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 -- 1474 2023-07-24 00:05:38 |
2 only format change Meta information modification 1474 2023-07-25 03:53:19 |

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.
Hogenesch, H.; Laera, D.; O’hagan, D.T. Physicochemical Characterization of Aluminum Adjuvants. Encyclopedia. Available online: https://encyclopedia.pub/entry/47151 (accessed on 02 July 2024).
Hogenesch H, Laera D, O’hagan DT. Physicochemical Characterization of Aluminum Adjuvants. Encyclopedia. Available at: https://encyclopedia.pub/entry/47151. Accessed July 02, 2024.
Hogenesch, Harm, Donatello Laera, Derek T. O’hagan. "Physicochemical Characterization of Aluminum Adjuvants" Encyclopedia, https://encyclopedia.pub/entry/47151 (accessed July 02, 2024).
Hogenesch, H., Laera, D., & O’hagan, D.T. (2023, July 24). Physicochemical Characterization of Aluminum Adjuvants. In Encyclopedia. https://encyclopedia.pub/entry/47151
Hogenesch, Harm, et al. "Physicochemical Characterization of Aluminum Adjuvants." Encyclopedia. Web. 24 July, 2023.
Physicochemical Characterization of Aluminum Adjuvants
Edit

Aluminum-based adjuvants will continue to be a key component of currently approved and next generation vaccines. Their large adsorptive capacity allows the combination with other immunostimulatory molecules to create a toolbox of adjuvants for selective vaccine applications. The widespread use of aluminum adjuvants is due to their excellent safety profile, which has been established through the use of hundreds of millions of doses in humans over many years, low cost, and extensive experience with licensure through regulatory agencies.

aluminum adjuvants physicochemical

1. Introduction

New-generation vaccines are being increasingly prepared with highly purified antigens, which improves their safety and tolerability, whilst enabling more simple characterization, but also typically results in decreased immunogenicity. Consequently, adjuvants are added to these vaccines, particularly those comprising recombinant protein subunits, to enhance their ability to induce robust immune responses. Although several new adjuvants have been introduced for specific vaccines over the past 20 years, aluminum adjuvants remain the most commonly used approach, even though they were first discovered nearly 100 years ago [1]. The two main types of aluminum adjuvants included in licensed human vaccines are aluminum hydroxide (AH) and aluminum phosphate (AP). The term ‘alum’ is often incorrectly used to refer to aluminum-containing adjuvants but can be an acceptable abbreviation if associated with a clarifying statement. Alum is a chemical solution of potassium aluminum sulfate, [KAl(SO4)2·12H2O], which is not used as a vaccine adjuvant. While the term ‘alum’ may be used for reasons of simplicity, it is important to define whether this refers to AH or AP, as these adjuvants have very different physical and chemical properties [2]. In addition to AH and AP, a product called ImjectTM Alum (ThermoFisher Scientific, Waltham, MA, USA) is sometimes used in preclinical studies. However, ImjectTM Alum comprises amorphous aluminum hydroxycarbonate and crystalline magnesium hydroxide [3]. Although it has immunostimulatory activity, it appears to be less potent than AH [4] and is not used in licensed human vaccines.
The molecular and physical nature of materials used in vaccine adjuvants is very diverse, often for historical reasons. In addition to aluminum salts, adjuvants include oil-in-water emulsions, liposomes, and various natural products, including saponins and Toll-like receptor (TLR) agonists. Consequently, the mechanisms by which these components enhance immune responses are very heterogeneous and often not completely understood, as discussed comprehensively elsewhere [5][6][7]. In general, adjuvants act only at the site of injection and the draining lymph nodes, while systemic effects should be minimized to reduce the potential for poor tolerability. Adjuvants typically increase the recruitment and activation of innate immune cells, enhance antigen uptake or processing and presentation, increase accumulation of lymphocytes in draining lymph nodes, and can promote the development and persistence of germinal centers, which are necessary for the development of a robust immune response [7]. Each individual adjuvant can contribute through some or all of these mechanisms, to a greater or lesser extent. The enhancement of the immune response by adjuvants typically requires coadministration of antigen and adjuvant, although physical association between adjuvants and antigens is often not required. Nevertheless, adsorption of antigens is particularly important for aluminum adjuvants. Hence, the adsorption of antigen to aluminum adjuvants requires the use of specialized analytical techniques. Once the need for an adjuvant in vaccine formulations is concluded, the choice of adjuvant in vaccine formulations is determined by multifactorial considerations and is highly dependent on the targeted disease, the chemical and physical nature of the vaccine antigens, and the population for which the vaccine is intended. Aluminum adjuvants should be the first choice, given their long history of safe and effective use in man, in addition to their wide availability, low cost, and extensive experience of licensure of aluminum-adsorbed vaccines through various regulatory agencies. However, inadequate performance or incompatibility with vaccine antigens when using aluminum adjuvants can necessitate the use of alternative adjuvants such as emulsions or combination adjuvants with TLR agonists.

2. Physicochemical Characterization of Aluminum Adjuvants

A number of established assays are available that can be used to characterize aluminum adjuvants and to ensure consistency between batches [8]. Structural information can be obtained using X-ray diffraction (for AH adjuvants only), spectroscopy (Fourier transform infrared, nuclear magnetic resonance, Raman), and transmission electron microscopy [8][9].

2.1. Particle Size

Aluminum adjuvants are typically composed of primary nanoparticles that form irregularly shaped aggregates with dimensions of 1 to 20 μm [8]. The particle size and shape have important implications for the efficiency of particle uptake by immune cells through phagocytosis [10][11][12]. However, the reported size of the aggregates varies widely and depends on the methodology used to measure particle size, the tonicity and pH of the dispersion medium, and any dilution factor. The particle size can be determined using laser diffraction, dynamic light scattering, or microflow imaging. The surface charge of aluminum adjuvants is pH-dependent, and larger particle sizes are observed when the pH approaches the point of zero charge as the electrostatic repulsion is decreased [13]. Similarly, the addition of sodium chloride can obscure surface charges and enhance particle aggregation [14]. However, dilution of aluminum adjuvants in saline caused a decrease in particle size [15].

2.2. Surface Charge

The aluminum ions at the surface of AH nanoparticles are coordinated with a hydroxyl that can accept or donate a proton depending on the pH of the dispersion medium. As a result, AH has a pH-dependent surface charge. Its point of zero charge (PZC) is 11.4 and it is positively charged at neutral pH [16]. In the case of AP, a proportion of the surface hydroxyls is replaced by phosphate because of the higher affinity of aluminum for phosphate. Commercial AP adjuvants have a P:Al ratio of 1.1–1.15:1 and a PZC of around 5, which gives them a negative surface charge at neutral pH [17]. A greater proportion of surface hydroxyls results in a higher PZC. Thus, AAHS with a P:Al ratio of 0.3 has a higher PZC and is neutral at pH 7. The surface charge can be determined using a Zetasizer instrument.

2.3. Surface Area

The primary nanoparticles that make up the aluminum aggregates afford the adjuvants a very large surface area, estimated at 514 m2/g for AH adjuvant, based on water adsorption measured using gravimetric FTIR spectroscopy [18]. The surface area can also be determined using the Brunauer–Emmett–Teller (BET) theory following nitrogen adsorption. However, a smaller surface area was reported for AH compared with water adsorption [14], because dehydration of the samples results in agglomeration of particles with loss of surface area. While these methods cannot be used for AP adjuvants, the ultrastructure of AP, composed of 50 nm nanoparticles, suggests that AP also has a very large surface area.

2.4. Adsorption

The large surface area of aluminum adjuvants allows for a high adsorptive capacity for antigens, which can be used as a key tool to allow characterization of the adjuvants. Importantly, adsorption of antigens can impact the quality and magnitude of the immune response and may enhance or decrease the stability of antigens [19]. It should be noted that the dose of antigens in vaccine formulations is typically low, and usually well below full adsorptive capacity. Adsorptive capacity is affected by the type of antigens, the buffer (pH, ionic strength, composition), and other excipients, including the presence of stabilizers or surfactants. The major mechanisms involved in adsorption are ligand exchange of phosphates on the antigen with surface hydroxyls on the adjuvants, along with electrostatic and hydrophobic interactions [2]. Since electrostatic mechanisms play an important role in adsorption, the adsorptive capacity of AH is often determined using a protein that is negatively charged at neutral pH, such as bovine serum albumin (BSA) or ovalbumin, while the adsorptive capacity of AP is typically evaluated with a positively charged protein, such as lysozyme [20]. However, recent studies have suggested that ligand exchange contributes to the adsorption of BSA in spite of the fact that only 0.6% of its serine and threonine residues are phosphorylated [21]. Marked differences were observed in the adsorption mechanisms between BSA and two different types of AH as revealed by changing the pH and tonicity of the BSA solution [21].
It has long been recognized that the strength of adsorption of protein antigens to aluminum adjuvants can increase over time [22][23][24], which likely reflects structural changes in the adsorbed antigens as discussed previously [19]. These structural changes may improve the immunogenicity of the vaccine formulation, but can also lead to deamidation and loss of epitopes, as demonstrated for recombinant protective antigen of the anthrax bacillus [25]. The stability and immunogenicity of adsorbed vaccine formulations should be studied over time.

2.5. Elemental Composition

The presence of impurities in aluminum adjuvants can be determined using inductively coupled plasma mass spectrometry (ICP-MS) [26]. Differences in the type and quantity of metal ions were reported between AH adjuvants obtained from different manufacturers and different batches, which are likely caused by differences in the sources of aluminum salts, chemicals, and water used during the production process [26]. The presence of sulfur likely reflects the use of alum as the starting material as discussed above. Some contaminants such as copper may affect the stability of adsorbed antigens [26].

References

  1. Glenny, A.T.; Pope, C.G.; Waddington, H.; Wallace, U. Immunological notes. XVI1.-XXIV. J. Pathol. Bacteriol. 1926, 29, 31–40.
  2. Hem, S.L.; HogenEsch, H. Relationship between physical and chemical properties of aluminum-containing adjuvants and immunopotentiation. Expert. Rev. Vaccines 2007, 6, 685–698.
  3. Hem, S.L.; Johnston, C.T.; HogenEsch, H. Imject Alum is not aluminum hydroxide adjuvant or aluminum phosphate adjuvant. Vaccine 2007, 25, 4985–4986.
  4. Cain, D.W.; Sanders, S.E.; Cunningham, M.M.; Kelsoe, G. Disparate adjuvant properties among three formulations of “alum”. Vaccine 2013, 31, 653–660.
  5. Del Giudice, G.; Rappuoli, R.; Didierlaurent, A.M. Correlates of adjuvanticity: A review on adjuvants in licensed vaccines. Semin. Immunol. 2018, 39, 14–21.
  6. 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.
  7. HogenEsch, H.; Orr, M.T.; Fox, C.B. Vaccine adjuvants: Mechanisms of action. In Vaccine Development: From Concept to Clinic; Prasad, A.K., Ed.; The Royal Society of Chemistry: London, UK, 2023; pp. 214–234.
  8. Hem, S.L.; Johnston, C.T. Production and characterization of aluminum-containing adjuvants. In Vaccine Development and Manufacturing; Wen, E.P., Ellis, R., Pujar, N.S., Eds.; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2015; pp. 319–346.
  9. Mei, C.; Deshmukh, S.; Cronin, J.; Cong, S.; Chapman, D.; Lazaris, N.; Sampaleanu, L.; Schacht, U.; Drolet-Vives, K.; Ore, M.; et al. Aluminum Phosphate Vaccine Adjuvant: Analysis of Composition and Size Using Off-Line and In-Line Tools. Comput. Struct. Biotechnol. J. 2019, 17, 1184–1194.
  10. Tabata, Y.; Ikada, Y. Effect of the size and surface charge of polymer microspheres on their phagocytosis by macrophage. Biomaterials 1988, 9, 356–362.
  11. Champion, J.A.; Mitragotri, S. Role of target geometry in phagocytosis. Proc. Natl. Acad. Sci. USA 2006, 103, 4930–4934.
  12. Champion, J.A.; Walker, A.; Mitragotri, S. Role of particle size in phagocytosis of polymeric microspheres. Pharm. Res. 2008, 25, 1815–1821.
  13. Langford, A.; Horwitz, T.; Adu-Gyamfi, E.; Wiley, C.; Holding, E.; Zimmermann, D.; Ignatius, A.A.; Ohtake, S. Impact of Formulation and Suspension Properties on Redispersion of Aluminum-Adjuvanted Vaccines. J. Pharm. Sci. 2020, 109, 1460–1466.
  14. Art, J.F.; Vander Straeten, A.; Dupont-Gillain, C.C. NaCl strongly modifies the physicochemical properties of aluminum hydroxide vaccine adjuvants. Int. J. Pharm. 2017, 517, 226–233.
  15. Badran, G.; Angrand, L.; Masson, J.D.; Crepeaux, G.; David, M.O. Physico-chemical properties of aluminum adjuvants in vaccines: Implications for toxicological evaluation. Vaccine 2022, 40, 4881–4888.
  16. Hem, S.L.; Klepak, P.B.; Lindblad, E.B. Aluminum hydroxide adjuvant. In Handbook of Pharmaceutical Excipients, 5th ed.; Rowe, R.C., Sheskey, P.J., Owen, S.C., Eds.; Pharmaceutical Press: London, UK, 2006; pp. 36–37.
  17. Hem, S.L.; Klepak, P.B.; Lindblad, E.B. Aluminum phosphate adjuvant. In Handbook of Pharmaceutical Excipients, 5th ed.; Rowe, R.C., Sheskey, P.J., Owen, S.C., Eds.; Pharmaceutical Press: London, UK, 2006; pp. 40–41.
  18. Wang, S.L.; Johnston, C.T.; Bish, D.L.; White, J.L.; Hem, S.L. Water-vapor adsorption and surface area measurement of poorly crystalline boehmite. J. Colloid Interface Sci. 2003, 260, 26–35.
  19. HogenEsch, H.; O’Hagan, D.T.; Fox, C.B. Optimizing the utilization of aluminum adjuvants in vaccines: You might just get what you want. NPJ Vaccines 2018, 3, 51.
  20. al-Shakhshir, R.H.; Regnier, F.E.; White, J.L.; Hem, S.L. Contribution of electrostatic and hydrophobic interactions to the adsorption of proteins by aluminium-containing adjuvants. Vaccine 1995, 13, 41–44.
  21. Art, J.F.; Soumillion, P.; Dupont-Gillain, C.C. Use of a quartz crystal microbalance platform to study protein adsorption on aluminum hydroxide vaccine adjuvants: Focus on phosphate-hydroxide ligand exchanges. Int. J. Pharm. 2020, 573, 118834.
  22. Gupta, R.K. Aluminum compounds as vaccine adjuvants. Adv. Drug. Deliv. Rev. 1998, 32, 155–172.
  23. Van Ramshorst, J.D. The adsorption of diphtheria toxoid on aluminium phosphate. Recl. Trav. Chim. Des Pays-Bas 1949, 68, 169–180.
  24. Barrett, B.S.; Markham, A.P.; Esfandiary, R.; Picking, W.L.; Picking, W.D.; Joshi, S.B.; Middaugh, C.R. Formulation and immunogenicity studies of type III secretion system needle antigens as vaccine candidates. J. Pharm. Sci. 2010, 99, 4488–4496.
  25. D’Souza, A.J.; Mar, K.D.; Huang, J.; Majumdar, S.; Ford, B.M.; Dyas, B.; Ulrich, R.G.; Sullivan, V.J. Rapid deamidation of recombinant protective antigen when adsorbed on aluminum hydroxide gel correlates with reduced potency of vaccine. J. Pharm. Sci. 2013, 102, 454–461.
  26. Schlegl, R.; Weber, M.; Wruss, J.; Low, D.; Queen, K.; Stilwell, S.; Lindblad, E.B.; Mohlen, M. Influence of elemental impurities in aluminum hydroxide adjuvant on the stability of inactivated Japanese Encephalitis vaccine, IXIARO. Vaccine 2015, 33, 5989–5996.
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: 335
Revisions: 2 times (View History)
Update Date: 25 Jul 2023
1000/1000
Video Production Service