Please wait a minute...
Frontiers of Chemical Science and Engineering

ISSN 2095-0179

ISSN 2095-0187(Online)

CN 11-5981/TQ

Postal Subscription Code 80-969

2018 Impact Factor: 2.809

Front. Chem. Sci. Eng.    2021, Vol. 15 Issue (5) : 1296-1311    https://doi.org/10.1007/s11705-020-2034-6
RESEARCH ARTICLE
Radial porous SiO2 nanoflowers potentiate the effect of antigen/adjuvant in antitumor immunotherapy
Chuangnian Zhang1, Ying Dong2, Jing Gao2, Xiaoli Wang1(), Yanjun Jiang2()
1. Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
2. School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
 Download: PDF(5369 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Here, we reported a cancer nanovaccine based on SiO2 nanoflowers with a special radial pore structure, which greatly enhanced cross-presentation and induced the production of cytotoxic T lymphocyte cells secreting granzymes B and interferon-γ. The antigen ovalbumin was covalently conjugated onto the as-synthesized hierarchical SiO2 nanoflowers, and the adjuvant cytosine-phosphate-guanine was electrostatically adsorbed into their radial pore by simple mixing before use. The nanovaccine exhibited excellent storage stability without antigen release after 27 days of incubation, negligible cytotoxicity to dendritic cells, and a high antigen loading capacity of 430 ± 66 mg·g−1 support. Besides, the nanovaccine could be internalized by dendritic cells via multiple pathways. And the enhancement of antigen/adjuvant uptake and lysosome escape of antigen were observed. Noteworthy, in vitro culture of bone marrow-derived dendritic cells in the presence of nanovaccine proved the activation of dendritic cells and antigen cross-presentation as well as secretion of proinflammatory cytokines. Besides, in vivo study verified the targeting of nanovaccine to draining lymph nodes, the complete suppression of tumor in six out of ten mice, and the triggering of notable tumor growth delay. Overall, the present results indicated that the nanovaccine can be served as a potential therapeutic vaccine to treat cancer.

Keywords silica nanoflower      antigen delivery      cancer immunotherapy      nanovaccine     
Corresponding Author(s): Xiaoli Wang,Yanjun Jiang   
Just Accepted Date: 03 March 2021   Online First Date: 12 April 2021    Issue Date: 30 August 2021
 Cite this article:   
Chuangnian Zhang,Ying Dong,Jing Gao, et al. Radial porous SiO2 nanoflowers potentiate the effect of antigen/adjuvant in antitumor immunotherapy[J]. Front. Chem. Sci. Eng., 2021, 15(5): 1296-1311.
 URL:  
https://academic.hep.com.cn/fcse/EN/10.1007/s11705-020-2034-6
https://academic.hep.com.cn/fcse/EN/Y2021/V15/I5/1296
Fig.1  (a) Schematic diagram for the synthetic process of the nanovaccine (OVA@fSiO2 + CpG); (b) SEM image; (c) TEM image; (d) the nitrogen adsorption-desorption isotherm; (e) the pore diameter distribution curve of fSiO2-NH2; (f) the effect of glutaraldehyde concentration on the OVA loading capacity in OVA@fSiO2; (g) the surface zeta potential of fSiO2 prior and after amination, activation and covalently conjugated with OVA; (h) dynamic light scattering size distribution of fSiO2-NH2 and OVA@fSiO2; (i) Fourier transform infrared spectrum of fSiO2-OH, fSiO2-NH2, fSiO2-CHO and OVA@fSiO2; (j–l) the stabilities of OVA@fSiO2 in different pH solution.
Fig.2  (a) The cell viability of DC2.4 and RAW264.7 after incubating these cells for 48 h with OVA@fSiO2, fresh culture medium was served as a negative control; (b) the internalization pathways of nanovaccine by DCs and the corresponding inhibitors; the uptake of (c, e) OVA and (d, f) CpG estimated by the MFI of DC2.4 cells after incubating the cells with OVA@fSiO2 + CpG in the presence of various inhibitors, and their representative FACS histograms (Data were expressed as the mean±SD of five individual experiments, and the differences between groups were determined by one-way ANOVA followed by Tukey’s multiple comparison test, *P<0.05, **P<0.01, ***P<0.001).
Fig.3  (a) Images observed by CLSM after incubating BMDCs with OVA+ CpG or OVA@fSiO2 + CpG for 24 h, the endo/lysosomes membrane was marked with Lamp-1-APC; (b) the colocalization analysis was conducted by Zeiss Zen 2008 Light Edition.
Fig.4  The expression of costimulatory markers: (a) CD40; (b) CD80; (c) CD86; (d) MHC II; (e) MHC I; (f) SIINFEKL-MHC I on pulsed BMDCs; (g) the cross-presentation of antigen OVA was appraised by the activity of b-galactosidase produced by B3Z cells using a colorimetric LacZ assay (OD405 nm); (h) the expression of CCR7 on pulsed BMDCs (Data were represented the mean±SD of three individual experiments, the differences between groups were determined by one-way ANOVA followed by Tukey’s multiple comparison test, ** P<0.01, *** P<0.001).
Fig.5  The released cytokine levels (IL-12, IL-1b, IL-6, TNF-a, IL-4 and IL-10) from BMDCs pulsed with PBS, OVA+ CpG or OVA@fSiO2 + CpG (Data were represented the mean±SD of three individual experiments, the differences between groups were determined by one-way ANOVA followed by Tukey’s multiple comparison test, *** P<0.001).
Fig.6  The fluorescent signal of Cy7 at the administration site and left lymph nodes of mice vaccinated by OVA-Cy7 or OVA-Cy7@fSiO2, lymph nodes indicated by red arrows.
Fig.7  (a) The images of fresh tumor tissue excised from each treated group at day 25; (b–d) the tumor growth curves of each mouse in each experimental group; (e) the average tumor volumes at the 25th day; (f) the average tumor weight of mice at the 25th day; (g) the body weight of mice over time in each experimental group (Data were shown as the mean±s. e. m of ten individual experiments and analyzed by unpaired student’s t-test, *P<0.05).
Fig.8  The percentages of (a) Ki67+, (b) CD44+CD62+ and (c) CD44+CD62 among CD8+ T cells in mouse splenocyte; (d) the percentages of CD3+CD8+Granzyme B+ CTLs in mouse splenocyte; (e) IFN-g level of splenocyte supernatant after three days of antigen restimulation; (f) the serum IFN-g level tested by ELISA assay; (g–i) IL-2, IL-4 and IL-10 levels of splenocyte supernatant after three days of antigen restimulation; OVA-specific total (j) IgG titer, (k) IgG1 titer, and (l) IgG2a titer on day 25 were measured by ELISA assay (The data were shown as the means±s. e. m (n = 10) and analyzed using one-way ANOVA followed by Newman-Keuls multiple comparison test. *P<0.05, **P<0.01, ***P<0.001).
1 P N Kelly. The cancer immunotherapy revolution. Science, 2018, 359(6382): 1344–1345
https://doi.org/10.1126/science.359.6382.1344
2 U Sahin, Ö Türeci. Personalized vaccines for cancer immunotherapy. Science, 2018, 359(6382): 1355–1360
https://doi.org/10.1126/science.aar7112
3 J Nam, S Son, K S Park, W Zou, L D Shea, J J Moon. Cancer nanomedicine for combination cancer immunotherapy. Nature Reviews. Materials, 2019, 4(6): 398–414
https://doi.org/10.1038/s41578-019-0108-1
4 R E Hollingsworth, K Jansen. Turning the corner on therapeutic cancer vaccines. NPJ Vaccines, 2019, 4(1): 7
https://doi.org/10.1038/s41541-019-0103-y
5 S K Wculek, F J Cueto, A M Mujal, I Melero, M F Krummel, D Sancho. Dendritic cells in cancer immunology and immunotherapy. Nature Reviews. Immunology, 2020, 20(1): 1–18
https://doi.org/10.1038/s41577-019-0210-z
6 Z Hu, P A Ott, C J Wu. Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nature Reviews. Immunology, 2017, 18(3): 168–182
https://doi.org/10.1038/nri.2017.131
7 R S Riley, C H June, R Langer, M J Mitchell. Delivery technologies for cancer immunotherapy. Nature Reviews. Drug Discovery, 2019, 18(3): 175–196
https://doi.org/10.1038/s41573-018-0006-z
8 P Huang, X Wang, X Liang, J Yang, C Zhang, D Kong, W Wang. Nano-, micro-, and macroscale drug delivery systems for cancer immunotherapy. Acta Biomaterialia, 2019, 85: 1–26
https://doi.org/10.1016/j.actbio.2018.12.028
9 J M Newton, A G Sikora, S Young. Chapter 41—Materials-Based Cancer Immunotherapies: Principles of Regenerative Medicine. 3rd ed. Boston: Academic Press, 2019, 715–739
10 K T Gause, A K Wheatley, J Cui, Y Yan, S J Kent, F Caruso. Immunological principles guiding the rational design of particles for vaccine delivery. ACS Nano, 2017, 11(1): 54–68
https://doi.org/10.1021/acsnano.6b07343
11 L Gu. Tailored silica nanomaterials for immunotherapy. ACS Central Science, 2018, 4(5): 527–529
https://doi.org/10.1021/acscentsci.8b00181
12 V Giglio, S Varelaaramburu, L Travaglini, F Fiorini, P H Seeberger, L Maggini, L de Cola. Reshaping silica particles: mesoporous nanodiscs for bimodal delivery and improved cellular uptake. Chemical Engineering Journal, 2018, 340: 148–154
https://doi.org/10.1016/j.cej.2018.01.059
13 Y Yang, Y Lu, P L Abbaraju, J Zhang, M Zhang, G Xiang, C Yu. Multi-shelled dendritic mesoporous organosilica hollow spheres: roles of composition and architecture in cancer immunotherapy. Angewandte Chemie International Edition, 2017, 56(29): 8446–8450
https://doi.org/10.1002/anie.201701550
14 P L Abbaraju, A K Meka, H Song, Y Yang, M Jambhrunkar, J Zhang, C Xu, M Yu, C Yu. Asymmetric silica nanoparticles with tunable headtail structures enhance hemocompatibility and maturation of immune cells. Journal of the American Chemical Society, 2017, 139(18): 6321–6328
https://doi.org/10.1021/jacs.6b12622
15 X Wang, X Li, A Ito, Y Watanabe, Y Sogo, N M Tsuji, T Ohno. Stimulation of in vivo antitumor immunity with hollow mesoporous silica nanospheres. Angewandte Chemie International Edition, 2016, 55(5): 1899–1903
https://doi.org/10.1002/anie.201506179
16 X Li, X Wang, Y Sogo, T Ohno, K Onuma, A Ito. Mesoporous silicacalcium phosphate-tuberculin purified protein derivative composites as an effective adjuvant for cancer immunotherapy. Advanced Healthcare Materials, 2013, 2(6): 863–871
https://doi.org/10.1002/adhm.201200149
17 W A Li, B Y Lu, L Gu, Y Choi, J Kim, D J Mooney. The effect of surface modification of mesoporous silica micro-rod scaffold on immune cell activation and infiltration. Biomaterials, 2016, 83: 249–256
https://doi.org/10.1016/j.biomaterials.2016.01.026
18 X Wang, X Li, K Yoshiyuki, Y Watanabe, Y Sogo, T Ohno, N M Tsuji, A Ito. Comprehensive mechanism analysis of mesoporous-silica-nanoparticle-induced cancer immunotherapy. Advanced Healthcare Materials, 2016, 5(10): 1169–1176
https://doi.org/10.1002/adhm.201501013
19 M An, M Li, J Xi, H Liu. Silica nanoparticle as a lymph node targeting platform for vaccine delivery. ACS Applied Materials & Interfaces, 2017, 9(28): 23466–23475
https://doi.org/10.1021/acsami.7b06024
20 B G Cha, J H Jeong, J Kim. Extra-large pore mesoporous silica nanoparticles enabling co-delivery of high amounts of protein antigen and toll-like receptor 9 agonist for enhanced cancer vaccine efficacy. ACS Central Science, 2018, 4(4): 484–492
https://doi.org/10.1021/acscentsci.8b00035
21 D S Moon, J K Lee. Tunable synthesis of hierarchical mesoporous silica nanoparticles with radial wrinkle structure. Langmuir, 2012, 28(33): 12341–12347
https://doi.org/10.1021/la302145j
22 X Du, S Qiao. Dendritic silica particles with center-radial pore channels: promising platforms for catalysis and biomedical applications. Small, 2015, 11(4): 392–413
https://doi.org/10.1002/smll.201401201
23 Z Shen, Y Li, H Wen, X Ren, J Liu, L Yang. Investigation on the role of surfactants in bubble-algae interaction in flotation harvesting of Chlorella vulgaris. Scientific Reports, 2018, 8(1): 3303
https://doi.org/10.1038/s41598-018-21629-x
24 R Kuai, L J Ochyl, K S Bahjat, A Schwendeman, J J Moon. Designer vaccine nanodiscs for personalized cancer immunotherapy. Nature Materials, 2016, 16(4): 489–496
https://doi.org/10.1038/nmat4822
25 W S Bowen, A K Svrivastava, L Batra, H Barsoumian, H Shirwan. Current challenges for cancer vaccine adjuvant development. Expert Review of Vaccines, 2018, 17(3): 207–215
https://doi.org/10.1080/14760584.2018.1434000
26 V U Warrier, A I Makandar, M Garg, G Sethi, R Kant, J K Pal, E Yuba, R K Gupta. Engineering anti-cancer nanovaccine based on antigen cross-presentation. Bioscience Reports, 2019, 39(10): BSR20193220
https://doi.org/10.1042/BSR20193220
27 C Casaravilla, Á Pittini, D Rückerl, P I Seoane, S J Jenkins, A S MacDonald, A M Ferreira, J E Allen, Á Díaz. Unconventional maturation of dendritic cells induced by particles from the laminated layer of larval Echinococcus granulosus. Infection and Immunity, 2014, 82(8): 3164–3176
https://doi.org/10.1128/IAI.01959-14
28 S Yoshida, I Gaeta, R Pacitto, L Krienke, O Alge, B Gregorka, J A Swanson. Differential signaling during macropinocytosis in response to M-CSF and PMA in macrophages. Frontiers in Physiology, 2015, 6: 8
https://doi.org/10.3389/fphys.2015.00008
29 H Kasai, K Inoue, K Imamura, C Yuvienco, J K Montclare, S Yamano. Efficient siRNA delivery and gene silencing using a lipopolypeptide hybrid vector mediated by a caveolae-mediated and temperature-dependent endocytic pathway. Journal of Nanobiotechnology, 2019, 17(1): 11
https://doi.org/10.1186/s12951-019-0444-8
30 A El-Sayed, H Harashima. Endocytosis of gene delivery vectors: from clathrin-dependent to lipid raft-mediated endocytosis. Molecular Therapy, 2013, 21(6): 1118–1130
https://doi.org/10.1038/mt.2013.54
31 K Kubiak Ossowska, B Jachimska, M Al Qaraghuli, P A Mulheran. Protein interactions with negatively charged inorganic surfaces. Current Opinion in Colloid & Interface Science, 2019, 41: 104–117
https://doi.org/10.1016/j.cocis.2019.02.001
32 I Slowing, B G Trewyn, V S Y Lin. Effect of surface functionalization of MCM-41-type mesoporous silica nanoparticles on the endocytosis by human cancer cells. Journal of the American Chemical Society, 2006, 128(46): 14792–14793
https://doi.org/10.1021/ja0645943
33 E Latz, A Schoenemeyer, A Visintin, K A Fitzgerald, B G Monks, C F Knetter, E Lien, N J Nilsen, T Espevik, D T Golenbock. TLR9 signals after translocating from the ER to CpG DNA in the lysosome. Nature Immunology, 2004, 5(2): 190–198
https://doi.org/10.1038/ni1028
34 G M Hjortø, O Larsen, A Steen, V Daugvilaite, C Berg, S Fares, M Hansen, S Ali, M M Rosenkilde. Differential CCR7 targeting in dendritic cells by three naturally occurring CC-chemokines. Frontiers in Immunology, 2016, 7: 568
https://doi.org/10.3389/fimmu.2016.00568
35 C Gerlach, E A Moseman, S M Loughhead, D Alvarez, A J Zwijnenburg, L Waanders, R Garg, J C de la Torre, U H von Andrian. The chemokine receptor CX3CR1 defines three antigen-experienced CD8+ T cell subsets with distinct roles in immune surveillance and homeostasis. Immunity, 2016, 45(6): 1270–1284
https://doi.org/10.1016/j.immuni.2016.10.018
36 M B Lutz, G Schuler. Immature, semi-mature and fully mature dendritic cells: Which signals induce tolerance or immunity? Trends in Immunology, 2002, 23(9): 445–449
https://doi.org/10.1016/S1471-4906(02)02281-0
37 S Tugues, S H Burkhard, I Ohs, M Vrohlings, K Nussbaum, J Vom Berg, P Kulig, B Becher. New insights into IL-12-mediated tumor suppression. Cell Death and Differentiation, 2015, 22(2): 237–246
https://doi.org/10.1038/cdd.2014.134
38 G Kaplanski, V Marin, F Montero Julian, A Mantovani, C Farnarier. IL-6: a regulator of the transition from neutrophil to monocyte recruitment during inflammation. Trends in Immunology, 2003, 24(1): 25–29
https://doi.org/10.1016/S1471-4906(02)00013-3
39 M Sixt, N Kanazawa, M Selg, T Samson, G Roos, D P Reinhardt, R Pabst, M B Lutz, L Sorokin. The conduit system transports soluble antigens from the afferent lymph to resident dendritic cells in the T cell area of the lymph node. Immunity, 2005, 22(1): 19–29
https://doi.org/10.1016/j.immuni.2004.11.013
40 F Sallusto, J Geginat, A Lanzavecchia. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annual Review of Immunology, 2004, 22(1): 745–763
https://doi.org/10.1146/annurev.immunol.22.012703.104702
41 F Nimmerjahn, P Bruhns, K Horiuchi, J V Ravetch. FcgRIV: a novel FcR with distinct IgG subclass specificity. Immunity, 2005, 23(1): 41–51
https://doi.org/10.1016/j.immuni.2005.05.010
[1] FCE-20092-OF-ZC_suppl_1 Download
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed