematic review of asymptomatic infections with COVID-19. J Microbiol Immunol Infect. 2020 doi: 10.1016/j.jmii.2020.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.He J, Ni Y, Liao L, Xiao Y, Guo Y. Proportion of asymptomatic coronavirus disease 2019: a systematic review and meta-analysis. J Med Virol. 2021 Feb;93((2)):820–30. doi: 10.1002/jmv.26326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Song Y, Zhang M, Yin L, Wang K, Zhou Y, Zhou M, et al. COVID-19 treatment: close to a cure? A rapid review of pharmacotherapies for the novel coronavirus (SARS-CoV-2) Int J Antimicrob Agents. 2020;56((2)):106080. doi: 10.1016/j.ijantimicag.2020.106080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.National Library of Medicine at the U.S. National Institutes of Health ClinicalTrials. Available from: www.clinicaltrials.gov.
- 23.Gasparyan AY, Ayvazyan L, Blackmore H, Kitas GD. Writing a narrative biomedical review: considerations for authors, peer reviewers, and editors. Rheumatol Int. 2011;31((11)):1409–17. doi: 10.1007/s00296-011-1999-3. [DOI] [PubMed] [Google Scholar]
- 24.Catanzaro M, Fagiani F, Racchi M, Corsini E, Govoni S, Lanni C. Immune response in COVID-19: addressing a pharmacological challenge by targeting pathways triggered by SARS-CoV-2. Signal Transduct Target Ther. 2020;5((1)):84. doi: 10.1038/s41392-020-0191-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Rizzo P, Vieceli Dalla Sega F, Fortini F, Marracino L, Rapezzi C, Ferrari R. COVID-19 in the heart and the lungs: could we “Notch” the inflammatory storm? Basic Res Cardiol. 2020;115((3)):31. doi: 10.1007/s00395-020-0791-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mason RJ. Pathogenesis of COVID-19 from a cell biology perspective. Eur Respir J. 2020;55((4)):2000607. doi: 10.1183/13993003.00607-2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Lou B, Li TD, Zheng SF, Su YY, Li ZY, Liu W, et al. Serology characteristics of SARS-CoV-2 infection after exposure and post-symptom onset. Eur Respir J. 2020;56((2)):2000763. doi: 10.1183/13993003.00763-2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Padoan A, Sciacovelli L, Basso D, Negrini D, Zuin S, Cosma C, et al. IgA-Ab response to spike glycoprotein of SARS-CoV-2 in patients with COVID-19: a longitudinal study. Clin Chim Acta. 2020;507:164–6. doi: 10.1016/j.cca.2020.04.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Tai W, He L, Zhang X, Pu J, Voronin D, Jiang S, et al. Characterization of the receptor-binding domain (RBD) of 2019 novel coronavirus: implication for development of RBD protein as a viral attachment inhibitor and vaccine. Cell Mol Immunol. 2020;17((6)):613–20. doi: 10.1038/s41423-020-0400-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ni L, Ye F, Cheng M-L, Feng Y, Deng Y-Q, Zhao H, et al. Detection of SARS-CoV-2-specific humoral and cellular immunity in COVID-19 convalescent individuals. Immunity. 2020;52((6)):971–7. doi: 10.1016/j.immuni.2020.04.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Grifoni A, Weiskopf D, Ramirez SI, Mateus J, Dan JM, Moderbacher CR, et al. Targets of T cell responses to SARS-CoV-2 coronavirus in humans with COVID-19 disease and unexposed individuals. Cell. 2020;181((7)):1489–1501.e15. doi: 10.1016/j.cell.2020.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Sekine T, Perez-Potti A, Rivera-Ballesteros O, Strålin K, Gorin JB, Olsson A, et al. Robust T cell immunity in convalescent individuals with asymptomatic or mild COVID-19. Cell. 2020;183((1)):158–68.e14. doi: 10.1016/j.cell.2020.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Jeyanathan M, Afkhami S, Smaill F, Miller MS, Lichty BD, Xing Z. Immunological considerations for COVID-19 vaccine strategies. Nat Rev Immunol. 2020;20((10)):615–32. doi: 10.1038/s41577-020-00434-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Poland GA, Ovsyannikova IG, Kennedy RB. SARS-CoV-2 immunity: review and applications to phase 3 vaccine candidates. Lancet. 2020 Nov 14;396((10262)):1595–606. doi: 10.1016/S0140-6736(20)32137-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Plotkin S. History of vaccination. Proc Natl Acad Sci U S A. 2014;111((34)):12283–7. doi: 10.1073/pnas.1400472111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Delrue I, Verzele D, Madder A, Nauwynck HJ. Inactivated virus vaccines from chemistry to prophylaxis: merits, risks and challenges. Expert Rev Vaccines. 2012;11((6)):695–719. doi: 10.1586/erv.12.38. [DOI] [PubMed] [Google Scholar]
- 37.Zepp F. Principles of vaccine design-lessons from nature. Vaccine. 2010;28((Suppl 3)):C14–24. doi: 10.1016/j.vaccine.2010.07.020. [DOI] [PubMed] [Google Scholar]
- 38.Vartak A, Sucheck SJ. Recent advances in subunit vaccine carriers. Vaccines. 2016 Apr 16;4((2)):12. doi: 10.3390/vaccines4020012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Bonam SR, Partidos CD, Halmuthur SKM, Muller S. An overview of novel adjuvants designed for improving vaccine efficacy. Trends Pharmacol Sci. 2017;38((9)):771–93. doi: 10.1016/j.tips.2017.06.002. [DOI] [PubMed] [Google Scholar]
- 40.Di Pasquale A, Preiss S, Tavares Da Silva F, Garçon N. Vaccine adjuvants: from 1920 to 2015 and beyond. Vaccines. 2015;3((2)):320–43. doi: 10.3390/vaccines3020320. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Honda-Okubo Y, Barnard D, Ong CH, Peng BH, Tseng CT, Petrovsky N. Severe acute respiratory syndrome-associated coronavirus vaccines formulated with delta inulin adjuvants provide enhanced protection while ameliorating lung eosinophilic immunopathology. J Virol. 2015;89((6)):2995–3007. doi: 10.1128/JVI.02980-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Nascimento IP, Leite LC. Recombinant vaccines and the development of new vaccine strategies. Braz J Med Biol Res. 2012;45((12)):1102–11. doi: 10.1590/S0100-879X2012007500142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Tiwari S, Verma PC, Singh PK, Tuli R. Plants as bioreactors for the production of vaccine antigens. Biotechnol Adv. 2009;27((4)):449–67. doi: 10.1016/j.biotechadv.2009.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Syomin BV, Ilyin YV. Virus-like particles as an instrument of vaccine production. Mol Biol. 2019;53((3)):323–34. doi: 10.1134/S0026893319030154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Hobernik D, Bros M. DNA vaccines-how far from clinical use? Int J Mol Sci. 2018;19((11)):3605. doi: 10.3390/ijms19113605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Donnelly JJ, Wahren B, Liu MA. DNA vaccines: progress and challenges. J Immunol. 2005;175((2)):633–9. doi: 10.4049/jimmunol.175.2.633. [DOI] [PubMed] [Google Scholar]
- 47.Li L, Petrovsky N. Molecular mechanisms for enhanced DNA vaccine immunogenicity. Expert Rev Vaccines. 2016;15((3)):313–29. doi: 10.1586/14760584.2016.1124762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Schalk JA, Mooi FR, Berbers GA, van Aerts LA, Ovelgönne H, Kimman TG. Preclinical and clinical safety studies on DNA vaccines. Hum Vaccin. 2006;2((2)):45–53. doi: 10.4161/hv.2.2.2620. [DOI] [PubMed] [Google Scholar]
- 49.Ross J. mRNA stability in mammalian cells. Microbiol Rev. 1995;59((3)):423–50. doi: 10.1128/mr.59.3.423-450.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zhang C, Maruggi G, Shan H, Li J. Advances in mRNA vaccines for infectious diseases. Front Immunol. 2019;10:594. doi: 10.3389/fimmu.2019.00594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Ablasser A, Poeck H, Anz D, Berger M, Schlee M, Kim S, et al. Selection of molecular structure and delivery of RNA oligonucleotides to activate TLR7 versus TLR8 and to induce high amounts of IL-12p70 in primary human monocytes. J Immunol. 2009;182((11)):6824–33. doi: 10.4049/jimmunol.0803001. [DOI] [PubMed] [Google Scholar]
- 52.Hua Z, Hou B. TLR signaling in B-cell development and activation. Cell Mol Immunol. 2013;10((2)):103–6. doi: 10.1038/cmi.2012.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kato H, Oh SW, Fujita T. RIG-I-like receptors and type I interferonopathies. J Interferon Cytokine Res. 2017;37((5)):207–13. doi: 10.1089/jir.2016.0095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Pollard C, Rejman J, De Haes W, Verrier B, Van Gulck E, Naessens T, et al. Type I IFN counteracts the induction of antigen-specific immune responses by lipid-based delivery of mRNA vaccines. Mol Ther. 2013;21((1)):251–9. doi: 10.1038/mt.2012.202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Vogel AB, Lambert L, Kinnear E, Busse D, Erbar S, Reuter KC, et al. Self-amplifying RNA vaccines give equivalent protection against influenza to mRNA vaccines but at much lower doses. Mol Ther. 2018;26((2)):446–55. doi: 10.1016/j.ymthe.2017.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Pardi N, Hogan MJ, Porter FW, Weissman D. mRNA vaccines: a new era in vaccinology. Nat Rev Drug Discov. 2018;17((4)):261–79. doi: 10.1038/nrd.2017.243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Ramezanpour B, Haan I, Osterhaus A, Claassen E. Vector-based genetically modified vaccines: exploiting Jenner's legacy. Vaccine. 2016;34((50)):6436–48. doi: 10.1016/j.vaccine.2016.06.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Fausther-Bovendo H, Kobinger GP. Pre-existing immunity against Ad vectors: humoral, cellular, and innate response, what's important? Hum Vaccin Immunother. 2014;10((10)):2875–84. doi: 10.4161/hv.29594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Humphreys IR, Sebastian S. Novel viral vectors in infectious diseases. Immunology. 2018;153((1)):1–9. doi: 10.1111/imm.12829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Rauch S, Jasny E, Schmidt KE, Petsch B. New vaccine technologies to combat outbreak situations. Front Immunol. 2018;9:1963. doi: 10.3389/fimmu.2018.01963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Zhang L, Wang W, Wang S. Effect of vaccine administration modality on immunogenicity and efficacy. Expert Rev Vaccines. 2015;14((11)):1509–23. doi: 10.1586/14760584.2015.1081067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Schnyder JL, De Pijper CA, Garcia Garrido HM, Daams JG, Goorhuis A, Stijnis C, et al. Fractional dose of intradermal compared to intramuscular and subcutaneous vaccination: a systematic review and meta-analysis. Travel Med Infect Dis. 2020 Sep− Oct;37:101868. doi: 10.1016/j.tmaid.2020.101868. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Li M, Wang Y, Sun Y, Cui H, Zhu SJ, Qiu HJ. Mucosal vaccines: strategies and challenges. Immunol Lett. 2020;217:116–25. doi: 10.1016/j.imlet.2019.10.013. [DOI] [PubMed] [Google Scholar]
- 64.Folegatti PM, Ewer KJ, Aley PK, Angus B, Becker S, Belij-Rammerstorfer S, et al. Safety and immunogenicity of the ChAdOx1 nCoV-19 vaccine against SARS-CoV-2: a preliminary report of a phase 1/2, single-blind, randomised controlled trial. Lancet. 2020;396((10249)):467–78. doi: 10.1016/S0140-6736(20)31604-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Voysey M, Clemens SAC, Madhi SA, Weckx LW, Folegatti PM, Aley PK, et al. Safety and efficacy of the ChAdOx1 nCoV-19 vaccine (AZD1222) against SARS-CoV-2: an interim analysis of four randomised controlled trials in Brazil, South Africa, and the UK. Lancet. 2020 Dec 8;397((10269)):99–111. doi: 10.1016/S0140-6736(20)32661-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Jackson LA, Anderson EJ, Rouphael NG, Roberts PC, Makhene M, Coler RN, et al. An mRNA vaccine against SARS-CoV-2: preliminary report. N Engl J Med. 2020;383((20)):1920–31. doi: 10.1056/NEJMoa2022483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Polack FP, Thomas SK, Kitchin N, Absalon J, Gurtman A, Lockhart S, et al. Safety and efficacy of the BNT162b2 mRNA Covid-19 vaccine. N Engl J Med. 2020 Dec 31;383((27)):2603–15. doi: 10.1056/NEJMoa2034577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Escobar LE, Molina-Cruz A, Barillas-Mury C. BCG vaccine protection from severe coronavirus disease 2019 (COVID-19) Proc Natl Acad Sci U S A. 2020;117((30)):17720–6. doi: 10.1073/pnas.2008410117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kamat S, Kumari M. BCG against SARS-CoV-2: second youth of an old age vaccine? Front Pharmacol. 2020;11:1050. doi: 10.3389/fphar.2020.01050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Moorlag SJCFM, van Deuren RC, van Werkhoven CH, Jaeger M, Debisarun P, Taks E, et al. Safety and COVID-19 symptoms in individuals recently vaccinated with BCG: a retrospective cohort study. Cell Rep Med. 2020;1((5)):100073. doi: 10.1016/j.xcrm.2020.100073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Rajarshi K, Chatterjee A, Ray S. BCG vaccination strategy implemented to reduce the impact of COVID-19: hype or hope? Med Drug Discov. 2020;7:100049. doi: 10.1016/j.medidd.2020.100049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Sharma A, Kumar Sharma S, Shi Y, Bucci E, Carafoli E, Melino G, et al. BCG vaccination policy and preventive chloroquine usage: do they have an impact on COVID-19 pandemic? Cell Death Dis. 2020;11((7)):516. doi: 10.1038/s41419-020-2720-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Anbarasu A, Ramaiah S, Livingstone P. Vaccine repurposing approach for preventing COVID 19: can MMR vaccines reduce morbidity and mortality? Hum Vaccin Immunother. 2020;16((9)):2217–8. doi: 10.1080/21645515.2020.1773141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ashford JW, Gold JE, Huenergardt MJA, Katz RBA, Strand SE, Bolanos J, et al. MMR vaccination: a potential strategy to reduce severity and mortality of COVID-19 Illness. Am J Med. 2020 Oct 23;S0002–9343((20)):30902–5. doi: 10.1016/j.amjmed.2020.10.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Aaby P, Roth A, Ravn H, Napirna BM, Rodrigues A, Lisse IM, et al. Randomized trial of BCG vaccination at birth to low-birth-weight children: beneficial nonspecific effects in the neonatal period? J Infect Dis. 2011;204((2)):245–52. doi: 10.1093/infdis/jir240. [DOI] [PubMed] [Google Scholar]
- 76.Moorlag SJCFM, Arts RJW, van Crevel R, Netea MG. Non-specific effects of BCG vaccine on viral infections. Clin Microbiol Infect. 2019;25((12)):1473–8. doi: 10.1016/j.cmi.2019.04.020. [DOI] [PubMed] [Google Scholar]
- 77.Rieckmann A, Villumsen M, Sørup S, Haugaard LK, Ravn H, Roth A, et al. Vaccinations against smallpox and tuberculosis are associated with better long-term survival: a Danish case-cohort study 1971–2010. Int J Epidemiol. 2017;46((2)):695–705. doi: 10.1093/ije/dyw120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Sorup S, Benn CS, Poulsen A, Krause TG, Aaby P, Ravn H. Live vaccine against measles, mumps, and rubella and the risk of hospital admissions for nontargeted infections. JAMA. 2014;311((8)):826–35. doi: 10.1001/jama.2014.470. [DOI] [PubMed] [Google Scholar]
- 79.Abdelrahman Z, Li M, Wang X. Comparative review of SARS-CoV-2, SARS-CoV, MERS-CoV, and influenza A respiratory viruses. Front Immunol. 2020;11:552909. doi: 10.3389/fimmu.2020.552909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Lewnard JA, Cobey S. Immune history and influenza vaccine effectiveness. Vaccines. 2018 May 21;6((2)):28. doi: 10.3390/vaccines6020028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Rosendahl Huber SK, Hendriks M, Jacobi RHJ, van de Kassteele J, Mandersloot-Oskam JC, van Boxtel RAJ, et al. Immunogenicity of influenza vaccines: evidence for differential effect of secondary vaccination on humoral and cellular immunity. Front Immunol. 2018;9:3103. doi: 10.3389/fimmu.2018.03103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Christenson B, Böttiger M. Measles antibody: comparison of long-term vaccination titres, early vaccination titres and naturally acquired immunity to and booster effects on the measles virus. Vaccine. 1994;12((2)):129–33. doi: 10.1016/0264-410x(94)90049-3. [DOI] [PubMed] [Google Scholar]
- 83.Kongsgaard M, Bassi MR, Rasmussen M, Skjødt K, Thybo S, Gabriel M, et al. Adaptive immune responses to booster vaccination against yellow fever virus are much reduced compared to those after primary vaccination. Sci Rep. 2017;7((1)):662. doi: 10.1038/s41598-017-00798-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Hanley KA. The double-edged sword: how evolution can make or break a live-attenuated virus vaccine. Evolution. 2011;4((4)):635–43. doi: 10.1007/s12052-011-0365-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Lauring AS, Jones JO, Andino R. Rationalizing the development of live attenuated virus vaccines. Nat Biotechnol. 2010;28((6)):573–9. doi: 10.1038/nbt.1635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Minor PD. Live attenuated vaccines: historical successes and current challenges. Virology. 2015;479–480:379–92. doi: 10.1016/j.virol.2015.03.032. [DOI] [PubMed] [Google Scholar]
- 87.Mok DZL, Chan KR. The effects of pre-existing antibodies on live-attenuated viral vaccines. Viruses. 2020 May 8;12((5)):520. doi: 10.3390/v12050520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Amanna IJ, Slifka MK. Wanted, dead or alive: new viral vaccines. Antiviral Res. 2009;84((2)):119–30. doi: 10.1016/j.antiviral.2009.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Delgado MF, Coviello S, Monsalvo AC, Melendi GA, Hernandez JZ, Batalle JP, et al. Lack of antibody affinity maturation due to poor Toll-like receptor stimulation leads to enhanced respiratory syncytial virus disease. Nat Med. 2009;15((1)):34–41. doi: 10.1038/nm.1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.U.S. Food and Drug Administration. Accessed 2020 Dec 11. Available from: https://www.fda.gov/emergency-preparedness-and-response/coronavirus-disease-2019-covid-19/covid-19-vaccines.
- 91.European Medicines Agency. Accessed 2020 Nov 6. Available from: https://www.ema.europa.eu/en/human-regulatory/overview/public-health-threats/coronavirus-disease-covid-19/treatments-vaccines-covid-19.
- 92.Milstien J, Costa A, Jadhav S, Dhere R. Reaching international GMP standards for vaccine production: challenges for developing countries. Expert Rev Vaccines. 2009;8((5)):559–66. doi: 10.1586/erv.09.23. [DOI] [PubMed] [Google Scholar]
- 93.Bonilla FA. Update: vaccines in primary immunodeficiency. J Allergy Clin Immunol. 2018;141((2)):474–81. doi: 10.1016/j.jaci.2017.12.980. [DOI] [PubMed] [Google Scholar]
- 94.Furer V, Rondaan C, Heijstek MW, Agmon-Levin N, van Assen S, Bijl M, et al. 2019 update of EULAR recommendations for vaccination in adult patients with autoimmune inflammatory rheumatic diseases. Ann Rheum Dis. 2020;79((1)):39–52. doi: 10.1136/annrheumdis-2019-215882. [DOI] [PubMed] [Google Scholar]
- 95.Friedmann D, Goldacker S, Peter H-H, Warnatz K. Preserved cellular immunity upon influenza vaccination in most patients with common variable immunodeficiency. J Allergy Clin Immunol Pract. 2020;8((7)):2332–40. doi: 10.1016/j.jaip.2020.04.019. [DOI] [PubMed] [Google Scholar]
- 96.Mieves JF, Wittke K, Freitag H, Volk HD, Scheibenbogen C, Hanitsch LG. Influenza vaccination in patients with common variable immunodeficiency (CVID) Curr Allergy Asthma Rep. 2017;17((11)):78. doi: 10.1007/s11882-017-0749-3. [DOI] [PubMed] [Google Scholar]
- 97.Florindo HF, Kleiner R, Vaskovich-Koubi D, Acúrcio RC, Carreira B, Yeini E, et al. Immune-mediated approaches against COVID-19. Nat Nanotechnol. 2020;15((8)):630–45. doi: 10.1038/s41565-020-0732-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Chams N, Chams S, Badran R, Shams A, Araji A, Raad M, et al. COVID-19: a multidisciplinary review. Front Public Health. 2020;8:383. doi: 10.3389/fpubh.2020.00383. [DOI] [PMC free article] [PubMed] [Google Scholar]

