Applications of nanotechnology in allergy and asthma

  • Saima Pervaiz
  • Sara Masood Cheema
  • Tahir Shaheen
  • Romeeza Tahir
  • Zeeshan Haider
  • Saba Shamim
Volume 2
|
Pages 4-14
|
2023
61 Views
|
7 Downloads
|
0 Crossref Citations
PDF

Abstract

The increasing incidence of allergic diseases requires effective curative strategies for their alleviation. Allergen-specific immunotherapy (AIT) is a causal treatment technique instead of a symptomatic treatment technique for allergies. At present, AIT is being offered utilizing immunization response modifiers (IRMs) or adjuvants. The adjuvant helps in vigorous inaugral as well as long-term IR (immune response), thus enhancing the efficacy of AIT. The successful development of novel additives requires complete knowledge regarding the underdevelopment of novel and conservative additives. Hence, this review describes the applications of nanotechnology in allergic disorders, namely, allergies and asthma. The development of vaccines based on nanoparticles (NPs) is an important strategy for AIT because of their innate physicochemical characteristics, production ease and capacity to incite inherent immunity.

Keywords

Nanotechnology; Allergens; Immunotherapy, allergen-specific; Asthma; Adjuvants, immunologic

1. Introduction

Allergic disorders are believed to be an increasing health issue worldwide, affecting more than 25% of the population in developed states [1,2,3], and a considerable increase in the prevalence of allergic rhinitis (AR) and asthma has been observed globally since 1960 [1,4]. Currently, allergies are an important health care issue [5]. According to the World Health Organization (WHO), the majority of the world population (10–30%) experiences AR [6,7]. AR occurs due to IgE-mediated nasal mucosa inflammation. Patients present in outdoor and emergency departments with signs and symptoms of nasal blockage, nasal itching, watery rhinorrhea, sneezing, watery and red eyes, and psychosocial problems such as emotional distress, sleeping disorders, and financial losses due to absence from work [8]. AR is an important risk factor for asthma [5]. It is a respiratory disease that is characterized by spasms in the airway passages leading to difficulty breathing. This is usually due to the response to allergens and occurs in combination with other forms of hypersensitivity reactions. Asthma is a very common leading world health problem that affects more than 330 million people globally [9]; however, it is not curable despite decades of combined efforts to diagnose, treat, and prevent the disease in due time [10,11].

2. Immunotherapy for allergies and asthma

2.1. Immunotherapy

To treat allergies, immunotherapy is utilized extensively. Despite its efficiency, its fundamental mechanisms are incompletely understood. The purpose of immunotherapy is to ‘act specifically’ (AIT) to facilitate host desensitization to a particular antigen and, in other words, to make the host 'compatible' with the antigen. Therefore, a flourishing IT (immunotherapy) will cause an increase in the amount of allergen needed to encourage allergic indications, along with immediate- and late-phase reductions in allergic inflammation [12,13].

Various mediators are involved in allergic reactions (Figure 1). Therefore, it is advisable to deactivate them by using antagonists, for example, antihistamines, antileukotrienes or agonists of their cellular receptor, such as the β2 adrenoreceptor. However, this approach is not an ongoing therapeutic technique, and treatment must be repeated after every antigen encounter. In contrast, immunotherapy has an effect beyond therapy duration since immunotherapy changes the IR to antigens (Figure 2). IT decreases the Th1/Th2 ratio and encourages T effector regulatory cells. Such modifications in the global balance of T-cell subsets are shown in the enhanced formation of IL-10 by allergen-presenting cells, B cells and T cells, which, along with transforming growth factor beta (TGFβ) production, activate B cells to IgA (Immunoglobulin-A) and IgG4 (Immunoglobulin-G4) and repress IgE (Immunoglobulin-E) production [14]. IgA and IgG oppose IgE in terms of antigen binding, reducing antigen capture by basophils. Moreover, subsequent reactions or cascades after immunotherapy decrease the amount of mast cells, basophils, neutrophils and eosinophils at allergen exposure sites, resulting in a decrease in inflammatory indications. Allergies may be restricted to a systematic or organ (intestinal) involved in anaphylactic reactions. The engagement of various effectors and triggering elements should be measured during rational IT (Figure 3) [12].

Figure 1. Allergy process.

Figure 2. Immunization.

Figure 3. Oral immunotherapy.

Currently, traditional immunotherapy depends on immunization with repeated doses of allergens for durations imitating what the host feels while repeatedly confronting an ecological challenge. Therefore, after repetitive contact with ‘substances’ and to evade a constant inflammatory reaction, the host increases its regulatory reaction due to an increase in Tregs and Th1 cells, which represses immunoglobulin E production. However, despite these benefits, an insignificant proportion of patients opt for IT as their treatment option for allergies [15]. Subcutaneous immunotherapy, despite its effectiveness, is not limited due to possible side effects after immunization, such as local allergic reactions, infrequent anaphylaxis and even near-lethal responses [16,17]. Keeping in mind such risks, subcutaneous IT should be administered only by specialists. In contrast, to achieve continuing immunological reactions, it is necessary to obtain a minimum of 3 to 5 years of treatment, necessitating good patient compliance. An additional limitation regarding conventional subcutaneous immunotherapy is a phobia among a few patients about needles and associated risks of needle use [18]. Thus, efforts to develop more convenient, safer and effective strategies are being explored, such as recombinant allergen generation that offers no IgE-mediated activation, novel adjuvants supporting particular Th1 responses, hypoallergenic glycoconjugates and, furthermore, various routes of specific immunotherapy (SIT) application, particularly sublingual, intralymphatic and epicutaneous delivery of antigens [19,20,21].

2.2. Mechanism of immunotherapy based on specific allergens

Allergen-specific immunotherapy (AIT) techniques are not fully understood. Recent studies have shown that efficient AIT successively activates several mechanisms and promotes molecular and cellular modifications (Figure 4) [22].

Figure 4. Allergan-specific immune therapy.

This complicated mode of action of AIT occurs in three phases: i) rapid desensitization, ii) early tolerance, and iii) sustained tolerance. This rapid desensitization is due to an initial decrease in the degranulation of basophils and mast cells, possibly caused by rapid upregulation of the histamine type 2 receptor. The 2nd phase, which is called early tolerance, comprises a reduction in interleukin-4 (IL-4)-releasing TH2 cells and an increase in interleukin-10-releasing Breg and Treg cells. A switch is found between TH1-type reactions and TH2-type reactions, with increases in interleukin-10 and TGFβ production. Moreover, there is an increase in Treg cells, which is associated with medical improvement. The final phase is sustained tolerance, in which Treg cells excite B cells to create allergen-specific immunoglobulin-G4, a tolerogenic elevated similarity blocking the antibody that competes with allergen-specific immunoglobulin-E, hence evading the allergen-induced secretion of the mediators through basophils and mast cells. These successively activated methods induce immune tolerance, which mitigates or even eliminates the allergic reaction in the early (acute) phase and any subsequent immunological events [22,23].

3. Applications of nanotechnology in allergies and asthma

3.1. Nanotechnology

Nanotechnology is believed to be an emerging technology because of the probability of making innovative products with total novel attributes and functions together with massive potential in a diverse variety of applications along with considerable impact on health care [24]. Nanotechnology is a multisectoral area of research that controls and manipulates molecules and atoms between 0.1 and 100 nm in size and manufactures relevant structures and materials [25,26]. As an influential motive for developing biomedicine, nanotechnology has expanded over the fields comprising early identification, prevention and treatment of diseases and bioengineering research, demonstrating hopeful prospects. The majority of nanomaterials (NMs) have been utilized in biomedical studies. Nanoparticles (NPs) are the most extensively studied medical delivery system and have been described in more than 25000 research articles during the last decade [27]. In 1995, the Doxil Nanotechnology (NT) platform was approved and became successful on the market. Since then, several nondrugs have also been approved by the Food and Drug Administration of the USA, while several other medicines are in clinical development [25].

3.2. Nanotechnology medicine applications

In medicine, the use of nanotechnology enhances the half-life of medications by enhancing drug incorporation, reducing drug degradation/clearance, and slowing the release of loaded drugs. The movement period of the nanoparticle body is controlled by the changing surface charge of the NPs, as per their application goal. However, a positive charge enhances the incorporation of NPs, and in the blood stream, negative charges increase the time of circulation. Additionally, they enhance drug bioavailability and increase the dissolvability of the hydrophobic preparation. They study pharmacokinetics and deliver medicines exactly as needed. Hence, they reduce the required dosages and side effects. They can be combined with one drug or more in combination. Decisions can be made autonomously by the new generation of nanorobotics according to circumstances and can assist surgeons during surgery [28,29,30,31,32]. The use of nanotechnology in medicine is summarized in Figure 5.

Figure 5. Overview of nanotechnology: applications of nanoparticles in the medical sciences, including immunization, therapeutics, and diagnostics.

3.3. Nanotechnology applied to immunotherapy

During the past decade, nanotechnologies have been utilized in immunomodulatory treatments to increase their effectiveness and to reduce possible side effects, mostly in addressing carcinoma and communicable diseases [1]. Over the past 30 years, numerous polymers have been utilized to design NPs that are able to load both nucleic acids and proteins for therapeutic applications [33,34]. However, no universally accepted NP definition is available that could be considered a basic component of NMs, having described physical boundaries and not less than one dimension/diameter in the nanoscale range [35]. In 2011, the definition of NMs suggested by the European Commission was based on size as a basic parameter to explain incidental, manufactured and natural materials having particles along with any external measurement in the “nano” range (1 to 100 nm) [36]. A very broad review, including current technological and scientific developments, is in progress [1].

NPs can take several different shapes and possess variable physical characteristics and chemical compositions, including size/hydrodynamic radius, surface chemistry, solubility, morphology and charge [37]. These characteristics could be exploited to make them appropriate for particular biomedical applications. The influence of physicochemical properties on NP biocompatibility is important for their use in biological applications and has been extensively investigated. In contrast, the biodistribution of NPs after they enter the human body through several routes, their cellular intake mechanisms and their probable toxicity are influenced mainly by the physicochemical characteristics of the particles, especially their surface and size characteristics [38].

3.4. Physical and chemical applications of NPs

Several physicochemical applications of NPs have been demonstrated to assist direct or indirect immune modulation. Short NPs can enhance tissue infiltration and thus increase the availability of particle-loaded allergens to lymph nodes and blood vessels [39]. The effects of transdermal administration of amorphous silicon dioxide were examined by Palmer and coworkers. In a model of allergic contact dermatitis, NPs were used, and they were found to have increased immunomodulatory potential. This research compares the epidermal patency of NPs to that of microparticles, indicating that NPs enhanced invasion (27.8 ± 3.4 nm) in mouse skin compared with microparticles (557.6 ± 35.1 nm) [40]. Similarly, Hirai and colleagues examined the epidermal penetration and subcellular localization of mono-dispersal silica in amorphous stat NPs 70 nm wide in rats and showed that the particles effectively invaded via epidemic barricades and were found in the lymph nodes present in local tissues [41]. Numerous studies have demonstrated the efficiency of injecting NPs into the epidermis barrier [42,43]. Good penetration and localization in tissue can actually enhance AIT through new administration routes, such as intranasal or percutaneous delivery. Moreover, their size, chemical composition, shape, solubility and surface charge make them appealing for AIT. Jatana and associates investigated the effect of the charge and size of various NPs (silica, silver, titanium dioxide and gold) on immunomodulation in a skin allergy mouse model and reported that negatively charged small NPs demonstrated immunosuppression [44]. Environmental polynameric NPs, which are biocompatible with cells and tissues, could be promising candidates for AIT because they could decrease unnecessary side effects related to the present therapy by utilizing alum as an additive. The nanoparticle surface and shape have a significant effect on cellular intake. Champion and teammates demonstrated in their study that one may control phagocytic intake through an effective structure. The authors found that globular particles are more effectively taken up by macrophages because of their elevated length, as indicated by the normalization curve [45]. Particle surface properties can promote uptake by inducing communication with receptors on the cell surface (CRS). Hence, this characteristic can be utilized through nanoparticle surface functions with preferred active groups binding to particular receptors present on the surface. Lectin-functionalized polylactic-co-glycolic acid particles were suggested to be favorable platforms for oral AIT. This technique of functionalization was effective in selecting enterocytes, hence improving the uptake and preventing the degradation of the delivered allergen via gastrointestinal enzymes [46].

3.5. Depot-forming ability

The depot effect is believed to be a significant mechanism of immune tolerance induction. Prolonged and constant discharge of antigens can increase the exposure time of immune cells and can cause immunomodulation [47]. Furthermore, depot formation at the target site concurrently decreases the therapeutic dosage. In the case of NPs, the allergen can be summarized within carrying systems, while their discharge can be adjusted to the desired functions by adjusting the surface particle along with the polymer. The coating of chitosan on the encapsulating drug polylactic-co-glycolic acid NPs demonstrated a measured discharge of ingredients in pharmaceutical formulations that are active in comparison with that of pristine polylactic-co-glycolic acid NPs [48]. Lacey and colleagues highlighted an immune-increasing depot effect of cationic tuberculosis (TB) containing a liposome vaccine allergen (Ag85B–ESAT-6) [49]. Therefore, the ability of NPs to form a depot may have curative benefits [39].

3.6. Protection from enzymatic degradation

NPs can defend against antigens via encapsulation by protecting them from protease enzymes within the body, which is desired during oral immune therapy (OIT), similar to vaccines that have to overcome the harsh conditions of the gastrointestinal tract [50]. OIT is an innovative strategy under study for food allergy treatment. It was demonstrated to be effective for almost 60-80% of the populace investigated [51]. Brotons-Canto and colleagues examined the ability of mannosylated NPs for oral immunotherapy against peanut allergy in mice and found that AIT was highly suitable. From an innovative polymer, the NP system was synthesized by covalent binding of mannosamine to a polyanhydride backbone [52]. Similarly, OIT with polyanhydride NPs was shown to have significant advantages in peanut allergy therapy. A study showed an increase in Treg and Th1-resistant responses and a decrease in the activation of Th2 cells in in vivo mouse models. Srivasta and coworkers also demonstrated the success of preclinical research on oral immunotherapy utilizing CpG-coated polylactic-co-glycolic acid NPs in murine peanut allergy models [53]. The authors observed a significant and sustained reduction in the levels of peanut-specific IgE/IgG1, along with Th2 cytokines, as well as an increase in the levels of peanut-specific IgG2a and IFN-ϒ. Additionally, this study investigated the safety of the use of CpG-poly(lactic-co-glycolic acid) NPs in oral immunotherapy by evaluating their inability to induce anaphylaxis by assessing the release of plasma histamine [53]. Therefore, the ability of NPs to prevent antigens together with the activation of the desired immune reaction enables them to be perfect for AIT [39].

3.7. Enhancement of allergen-specific tolerance

The ultimate goal of immunotherapy for allergic disorders is to increase tolerance to allergen-specific immune agents. The innate ability of NPs to target antigen-presenting cells and their ability to transmit signals, which evoke antigen-specific immune reactions, make them feasible tools for modulating immune responses [54]. The NPs that can attain this objective of resistance tolerance induction can collectively be described as tolerogenic NPs. The application of AIT tolerogenic NPs would be suitable for preventing unnecessary immune reactions, which tend to occur in present therapeutic practice. Various sets of NPs have been used to induce tolerogenic immune reactions. Metalic oxide NPs, synthetic polymers and liposomes are being investigated; however, biodegradable polymeric NPs are the most commonly studied NMs [55,56]. Maldondo and collaborators reported the potential of a tolerogenic nanocarrier system (encapsulated with peptide protein/antigen) to neutralize and prevent a pathological resistance response [57]. Furthermore, polymeric NPs combined with CpG ODNs were confirmed to be efficient at treating allergic airway illness caused by house dust mites [58]. Nanoscale bronchial interventions combined with CpG markedly improved dendritic cell (DC) recruitment and initialization, causing Th1 immune resistance in a model of household dust mite allergy and decreasing allergy-associated indications. These studies certainly recommend further nanomaterial expansion as a potential platform technology to mitigate allergic diseases [39].

3.8. Nanoparticle therapy for asthma

Asthma occurs due to chronic airway inflammatory disorders related to airway hyperresponsiveness [59]. In asthma, chronic inflammation can cause ultrastructural modifications in airways related to airway remodeling. These modifications are incompletely transposed by recently available treatment strategies, such as steroids [60].

Asthma is thought to be best controlled by the use of steroid inhalation; however, the effects of this intervention are likely to be small, not >1–2 hours. Current research has demonstrated that stealth steroids compressed with NPs achieve high and prolonged benefits at airway inflammation sites compared with free steroids [61]. Moreover, budesonide NP agglomerates have shown an attractive microstructure for effective lung deposition, as do nanostructures for the swift dissolution of inadequately water-soluble drugs [62]. In addition, smaller NPs have more effective effects on bronchodilation [63]. Bhavna and associates (2009) highlighted that NPs compressed with salbutamol act together more with the lung layer, as mucociliary movement and peripheral deposition back to the tracheo-bronchial area are more severe, leading to higher and further sustained medicine concentrations within the target area [64].

During gene transfer, chitosan (a biocompatible cationic polysaccharide obtained from crustacean shell chitin) with an NP shape (100 to 200 nm) may be utilized to deliver plasmids. Kumar and colleagues (2003) indicated that chitosan interferon (IFN)-γ-plasmid deoxyribonucleic acid (pDNA) NP treatment successfully decreased functional and immunological anomalies related to antigen sensitization and challenge. This impact was primarily mediated through the STAT4 signaling pathway. Furthermore, due to similarities between humans and mice in terms of the T-cell demarcation pathway, these findings highlighted that chitosan IFN-γ-pDNA NPs could reverse allergic asthma in humans. Additionally, the outcomes obtained by Kumar and colleagues demonstrated that intranasal chitosan IFN-γ-pDNA nanoparticle treatment could be helpful in both prophylaxis and asthma treatment [65].

Additionally, Kong and Teammates (2008) reported that this treatment caused in situ production of IFN-γ, decreased airway reactivity and inflammation, decreased numbers of proinflammatory CD8+ T cells and inhibited antigen-presenting activity in dendritic cells in mice [66].

4. Conclusions

For AIT, a prospective vaccine should have the ability to achieve (i) efficacy and safety with an appropriate allergen dose, (ii) patient compliance, which is promoted through a well-tolerated regimen, self-administration, and cost-effectiveness, and (c) the minimization of adverse effects related to treatment. NPs can technically fulfill the abovementioned criteria, and their physiochemical properties and distinctive ability to induce immune tolerance might qualify them as an ideal type of adjuvant for AIT. Nanoparticle drug formulations offer numerous advantages over conventional formulations.

Thus, development in nanoscience will have a very significant impact because it has the potential to significantly transform healthcare in the coming years. Future studies and clinical trials are recommended to evaluate the exact role, efficacy, side effects and safety in the management of AR and asthma using robust new techniques.

Author contributions

SP, SMC, TS were involved in the conception and design of the study. RT and ZH were involved in the investigation and screening of articles and data extraction. RT, SMC, ZH, and SS were involved in the analysis and interpretation of data. SP, SMC, SMC, TS, RT, ZH and SS were involved in the drafting and critical revision of the manuscript. All authors have read and agreed to the published version of the manuscript.

Publication history

Received Revised Accepted Published
04 May 2023 07 July 2023 14 July 2023 29 July 2023

Funding

This research received no specific grant from the public, commercial, or not-for-profit funding agencies.

Ethics statement

Not applicable.

Consent to participate

Not applicable.

Data availability

Not applicable.

Acknowledgments

None.

Conflicts of interest

The authors declare no conflicts of interest.

Copyright

© 2023 The Author(s). This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY) Li­cense. The use, distribution, or re­production in other forums is per­mitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or re­production is permitted which does not comply with these terms.

Publisher's note

Logixs Journals re­mains neutral concerning jurisdic­tional claims in its published subject matter, including maps and institutional affiliations.

References

[1]

Di Felice G, Colombo P. Nanoparticle–allergen complexes for allergen immunotherapy. Int J Nanomedicine. 2017;12:4493-504. https://doi.org/10.2147/ijn.s134630

[2]

Bousquet J, Anto J, Auffray C, Akdis M, Cambon-Thomsen A, Keil T, et al. MeDALL (Mechanisms of the Development of ALLergy): an integrated approach from phenotypes to systems medicine. Allergy. 2011;66(5):596-604. https://doi.org/10.1111/j.1398-9995.2010.02534.x

[3]

Newson RB, Van Ree R, Forsberg B, Janson C, Lötvall J, Dahlén SE, et al. Geographical variation in the prevalence of sensitization to common aeroallergens in adults: the GA(2) LEN survey. Allergy. 2014;69(5):643-51. https://doi.org/10.1111/all.12397

[4]

Devereux G. The increase in the prevalence of asthma and allergy: food for thought. Nat Rev Immunol. 2006;6:869-74. https://doi.org/10.1038/nri1958

[5]

Garaczi E, Szabó K, Francziszti L, Csiszovszki Z, Lőrincz O, Tőke ER, et al. DermAll nanomedicine for allergen-specific immunotherapy. Nanomedicine. 2013;9(8):1245-54. https://doi.org/10.1016/j.nano.2013.05.011

[6]

Compalati E, Penagos M, Henley K, Canonica GW. Allergy prevalence survey by the world allergy organization. J World Allergy Organ. 2007;19(3):82-90. 

[7]

Pawankar R, Canonica GW, Holgate ST, Lockey RF, editors. World Allergy Organization (WAO) white book on allergy. United Kingdom;2011. 

PDF
[8]

Bousquet J, Khaltaev N, Cruz AA, Denburg J, Fokkens WJ, Togias A, et al. Allergic rhinitis and its impact on asthma (ARIA) 2008. Allergy. 2008;63(s86):8-160. 

[9]

Cheng WC, Chen CH. Nanotechnology bring a new hope for asthmatics. Ann Transl Med. 2019;7(20):516. https://doi.org/10.21037/atm.2019.09.153

[10]

Da Silva AL, De Oliveira GP, Kim N, Cruz FF, Kitoko JZ, Blanco NG, et al. Nanoparticle-based thymulin gene therapy therapeutically reverses key pathology of experimental allergic asthma. Sci Adv. 2020;6(24):eaay7973. https://doi.org/10.1126/sciadv.aay7973

[11]

Gliklich RE, Castro M, Leavy MB, Press VG,  Barochia A, Carroll CL, et al. Harmonized outcome measures for use in asthma patient registries and clinical practice. J Allergy Clin Immunol. 2019;144(3):671-81. https://doi.org/10.1016/j.jaci.2019.02.025

[12]

Gamazo C, Gastaminza G, Ferrer M, Sanz ML, Irache JM. Nanoparticle based-immunotherapy against allergy. Immunotherapy. 2014;6(7):885-97. https://doi.org/10.2217/imt.14.63

[13]

Akdis CA, Akdis M. Mechanisms and treatment of allergic disease in the big picture of regulatory T cells. J Allergy Clin Immunol. 2009;123(4):735-46. https://doi.org/10.1016/j.jaci.2009.02.030

[14]

Steinke JW, Lawrence MG. T-cell biology in immunotherapy. Ann Allergy Asthma Immunol. 2014;112(3):195-9. https://doi.org/10.1016/j.anai.2013.12.020

[15]

Guenecehea-Sola M, Hariri SR, Galoosian A, Yusin JS. A retrospective review of veterans’ adherence to allergen immunotherapy over 10 years. Ann Allergy Asthma Immunol. 2014;112(1):79-81. https://doi.org/10.1016/j.anai.2013.10.018

[16]

Ragusa VF, Massolo A. Non-fatal systemic reactions to subcutaneous immunotherapy: a 20-year experience comparison of two 10-year periods. Eur Ann Allergy Clin Immunol. 2004;36(2):52-5. 

[17]

Amin HS, Liss GM, Bernstein DI. Evaluation of near-fatal reactions to allergen immunotherapy injections. J Allergy Clin Immunol. 2006; 117(1):169-75. https://doi.org/10.1016/j.jaci.2005.10.010

[18]

National Center for Immunization and Respiratory Diseases. General recommendations on immunization. Recommendations of the Advisory Committee on Immunization Practices (ACIP). MMWR Recomm Rep. 2011;60(2):1-64.

[19]

Niederberger V, Horak F, Vrtala S, Spitzauer S, Krauth MT, Valent P, et al. Vaccination with genetically engineered allergens prevents progression of allergic disease. Proc Natl Acad Sci U S A. 2004;101(Suppl 2):14677-82. https://doi.org/10.1073/pnas.0404735101

[20]

Weiss R, Scheiblhofer S, Machado Y, Thalhamer J. New approaches to transcutaneous immunotherapy: targeting dendritic cells with novel allergen conjugates. Curr Opin Allergy Clin Immunol. 2013;13(6):669-76. https://doi.org/10.1097/ACI.0b013e328364f4df

[21]

Weinberger EE, Himly M, Myschik J, Hauser M, Altmann F, Isakovic A, et al. Generation of hypoallergenic neoglycoconjugates for dendritic cell targeted vaccination: a novel tool for specific immunotherapy. J Control Release. 2013;165(2):101-9. https://doi.org/10.1016/j.jconrel.2012.11.002

[22]

Zubeldia JM, Ferrer M, Dávila I, Justicia JL. Adjuvants in allergen-specific immunotherapy: modulating and enhancing the immune response. J Investig Allergol Clin Immunol. 2019;29(2):103-11. https://doi.org/10.18176/jiaci.0349

[23]

Jutel M, Agache I, Bonini S, Burks AW, Calderon M, Canonica W, et al. International consensus on allergen immunotherapy II: mechanisms, standardization, and pharmacoeconomics. J Allergy Clin Immunol. 2016;137(2):358-68. https://doi.org/10.1016/j.jaci.2015.12.1300

[24]

Ali SA, Tariq M. Nanotechnology and its implication in medical science. J Pak Med Assoc. 2014;64(9):984-6.

[25]

Wang L, Feng M, Li Q, Qiu C, Chen R. Advances in nanotechnology and asthma. Ann Transl Med. 2019;7(8):180. https://doi.org/10.21037/atm.2019.04.62

[26]

Heidel JD, Davis ME. Clinical developments in nanotechnology for cancer therapy. Pharm Res. 2011;28:187-99. https://doi.org/10.1007/s11095-010-0178-7

[27]

Anselmo AC, Mitragotri S. An overview of clinical and commercial impact of drug delivery systems. J Control Release. 2014;190:15-28. https://doi.org/10.1016/j.jconrel.2014.03.053

[28]

El-Sayed A, Kamel M. Advances in nanomedical applications: diagnostic, therapeutic, immunization, and vaccine production. Environ Sci Pollut Res. 2020;27(16):19200-13. https://doi.org/10.1007/s11356-019-06459-2

[29]

Freitas RA Jr. Microbivores: artificial mechanical phagocytes using digest and discharge protocol. J Evol Technol. 2005;14(1):54-106.

PDF
[30]

Freitas RA. Current status of nanomedicine and medical nanorobotics. J Comput Theor Nanosci. 2005;2(1):1-25. https://doi.org/10.1166/jctn.2005.01

[31]

Jurj A, Braicu C, Pop LA, Tomuleasa C, Gherman CD, Berindan-Neagoe I. The new era of nanotechnology, an alternative to change cancer treatment. Drug Des Devel Ther. 2017;11:2871-90. https://doi.org/10.2147/dddt.s142337

[32]

Troncarelli MZ, Brandão HM, Gern JC, Guimarães AS, Langoni H. Nanotechnology and antimicrobials in veterinary medicine. Formatex: Badajoz, Spain. 2013;13:543-56.

[33]

Etheridge ML, Campbell SA, Erdman AG, Haynes CL, Wolf SM, McCullough J. The big picture on nanomedicine: the state of investigational and approved nanomedicine products. Nanomedicine. 2013;9(1):1-14. https://doi.org/10.1016/j.nano.2012.05.013

[34]

Weissig V, Pettinger TK, Murdock N. Nanopharmaceuticals (part 1): products on the market. Int J Nanomedicine. 2014;9(1):4357-73. https://doi.org/10.2147/ijn.s46900

[35]

Duncan R, Gaspar R. Nanomedicine(s) under the microscope. Mol Pharmaceutics. 2011;8(6):2101-41. https://doi.org/10.1021/mp200394t

[36]

Official Journal of the European Union. Commission Recommendation of 18 October 2011 on the definition of nanomaterial. European Commission; 2011. 2023 [cited 10 March 2020]. Available from: http://eur-lex.europa.eu/lexuriserv/lexuriserv.do?uri=oj:l:2011:275:0038:0040:en:pdf.

[37]

Wu L, Zhang J, Watanabe W. Physical and chemical stability of drug nanoparticles. Adv Drug Deliv Rev. 2011;63(6):456-69. https://doi.org/10.1016/j.addr.2011.02.001

[38]

Nyström AM, Fadeel B. Safety assessment of nanomaterials: implications for nanomedicine. J Control Release. 2012;161(2):403-8. https://doi.org/10.1016/j.jconrel.2012.01.027

[39]

Johnson L, Duschl A, Himly M. Nanotechnology-based vaccines for allergen-specific immunotherapy: potentials and challenges of conventional and novel adjuvants under research. Vaccines. 2020;8(2):237. https://doi.org/10.3390/vaccines8020237

[40]

Palmer BC, Jatana S, Phelan-Dickinson SJ, DeLouise LA. Amorphous silicon dioxide nanoparticles modulate immune responses in a model of allergic contact dermatitis. Sci Rep. 2019;9:5085. https://doi.org/10.1038/s41598-019-41493-7

[41]

Hirai T, Yoshikawa T, Nabeshi H, Yoshida T, Akase T, Yoshioka Y, et al. Dermal absorption of amorphous nanosilica particles after topical exposure for three days. Pharmazie. 2012;67(8):742-3. https://doi.org/10.1691/ph.2012.1853

[42]

Palmer BC, DeLouise LA. Nanoparticle-enabled transdermal drug delivery systems for enhanced dose control and tissue targeting. Molecules. 2016;21(12):1719. https://doi.org/10.3390/molecules21121719

[43]

Yokota J, Kyotani S. Influence of nanoparticle size on the skin penetration, skin retention and anti-inflammatory activity of non-steroidal anti-inflammatory drugs. J Chin Med Assoc. 2018;81(6):511-9. https://doi.org/10.1016/j.jcma.2018.01.008

[44]

Jatana S, Palmer BC, Phelan SJ, DeLouise LA. Immunomodulatory effects of nanoparticles on skin allergy. Sci Rep. 2017;7:3979. https://doi.org/10.1038/s41598-017-03729-2

[45]

Champion JA, Mitragotri S. Shape induced inhibition of phagocytosis of polymer particles. Pharm Res. 2009;26:244-9. https://doi.org/10.1007/s11095-008-9626-z

[46]

Diesner SC, Wang XY, Jensen-Jarolim E, Untersmayr E, Gabor F. Use of lectin-functionalized particles for oral immunotherapy. Ther Deliv. 2012;3(2):277-90. https://doi.org/10.4155/tde.11.146 

[47]

Yoshioka Y, Kuroda E, Hirai T, Tsutsumi Y, Ishii KJ. Allergic responses induced by the immunomodulatory effects of nanomaterials upon skin exposure. Front Immunol. 2017;8:169. https://doi.org/10.3389/fimmu.2017.00169

[48]

Martínez-Pérez B, Quintanar-Guerrero D, Tapia-Tapia M, Cisneros-Tamayo R, Zambrano-Zaragoza ML, Alcalá-Alcalá S, et al. Controlled-release biodegradable nanoparticles: from preparation to vaginal applications. Eur J Pharm Sci. 2018;115:185-95. https://doi.org/10.1016/j.ejps.2017.11.029

[49]

Henriksen-Lacey M, Bramwell VW, Christensen D, Agger EM, Andersen P, Perrie Y. Liposomes based on dimethyldioctadecylammonium promote a depot effect and enhance immunogenicity of soluble antigen. J Control Release. 2010;142(2):180-6. https://doi.org/10.1016/j.jconrel.2009.10.022

[50]

Virkud YV, Wang J, Shreer WG. Enhancing the safety and efficacy of food allergy immunotherapy: a review of adjunctive therapies. Clin Rev Allergy Immunol. 2018;55:172-89. https://doi.org/10.1007/s12016-018-8694-z

[51]

American Academy of Allergy Asthma & Immunology. The current state of oral immunotherapy. 2021 [cited 10 March 2021]. Available from: https://www.aaaai.org/tools-for-the-public/conditions-library/allergies/the-current-state-of-oral-immunotherapy.

[52]

Brotons-Canto A, Gamazo C, Martín-Arbella N, Abdulkarim M, Gumbleton M, Quincoces G, et al. Mannosylated nanoparticles for oral immunotherapy in a murine model of peanut allergy. J Pharm Sci. 2019;108(7):2421-29. https://doi.org/10.1016/j.xphs.2019.02.022

[53]

Srivastava KD, Siefert A, Fahmy TM, Caplan MJ, Li XM, Sampson HA. Investigation of peanut oral immunotherapy using CpG/peanut-nanoparticles in a murine model of peanut allergy. J Allergy Clin Immunol. 2016;138(2):536-43. https://doi.org/10.1016/j.jaci.2016.01.047

[54]

Manolova V, Flace A, Bauer M, Schwarz K, Saudan P, Bachmann MF. Nanoparticles target distinct dendritic cell populations according to their size. Eur J Immunol. 2008;38(5):1404-13. https://doi.org/10.1002/eji.200737984

[55]

Smarr CB, Miller SD. The use of biodegradable nanoparticles for tolerogenic therapy of allergic inflammation. Methods Mol Biol. 2018;1799:353-58. https://doi.org/10.1007/978-1-4939-7896-0_25

[56]

Kishimoto TK, Maldonado RA. Nanoparticles for the induction of antigen-specific immunological tolerance. Front Immunol. 2018;9:230. https://doi.org/10.3389/fimmu.2018.00230 

[57]

Maldonado RA, LaMothe RA, Ferrari JD, Zhang AH, Rossi RJ, Kolte PN, et al. Polymeric synthetic nanoparticles for the induction of antigen-specific immunological tolerance. Proc Natl Acad Sci USA. 2015;112(2):e156-65. https://doi.org/10.1073/pnas.1408686111

[58]

Ballester M, Jeanbart L, De Titta A, Nembrini C, Marsland BJ, Hubbell JA, et al. Nanoparticle conjugation enhances the immunomodulatory effects of intranasally delivered CpG in house dust mite-allergic mice. Sci Rep. 2015;5:14274. https://doi.org/10.1038/srep14274

[59]

Da Silva AL, Santos RS, Xisto DG, Alonso SDV, Morales MM, Rocco PRM. Nanoparticle-based therapy for respiratory diseases. An Acad Bras Cienc. 2013; 85(1):137-46. https://doi.org/10.1590/S0001-37652013005000018

[60]

Kroegel C. Global Initiative for Asthma (GINA) guidelines: 15 years of application. Expert Rev Clin Immunol. 2009;5(3):239-49. https://doi.org/10.1586/eci.09.1

[61]

Matsuo Y, Ishihara T, Ishizaki J, Miyamoto K, Higaki M, Yamashita N. Effect of betamethasone phosphate loaded polymeric nanoparticles on a murine asthma model. Cell Immunol. 2009;260(1):33-8. https://doi.org/10.1016/j.cellimm.2009.07.004

[62]

El-Gendy N, Gorman EM, Munson EJ, Berkland C. Budesonide nanoparticle agglomerates as dry powder aerosols with rapid dissolution. J Pharm Sci. 2009;98(8):2731-46. https://doi.org/10.1002/jps.21630

[63]

Usmani OS, Biddiscombe MF, Barnes PJ. Regional lung deposition and bronchodilator response as a function of beta2-agonist particle size. Am J Respir Crit Care Med. 2005;172(12):1497-1504. https://doi.org/10.1164/rccm.200410-1414oc

[64]

Bhavna, Ahmad FJ, Mittal G, Jain GK, Malhotra G, Khar RK, et al. Nano-salbutamol dry powder inhalation: a new approach for treating broncho-constrictive conditions. Eur J Pharm Biopharm. 2009;71(2):282-91. https://doi.org/10.1016/j.ejpb.2008.09.018

[65]

Kumar M, Kong X, Behera AK, Hellermann GR, Lockey RF, Mohapatra SS. Chitosan IFN-γ-pDNA nanoparticle (CIN) therapy for allergic asthma. Genet Vaccines Ther. 2003;1:3. https://doi.org/10.1186/1479-0556-1-3

[66]

Kong X, Hellermann GR, Zhang W, Jena P, Kumar M, Behera A, et al. Chitosan interferon-γ nanogene therapy for lung disease: modulation of T-cell and dendritic cell immune responses. Allergy Asthma Clin Immunol. 2008;4:95-105. https://doi.org/10.1186/1710-1492-4-3-95