Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3295 2023-09-28 17:57:43 |
2 format Meta information modification 3295 2023-10-07 04:37:28 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Lee, B.; Nanishi, E.; Levy, O.; Dowling, D.J. Precision Vaccinology Approaches for Adjuvanted Vaccines. Encyclopedia. Available online: https://encyclopedia.pub/entry/49780 (accessed on 29 April 2024).
Lee B, Nanishi E, Levy O, Dowling DJ. Precision Vaccinology Approaches for Adjuvanted Vaccines. Encyclopedia. Available at: https://encyclopedia.pub/entry/49780. Accessed April 29, 2024.
Lee, Branden, Etsuro Nanishi, Ofer Levy, David J. Dowling. "Precision Vaccinology Approaches for Adjuvanted Vaccines" Encyclopedia, https://encyclopedia.pub/entry/49780 (accessed April 29, 2024).
Lee, B., Nanishi, E., Levy, O., & Dowling, D.J. (2023, September 28). Precision Vaccinology Approaches for Adjuvanted Vaccines. In Encyclopedia. https://encyclopedia.pub/entry/49780
Lee, Branden, et al. "Precision Vaccinology Approaches for Adjuvanted Vaccines." Encyclopedia. Web. 28 September, 2023.
Precision Vaccinology Approaches for Adjuvanted Vaccines
Edit

Infection persists as one of the leading global causes of morbidity and mortality, with particular burden at the extremes of age and in populations who are immunocompromised or suffer chronic co-morbid diseases. By focusing discovery and innovation efforts to better understand the phenotypic and mechanistic differences in the immune systems of diverse vulnerable populations, emerging research in precision vaccine discovery and development has explored how to optimize immunizations across the lifespan. 

adjuvant systems immune potentiators immunomodulators toll-like receptor (TLR) agonists

1. Introduction

For several decades beginning in the 1920s, insoluble aluminum salts were the first and only adjuvants used to enhance vaccine efficacy against infectious diseases [1]. Since that period, several additional adjuvants have been included in approved vaccines, from the oil-in-water (OIW) emulsion MF59 to the GSK adjuvant systems (AS). These developments signify a nearly-100-year timeline of interconnected developments in the fields of vaccinology and immunology [1][2].
Recently, the notable widespread employment of mRNA, viral vector, and adjuvanted vaccine technologies have had a profound global impact on reducing the impact of the COVID-19 pandemic. An estimated 10+ million deaths were prevented globally due to SARS-CoV-2 vaccinations, with a significant contribution attributed to the speed of pathogen-specific mRNA vaccine development [3]. However, while this innovation signifies a breakthrough in vaccinology and modern medicine, it also highlights an important challenge in the field. Different age groups presented with distinct responses to the mRNA vaccines. In particular, the mRNA vaccines demonstrate waning effectiveness, diminished protection, and heterogeneous efficacy, a phenomenon that is not restricted to but is especially pronounced in older adults [4][5][6][7]. This is likely a result of several factors, which include the variability in the magnitude and persistence of antigen expression and subsequent clearance from the encoded mRNA [8][9][10]. Ultimately, these heterogeneous responses invite further research into the biodistribution and pharmacometrics of this novel vaccine platform as gene therapies begin to be integrated into vaccinology. Nevertheless, a diminished protection for older adults is also seen in both non-mRNA vaccines against SARS-CoV-2 [11] and seasonal influenza vaccines [12][13]. A number of factors appear to be driving this differential response, from age-specific response to adjuvants [14][15], immune senescence, to potential diminishing immunization efficacy over time as a result of immune imprinting [16][17][18]. Considering these factors to inform rationally designed vaccines that effectively protect immunocompromised individuals and/or older adults remains an active area for improvement in current vaccine development streams.
A related paradigm also exists for immunizing younger populations. As the immune system undergoes dramatic changes in the early years of life, significant phenotypic differences emerge between pediatric and adult immune responses [19][20][21]. These differences manifest in a higher pediatric susceptibility and vulnerability to certain infectious diseases [22][23][24]. In fact, infectious diseases are amongst the leading global causes of pediatric mortality [25], which is at least partially due to their distinct immune responses. Further, the more distinct immunological phenotypes of young children as well the impact of transplacental maternal immunity contribute to significant differences in vaccine response in early life [26][27][28]. Thus, an important objective of modern vaccinology is defining safe approaches to shape immune responses in early life [29][30][31][32].
Similarly, individuals with weakened immune systems, such as those living with metabolic disorders and diabetes, immunocompromised populations, and people living with chronic viral infection and/or cancer, experience heightened susceptibility to infectious diseases [33] and suboptimal responses to immunizations [34][35]. While immunocompromised status represents a large and heterogenous population with diverse phenotypes and severity, there is an unmet need to enhance vaccine efficacy for this diverse group [36]. Defining immune pathways to trigger protective immunity in these populations, including optimal adjuvants, may be key to protection of these groups.

2. Precision Vaccinology Principles in Adjuvant Innovation, Discovery, and Development

2.1. Emerging Tools for Adjuvant Discovery

High-throughput screening (HTS) of small molecules has been leveraged by pharmaceutical and academic centers to expand scientific toolbox. HTS has contributed to ~20–30% of late-stage clinical development candidates at several prominent pharmaceutical companies [37]. However, the same analyses suggest that ~60% of early-stage candidates discovered via HTS fail to reach later-stage applications. Several approaches have been employed to overcome this modest yield, including (1) developing more curated and druggable chemical libraries to optimize the source of testable drug candidates [38] and, (2) optimizing the screening platform itself—finding more applicable or translational in vitro systems to screen the chemical libraries. From engineered constructs for target-based screening to broad phenotypic screens for functional discovery, there has been a significant expansion in available tools to screen compounds. Some notable developments include the application of organ-on-chip technologies as screening tools and incorporation of microfluidic screening to enable ultra-high-throughput screening [39][40]. Extending to vaccine adjuvant discovery, innovation in phenotypic and targeted screens have identified multiple novel adjuvants [41][42][43].
The expanded menu of adjuvant discovery tools can advance precision vaccinology. In regard to the growth in target-based screening technologies, targeted screens can be steered towards addressing gaps in vaccine efficacy for distinct vulnerable populations. For example, infant populations often skew towards Th2-like immune responses and fail to mount sufficient Th1 responses [44][45]. This Th2-skewed response leaves pediatric populations at higher susceptibility and vulnerability to many serious complications arising from intracellular pathogens, such as viral respiratory infections [46][47]. Of note, activation of endosomal TLRs, such as TLR7 and/or -8, can shape the infant immune response towards a more adult-like Th1 response [48][49], which can enable effective protection against many respiratory viruses. Thus, with more advanced targeting screening tools for these endosomal targets, the arsenal of precision adjuvants available to vaccinate this vulnerable population can substantially advance.
The expansion of phenotypic screening tools can enhance translation to advance precision-adjuvant discovery. With human in vitro models being developed for HTS, it is now possible to screen for molecules with optimal activity towards distinct populations [50]. Indeed, human primary immune cells have been directly used to screen for adjuvant discovery [51]. Using human primary cells from individuals from distinct populations (e.g., by age, sex, and/or co-morbidity), may enable discovery and development of bespoke adjuvants for vulnerable populations.

2.2. Innovations through Advanced Adjuvant Compositions

One of the largest bottlenecks in advancing novel lead candidates from discovery to clinical consideration is optimizing the function of the compounds via medicinal chemistry. Altering compound structure can improve on-target efficacy of compounds in addition to their pharmacodynamic and pharmacokinetic properties [52][53][54]. These types of medicinal chemistry approaches can correspondingly be applied to develop more ideal adjuvant compositions [55][56]. Evolving objectives for these optimized compounds include improving potency, efficacy, safety, and biological clearance. Additionally, an emerging focus of adjuvant medicinal chemistry involves ligating chemical bridges onto novel adjuvants to optimize integration into antigen formulation [57][58]. Thus, characterizing the active moiety of a molecule and adding bridges, such as phosphonate groups for alum absorption, that do not interfere with this moiety can be a powerful tool for translation/application. This impact is heightened with emerging drug-delivery technologies that enable targeting of and integration for specific tissues and even cells [59][60][61]. Thus, the inherent functionality of novel small-molecule adjuvants can be optimized and they can be accommodated into sophisticated delivery technologies.
In addition to improving the functionality of compounds, another important focus of medicinal chemistry effort for novel adjuvants is to remove any off-target effects, sometimes referred to as “wasted immunity”. Many discovered leads are compounds with pleiotropic activities. This can be the basis of drug-repurposing efforts that have come to define the growing field of polypharmacology. More often, however, this pleiotropic nature, which is notably linked to larger and unrefined biologics [62][63][64], accompanies adverse side-effects and safety concerns that contribute to the high attrition rates seen in the drug-development pipeline [65]. Indeed, several early- and late-stage vaccine adjuvant candidates, such as water in oil/squalene or poly(I:C), demonstrate potential adverse reactogenicity [66]. Potential reactogenicity can be addressed by reengineering biologic or chemically altering small molecule candidates to yield more precise functionalities [67][68]. Indeed, a precision medicine approach is key for discovery and development of safe and effective vaccines for distinct populations that vary in susceptibility to vaccine-associated adverse events [68].

3. Exploration of Immunization Sites to Unleash Maximal Effects in Target Populations

While significant research has been focused on expanding the tools available to formulate a vaccine, equally important is defining the immunogenicity needed to achieve protection [69]. To this end, all licensed vaccines have targeted routes of administration that maintain a localized and controlled antigen presentation, thereby avoiding the potential dangers of a systemic vaccine and the added potential benefit that some vaccines provide via depot effects [70][71][72]. Depending on route of administration, vaccines can unleash unique benefits reflecting distinct immunity mounted by different target tissues.

3.1. Intradermal, Subcutaneuous, Intramuscular—A Layered Lesson in Precision Vaccinology

The layers of tissue from skin to muscle provide several targets for immunization. Considering the different immune cell compositions found at each tissue layer, there are characteristic differences in immunity induced between intradermal (ID), subcutaneous (SC), and intramuscular (IM) vaccination [73].
ID injections enable preferential access to dermal dendritic cells (DCs) and Langerhans cells (LCs). Targeting these dermal sentinels facilitates highly-effective antigen presentation to skin resident and lymph-node-resident memory T cells via migration to lymphoid organs [74][75]. Furthermore, with >20 times the myeloid DCs and double the T cells spanning the human dermis compared to circulating blood [76], ID injections are an attractive approach to optimize vaccine immunogenicity. In fact, at an equivalent antigen dose, ID vaccination often induces greater immunogenicity than IM or SC vaccination [77]. Further, LCs are maintained as a steady-state cellular population, rendering them a highly promising target for precision vaccines for both older and younger populations [78][79]. However, the drawback of this route of administration is the inherent difficulty of physically injecting at the proper depth, further compounded by the heterogeneity of the dermal thickness across age groups, body types, and demographics [80][81][82][83]. As innovations move the field from the canonical needle-and-syringe approach to standardizable novel systems, such as dermal patches or laser-guided injection systems that can precisely deliver vaccine components, ID immunizations become an even more appealing option in precision vaccinology [84][85][86].
SC vaccinations, while historically considered a safe and efficacious route of administration, have been questioned of late. Concerns about safety and poor mobilization and ultimate presentation of antigen have highlighted some shortcomings of this route [87][88][89]. Further, new lines of research indicate that other routes may be more efficacious. Using the recent monkeypox public health emergency as a case-study, the Vaccinia Ankara vaccine was deployed for vulnerable populations. Historically, this vaccine was given with a two-dose regimen via SC immunization. However, it was retooled as a single dose ID immunization at 1/5th of the antigen dose and still maintained comparable protection [90][91][92][93].
IM injections remain the most common way to deliver vaccines. With a lower density of pain fibers compared to ID or SC layers and ample blood flow, IM is an ideal candidate for many types of formulations [94][95]. While the IM route may not proffer the same density of immune cells that ID layers do, the resiliency of muscle may enable a higher threshold of tolerance for a range of vaccine formulations.

3.2. Nose to Gut—Mucosal Immunity

While the IM, SC, and ID routes of vaccine delivery are effective for inducing systemic immunity to prevent severe illness, they may be suboptimal in preventing infection at the site of invasion. These considerations highlight a significant area for improvement in current vaccine development efforts, which has become growingly visible due to the ongoing COVID-19 pandemic: mucosal immunity [69][96][97]. Mucosal layers cover much of respiratory, gastroenterological, and genital tracts and serve as a barrier against respiratory, enteric, and genital infections, respectively. Establishing mucosal vaccination approaches that can potentiate protection at this layer and can prevent infection from occurring in the first place is a key area for research and translation. From a public-health framework, developing a mucosal vaccine can be a future tool to better curb transmission of highly virulent strains and even eradicate the presence of malignant viruses. Pertinently, this is an even higher priority precision-vaccine objective, when routinely protecting vulnerable populations via this approach is still mostly technocritical.
There have been many modern advancements in mucosal vaccines, in both oral- and nasal-delivery systems. There have been several mucosal vaccine candidates with exciting preclinical results [98][99] and even some promising indications in clinical trials [100][101][102]. However, most of the candidates have mixed to moderate levels of protection, with observed variability across different age groups [103][104][105]. As a result, there are less than 10 licensed enteric vaccines and less than five licensed nasal vaccines, which includes two recently approved intranasal candidates against SARS-CoV-2 [106][107].
One of the major challenges in developing mucosal vaccines, which may explain the limited number of approved candidates, is ensuring a sufficiently immunogenic and durable response. Considering that the mucosal layer constitutes unique subsets of immune sentinels and a distinct mechanism of protection [108], growing research is focused on establishing important targets to stimulate in this context. Novel adjuvanted approaches have been described that may be unique or optimal to mucosal immunizations and understand the correlates of protection and mucosal triggers, the efficacy of these vaccines will substantially improve [109]. Similarly, vaccine immunogenicity, especially that of mucosal vaccines, is effected by microbiome in complex ways [110][111][112]. Characterizing how the microbiome impacts vaccine immunogenicity, may inform precision vaccination approaches. Overall, developing effective mucosal adjuvants may enable overcoming generally poor immunogenicity and heterogeneity of mucosal vaccines across different age groups.
However, adjuvanted approaches for mucosal vaccines, especially via a nasal route, need to be accompanied with a particular emphasis on safety. Intranasal influenza vaccine with Escherichia coli heat-labile toxin as a mucosal adjuvant has been associated with higher risk of developing Bell’s palsy [113]. While the association may be specific to this adjuvant, not with the specific route of administration, with the evidence that Bell’s palsy is also associated with parental SARS-CoV-2 mRNA vaccine administration [114], it is of heightened concern in intranasal contexts, considering its proximity to cranial nerves and the larger central nervous system.

3.3. Crossing the Aisle—Heterologous Vaccines

While steering the immune response towards either mucosal or systemic immunity draws upon distinct mechanisms, the two can be combined via heterologous vaccination such as via intramuscular (IM) prime for systemic protection and intranasal (IN) pull for mucosal protection. For example, an IM prime with an mRNA-LNP vaccine followed by an IN pull with unadjuvanted spike protein generated robust mucosal and systemic immunogenicity [115]. In contrast, IM + IM or IN + IN schemes lacked effective coverage in at least one of the forms of immunity. As a proof of concept, this shows the immense potential in converging these two streams for polyfunctional immunization schemes. Similarly, while IM + IN schemes have been the key focus in heterologous schemes [116][117], future research should explore combining other routes of administration (e.g., intradermal, subcutaneous, and enteric). Considering how some of these other routes may provide more robust and/or durable protection in certain antigen/adjuvant combinations, other combinations may prove more effective.
From a precision medicine perspective, these heterologous schemes can be ideal for immunizing vulnerable populations by inducing both mucosal and systemic protection. One possible limitation of mucosal vaccines is the partly compartmentalization of the mucosal immune response from the larger systemic response [118], as the distribution of the responses depends on the actual route of induction. This separation of immune apparatuses leads to gaps in potential protection, which can be further unleased via heterologous schemes. Future studies should investigate novel heterologous combinations, including the role of adjuvantation to optimize immunogenicity and protection.

4. Putting It Together—Formulating Vaccine Compositions

4.1. Back to Basics: Alum, Polymers, and Lipids

A key aspect of vaccine development is matching an appropriate antigen and adjuvant combination with a formulation for optimal delivery and efficacy. Interestingly, the most common historical vaccine platforms (alum, polymerized formulations, and oil-in-water combinations) can be inherently immunogenic, while also enhancing the delivery efficiency of the vaccines [119][120][121]. This translates to a significant enhancement in vaccine efficacy and durability via established models that can be coupled with other novel adjuvants to boost responses, generating combination adjuvant platforms such as GSK’s adjuvantation systems [122][123].
The framework of matching antigen, adjuvant and formulation can be directly combined with precision medicine approaches to create novel combinations that enhance responses from targeted populations. As disruptive technologies and innovations such as artificial intelligence and multifactorial immune-monitoring systems elucidate insights into systemic immune responses [124][125][126], this information can help guide precision and rational vaccine design [127]. In fact, many established adjuvant-delivery systems have already been combined with novel candidates to overcome hypo-responsiveness from both early- and later-life populations [128][129][130]. These efforts are developing in parallel to existing efforts to modulate the chemo–physical properties of these canonical platforms for improved immunogenicity [120][131]. Further, the functionality of certain adjuvants is only unleashed when directly combined with appropriate immune enhancers, antigens, and/or other adjuvants. Thus, rational combinations of novel with existing systems can help integrate novel adjuvant discoveries, thereby accelerating vaccine development for vulnerable populations.

4.2. Strength in Numbers: Multimeric Antigenic Systems for Precision Vaccines

One of the key innovations in recent vaccine research includes advances in structure-based antigen design which can enhance immunogenicity of otherwise weak immunogens, such as fusion glycoprotein of respiratory syncytial virus [132][133][134]. Structure-based antigen optimization has been widely applied for vaccines against SARS-CoV-2, most commonly by stabilizing spike protein in trimeric prefusion conformation to improve immunogenicity [133][135]. More recently, high-density, multimeric antigens displayed onto protein or synthetic nanoparticles have demonstrated the advantage of enhancing antigen trafficking to draining lymph nodes and promoting clustering and activation of the B cell receptor, thus strengthening the magnitude and breadth of immunogenicity [136][137][138]. This is of particular importance as waning immunity is a significant concern, especially for younger children and older adults [6][139]. Thus, developing novel multimeric antigenic vaccines can be a powerful strategy to enhance and elongate protection for many vulnerable populations.
While structure-based antigen optimization is a promising approach to enhance vaccine efficacy especially in vulnerable populations, vaccines comprised of optimized antigen alone are generally insufficiently immunogenic. Clinical and preclinical studies which employed a self-assembling, two-component SARS-CoV-2 receptor-binding protein nanoparticle antigen demonstrated limited immunogenicity in the non-adjuvanted group while adjuvants such as AS03 (GSK) conferred robust protection [140][141]. Although it is possible to generalize desirable properties for an adjuvant, a proper match between an adjuvant and a specific antigen must be empirically evaluated. Evaluation of candidate antigen–adjuvant combination in pre-clinical models, such as huma in vitro and animal models, that take into account population (e.g., age, sex, and co-morbidity)-dependent vaccine immunogenicity may enable down-selecting and prioritizing adjuvanted RBD antigen-based vaccines [142].

4.3. Adjuvantation via mRNA Delivery Systems—Welcoming LNPs into the Club

While mRNA vaccines do not contain an adjuvant per se, they appear to be self-adjuvated. Indeed their lipid nanoparticle (LNP) delivery system not only aids in stability and delivery of the vaccine, but also stimulates innate immune responses that boost the immunogenicity of the encoded mRNA [143][144][145][146]. However, in addition to the LNP adjuvant, additional adjuvants can be utilized in mRNA-LNP vaccines to boost vaccine efficacy. From canonical adjuvants, such as cGAMP, to manganese nanoparticles, the stimulator of interferon genes (STING) pathway has been a key target to boost the innate immune response of an mRNA-LNP vaccine [147][148][149]. Further, more advanced LNP platforms containing encoding key immunological agents such as IL-12 have been used to enhance onco-immunology treatments [150][151]. These developments can be applied to enhance vaccines against infectious disease in the near future. Further, both biologic and small-molecule adjuvants can be tethered to mRNA-LNP platforms without loss of function of antigen or adjuvant recognition [152]. Thus, traditional adjuvant strategies can be powerfully combined with novel mRNA-LNP technologies to magnify quantity and quality of immune responses and extend durability of protection for vulnerable populations. Thus, as the novel mRNA-LNP technology becomes more broad ly applied to immunize against different pathogens, further adjuvantation is a practical possi bility to enhance these vaccines for populations that were less responsive to the original vaccine composition. For key components of LNPs, such as ionic lipids and pegylated polymers, rational design, including, as may be needed, optimization of these vaccine components, not just for their antigen delivery capacity but also for their inherently self-adjuvanting nature, can inform future vaccine design [123][153].

References

  1. Garçon, N.; Di Pasquale, A. From discovery to licensure, the Adjuvant System story. Hum. Vaccines Immunother. 2017, 13, 19–33.
  2. Pulendran, B.; Arunachalam, P.S.; O’Hagan, D.T. Emerging concepts in the science of vaccine adjuvants. Nat. Rev. Drug Discov. 2021, 20, 454–475.
  3. Watson, O.J.; Barnsley, G.; Toor, J.; Hogan, A.B.; Winskill, P.; Ghani, A.C. Global impact of the first year of COVID-19 vaccination: A mathematical modelling study. Lancet Infect. Dis. 2022, 22, 1293–1302.
  4. Collier, D.A.; Ferreira, I.A.T.M.; Kotagiri, P.; Datir, R.P.; Lim, E.Y.; Touizer, E.; Meng, B.; Abdullahi, A.; Baker, S.; Dougan, G.; et al. Age-related immune response heterogeneity to SARS-CoV-2 vaccine BNT162b2. Nature 2021, 596, 417–422.
  5. Palacios-Pedrero, M.Á.; Jansen, J.M.; Blume, C.; Stanislawski, N.; Jonczyk, R.; Molle, A.; Hernandez, M.G.; Kaiser, F.K.; Jung, K.; Osterhaus, A.D.M.E.; et al. Signs of immunosenescence correlate with poor outcome of mRNA COVID-19 vaccination in older adults. Nat. Aging 2022, 2, 896–905.
  6. Nanishi, E.; Levy, O.; Ozonoff, A. Waning effectiveness of SARS-CoV-2 mRNA vaccines in older adults: A rapid review. Hum. Vaccines Immunother. 2022, 18, 2045857.
  7. Nanishi, E.; McGrath, M.E.; O’Meara, T.R.; Barman, S.; Yu, J.; Wan, H.; Dillen, C.A.; Menon, M.; Seo, H.S.; Song, K.; et al. mRNA booster vaccination protects aged mice against the SARS-CoV-2 Omicron variant. Commun. Biol. 2022, 5, 790.
  8. Sahin, U.; Karikó, K.; Türeci, Ö. mRNA-based therapeutics—Developing a new class of drugs. Nat. Rev. Drug Discov. 2014, 13, 759–780.
  9. Fertig, T.E.; Chitoiu, L.; Marta, D.S.; Ionescu, V.S.; Cismasiu, V.B.; Radu, E.; Angheluta, G.; Dobre, M.; Serbanescu, A.; Hinescu, M.E.; et al. Vaccine mRNA Can Be Detected in Blood at 15 Days Post-Vaccination. Biomedicines 2022, 10, 1538.
  10. Hirota, M.; Tamai, M.; Yukawa, S.; Taira, N.; Matthews, M.M.; Toma, T.; Seto, Y.; Yoshida, M.; Toguchi, S.; Miyagi, M.; et al. Human immune and gut microbial parameters associated with inter-individual variations in COVID-19 mRNA vaccine-induced immunity. Commun. Biol. 2023, 6, 368.
  11. Liang, C.-K.; Lee, W.-J.; Peng, L.-N.; Meng, L.-C.; Hsiao, F.-Y.; Chen, L.-K. COVID-19 Vaccines in Older Adults: Challenges in Vaccine Development and Policy Making. Clin. Geriatr. Med. 2022, 38, 605–620.
  12. Goodwin, K.; Viboud, C.; Simonsen, L. Antibody response to influenza vaccination in the elderly: A quantitative review. Vaccine 2006, 24, 1159–1169.
  13. Osterholm, M.T.; Kelley, N.S.; Sommer, A.; Belongia, E.A. Efficacy and effectiveness of influenza vaccines: A systematic review and meta-analysis. Lancet Infect. Dis. 2012, 12, 36–44.
  14. Nanishi, E.; Angelidou, A.; Rotman, C.; Dowling, D.J.; Levy, O.; Ozonoff, A. Precision Vaccine Adjuvants for Older Adults: A Scoping Review. Clin. Infect. Dis. 2022, 75, S72–S80.
  15. Barman, S.; Soni, D.; Brook, B.; Nanishi, E.; Dowling, D.J. Precision Vaccine Development: Cues From Natural Immunity. Front. Immunol. 2021, 12, 662218.
  16. Haynes, L. Aging of the Immune System: Research Challenges to Enhance the Health Span of Older Adults. Front. Aging 2020, 1, 602108.
  17. Panda, A.; Qian, F.; Mohanty, S.; van Duin, D.; Newman, F.K.; Zhang, L.; Chen, S.; Towle, V.; Belshe, R.B.; Fikrig, E.; et al. Age-associated decrease in TLR function in primary human dendritic cells predicts influenza vaccine response. J. Immunol. 2010, 184, 2518–2527.
  18. Wheatley, A.K.; Fox, A.; Tan, H.X.; Juno, J.A.; Davenport, M.P.; Subbarao, K.; Kent, S.J. Immune imprinting and SARS-CoV-2 vaccine design. Trends Immunol. 2021, 42, 956–959.
  19. Simon, A.K.; Hollander, G.A.; McMichael, A. Evolution of the immune system in humans from infancy to old age. Proc. Biol. Sci. 2015, 282, 20143085.
  20. Basha, S.; Surendran, N.; Pichichero, M. Immune responses in neonates. Expert Rev. Clin. Immunol. 2014, 10, 1171–1184.
  21. Kollmann, T.R.; Crabtree, J.; Rein-Weston, A.; Blimkie, D.; Thommai, F.; Wang, X.Y.; Lavoie, P.M.; Furlong, J.; Fortuno, E.S., 3rd; Hajjar, A.M.; et al. Neonatal innate TLR-mediated responses are distinct from those of adults. J. Immunol. 2009, 183, 7150–7160.
  22. Weinberg, G.A. Respiratory syncytial virus mortality among young children. Lancet Glob. Health 2017, 5, e951–e952.
  23. Hwang, J.K.; Na, J.Y.; Kim, J.; Oh, J.-W.; Kim, Y.J.; Choi, Y.-J. Age-Specific Characteristics of Adult and Pediatric Respiratory Viral Infections: A Retrospective Single-Center Study. J. Clin. Med. 2022, 11, 3197.
  24. Ikuta, K.S.; Swetschinski, L.R.; Robles Aguilar, G.; Sharara, F.; Mestrovic, T.; Gray, A.P.; Davis Weaver, N.; Wool, E.E.; Han, C.; Gershberg Hayoon, A.; et al. Global mortality associated with 33 bacterial pathogens in 2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 2022, 400, 2221–2248.
  25. Liu, L.; Oza, S.; Hogan, D.; Chu, Y.; Perin, J.; Zhu, J.; Lawn, J.E.; Cousens, S.; Mathers, C.; Black, R.E. Global, regional, and national causes of under-5 mortality in 2000-15: An updated systematic analysis with implications for the Sustainable Development Goals. Lancet 2016, 388, 3027–3035.
  26. Cadoz, M. Potential and limitations of polysaccharide vaccines in infancy. Vaccine 1998, 16, 1391–1395.
  27. Barrett, D.J. Human immune responses to polysaccharide antigens: An analysis of bacterial polysaccharide vaccines in infants. Adv. Pediatr. 1985, 32, 139–158.
  28. Niewiesk, S. Maternal antibodies: Clinical significance, mechanism of interference with immune responses, and possible vaccination strategies. Front. Immunol. 2014, 5, 446.
  29. Dowling, D.J.; Levy, O. Pediatric Vaccine Adjuvants: Components of the Modern Vaccinologist’s Toolbox. Pediatr. Infect. Dis. J. 2015, 34, 1395–1398.
  30. Levy, O.; Netea, M.G. Innate immune memory: Implications for development of pediatric immunomodulatory agents and adjuvanted vaccines. Pediatr. Res. 2014, 75, 184–188.
  31. Soni, D.; Bobbala, S.; Li, S.; Scott, E.A.; Dowling, D.J. The sixth revolution in pediatric vaccinology: Immunoengineering and delivery systems. Pediatr. Res. 2021, 89, 1364–1372.
  32. Semmes, E.C.; Chen, J.L.; Goswami, R.; Burt, T.D.; Permar, S.R.; Fouda, G.G. Understanding Early-Life Adaptive Immunity to Guide Interventions for Pediatric Health. Front. Immunol. 2020, 11, 595297.
  33. Englund, J.; Feuchtinger, T.; Ljungman, P. Viral infections in immunocompromised patients. Biol. Blood Marrow Transpl. 2011, 17, S2–S5.
  34. Ceravolo, A.; Orsi, A.; Parodi, V.; Ansaldi, F. Influenza vaccination in HIV-positive subjects: Latest evidence and future perspective. J. Prev. Med. Hyg. 2013, 54, 1–10.
  35. Ferreira, V.H.; Solera, J.T.; Hu, Q.; Hall, V.G.; Arbol, B.G.; Rod Hardy, W.; Samson, R.; Marinelli, T.; Ierullo, M.; Virk, A.K.; et al. Homotypic and heterotypic immune responses to Omicron variant in immunocompromised patients in diverse clinical settings. Nat. Commun. 2022, 13, 4489.
  36. Di Fusco, M.; Lin, J.; Vaghela, S.; Lingohr-Smith, M.; Nguyen, J.L.; Scassellati Sforzolini, T.; Judy, J.; Cane, A.; Moran, M.M. COVID-19 vaccine effectiveness among immunocompromised populations: A targeted literature review of real-world studies. Expert Rev. Vaccines 2022, 21, 435–451.
  37. Macarron, R.; Banks, M.N.; Bojanic, D.; Burns, D.J.; Cirovic, D.A.; Garyantes, T.; Green, D.V.S.; Hertzberg, R.P.; Janzen, W.P.; Paslay, J.W.; et al. Impact of high-throughput screening in biomedical research. Nat. Rev. Drug Discov. 2011, 10, 188–195.
  38. Fox, S.; Farr-Jones, S.; Sopchak, L.; Boggs, A.; Nicely, H.W.; Khoury, R.; Biros, M. High-throughput screening: Update on practices and success. J. Biomol. Screen. 2006, 11, 864–869.
  39. Autour, A.; Ryckelynck, M. Ultrahigh-Throughput Improvement and Discovery of Enzymes Using Droplet-Based Microfluidic Screening. Micromachines 2017, 8, 128.
  40. Azizgolshani, H.; Coppeta, J.R.; Vedula, E.M.; Marr, E.E.; Cain, B.P.; Luu, R.J.; Lech, M.P.; Kann, S.H.; Mulhern, T.J.; Tandon, V.; et al. High-throughput organ-on-chip platform with integrated programmable fluid flow and real-time sensing for complex tissue models in drug development workflows. Lab Chip 2021, 21, 1454–1474.
  41. Salyer, A.C.; Caruso, G.; Khetani, K.K.; Fox, L.M.; Malladi, S.S.; David, S.A. Identification of Adjuvantic Activity of Amphotericin B in a Novel, Multiplexed, Poly-TLR/NLR High-Throughput Screen. PLoS ONE 2016, 11, e0149848.
  42. Guan, Y.; Omueti-Ayoade, K.; Mutha, S.K.; Hergenrother, P.J.; Tapping, R.I. Identification of novel synthetic toll-like receptor 2 agonists by high throughput screening. J. Biol. Chem. 2010, 285, 23755–23762.
  43. Martínez-Gil, L.; Ayllon, J.; Ortigoza, M.B.; García-Sastre, A.; Shaw, M.L.; Palese, P. Identification of Small Molecules with Type I Interferon Inducing Properties by High-Throughput Screening. PLoS ONE 2012, 7, e49049.
  44. Zaghouani, H.; Hoeman, C.M.; Adkins, B. Neonatal immunity: Faulty T-helpers and the shortcomings of dendritic cells. Trends Immunol. 2009, 30, 585–591.
  45. Barrios, C.; Brawand, P.; Berney, M.; Brandt, C.; Lambert, P.H.; Siegrist, C.A. Neonatal and early life immune responses to various forms of vaccine antigens qualitatively differ from adult responses: Predominance of a Th2-biased pattern which persists after adult boosting. Eur. J. Immunol. 1996, 26, 1489–1496.
  46. Becker, Y. Respiratory syncytial virus (RSV) evades the human adaptive immune system by skewing the Th1/Th2 cytokine balance toward increased levels of Th2 cytokines and IgE, markers of allergy—A review. Virus Genes 2006, 33, 235–252.
  47. Pinto, R.A.; Arredondo, S.M.; Bono, M.R.; Gaggero, A.A.; Díaz, P.V. T helper 1/T helper 2 cytokine imbalance in respiratory syncytial virus infection is associated with increased endogenous plasma cortisol. Pediatrics 2006, 117, e878–e886.
  48. Kim, S.-Y.; Heo, M.B.; Hwang, G.-S.; Jung, Y.; Choi, D.Y.; Park, Y.-M.; Lim, Y.T. Multivalent Polymer Nanocomplex Targeting Endosomal Receptor of Immune Cells for Enhanced Antitumor and Systemic Memory Response. Angew. Chem. Int. Ed. 2015, 54, 8139–8143.
  49. van Haren, S.D.; Dowling, D.J.; Foppen, W.; Christensen, D.; Andersen, P.; Reed, S.G.; Hershberg, R.M.; Baden, L.R.; Levy, O. Age-Specific Adjuvant Synergy: Dual TLR7/8 and Mincle Activation of Human Newborn Dendritic Cells Enables Th1 Polarization. J. Immunol. 2016, 197, 4413–4424.
  50. Geng, H.; Whiteley, G.; Ribbens, J.; Zheng, W.; Southall, N.; Hu, X.; Marugan, J.J.; Ferrer, M.; Maegawa, G.H.B. Novel Patient Cell-Based HTS Assay for Identification of Small Molecules for a Lysosomal Storage Disease. PLoS ONE 2011, 6, e29504.
  51. Chew, K.; Lee, B.; van Haren, S.D.; Nanishi, E.; O’Meara, T.; Splaine, J.B.; DeLeon, M.; Soni, D.; Seo, H.S.; Dhe-Paganon, S.; et al. Adjuvant Discovery via a High Throughput Screen using Human Primary Mononuclear Cells. bioRxiv 2022.
  52. Dorr, P.; Westby, M.; Dobbs, S.; Griffin, P.; Irvine, B.; Macartney, M.; Mori, J.; Rickett, G.; Smith-Burchnell, C.; Napier, C.; et al. Maraviroc (UK-427,857), a potent, orally bioavailable, and selective small-molecule inhibitor of chemokine receptor CCR5 with broad-spectrum anti-human immunodeficiency virus type 1 activity. Antimicrob. Agents Chemother. 2005, 49, 4721–4732.
  53. Gabrielsson, J.; Fjellström, O.; Ulander, J.; Rowley, M.; Van Der Graaf, P.H. Pharmacodynamic-pharmacokinetic integration as a guide to medicinal chemistry. Curr. Top. Med. Chem. 2011, 11, 404–418.
  54. Awale, M.; Hert, J.; Guasch, L.; Riniker, S.; Kramer, C. The Playbooks of Medicinal Chemistry Design Moves. J. Chem. Inf. Model. 2021, 61, 729–742.
  55. Wang, P.; Ding, X.; Kim, H.; Škalamera, Đ.; Michalek, S.M.; Zhang, P. Vaccine Adjuvants Derivatized from Momordica Saponins I and II. J. Med. Chem. 2019, 62, 9976–9982.
  56. Ryter, K.T.; Ettenger, G.; Rasheed, O.K.; Buhl, C.; Child, R.; Miller, S.M.; Holley, D.; Smith, A.J.; Evans, J.T. Aryl Trehalose Derivatives as Vaccine Adjuvants for Mycobacterium tuberculosis. J. Med. Chem. 2020, 63, 309–320.
  57. Wilson, D.S.; Hirosue, S.; Raczy, M.M.; Bonilla-Ramirez, L.; Jeanbart, L.; Wang, R.; Kwissa, M.; Franetich, J.F.; Broggi, M.A.S.; Diaceri, G.; et al. Antigens reversibly conjugated to a polymeric glyco-adjuvant induce protective humoral and cellular immunity. Nat. Mater. 2019, 18, 175–185.
  58. Cortez, A.; Li, Y.; Miller, A.T.; Zhang, X.; Yue, K.; Maginnis, J.; Hampton, J.; Hall de, S.; Shapiro, M.; Nayak, B.; et al. Incorporation of Phosphonate into Benzonaphthyridine Toll-like Receptor 7 Agonists for Adsorption to Aluminum Hydroxide. J. Med. Chem. 2016, 59, 5868–5878.
  59. Hartwell, B.L.; Melo, M.B.; Xiao, P.; Lemnios, A.A.; Li, N.; Chang, J.Y.H.; Yu, J.; Gebre, M.S.; Chang, A.; Maiorino, L.; et al. Intranasal vaccination with lipid-conjugated immunogens promotes antigen transmucosal uptake to drive mucosal and systemic immunity. Sci. Transl. Med. 2022, 14, eabn1413.
  60. Bern, M.; Nilsen, J.; Ferrarese, M.; Sand, K.M.K.; Gjølberg, T.T.; Lode, H.E.; Davidson, R.J.; Camire, R.M.; Bækkevold, E.S.; Foss, S.; et al. An engineered human albumin enhances half-life and transmucosal delivery when fused to protein-based biologics. Sci. Transl. Med. 2020, 12, eabb0580.
  61. Shields, C.W.t.; Evans, M.A.; Wang, L.L.; Baugh, N.; Iyer, S.; Wu, D.; Zhao, Z.; Pusuluri, A.; Ukidve, A.; Pan, D.C.; et al. Cellular backpacks for macrophage immunotherapy. Sci. Adv. 2020, 6, eaaz6579.
  62. Leeson, P.D.; Springthorpe, B. The influence of drug-like concepts on decision-making in medicinal chemistry. Nat. Rev. Drug Discov. 2007, 6, 881–890.
  63. Wenlock, M.C.; Austin, R.P.; Barton, P.; Davis, A.M.; Leeson, P.D. A Comparison of Physiochemical Property Profiles of Development and Marketed Oral Drugs. J. Med. Chem. 2003, 46, 1250–1256.
  64. Robinson, R.A.; DeVita, V.T.; Levy, H.B.; Baron, S.; Hubbard, S.P.; Levine, A.S. A Phase I–II Trial of Multiple-Dose Polyriboinosinic-Polyribocytidylic Acid in Patients with Leukemia or Solid Tumors. JNCI J. Natl. Cancer Inst. 1976, 57, 599–602.
  65. Waring, M.J.; Arrowsmith, J.; Leach, A.R.; Leeson, P.D.; Mandrell, S.; Owen, R.M.; Pairaudeau, G.; Pennie, W.D.; Pickett, S.D.; Wang, J.; et al. An analysis of the attrition of drug candidates from four major pharmaceutical companies. Nat. Rev. Drug Discov. 2015, 14, 475–486.
  66. Kuroda, Y.; Nacionales, D.C.; Akaogi, J.; Reeves, W.H.; Satoh, M. Autoimmunity induced by adjuvant hydrocarbon oil components of vaccine. Biomed. Pharmacother. 2004, 58, 325–337.
  67. Flower, D.R. Systematic identification of small molecule adjuvants. Expert Opin. Drug Discov. 2012, 7, 807–817.
  68. Sathish, J.G.; Sethu, S.; Bielsky, M.-C.; de Haan, L.; French, N.S.; Govindappa, K.; Green, J.; Griffiths, C.E.M.; Holgate, S.; Jones, D.; et al. Challenges and approaches for the development of safer immunomodulatory biologics. Nat. Rev. Drug Discov. 2013, 12, 306–324.
  69. Knisely, J.M.; Buyon, L.E.; Mandt, R.; Farkas, R.; Balasingam, S.; Bok, K.; Buchholz, U.J.; D’Souza, M.P.; Gordon, J.L.; King, D.F.L.; et al. Mucosal vaccines for SARS-CoV-2: Scientific gaps and opportunities-workshop report. NPJ Vaccines 2023, 8, 53.
  70. Li, C.; Chen, Y.; Zhao, Y.; Lung, D.C.; Ye, Z.; Song, W.; Liu, F.-F.; Cai, J.-P.; Wong, W.-M.; Yip, C.C.-Y.; et al. Intravenous Injection of Coronavirus Disease 2019 (COVID-19) mRNA Vaccine Can Induce Acute Myopericarditis in Mouse Model. Clin. Infect. Dis. 2021, 74, 1933–1950.
  71. Tam, H.H.; Melo, M.B.; Kang, M.; Pelet, J.M.; Ruda, V.M.; Foley, M.H.; Hu, J.K.; Kumari, S.; Crampton, J.; Baldeon, A.D.; et al. Sustained antigen availability during germinal center initiation enhances antibody responses to vaccination. Proc. Natl. Acad. Sci. USA 2016, 113, E6639–E6648.
  72. Pedersen, G.K.; Wørzner, K.; Andersen, P.; Christensen, D. Vaccine Adjuvants Differentially Affect Kinetics of Antibody and Germinal Center Responses. Front. Immunol. 2020, 11, 579761.
  73. Rosenbaum, P.; Tchitchek, N.; Joly, C.; Rodriguez Pozo, A.; Stimmer, L.; Langlois, S.; Hocini, H.; Gosse, L.; Pejoski, D.; Cosma, A.; et al. Vaccine Inoculation Route Modulates Early Immunity and Consequently Antigen-Specific Immune Response. Front. Immunol. 2021, 12, 645210.
  74. Romani, N.; Flacher, V.; Tripp, C.H.; Sparber, F.; Ebner, S.; Stoitzner, P. Targeting skin dendritic cells to improve intradermal vaccination. Curr. Top. Microbiol. Immunol. 2012, 351, 113–138.
  75. Clayton, K.; Vallejo, A.F.; Davies, J.; Sirvent, S.; Polak, M.E. Langerhans Cells-Programmed by the Epidermis. Front. Immunol. 2017, 8, 1676.
  76. Wang, X.N.; McGovern, N.; Gunawan, M.; Richardson, C.; Windebank, M.; Siah, T.W.; Lim, H.Y.; Fink, K.; Yao Li, J.L.; Ng, L.G.; et al. A three-dimensional atlas of human dermal leukocytes, lymphatics, and blood vessels. J. Investig. Dermatol. 2014, 134, 965–974.
  77. Schnyder, J.L.; Garcia Garrido, H.M.; De Pijper, C.A.; Daams, J.G.; Stijnis, C.; Goorhuis, A.; Grobusch, M.P. Comparison of equivalent fractional vaccine doses delivered by intradermal and intramuscular or subcutaneous routes: A systematic review. Travel Med. Infect. Dis. 2021, 41, 102007.
  78. Merad, M.; Ginhoux, F.; Collin, M. Origin, homeostasis and function of Langerhans cells and other langerin-expressing dendritic cells. Nat. Rev. Immunol. 2008, 8, 935–947.
  79. Drijkoningen, M.; De Wolf-Peeters, C.; Van Der Steen, K.; Moerman, P.; Desmet, V. Epidermal Langerhans’ Cells and Dermal Dendritic Cells in Human Fetal and Neonatal Skin: An Immunohistochemical Study. Pediatr. Dermatol. 1987, 4, 11–17.
  80. Groswasser, J.; Kahn, A.; Bouche, B.; Hanquinet, S.; Perlmuter, N.; Hessel, L. Needle length and injection technique for efficient intramuscular vaccine delivery in infants and children evaluated through an ultrasonographic determination of subcutaneous and muscle layer thickness. Pediatrics 1997, 100, 400–403.
  81. Shaw, F.E., Jr.; Guess, H.A.; Roets, J.M.; Mohr, F.E.; Coleman, P.J.; Mandel, E.J.; Roehm, R.R., Jr.; Talley, W.S.; Hadler, S.C. Effect of anatomic injection site, age and smoking on the immune response to hepatitis B vaccination. Vaccine 1989, 7, 425–430.
  82. Poland, G.A.; Borrud, A.; Jacobson, R.M.; McDermott, K.; Wollan, P.C.; Brakke, D.; Charboneau, J.W. Determination of deltoid fat pad thickness. Implications for needle length in adult immunization. JAMA 1997, 277, 1709–1711.
  83. Hettinga, J.; Carlisle, R. Vaccination into the Dermal Compartment: Techniques, Challenges, and Prospects. Vaccines 2020, 8, 534.
  84. Caudill, C.; Perry, J.L.; Iliadis, K.; Tessema, A.T.; Lee, B.J.; Mecham, B.S.; Tian, S.; DeSimone, J.M. Transdermal vaccination via 3D-printed microneedles induces potent humoral and cellular immunity. Proc. Natl. Acad. Sci. USA 2021, 118, e2102595118.
  85. Hogan, N.C.; Anahtar, M.N.; Taberner, A.J.; Hunter, I.W. Delivery of immunoreactive antigen using a controllable needle-free jet injector. J. Control. Release 2017, 258, 73–80.
  86. Menon, I.; Kang, S.M.; Braz Gomes, K.; Uddin, M.N.; D’Souza, M. Laser-assisted intradermal delivery of a microparticle vaccine for respiratory syncytial virus induces a robust immune response. Vaccine 2023, 41, 1209–1222.
  87. Cook, I.F. Subcutaneous vaccine administration—An outmoded practice. Hum. Vaccines Immunother. 2021, 17, 1329–1341.
  88. Haramati, N.; Lorans, R.; Lutwin, M.; Kaleya, R.N. Injection granulomas. Intramuscle or intrafat? Arch. Fam. Med. 1994, 3, 146–148.
  89. Zuckerman, J.N. The importance of injecting vaccines into muscle. Different patients need different needle sizes. BMJ 2000, 321, 1237–1238.
  90. Hazra, A.; Rusie, L.; Hedberg, T.; Schneider, J.A. Human Monkeypox Virus Infection in the Immediate Period after Receiving Modified Vaccinia Ankara Vaccine. JAMA 2022, 328, 2064–2067.
  91. Brooks, J.T.; Marks, P.; Goldstein, R.H.; Walensky, R.P. Intradermal Vaccination for Monkeypox—Benefits for Individual and Public Health. N. Engl. J. Med. 2022, 387, 1151–1153.
  92. Wolff Sagy, Y.; Zucker, R.; Hammerman, A.; Markovits, H.; Arieh, N.G.; Abu Ahmad, W.; Battat, E.; Ramot, N.; Carmeli, G.; Mark-Amir, A.; et al. Real-world effectiveness of a single dose of mpox vaccine in males. Nat. Med. 2023, 29, 748–752.
  93. del Rio, C.; Malani, P.N. Update on the Monkeypox Outbreak. JAMA 2022, 328, 921–922.
  94. Greenblatt, D.J.; Allen, M.D. Intramuscular injection-site complications. JAMA 1978, 240, 542–544.
  95. Michaels, L.; Poole, R.W. Injection granuloma of the buttock. Can. Med. Assoc. J. 1970, 102, 626–628.
  96. Russell, M.W.; Mestecky, J. Mucosal immunity: The missing link in comprehending SARS-CoV-2 infection and transmission. Front. Immunol. 2022, 13, 957107.
  97. Morens, D.M.; Taubenberger, J.K.; Fauci, A.S. Rethinking next-generation vaccines for coronaviruses, influenzaviruses, and other respiratory viruses. Cell Host Microbe 2023, 31, 146–157.
  98. Horvath, D.; Temperton, N.; Mayora-Neto, M.; Da Costa, K.; Cantoni, D.; Horlacher, R.; Günther, A.; Brosig, A.; Morath, J.; Jakobs, B.; et al. Novel intranasal vaccine targeting SARS-CoV-2 receptor binding domain to mucosal microfold cells and adjuvanted with TLR3 agonist Riboxxim™ elicits strong antibody and T-cell responses in mice. Sci. Rep. 2023, 13, 4648.
  99. Le Nouën, C.; Nelson, C.E.; Liu, X.; Park, H.-S.; Matsuoka, Y.; Luongo, C.; Santos, C.; Yang, L.; Herbert, R.; Castens, A.; et al. Intranasal pediatric parainfluenza virus-vectored SARS-CoV-2 vaccine is protective in monkeys. Cell 2022, 185, 4811–4825.e17.
  100. Kim, L.; Liebowitz, D.; Lin, K.; Kasparek, K.; Pasetti, M.F.; Garg, S.J.; Gottlieb, K.; Trager, G.; Tucker, S.N. Safety and immunogenicity of an oral tablet norovirus vaccine, a phase I randomized, placebo-controlled trial. JCI Insight 2018, 3, e121077.
  101. Johnson, S.; Martinez, C.I.; Jegede, C.B.; Gutierrez, S.; Cortese, M.; Martinez, C.J.; Garg, S.J.; Peinovich, N.; Dora, E.G.; Tucker, S.N. SARS-CoV-2 oral tablet vaccination induces neutralizing mucosal IgA in a phase 1 open label trial. medRxiv 2022.
  102. Wu, S.; Huang, J.; Zhang, Z.; Wu, J.; Zhang, J.; Hu, H.; Zhu, T.; Zhang, J.; Luo, L.; Fan, P.; et al. Safety, tolerability, and immunogenicity of an aerosolised adenovirus type-5 vector-based COVID-19 vaccine (Ad5-nCoV) in adults: Preliminary report of an open-label and randomised phase 1 clinical trial. Lancet Infect. Dis. 2021, 21, 1654–1664.
  103. Madhavan, M.; Ritchie, A.J.; Aboagye, J.; Jenkin, D.; Provstgaad-Morys, S.; Tarbet, I.; Woods, D.; Davies, S.; Baker, M.; Platt, A.; et al. Tolerability and immunogenicity of an intranasally-administered adenovirus-vectored COVID-19 vaccine: An open-label partially-randomised ascending dose phase I trial. EBioMedicine 2022, 85, 104298.
  104. Belshe, R.B.; Edwards, K.M.; Vesikari, T.; Black, S.V.; Walker, R.E.; Hultquist, M.; Kemble, G.; Connor, E.M. Live Attenuated versus Inactivated Influenza Vaccine in Infants and Young Children. N. Engl. J. Med. 2007, 356, 685–696.
  105. Trombetta, C.M.; Gianchecchi, E.; Montomoli, E. Influenza vaccines: Evaluation of the safety profile. Hum. Vaccines Immunother. 2018, 14, 657–670.
  106. Waltz, E. China and India approve nasal COVID vaccines—Are they a game changer? Nature 2022, 609, 450.
  107. Vela Ramirez, J.E.; Sharpe, L.A.; Peppas, N.A. Current state and challenges in developing oral vaccines. Adv. Drug Deliv. Rev. 2017, 114, 116–131.
  108. McGhee, J.R.; Fujihashi, K. Inside the Mucosal Immune System. PLoS Biol. 2012, 10, e1001397.
  109. Alu, A.; Chen, L.; Lei, H.; Wei, Y.; Tian, X.; Wei, X. Intranasal COVID-19 vaccines: From bench to bed. EBioMedicine 2022, 76, 103841.
  110. Zimmermann, P. The immunological interplay between vaccination and the intestinal microbiota. NPJ Vaccines 2023, 8, 24.
  111. Gonçalves, J.I.B.; Borges, T.J.; de Souza, A.P.D. Microbiota and the Response to Vaccines against Respiratory Virus. Front. Immunol. 2022, 13, 889945.
  112. Thomson, C.A.; Morgan, S.C.; Ohland, C.; McCoy, K.D. From germ-free to wild: Modulating microbiome complexity to understand mucosal immunology. Mucosal Immunol. 2022, 15, 1085–1094.
  113. Mutsch, M.; Zhou, W.; Rhodes, P.; Bopp, M.; Chen, R.T.; Linder, T.; Spyr, C.; Steffen, R. Use of the inactivated intranasal influenza vaccine and the risk of Bell’s palsy in Switzerland. N. Engl. J. Med. 2004, 350, 896–903.
  114. Ozonoff, A.; Nanishi, E.; Levy, O. Bell’s palsy and SARS-CoV-2 vaccines. Lancet Infect. Dis. 2021, 21, 450–452.
  115. Mao, T.; Israelow, B.; Peña-Hernández, M.A.; Suberi, A.; Zhou, L.; Luyten, S.; Reschke, M.; Dong, H.; Homer, R.J.; Saltzman, W.M.; et al. Unadjuvanted intranasal spike vaccine elicits protective mucosal immunity against sarbecoviruses. Science 2022, 378, eabo2523.
  116. Wang, Q.; Yang, C.; Yin, L.; Sun, J.; Wang, W.; Li, H.; Zhang, Z.; Chen, S.; Liu, B.; Liu, Z.; et al. Intranasal booster using an Omicron vaccine confers broad mucosal and systemic immunity against SARS-CoV-2 variants. Signal Transduct. Target. Ther. 2023, 8, 167.
  117. Li, X.; Wang, L.; Liu, J.; Fang, E.; Liu, X.; Peng, Q.; Zhang, Z.; Li, M.; Liu, X.; Wu, X.; et al. Combining intramuscular and intranasal homologous prime-boost with a chimpanzee adenovirus-based COVID-19 vaccine elicits potent humoral and cellular immune responses in mice. Emerg. Microbes Infect. 2022, 11, 1890–1899.
  118. Holmgren, J.; Czerkinsky, C. Mucosal immunity and vaccines. Nat. Med. 2005, 11, S45–S53.
  119. Smith, A.; Perelman, M.; Hinchcliffe, M. Chitosan: A promising safe and immune-enhancing adjuvant for intranasal vaccines. Hum. Vaccines Immunother. 2014, 10, 797–807.
  120. Jansen, T.; Hofmans, M.P.; Theelen, M.J.; Schijns, V.E. Structure-activity relations of water-in-oil vaccine formulations and induced antigen-specific antibody responses. Vaccine 2005, 23, 1053–1060.
  121. Peleteiro, M.; Presas, E.; González-Aramundiz, J.V.; Sánchez-Correa, B.; Simón-Vázquez, R.; Csaba, N.; Alonso, M.J.; González-Fernández, Á. Polymeric Nanocapsules for Vaccine Delivery: Influence of the Polymeric Shell on the Interaction with the Immune System. Front. Immunol. 2018, 9, 791.
  122. Coccia, M.; Collignon, C.; Hervé, C.; Chalon, A.; Welsby, I.; Detienne, S.; van Helden, M.J.; Dutta, S.; Genito, C.J.; Waters, N.C.; et al. Cellular and molecular synergy in AS01-adjuvanted vaccines results in an early IFNγ response promoting vaccine immunogenicity. NPJ Vaccines 2017, 2, 25.
  123. Dowling, D.J.; Scott, E.A.; Scheid, A.; Bergelson, I.; Joshi, S.; Pietrasanta, C.; Brightman, S.; Sanchez-Schmitz, G.; Van Haren, S.D.; Ninković, J.; et al. Toll-like receptor 8 agonist nanoparticles mimic immunomodulating effects of the live BCG vaccine and enhance neonatal innate and adaptive immune responses. J. Allergy Clin. Immunol. 2017, 140, 1339–1350.
  124. Stafford, I.S.; Kellermann, M.; Mossotto, E.; Beattie, R.M.; MacArthur, B.D.; Ennis, S. A systematic review of the applications of artificial intelligence and machine learning in autoimmune diseases. NPJ Digit. Med. 2020, 3, 30.
  125. Pavlović, M.; Scheffer, L.; Motwani, K.; Kanduri, C.; Kompova, R.; Vazov, N.; Waagan, K.; Bernal, F.L.M.; Costa, A.A.; Corrie, B.; et al. The immuneML ecosystem for machine learning analysis of adaptive immune receptor repertoires. Nat. Mach. Intell. 2021, 3, 936–944.
  126. Brusic, V.; Gottardo, R.; Kleinstein, S.H.; Davis, M.M.; Davis, M.M.; Hafler, D.A.; Quill, H.; Palucka, A.K.; Poland, G.A.; Pulendran, B.; et al. Computational resources for high-dimensional immune analysis from the Human Immunology Project Consortium. Nat. Biotechnol. 2014, 32, 146–148.
  127. Bramwell, V.W.; Perrie, Y. The rational design of vaccines. Drug Discov. Today 2005, 10, 1527–1534.
  128. Dowling, D.J.; Barman, S.; Smith, A.J.; Borriello, F.; Chaney, D.; Brightman, S.E.; Melhem, G.; Brook, B.; Menon, M.; Soni, D.; et al. Development of a TLR7/8 agonist adjuvant formulation to overcome early life hyporesponsiveness to DTaP vaccination. Sci. Rep. 2022, 12, 16860.
  129. Nanishi, E.; Borriello, F.; O’Meara, T.R.; McGrath, M.E.; Saito, Y.; Haupt, R.E.; Seo, H.S.; van Haren, S.D.; Cavazzoni, C.B.; Brook, B.; et al. An aluminum hydroxide:CpG adjuvant enhances protection elicited by a SARS-CoV-2 receptor binding domain vaccine in aged mice. Sci. Transl. Med. 2022, 14, eabj5305.
  130. Yang, J.; Kim, J.; Kwak, C.; Poo, H. Poly-γ-glutamic acid/Alum adjuvanted pH1N1 vaccine-immunized aged mice exhibit a significant increase in vaccine efficacy with a decrease in age-associated CD8+ T cell proportion in splenocytes. Immun. Ageing 2022, 19, 22.
  131. Dmour, I.; Islam, N. Recent advances on chitosan as an adjuvant for vaccine delivery. Int. J. Biol. Macromol. 2022, 200, 498–519.
  132. Falsey, A.R.; Williams, K.; Gymnopoulou, E.; Bart, S.; Ervin, J.; Bastian, A.R.; Menten, J.; De Paepe, E.; Vandenberghe, S.; Chan, E.K.H.; et al. Efficacy and Safety of an Ad26.RSV.preF-RSV preF Protein Vaccine in Older Adults. N. Engl. J. Med. 2023, 388, 609–620.
  133. Hsieh, C.L.; Goldsmith, J.A.; Schaub, J.M.; DiVenere, A.M.; Kuo, H.C.; Javanmardi, K.; Le, K.C.; Wrapp, D.; Lee, A.G.; Liu, Y.; et al. Structure-based design of prefusion-stabilized SARS-CoV-2 spikes. Science 2020, 369, 1501–1505.
  134. Graham, B.S.; Gilman, M.S.A.; McLellan, J.S. Structure-Based Vaccine Antigen Design. Annu. Rev. Med. 2019, 70, 91–104.
  135. Corbett, K.S.; Edwards, D.K.; Leist, S.R.; Abiona, O.M.; Boyoglu-Barnum, S.; Gillespie, R.A.; Himansu, S.; Schäfer, A.; Ziwawo, C.T.; DiPiazza, A.T.; et al. SARS-CoV-2 mRNA vaccine design enabled by prototype pathogen preparedness. Nature 2020, 586, 567–571.
  136. Boyoglu-Barnum, S.; Ellis, D.; Gillespie, R.A.; Hutchinson, G.B.; Park, Y.J.; Moin, S.M.; Acton, O.J.; Ravichandran, R.; Murphy, M.; Pettie, D.; et al. Quadrivalent influenza nanoparticle vaccines induce broad protection. Nature 2021, 592, 623–628.
  137. Arunachalam, P.S.; Walls, A.C.; Golden, N.; Atyeo, C.; Fischinger, S.; Li, C.; Aye, P.; Navarro, M.J.; Lai, L.; Edara, V.V.; et al. Adjuvanting a subunit COVID-19 vaccine to induce protective immunity. Nature 2021, 594, 253–258.
  138. Walls, A.C.; Fiala, B.; Schäfer, A.; Wrenn, S.; Pham, M.N.; Murphy, M.; Tse, L.V.; Shehata, L.; O’Connor, M.A.; Chen, C.; et al. Elicitation of Potent Neutralizing Antibody Responses by Designed Protein Nanoparticle Vaccines for SARS-CoV-2. Cell 2020, 183, 1367–1382.e17.
  139. Stich, M.; Benning, L.; Speer, C.; Garbade, S.F.; Bartenschlager, M.; Kim, H.; Gleich, F.; Jeltsch, K.; Haase, B.; Janda, A.; et al. Waning Immunity 14 Months after SARS-CoV-2 Infection. Pediatrics 2022, 150, e2022057151.
  140. Song, J.Y.; Choi, W.S.; Heo, J.Y.; Lee, J.S.; Jung, D.S.; Kim, S.W.; Park, K.H.; Eom, J.S.; Jeong, S.J.; Lee, J.; et al. Safety and immunogenicity of a SARS-CoV-2 recombinant protein nanoparticle vaccine (GBP510) adjuvanted with AS03: A randomised, placebo-controlled, observer-blinded phase 1/2 trial. EClinicalMedicine 2022, 51, 101569.
  141. Nanishi, E.; Borriello, F.; Seo, H.S.; O’Meara, T.R.; McGrath, M.E.; Saito, Y.; Chen, J.; Diray-Arce, J.; Song, K.; Xu, A.Z.; et al. Carbohydrate fatty acid monosulphate: Oil-in-water adjuvant enhances SARS-CoV-2 RBD nanoparticle-induced immunogenicity and protection in mice. NPJ Vaccines 2023, 8, 18.
  142. Dowling, D.J.; Levy, O. A Precision Adjuvant Approach to Enhance Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Vaccines Optimized for Immunologically Distinct Vulnerable Populations. Clin. Infect. Dis. 2022, 75, S30–S36.
  143. Li, C.; Lee, A.; Grigoryan, L.; Arunachalam, P.S.; Scott, M.K.D.; Trisal, M.; Wimmers, F.; Sanyal, M.; Weidenbacher, P.A.; Feng, Y.; et al. Mechanisms of innate and adaptive immunity to the Pfizer-BioNTech BNT162b2 vaccine. Nat. Immunol. 2022, 23, 543–555.
  144. Tahtinen, S.; Tong, A.-J.; Himmels, P.; Oh, J.; Paler-Martinez, A.; Kim, L.; Wichner, S.; Oei, Y.; McCarron, M.J.; Freund, E.C.; et al. IL-1 and IL-1ra are key regulators of the inflammatory response to RNA vaccines. Nat. Immunol. 2022, 23, 532–542.
  145. Kobiyama, K.; Ishii, K.J. Making innate sense of mRNA vaccine adjuvanticity. Nat. Immunol. 2022, 23, 474–476.
  146. Verbeke, R.; Hogan, M.J.; Loré, K.; Pardi, N. Innate immune mechanisms of mRNA vaccines. Immunity 2022, 55, 1993–2005.
  147. Tse, S.W.; McKinney, K.; Walker, W.; Nguyen, M.; Iacovelli, J.; Small, C.; Hopson, K.; Zaks, T.; Huang, E. mRNA-encoded, constitutively active STING(V155M) is a potent genetic adjuvant of antigen-specific CD8(+) T cell response. Mol. Ther. 2021, 29, 2227–2238.
  148. Zhu, W.; Wei, L.; Dong, C.; Wang, Y.; Kim, J.; Ma, Y.; Gonzalez, G.X.; Wang, B.Z. cGAMP-adjuvanted multivalent influenza mRNA vaccines induce broadly protective immunity through cutaneous vaccination in mice. Mol. Nucleic Acids 2022, 30, 421–437.
  149. Fan, N.; Chen, K.; Zhu, R.; Zhang, Z.; Huang, H.; Qin, S.; Zheng, Q.; He, Z.; He, X.; Xiao, W.; et al. Manganese-coordinated mRNA vaccines with enhanced mRNA expression and immunogenicity induce robust immune responses against SARS-CoV-2 variants. Sci. Adv. 2022, 8, eabq3500.
  150. Liu, J.Q.; Zhang, C.; Zhang, X.; Yan, J.; Zeng, C.; Talebian, F.; Lynch, K.; Zhao, W.; Hou, X.; Du, S.; et al. Intratumoral delivery of IL-12 and IL-27 mRNA using lipid nanoparticles for cancer immunotherapy. J. Control. Release 2022, 345, 306–313.
  151. Li, Y.; Su, Z.; Zhao, W.; Zhang, X.; Momin, N.; Zhang, C.; Wittrup, K.D.; Dong, Y.; Irvine, D.J.; Weiss, R. Multifunctional oncolytic nanoparticles deliver self-replicating IL-12 RNA to eliminate established tumors and prime systemic immunity. Nat. Cancer 2020, 1, 882–893.
  152. Loomis, K.H.; Lindsay, K.E.; Zurla, C.; Bhosle, S.M.; Vanover, D.A.; Blanchard, E.L.; Kirschman, J.L.; Bellamkonda, R.V.; Santangelo, P.J. In Vitro Transcribed mRNA Vaccines with Programmable Stimulation of Innate Immunity. Bioconjug. Chem. 2018, 29, 3072–3083.
  153. Barman, S.; Borriello, F.; Brook, B.; Pietrasanta, C.; De Leon, M.; Sweitzer, C.; Menon, M.; van Haren, S.D.; Soni, D.; Saito, Y.; et al. Shaping Neonatal Immunization by Tuning the Delivery of Synergistic Adjuvants via Nanocarriers. ACS Chem. Biol. 2022, 17, 2559–2571.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 177
Revisions: 2 times (View History)
Update Date: 07 Oct 2023
1000/1000