DOI QR코드

DOI QR Code

New opportunities for nanoparticles in cancer immunotherapy

  • Park, Wooram (Department of Biomedical Science, CHA University) ;
  • Heo, Young-Jae (Department of Biomedical Science, CHA University) ;
  • Han, Dong Keun (Department of Biomedical Science, CHA University)
  • 투고 : 2018.05.08
  • 심사 : 2018.08.24
  • 발행 : 2018.12.31

초록

Background: Recently, cancer immunotherapy has become standard for cancer treatment. Immunotherapy not only treats primary tumors, but also prevents metastasis and recurrence, representing a major advantage over conventional cancer treatments. However, existing cancer immunotherapies have limited clinical benefits because cancer antigens are often not effectively delivered to immune cells. Furthermore, unlike lymphoma, solid tumors evade anti-cancer immunity by forming an immune-suppressive tumor microenvironment (TME). One approach for overcoming these limitations of cancer immunotherapy involves nanoparticles based on biomaterials. Main body: Here, we review in detail recent trends in the use of nanoparticles in cancer immunotherapy. First, to illustrate the unmet needs for nanoparticles in this field, we describe the mechanisms underlying cancer immunotherapy. We then explain the role of nanoparticles in the delivery of cancer antigens and adjuvants. Next, we discuss how nanoparticles can be helpful within the immune-suppressive TME. Finally, we summarize current and future uses of nanoparticles with image-guided interventional techniques in cancer immunotherapy. Conclusion: Recently developed approaches for using nanoparticles in cancer immunotherapy have enormous potential for improving cancer treatment. Cancer immunotherapy based on nanoparticles is anticipated not only to overcome the limitations of existing immunotherapy, but also to generate synergistic effects via cooperation between nanoparticles and immune cells.

키워드

과제정보

연구 과제 주관 기관 : National Research Foundation of Korea (NRF)

참고문헌

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7-30. https://doi.org/10.3322/caac.21442
  2. Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J Clin Oncol. 2015;33(17):1974-82. https://doi.org/10.1200/JCO.2014.59.4358
  3. Phan GQ, et al. Cancer regression and autoimmunity induced by cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma. Proc Natl Acad Sci. 2003;100(14):8372-7. https://doi.org/10.1073/pnas.1533209100
  4. Hodi FS, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711-23. https://doi.org/10.1056/NEJMoa1003466
  5. Kroemer G, Zitvogel L. Cancer immunotherapy in 2017: the breakthrough of the microbiota. Nat Rev Immunol. 2018;18(2):87-8. https://doi.org/10.1038/nri.2018.4
  6. Byun DJ, et al. Cancer immunotherapy-immune checkpoint blockade and associated endocrinopathies. Nat Rev Endocrinol. 2017;13(4):195. https://doi.org/10.1038/nrendo.2016.205
  7. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015;27(4):450-61. https://doi.org/10.1016/j.ccell.2015.03.001
  8. Rosenberg SA, et al. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer. 2008;8(4):299-308. https://doi.org/10.1038/nrc2355
  9. Melero I, et al. T-cell and NK-cell infiltration into solid tumors: a key limiting factor for efficacious cancer immunotherapy. Cancer Discov. 2014;4(5):522-6. https://doi.org/10.1158/2159-8290.CD-13-0985
  10. Zou W. Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat Rev Cancer. 2005;5(4):263-74. https://doi.org/10.1038/nrc1586
  11. Chen F, et al. New horizons in tumor microenvironment biology: challenges and opportunities. BMC Med. 2015;13(1):45. https://doi.org/10.1186/s12916-015-0278-7
  12. Munn DH, Bronte V. Immune suppressive mechanisms in the tumor microenvironment. Curr Opin Immunol. 2016;39:1-6. https://doi.org/10.1016/j.coi.2015.10.009
  13. Marvel D, Gabrilovich DI. Myeloid-derived suppressor cells in the tumor microenvironment: expect the unexpected. J Clin Invest. 2015;125(9):3356-64. https://doi.org/10.1172/JCI80005
  14. Jo SD, et al. Harnessing designed nanoparticles: current strategies and future perspectives in cancer immunotherapy. Nano Today. 2017;17:23-37. https://doi.org/10.1016/j.nantod.2017.10.008
  15. Yoon HY, et al. Engineering nanoparticle strategies for effective cancer immunotherapy. Biomaterials. 2018;178:597-607. https://doi.org/10.1016/j.biomaterials.2018.03.036
  16. Shao K, et al. Nanoparticle-based immunotherapy for cancer. ACS Nano. 2014;9(1):16-30. https://doi.org/10.1021/nn5062029
  17. Velpurisiva P, et al. Nanoparticle design strategies for effective cancer immunotherapy. J Biomed. 2017;2(2):64-77. https://doi.org/10.7150/jbm.18877
  18. Santoni M, Cascinu S, Mills CD. Altering macrophage polarization in the tumor environment: the role of response gene to complement 32. Cell Mol Immunol. 2015;12(6):783-4. https://doi.org/10.1038/cmi.2014.115
  19. Rajendrakumar SK, et al. Nanoparticle-based phototriggered cancer immunotherapy and its domino effect in the tumor microenvironment. Biomacromolecules. 2018;19(6):1869-87. https://doi.org/10.1021/acs.biomac.8b00460
  20. Weigert A, Brune B. Nitric oxide, apoptosis and macrophage polarization during tumor progression. Nitric Oxide. 2008;19(2):95-102. https://doi.org/10.1016/j.niox.2008.04.021
  21. Nesbit M, et al. Low-level monocyte chemoattractant protein-1 stimulation of monocytes leads to tumor formation in nontumorigenic melanoma cells. J Immunol. 2001;166(11):6483-90. https://doi.org/10.4049/jimmunol.166.11.6483
  22. Ruff M, et al. Neuropeptides are chemoattractants for human tumor cells and monocytes: a possible mechanism for metastasis. Clin Immunol Immunopathol. 1985;37(3):387-96. https://doi.org/10.1016/0090-1229(85)90108-4
  23. Mantovani A, et al. Role of tumor-associated macrophages in tumor progression and invasion. Cancer Metastasis Rev. 2006;25(3):315-22. https://doi.org/10.1007/s10555-006-9001-7
  24. Bear AS, et al. Elimination of metastatic melanoma using gold nanoshellenabled photothermal therapy and adoptive T cell transfer. PLoS One. 2013;8(7):e69073. https://doi.org/10.1371/journal.pone.0069073
  25. Wilkerson A, et al. Nanoparticle systems modulating myeloid-derived suppressor cells for cancer immunotherapy. Curr Top Med Chem. 2017;17(16):1843-57. https://doi.org/10.2174/1568026617666161122121412
  26. Kennedy LC, et al. T cells enhance gold nanoparticle delivery to tumors in vivo. Nanoscale Res Lett. 2011;6(1):283. https://doi.org/10.1186/1556-276X-6-283
  27. Intlekofer AM, Thompson CB. At the bench: preclinical rationale for CTLA-4 and PD-1 blockade as cancer immunotherapy. J Leukoc Biol. 2013;94(1):25-39. https://doi.org/10.1189/jlb.1212621
  28. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252-64. https://doi.org/10.1038/nrc3239
  29. Mocellin S, Nitti D. CTLA-4 blockade and the renaissance of cancer immunotherapy. Biochim Biophys Acta. 2013;1836(2):187-96.
  30. Ostrand-Rosenberg S. Tolerance and immune suppression in the tumor microenvironment. Cell Immunol. 2016;299:23-9. https://doi.org/10.1016/j.cellimm.2015.09.011
  31. L-x Z, et al. Efficient co-delivery of neo-epitopes using dispersion-stable layered double hydroxide nanoparticles for enhanced melanoma immunotherapy. Biomaterials. 2018;174:54-66. https://doi.org/10.1016/j.biomaterials.2018.05.015
  32. Tran TH, et al. Nanoparticles for dendritic cell-based immunotherapy. Int J Pharm. 2018;542(1-2):253-65. https://doi.org/10.1016/j.ijpharm.2018.03.029
  33. Thomas SN, et al. Targeting the tumor-draining lymph node with adjuvanted nanoparticles reshapes the anti-tumor immune response. Biomaterials. 2014;35(2):814-24. https://doi.org/10.1016/j.biomaterials.2013.10.003
  34. Irvine DJ, et al. Synthetic nanoparticles for vaccines and immunotherapy. Chem Rev. 2015;115(19):11109-46. https://doi.org/10.1021/acs.chemrev.5b00109
  35. Reddy ST, et al. Exploiting lymphatic transport and complement activation in nanoparticle vaccines. Nat Biotechnol. 2007;25(10):1159-64. https://doi.org/10.1038/nbt1332
  36. Zhu G, et al. Efficient nanovaccine delivery in cancer immunotherapy. ACS Nano. 2017;11(3):2387-92. https://doi.org/10.1021/acsnano.7b00978
  37. Reddy ST, et al. In vivo targeting of dendritic cells in lymph nodes with poly (propylene sulfide) nanoparticles. J Control Release. 2006;112(1):26-34. https://doi.org/10.1016/j.jconrel.2006.01.006
  38. Toy R, et al. The effects of particle size, density and shape on margination of nanoparticles in microcirculation. Nanotechnology. 2011;22(11):115101. https://doi.org/10.1088/0957-4484/22/11/115101
  39. Carboni E, et al. Particle margination and its implications on intravenous anticancer drug delivery. AAPS PharmSciTech. 2014;15(3):762-71. https://doi.org/10.1208/s12249-014-0099-6
  40. Gentile F, et al. The effect of shape on the margination dynamics of non-neutrally buoyant particles in two-dimensional shear flows. J Biomech. 2008;41(10):2312-8. https://doi.org/10.1016/j.jbiomech.2008.03.021
  41. Gratton SE, et al. The effect of particle design on cellular internalization pathways. Proc Natl Acad Sci. 2008;105(33):11613-8. https://doi.org/10.1073/pnas.0801763105
  42. Manolova V, et al. 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
  43. Foged C, et al. Particle size and surface charge affect particle uptake by human dendritic cells in an in vitro model. Int J Pharm. 2005;298(2):315-22. https://doi.org/10.1016/j.ijpharm.2005.03.035
  44. Goodman CM, et al. Toxicity of gold nanoparticles functionalized with cationic and anionic side chains. Bioconjug Chem. 2004;15(4):897-900. https://doi.org/10.1021/bc049951i
  45. Dewitte H, et al. Nanoparticle design to induce tumor immunity and challenge the suppressive tumor microenvironment. Nano Today. 2014;9(6):743-58. https://doi.org/10.1016/j.nantod.2014.10.001
  46. Park J, Babensee JE. Differential functional effects of biomaterials on dendritic cell maturation. Acta Biomater. 2012;8(10):3606-17. https://doi.org/10.1016/j.actbio.2012.06.006
  47. Da Silva CA, et al. Chitin is a size-dependent regulator of macrophage TNF and IL-10 production. J Immunol. 2009;182(6):3573-82. https://doi.org/10.4049/jimmunol.0802113
  48. Shima F, et al. Manipulating the antigen-specific immune response by the hydrophobicity of amphiphilic poly(${\gamma}$-glutamic acid) nanoparticles. Biomaterials. 2013;34(37):9709-16. https://doi.org/10.1016/j.biomaterials.2013.08.064
  49. Shekarian T, et al. Pattern recognition receptors: immune targets to enhance cancer immunotherapy. Ann Oncol. 2017;28(8):1756-66. https://doi.org/10.1093/annonc/mdx179
  50. Hanson MC, et al. Nanoparticulate STING agonists are potent lymph node-targeted vaccine adjuvants. J Clin Invest. 2015;125(6):2532-46. https://doi.org/10.1172/JCI79915
  51. Chen S, et al. Microfluidic generation of chitosan/CpG oligodeoxynucleotide nanoparticles with enhanced cellular uptake and immunostimulatory properties. Lab Chip. 2014;14(11):1842-9. https://doi.org/10.1039/c4lc00015c
  52. Luo M, et al. A STING-activating nanovaccine for cancer immunotherapy. Nat Nanotechnol. 2017;12(7):648-54. https://doi.org/10.1038/nnano.2017.52
  53. Wilson JT, et al. pH-responsive nanoparticle vaccines for dual-delivery of antigens and immunostimulatory oligonucleotides. ACS Nano. 2013;7(5):3912-25. https://doi.org/10.1021/nn305466z
  54. Noh YW, et al. Multifaceted immunomodulatory nanoliposomes: reshaping tumors into vaccines for enhanced Cancer immunotherapy. Adv Funct Mater. 2017;27(8):1605398. https://doi.org/10.1002/adfm.201605398
  55. Jeanbart L, Swartz MA. Engineering opportunities in cancer immunotherapy. Proc Natl Acad Sci. 2015;112(47):14467-72. https://doi.org/10.1073/pnas.1508516112
  56. Ou W, et al. Regulatory T cell-targeted hybrid nanoparticles combined with immuno-checkpoint blockage for cancer immunotherapy. J Control Release. 2018;281:84-96. https://doi.org/10.1016/j.jconrel.2018.05.018
  57. Sacchetti C, et al. In vivo targeting of intratumor regulatory T cells using PEG-modified single-walled carbon nanotubes. Bioconjug Chem. 2013;24(6):852-8. https://doi.org/10.1021/bc400070q
  58. Lu K, et al. Low-dose X-ray radiotherapy-radiodynamic therapy via nanoscale metal-organic frameworks enhances checkpoint blockade immunotherapy. Nat Biomed Eng. 2018;2:600-10. https://doi.org/10.1038/s41551-018-0203-4
  59. Lu J, et al. Nano-enabled pancreas cancer immunotherapy using immunogenic cell death and reversing immunosuppression. Nat Commun. 2017;8(1):1811. https://doi.org/10.1038/s41467-017-01651-9
  60. Postow MA, et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N Engl J Med. 2012;366(10):925-31. https://doi.org/10.1056/NEJMoa1112824
  61. Demaria S, et al. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int J Radiat Oncol Biol Phys. 2004;58(3):862-70. https://doi.org/10.1016/j.ijrobp.2003.09.012
  62. Dewan MZ, et al. Fractionated but not single-dose radiotherapy induces an immune-mediated abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res. 2009;15(17):5379-88. https://doi.org/10.1158/1078-0432.CCR-09-0265
  63. Wilhelm S, et al. Analysis of nanoparticle delivery to tumours. Nat Rev Mater. 2016;1:16014. https://doi.org/10.1038/natrevmats.2016.14
  64. Phillips WT, et al. Image-guided interventional therapy for cancer with radiotherapeutic nanoparticles. Adv Drug Deliv Rev. 2014;76:39-59. https://doi.org/10.1016/j.addr.2014.07.001
  65. Park W, et al. Acidic pH-triggered drug-eluting nanocomposites for magnetic resonance imagingmonitored intra-arterial drug delivery to hepatocellular carcinoma. ACS Appl Mater Interfaces. 2016;8(20):12711-9. https://doi.org/10.1021/acsami.6b03505
  66. Park W, et al. Immunomodulatory magnetic microspheres for augmenting tumor-specific infiltration of natural killer (NK) cells. ACS Appl Mater Interfaces. 2017;9(16):13819-24. https://doi.org/10.1021/acsami.7b02258
  67. Ahmed M, et al. Image-guided tumor ablation: standardization of terminology and reporting criteria-a 10-year update. J Vasc Interv Radiol. 2014;25(11):1691-1705.e4. https://doi.org/10.1016/j.jvir.2014.08.027
  68. Kim D-H. Image-guided Cancer nanomedicine. J Imaging. 2018;4(1):18. https://doi.org/10.3390/jimaging4010018
  69. Ling D, Lee N, Hyeon T. Chemical synthesis and assembly of uniformly sized iron oxide nanoparticles for medical applications. Acc Chem Res. 2015;48(5):1276-85. https://doi.org/10.1021/acs.accounts.5b00038
  70. Lu Y, et al. Iron oxide nanoclusters for T 1 magnetic resonance imaging of non-human primates. Nat Biomed Eng. 2017;1(8):637-43. https://doi.org/10.1038/s41551-017-0116-7
  71. Ling D, et al. Multifunctional tumor pH-sensitive self-assembled nanoparticles for bimodal imaging and treatment of resistant heterogeneous tumors. J Am Chem Soc. 2014;136(15):5647-55. https://doi.org/10.1021/ja4108287
  72. Grifantini R, et al. Magnetically driven drug delivery systems improving targeted immunotherapy for colon-rectal cancer. J Control Release. 2018;280:76-86. https://doi.org/10.1016/j.jconrel.2018.04.052
  73. Park W, et al. Branched Gold Nanoparticle Coating of Clostridium novyi-NT Spores for CT-Guided Intratumoral Injection. Small. 2017;13(5):1602722. https://doi.org/10.1002/smll.201602722
  74. Park JS, et al. Multi-functional nanotracers for image-guided stem cell gene therapy. Nanoscale. 2017;9(14):4665-76. https://doi.org/10.1039/C6NR09090G
  75. van Hooren L, et al. Local checkpoint inhibition of CTLA-4 as a monotherapy or in combination with anti-PD1 prevents the growth of murine bladder cancer. Eur J Immunol. 2017;47(2):385-93. https://doi.org/10.1002/eji.201646583
  76. Fransen MF, et al. Controlled local delivery of CTLA-4 blocking antibody induces CD8+ T cell dependent tumor eradication and decreases risk of toxic side-effects. Clin Cancer Res. 2013;2012:0781.
  77. Sandin LC, et al. Local CTLA4 blockade effectively restrains experimental pancreatic adenocarcinoma growth in vivo. Oncoimmunology. 2014;3(1):e27614. https://doi.org/10.1186/s40824-018-0133-y.
  78. Weiden J, Tel J, Figdor CG. Synthetic immune niches for cancer immunotherapy. Nat Rev Immunol. 2018;18(3):212. https://doi.org/10.1038/nri.2017.89
  79. Sandin LC, et al. Locally delivered CD40 agonist antibody accumulates in secondary lymphoid organs and eradicates experimental disseminated bladder cancer. Cancer Immunol Res. 2014;2(1):80-90. https://doi.org/10.1158/2326-6066.CIR-13-0067
  80. Fransen MF, et al. Local reprogramming of CD8 T cells and systemic tumor eradication without toxicity via slow release and local delivery of agonistic CD40 antibody. Clin Cancer Res. 2011;17(8):2270-80. https://doi.org/10.1158/1078-0432.CCR-10-2888
  81. Pardi N, et al. mRNA vaccines-a new era in vaccinology. Nat Rev Drug Discov. 2018;17(4):261-79. https://doi.org/10.1038/nrd.2017.243
  82. Kreiter S, et al. Mutant MHC class II epitopes drive therapeutic immune responses to cancer. Nature. 2015;520(7549):692-6. https://doi.org/10.1038/nature14426
  83. Oberli MA, et al. Lipid nanoparticle assisted mRNA delivery for potent cancer immunotherapy. Nano Lett. 2016;17(3):1326-35. https://doi.org/10.1021/acs.nanolett.6b03329
  84. Sau S, et al. Multifunctional nanoparticles for cancer immunotherapy: a groundbreaking approach for reprogramming malfunctioned tumor environment. J Control Release. 2018;274:24-34. https://doi.org/10.1016/j.jconrel.2018.01.028
  85. Kuai R, et al. Elimination of established tumors with nanodisc-based combination chemoimmunotherapy. Sci Adv. 2018;4(4):eaao1736. https://doi.org/10.1126/sciadv.aao1736
  86. Kang TH, et al. Chemotherapy acts as an adjuvant to convert the tumor microenvironment into a highly permissive state for vaccination-induced antitumor immunity. Cancer Res. 2013;73(8):2493-504. https://doi.org/10.1158/0008-5472.CAN-12-4241
  87. Min Y, et al. Antigen-capturing nanoparticles improve the abscopal effect and cancer immunotherapy. Nat Nanotechnol. 2017;12(9):877-82. https://doi.org/10.1038/nnano.2017.113
  88. He C, et al. Core-shell nanoscale coordination polymers combine chemotherapy and photodynamic therapy to potentiate checkpoint blockade cancer immunotherapy. Nat Commun. 2016;7:12499. https://doi.org/10.1038/ncomms12499
  89. Lu K, et al. Chlorin-based nanoscale metal-organic framework systemically rejects colorectal cancers via synergistic photodynamic therapy and checkpoint blockade immunotherapy. J Am Chem Soc. 2016;138(38):12502-10. https://doi.org/10.1021/jacs.6b06663
  90. Nam J, et al. Chemo-photothermal therapy combination elicits anti-tumor immunity against advanced metastatic cancer. Nat Commun. 2018;9(1):1074. https://doi.org/10.1038/s41467-018-03473-9
  91. Chen Q, et al. Photothermal therapy with immune-adjuvant nanoparticles together with checkpoint blockade for effective cancer immunotherapy. Nat Commun. 2016;7:13193. https://doi.org/10.1038/ncomms13193
  92. Meir R, et al. Fast image-guided stratification using anti-programmed death ligand 1 gold nanoparticles for cancer immunotherapy. ACS Nano. 2017;11(11):11127-34. https://doi.org/10.1021/acsnano.7b05299
  93. Rhodes KR, Green JJ. Nanoscale artificial antigen presenting cells for cancer immunotherapy. Mol Immunol. 2018;98:13-8. https://doi.org/10.1016/j.molimm.2018.02.016
  94. Hickey JW, et al. Biologically inspired design of nanoparticle artificial antigen-presenting cells for immunomodulation. Nano Lett. 2017;17(11):7045-54. https://doi.org/10.1021/acs.nanolett.7b03734

피인용 문헌

  1. New opportunities for nanoparticles in cancer immunotherapy vol.22, pp.1, 2018, https://doi.org/10.1186/s40824-018-0133-y
  2. Bioactive Nanoparticles for Cancer Immunotherapy vol.19, pp.12, 2018, https://doi.org/10.3390/ijms19123877
  3. Immunomodulatory Nanosystems vol.6, pp.17, 2018, https://doi.org/10.1002/advs.201900101
  4. Recent Advances in Nanovaccines Using Biomimetic Immunomodulatory Materials vol.11, pp.10, 2018, https://doi.org/10.3390/pharmaceutics11100534
  5. Interactions of Nanoparticles and Biosystems: Microenvironment of Nanoparticles and Biomolecules in Nanomedicine vol.9, pp.10, 2019, https://doi.org/10.3390/nano9101365
  6. Host immune response to anti-cancer camptothecin conjugated cyclodextrin-based polymers vol.26, pp.1, 2018, https://doi.org/10.1186/s12929-019-0583-0
  7. Immunotherapy: A Challenge of Breast Cancer Treatment vol.11, pp.12, 2018, https://doi.org/10.3390/cancers11121822
  8. Precision Cancer Nanotherapy: Evolving Role of Multifunctional Nanoparticles for Cancer Active Targeting vol.62, pp.23, 2019, https://doi.org/10.1021/acs.jmedchem.9b00511
  9. Recognition Sites for Cancer-targeting Drug Delivery Systems vol.20, pp.10, 2019, https://doi.org/10.2174/1389200220666191003161114
  10. Peptide-based targeting of immunosuppressive cells in cancer vol.5, pp.1, 2018, https://doi.org/10.1016/j.bioactmat.2020.01.006
  11. Nano-immunoimaging vol.5, pp.4, 2018, https://doi.org/10.1039/c9nh00514e
  12. Materials for Immunotherapy vol.32, pp.13, 2018, https://doi.org/10.1002/adma.201901633
  13. Application of Nanomaterials in Biomedical Imaging and Cancer Therapy vol.10, pp.9, 2018, https://doi.org/10.3390/nano10091700
  14. Combination of Irreversible Electroporation and STING Agonist for Effective Cancer Immunotherapy vol.12, pp.11, 2018, https://doi.org/10.3390/cancers12113123
  15. Emerging nanomedicines for effective breast cancer immunotherapy vol.18, pp.1, 2020, https://doi.org/10.1186/s12951-020-00741-z
  16. Cationic Nanoparticle-Mediated Activation of Natural Killer Cells for Effective Cancer Immunotherapy vol.12, pp.51, 2018, https://doi.org/10.1021/acsami.0c16357
  17. Enhancing Cancer Immunotherapy Treatment Goals by Using Nanoparticle Delivery System vol.16, pp.None, 2021, https://doi.org/10.2147/ijn.s295300
  18. A Critical Review of the Use of Surfactant-Coated Nanoparticles in Nanomedicine and Food Nanotechnology vol.16, pp.None, 2021, https://doi.org/10.2147/ijn.s298606
  19. Designing and Immunomodulating Multiresponsive Nanomaterial for Cancer Theranostics vol.8, pp.None, 2021, https://doi.org/10.3389/fchem.2020.631351
  20. External and Internal Stimuli-Responsive Metallic Nanotherapeutics for Enhanced Anticancer Therapy vol.7, pp.None, 2018, https://doi.org/10.3389/fmolb.2020.597634
  21. Nanotechnology-based platforms to improve immune checkpoint blockade efficacy in cancer therapy vol.17, pp.6, 2018, https://doi.org/10.2217/fon-2020-0720
  22. Magnetic Nanostructures as Emerging Therapeutic Tools to Boost Anti-Tumour Immunity vol.13, pp.11, 2018, https://doi.org/10.3390/cancers13112735
  23. Recent Advances in Nanoscale Metal-Organic Frameworks Towards Cancer Cell Cytotoxicity: An Overview vol.31, pp.7, 2021, https://doi.org/10.1007/s10904-021-02011-3
  24. Nanoparticles in nanomedicine: a comprehensive updated review on current status, challenges and emerging opportunities vol.38, pp.6, 2018, https://doi.org/10.1080/02652048.2021.1942275
  25. Iron oxide nanoparticles for immune cell labeling and cancer immunotherapy vol.6, pp.9, 2018, https://doi.org/10.1039/d1nh00179e
  26. Thermoresponsive Chitosan-Grafted-Poly(N-vinylcaprolactam) Microgels via Ionotropic Gelation for Oncological Applications vol.13, pp.10, 2018, https://doi.org/10.3390/pharmaceutics13101654
  27. Single, binary and successive patterning of charged nanoparticles by electrophoretic deposition vol.23, pp.11, 2018, https://doi.org/10.1007/s11051-021-05368-1
  28. Nanotechnology-enhanced immunotherapy for metastatic cancer vol.2, pp.4, 2021, https://doi.org/10.1016/j.xinn.2021.100174
  29. Nanomedicine for Immunotherapy Targeting Hematological Malignancies: Current Approaches and Perspective vol.11, pp.11, 2021, https://doi.org/10.3390/nano11112792
  30. Nanoparticles for Cancer Therapy: Current Progress and Challenges vol.16, pp.1, 2018, https://doi.org/10.1186/s11671-021-03628-6
  31. Nanoparticles derived from insect exoskeleton modulates NLRP3 inflammasome complex activation in cervical cancer cell line model vol.12, pp.1, 2018, https://doi.org/10.1186/s12645-021-00090-y
  32. Identification of tumor antigens and immune subtypes in lower grade gliomas for mRNA vaccine development vol.19, pp.1, 2021, https://doi.org/10.1186/s12967-021-03014-x
  33. Systematic Review of Cancer Targeting by Nanoparticles Revealed a Global Association between Accumulation in Tumors and Spleen vol.22, pp.23, 2021, https://doi.org/10.3390/ijms222313011