Crimson Publishers Publish With Us Reprints e-Books Video articles

Full Text

Research & Development in Material Science

Metal Nanoparticles in Immunotherapy: Applications, Limitations and Pespectives

Dukhinova M1*, Shestovskaya M1 and Shtil AA1,2

1SCAMT laboratory (Solution Chemistry of Advanced Materials and Technologies), University of Information Technologies, Saint-Petersburg, Russia

2Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia

*Corresponding author: Dukhinova M, SCAMT laboratory (Solution Chemistry of Advanced Materials and Technologies), University of Information Technologies, Mechanics and Optics, Russia

Submission: December 20, 2019;Published: January 09, 2020

DOI: 10.31031/RDMS.2020.12.000793

ISSN: 2576-8840
Volume 12 Issue 4

Abstract

Metal oxide nanoparticles are becoming essential in various biomedical applications, from diagnostics and imaging to anticancer therapy, bacterial infections, wound healing, and others. The application these nanostructures for immunotherapy is also emerging, particularly because they undergo phagocytosis and transmission by macrophages, and exhibit pro- or anti-inflammatory features depending on the nanoparticle’ content and pathological context. However, there is still a number of limitations that have to be addressed before the nanoparticles enter the clinic. First, metal oxide nanoparticle may evoke cytotoxicity in various cell types including immune cells. Second, the immunogenic potential of nanoparticles may affect the immune populations outside the tumours or inflammatory sites. Third, the effects of nanoparticles differ in respect to cell microenvironment and can be hardly predictable and controllable. Nevertheless, most of these limitations can be addressed by combining other approaches such as chemotherapy, antibody-mediated targeted delivery and / or RNA-loaded machines.

Introduction

Metal nanoparticles (NPs) functionalize numerous cell populations critical for combating cancer, bacterial and viral infections, parasites, etc. Essentially, among the target cells that encounter NPs are the immune cells, one of the first responders and transmitters of external signals in the organism. Heterogeneity of the immune cell populations complicate the systemic analysis and NPs adjustment for immunomodulation; also, severe complications should be taken into consideration since the cells can respond unpredictably to the treatment. Nevertheless, the application of NP in medicine is rapidly emerging, therefore the above limitations must be carefully addressed.

Metal nanoparticles and immune cell heterogeneity

Currently, various metal NPs are used for diagnostics and therapy. The metal component can be represented by Ag, Au, Pt, rare earth metals, oxides of iron, nickel, copper, and others. These NPs can interact with the immune cells depending on the delivery root (local or systemic, intravenous or dietary or intranasal) [1,2].

Metal nanoparticles and inborn immunity

Once NPs enter the body, they encounter circulating monocytes or tissue resident macrophages that actively engulf and transfer NPs to different sites. NPs can polarize macrophages towards pro- (M1) or anti-inflammatory (M2) profile with respect to microenvironment and the type of metal [3]. M1 and M2 macrophages release a variety of cytokines which, in turn, recruit other immune cells that suppress (regulatory T cells) or prolong (cytotoxic T cells, T helpers, B lymphocytes) inflammation and the immune reaction triggered by NPs. Simultaneously with macrophages, NPs target neutrophils, the innate immune cells that fight against pathogens via extracellular trap formation and subsequent specific cell death termed netoptosis, for pathogen restriction and killing. A number of metal NPs up-regulate neutrophil extracellular trap activity and stimulate antimicrobial potential [4,5]. In some cases, nanosilver can also cause non-classical activation of neutrophils with IL8 release but not netoptosis, and the effect depends on NP size [6,7].

Metal nanoparticles and adaptive immunity

Following activation of macrophages and neutrophils, metal NPs involve evoke an adaptive immune system response driving the potential of T- and B-lymphocytes [8]. This stage is mediated directly by NPs as well as by the release of pro (TNFa, IL1b, IL6, IL8, chemokines CXCL10, CCL3,7) and anti (IL10, CXCL6) inflammatory cytokines to regulate lymphocyte activity [9,10]. Gold NPs are known to stimulate antibody production by B cells via modulation of Blimp1/Pax5 signaling pathway [11]. Interestingly, aluminum oxide NPs increase neutrophil and monocyte counts together with pro-inflammatory cytokines in systemic circulation; however, lymphocyte activity and antigen presentation are suppressed suggesting complex and indirect effects of NPs [9].

Application of metal NPs, alone or in combination with other approaches, allows to modulate other immune cell populations although these effects are less studied. For example, natural killer cells can be guided with iron NPs and magnetic field to reveal their cytotoxic potential specifically against the tumor [12]. Titanium, silver and gold NPs are known to activate dendritic cells, particularly against viral infections, such as HSV-2 [8,13,14].

Metal nanoparticles within immune microenvironment

Except the direct effect on cells within inflammatory sites, metal NPs can modify immune microenvironment.

Non-specific interactions with extracellular proteins

Due to their chemical and physical properties metal NPs interact with extracellular molecules. In the body NPs form so-called surface corona consisting of extracellular proteins [15]. Circulating NPs collect immunomodulatory proteins including the components of the complement system, hemoglobin, immunoglobulins, as well as cell growth/differentiation factors [16-18]. The corona changes the immunogenic activity of metal NPs but can also cause an immunosuppressive response in target cells [19].

Extracellular matrix

Metal NPs also interact with Extracellular Matrix (ECM). ECM is a non-cellular scaffold presented of all tissues and organs that provides an essential signalling for initiation of biochemical and biomechanical cues for morphogenesis, differentiation and homeostasis of all cells, including immune. Although, fundamentally, ECM is composed of proteins and polysaccharides, each tissue has the unique ECM composition and topology that can be associated with certain pathological events as inflammation or tumor growth and metastasis [20]. ECM variations also lead to heterogeneity of NP effects in various contexts. Importantly, ECM is an indispensable part of biological barriers and modulates NP delivery to the required therapeutic sites. NPs, including metal NPs, are influenced by ECM components via three directions: (i) physical localization of along ECM fibers; (ii) NPs restrict thermal motion of water molecules due to the proximity to the fibers (which also influences hydrodynamic interactions and slow NP diffusion (hydrodynamic interactions), and (iii) charged particles interact with polarized ECM components via electrostatic interactions to stabilize the system [21].

The predominant components of ECM are collagen and laminin produced by fibroblasts. Metal NPs change the cell - matrix interaction. Flow cytometry analysis showed that treatment with metal NPs reduced the percentage of cells expressing the collagen receptor, α2β1 integrin (VLA-2) and the laminin receptor α6β1 integrin (VLA-6). In contrast, NPs can increase and decrease the percentages of VLA-2- and VLA-6-positive cells, respectively. Besides, metal NPs can cause cytoskeletal reorganization, namely, increase the formation of stress fibers and cell protrusions, thereby impairing cell polarity [22].

Matrix metalloproteinases

ECM matrix metalloproteinases (MMPs) are a family of zincdependent neutral endopeptidases. Partially, MMPs are produced and controlled by macrophages and neutrophils [23-25]. MMPs are designated to modulate ECM and are thereby involved in important physiological and pathological events such as wound healing, cancer progression, inflammation, and drug / NP delivery [21]. With that, the immune cells activated by NPs interfere the ECM structure and function, cause tissue remodeling and change the delivery routes of drugs, tumor metastatic potential and CO2/oxygen exchange in tissues [26].

Limitations of nano therapy for immunomodulation

A plethora of biological stimuli induce reactive oxygen and nitrogen species (ROS and RNS) generation by immune cells, particularly macrophages and neutrophils, as ROS production is considered as a part of inborn immune response. ROS/RNS promote inflammation and increase death of pathogens or tumor cells [27-29]. Similarly, NP exposure often leads to ROS delivery from various cells, which, in this case, may also serve as a hallmark of cytotoxicity. Indeed, a number of studies have shown that NPs induce endoplasmic reticulum stress, mitochondrial damage and cell death in macrophages, neutrophils and lymphocytes [30-35]. This suggests that metal NPs may suppress immune cell function, and this factor is difficult to detect and control. On the contrary, in some cases metal NPs can induce strong inflammation and be harmful for normal tissues [36,37]. This may arise from systemic effects of NPs, e.g., i.v. delivery and accumulation in non-specific sites [38].

Combination approaches to improve efficiency of metal NPs

Considering the diversity of immune cell populations and the complexity of NP-mediated effects, it is recommended to use NPs in combination with other therapeutic approaches keeping in mind biological, physical or chemical properties of metal NPs [39]. This will provide the specificity of action and allow to modulate immunity in a more precise mode. Metal NPs can be combined with antigens or DNA vaccines for targeted delivery to the pathological site or specific cancer cell/pathogen elimination [13,39,40]. Zinc, aluminum, gold NPs act as adjuvants for therapy and result in increased number of antigen-specific T cells and more efficient antibody production, so that the immune response can be driven against tumor or bacteria without any self-toxicity [41-45] antigen.

In allergic conditions iron oxide NPs can eliminate the reactive T lymphocytes [46,47]. Iron magnetic NPs combined with antigens can be used for control delivery of immune cells to the damage/ infection site or any other specific location by external magnetic field [48,12]. Magnetic and antigen-specific nanoparticles allow to isolate specific lymphocyte clones in a fast and reproducible way suitable for experimental and therapeutic applications, such as delivery tumor antigen engineered T cells to the patient [14,49].

For immunomodulatory purposes metal NPs can be used together with pro- or anti-inflammatory cytokines that can guide macrophage polarization and T lymphocyte differentiation towards the cytotoxic or regulatory profiles. For modulation of macrophage polarization metal NPs loaded with pro- or anti-inflammatory cytokines can be used [50]. Metal NPs can be modified with other nanostructures such as hyaluronic or tannic acids for modulation of the immune cell survival and activity within the tumor [51,52]. Finally, it is possible to use the combinations of metals to achieve the controllable systemic immune activity at different stages of the disease. For example, titanium stimulates pro-inflammatory activity of macrophages that can be used against bacterial infection in the wound, while lithium exhibits an opposite effect attenuating inflammation and can be used to accelerate regeneration process [53].

Conclusion and Future Directions

Biomedical and therapeutic application of metal NPs in immunotherapy remain a challenge due to (i) risk of cytotoxicity and (ii) non-specific activation of immune subpopulations and redundant inflammatory outbreak. However, all these limitations can be minimized or even avoided if NPs are used together with other medical strategies. Overall, metal NPs represent a promising tool for controllable immunomodulation.

Acknowledgment

This work was supported by Russian Foundation for Basic Research grant N. 19-315-60012.

References

  1. Roach KA, Stefaniak AB, Roberts JR (2019) Metal nanomaterials: Immune effects and implications of physicochemical properties on sensitization, elicitation, and exacerbation of allergic disease. J Immunotoxicol 16: 87-124.
  2. Park EJ, Oh SY, Kim Y, Yoon C, Lee BS, et al. (2016) Distribution and immunotoxicity by intravenous injection of iron nanoparticles in a murine model. J Appl Toxicol 36(3): 414-423.
  3. Lategan K, Walters C, Pool E (2019) The effects of silver nanoparticles on RAW 264.7. Macrophages and human whole blood cell cultures. Front Biosci (Landmark Ed) 24: 347-365.
  4. Wang C, Liu X, Han Z, Zhang X, Wang J, et al. (2019) Nanosilver induces the formation of neutrophil extracellular traps in mouse neutrophil granulocytes. Ecotoxicol Environ Saf 183: 109508.
  5. Bilyy R, Unterweger H, Weigel B, Dumych T, Paryzhak S, et al. (2018) Inert coats of magnetic nanoparticles prevent formation of occlusive intravascular co-aggregates with neutrophil extracellular traps. Front Immunol 9: 2266.
  6. Poirier M, Simard J, Girard D (2016) Silver nanoparticles of 70nm and 20nm affect differently the biology of human neutrophils. J Immunotoxicol 13(3): 375-3
  7. Fraser JA, Kemp S, Young L, Ross M, Prach M, et al. (2018) Silver nanoparticles promote the emergence of heterogeneic human neutrophil sub-populations. Sci Rep 8(1): 7506.
  8. Schanen BC, Das S, Reilly CM, Warren WL, Self WT, et al. (2013) Immunomodulation and T helper TH₁/TH₂ response polarization by CeO₂ and TiO₂ PLoS One 8(5): e62816-e62816.
  9. Park E, Kim S, Kang M, Lee B, Yoon C, et al. (2016) A higher aspect ratio enhanced bioaccumulation and altered immune responses due to intravenously-injected aluminum oxide nanoparticles. J Immunotoxicol 13: 439-448.
  10. Park EJ, Kim SW, Yoon C, Kim Y, Kim JS (2016) Disturbance of ion environment and immune regulation following biodistribution of magnetic iron oxide nanoparticles injected intravenously. Toxicol Lett 243: 67-77.
  11. Lee CH, Syu SH, Chen YS, Hussain SM, Aleksandrovich OA, et al. (2014) Gold nanoparticles regulate the blimp1/pax5 pathway and enhance antibody secretion in B-cells. Nanotechnology 25(12): 125103.
  12. Burga RA, Khan DH, Agrawal N, Bollard CM, Fernandes R (2019) Designing magnetically responsive biohybrids composed of cord blood-derived natural killer cells and iron oxide nanoparticles. Bioconjug Chem 30(3): 552-560.
  13. Gulla SK, Rao BR, Moku G, Jinka S, Nimmu NV, et al. (2019) In vivo targeting of DNA vaccines to dendritic cells using functionalized gold nanoparticles. Biomater Sci 7(3): 773-788.
  14. Orlowski P, Tomaszewska E, Ranoszek-Soliwoda K, Gniadek M, Labedz O, et al. (2018) Tannic acid-modified silver and gold nanoparticles as novel stimulators of dendritic cells activation. Front Immunol 9: 1115.
  15. Gan N, Sun Q, Zhao L, Tang P, Suo Z, et al. (2019) Protein corona of metal-organic framework nanoparticals: Study on the adsorption behavior of protein and cell interaction. Int J Biol Macromol 140: 709-718.
  16. Zhu Y, Jiang P, Luo B, Lan F, He J, et al. (2019) Dynamic protein corona influences immune-modulating osteogenesis in magnetic nanoparticle (MNP)-infiltrated bone regeneration scaffolds in vivo. Nanoscale 11: 6817-6827.
  17. Del Pilar Chantada-Vázquez M, López AC, Bravo SB, Vázquez-Estévez S, Acea-Nebril B, et al. (2019) Proteomic analysis of the bio-corona formed on the surface of (Au, Ag, Pt)-nanoparticles in human serum. Colloids Surfaces B Biointerfaces 177: 141-148.
  18. Mirzaei S, Hadadi Z, Attar F, Mousavi S, Zargar S, et al. (2018) ROS-mediated heme degradation and cytotoxicity induced by iron nanoparticles: hemoglobin and lymphocyte cells as targets. J Biomol Struct Dyn 36: 4235-4245.
  19. Persaud I, Shannahan JH, Raghavendra AJ, Alsaleh NB, Podila R, et al. (2019) Biocorona formation contributes to silver nanoparticle induced endoplasmic reticulum stress. Ecotoxicol Environ Saf 170: 77-86.
  20. Frantz C, Stewart KM, Weaver VM (2010) The extracellular matrix at a glance. Journal of Cell Science 123: 4195-4200.
  21. Engin AB, Nikitovic D, Neagu M, Henrich-Noack P, Doceaet AO, et al. (2017) Mechanistic understanding of nanoparticles’ interactions with extracellular matrix: The cell and immune system. Particle and Fibre Toxicology 14(22).
  22. Vieira, Lins MP, Viana IMMN, dos Santos JE, Smaniotto S, et al. (2017) Metallic nanoparticles reduce the migration of human fibroblasts in vitro. Nanoscale Res Lett 12: 200.
  23. Yu-Ju Wu C, Chen CH, Lin CY, Feng LY, Lin YC, et al. (2019) CCL5 of glioma-associated microglia/macrophages regulates glioma migration and invasion via calcium-dependent matrix metalloproteinase-2. Neuro Oncol.
  24. Wang H, Gao M, Li J, Sun J, Wu R, et al. (2019) MMP-9-positive neutrophils are essential for establishing profibrotic microenvironment in the obstructed kidney of UUO mice. Acta Physiol 227(2): 313-317.
  25. Sakhno LV, Shevela EY, Lykov AP, Poveshchenko OV, Ostanin AA, et al. (2019) Effect of apoptotic neutrophils on the production of erythropoietin, MMP-9, and TIMP-1 in cultures of human macrophages. Bull Exp Biol Med 167: 755-758.
  26. Wan R, Mo Y, Zhang X, Chien S, Tollerud DJ, et al. (2008) Matrix metalloproteinase-2 and -9 are induced differently by metal nanoparticles in human monocytes: The role of oxidative stress and protein tyrosine kinase activation. Toxicol Appl Pharmacol 233: 276-285.
  27. Gan J, Liu C, Li H, Wang S, Wang Z, et al. (2019) Accelerated wound healing in diabetes by reprogramming the macrophages with particle-induced clustering of the mannose receptors. Biomaterials 219: 119340.
  28. Gambhir L, Sharma V, Kandwal P, Saxena S (2019) Perturbation in cellular redox homeostasis: Decisive regulator of T cell mediated immune responses. Int Immunopharmacol 67: 449-457.
  29. Paardekooper LM, Dingjan I, Linders PTA, Staal AHJ, Cristescu SM, et al. (2019) Human monocyte-derived dendritic cells produce millimolar concentrations of ros in phagosomes per second. Front Immunol 10: 1216.
  30. Simón-Vázquez R, Lozano-Fernández T, Dávila-Grana A, González-Fernández A (2016) Metal oxide nanoparticles interact with immune cells and activate different cellular responses. Int J Nanomedicine 11: 4657-4668.
  31. Noël C, Simard JC, Girard D (2016) Gold nanoparticles induce apoptosis, endoplasmic reticulum stress events and cleavage of cytoskeletal proteins in human neutrophils. Toxicol Vitr 31: 12-22.
  32. Soares T, Ribeiro D, Proença C, Chisté RC, Fernandes E, Freitas M (2016) Size-dependent cytotoxicity of silver nanoparticles in human neutrophils assessed by multiple analytical approaches. Life Sci 145: 247-254.
  33. Shah A, Mankus CI, Vermilya AM, Soheilian F, Clogston JD, et al. (2018) Feraheme® suppresses immune function of human T lymphocytes through mitochondrial damage and mitoROS production. Toxicol Appl Pharmacol 350: 52-63.
  34. Devanabanda M, Latheef S, Madduri R (2016) Immunotoxic effects of gold and silver nanoparticles: Inhibition of mitogen-induced proliferative responses and viability of human and murine lymphocytes in vitro. J Immunotoxicol 13: 897-902.
  35. Chang X, Zhu A, Liu F, Zou L, Su L, et al. (2017) Role of NF-κB activation and Th1/Th2 imbalance in pulmonary toxicity induced by nanoNiO. Environ Toxicol 32: 1354-1362.
  36. Watson CY, Molina RM, Louzada A, Murdaugh KM, Donaghey TC, et al. (2015) Effects of zinc oxide nanoparticles on Kupffer cell phagosomal motility, bacterial clearance, and liver function. Int J Nanomedicine 10: 4173-4184.
  37. Xu J, Yang J, Nyga A, Ehteramyan M, Moraga, et al. (2018) Cobalt (II) ions and nanoparticles induce macrophage retention by ROS-mediated down-regulation of RhoA expression. Acta Biomater. 72: 434-446.
  38. Han Y, Lee D, Kim S, Lee S, Jeon S, et al. (2018) High inflammogenic potential of rare earth oxide nanoparticles: the New Hazardous Entity. Nanotoxicology 12: 712-728.
  39. Yang Y, Chen Q, Wu JP, Kirk TB, Xu J, et al. (2018) Reduction-responsive codelivery system based on a metal-organic framework for eliciting potent cellular immune response. ACS Appl Mater Interfaces 10: 12463-12473.
  40. Wang T, Zhang H, Han Y, Liu H, Ren F, et al. (2019) Light-enhanced O2-evolving nanoparticles boost photodynamic therapy to elicit antitumor immunity. ACS Appl Mater Interfaces 11(18): 16367-16379.
  41. Sun B, Ji Z, Liao YP, Wang M, Wang X, et al. (2013) Engineering an effective immune adjuvant by designed control of shape and crystallinity of aluminum oxyhydroxide nanoparticles. ACS Nano 7: 10834-10849.
  42. Dakterzada F, Mohabati Mobarez A, Habibi Roudkenar M, Mohsenifar A (2016) Induction of humoral immune response against Pseudomonas aeruginosa flagellin(1-161) using gold nanoparticles as an adjuvant. Vaccine 34: 1472-1479.
  43. Roy R, Kumar S, Verma AK, Sharma A, Chaudhari BP, et al. (2014) Zinc oxide nanoparticles provide an adjuvant effect to ovalbumin via a Th2 response in Balb/c mice. Int Immunol 26: 159-172.
  44. Zhong X, Zhang Y, Tan L, Zheng T, Hou Y, et al. (2019) An aluminum adjuvant-integrated nano-MOF as antigen delivery system to induce strong humoral and cellular immune responses. J Contriol Release 300: 81-92.
  45. Makowski M, Silva ÍC, Do Amaral CP, Gonçalves S, Santos NC (2019) Advances in lipid and metal nanoparticles for antimicrobial peptide delivery. Pharmaceutics: 11(11): 588.
  46. Ban M Langonné I, Huguet N, Guichard Y, Goutet M (2013) Iron oxide particles modulate the ovalbumin-induced Th2 immune response in mice. Toxicol Lett 216(1): 31-39.
  47. Dul M, Nikolic T, Stefanidou M, McAteer M, Williams P (2019) Conjugation of a peptide autoantigen to gold nanoparticles for intradermally administered antigen specific immunotherapy. Int J Pharm 562: 303-312.
  48. Sanz-Ortega L, Rojas JM, Marcos A, Portilla Y, Stein JV, et al. (2019) T cells loaded with magnetic nanoparticles are retained in peripheral lymph nodes by the application of a magnetic field. J Nanobiotechnology 17(1): 14.
  49. Li CX, Zhang Y, Dong X, Zhang L, Liu MD, et al. (2019) Artificially reprogrammed macrophages as tumor-tropic immunosuppression-resistant biologics to realize therapeutics production and immune activation. Adv Mater 31(15): 1805867.
  50. Dukhinova MS, Prilepskii AY, Shtil AA, Vinogradov VV (2019) Metal oxide nanoparticles in therapeutic regulation of macrophage functions. Nanomaterials 9(11): 1631.
  51. Inturi S, Wang G, Chen F, Banda NK, Holers VM (2015) Modulatory role of surface coating of superparamagnetic iron oxide nanoworms in complement opsonization and leukocyte uptake. ACS Nano 9: 10758-10768.
  52. Hickey JW, Schneck JP (2018) Enrich and expand rare antigen-specific T cells with magnetic nanoparticles. J Vis Exp (141): e58640.
  53. Yang C, Wang W, Zhu K, Liu W, Luo Y, et al. (2019) Lithium chloride with immunomodulatory function for regulating titanium nanoparticle-stimulated inflammatory response and accelerating osteogenesis through suppression of MAPK signaling pathway. Int J Nanomedicine 14: 7475-7488.

© 2019 Dukhinova M. This is an open access article distributed under the terms of the Creative Commons Attribution License , which permits unrestricted use, distribution, and build upon your work non-commercially.