Nanoparticle-Based Therapeutics to Overcome Obstacles in the Tumor Microenvironment of Hepatocellular Carcinoma
Abstract
:1. Introduction
2. Major Constituents of the Tumor Microenvironment
2.1. Abnormal Vasculature of TME
2.2. Cancer-Associated Fibroblasts and ECM
2.3. Immunosuppressive Immune Cells in TME
2.3.1. Myeloid-Derived Suppressor Cells
2.3.2. Regulatory T Cells
2.3.3. M2-Polarized Macrophages
2.4. Crosstalk in the Dynamic TME
3. Nanomedicine-Based Strategies for TME Modulation
3.1. Anti-Angiogenesis Nanotherapy
3.2. Nanomedicines Designed to Overcome Tumor Physiological Barrier
3.3. Nanomedicine for Immunosuppressive Cells Inhibition
3.3.1. MDSCs Regulating Nanomedicine
3.3.2. T Cell-Modulating Nanoparticles
3.3.3. TAM Modulating Nanoparticles
Target | NP | Size (nm) | Mechanism | Animal Model | Cell Lines | Ref |
---|---|---|---|---|---|---|
Anti-angiogenesis | encapsulating sorafenib with PAM-PBLG-b-TPGS | 118.3 ± 5.1 | release sorafenib target angiogenic pathways | Balb/C nude mice | HepG2 and LO2 | [89] |
galactose modified GTC co-delivery iSur-Pdna and siVEGF | 130−160 | VEGF was depleted with siVEGF | female Balb/c nude mice and female Kunming mice | BEL-7402 | [91] | |
co-delivery of sorafenib and siVEGF based on mesoporous silica nanoparticles | 148.5 ± 3.5 | sustained release of sorafenib and siVEGF | NA | HepG2, Huh, HeLa and A549 | [92] | |
MTX and CA4 loaded N-urocanyl pullulan | 187.1 ± 15.2 | Release anti-tumor drug MTX and vascular disruption agents CA4 | Balb/c and nude mice | HepG2, PLC/PRF/5 and A549 | [94] | |
ECM/CAF | loaded MMF based on 1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N-poly | 156.23 ± 60.38 | MMF inhibited fibroblasts proliferation and tubulin expression; reduced CAF density | C57BL/6 mice, nude mice | Huh7, SUN 449, LM3, LX2, Hep1-6, NIH-3T3 | [108] |
DOX-KGFRWR | long nanofibers with average widths of 10.51 nm | MMP inhibition and antiproliferative effects | male Sprague–Dawley rats; male Institute of Cancer Research mice | SMMC7721 | [111] | |
RA- and DOX-loaded lipid nanoparticles modified with chondroitin sulfate | smaller than 100 | RA disrupted the ECM barrier by destroying the Golgi structure of hepatoma cells and HSCs, while DOX-induced cell death. | Male Kunming mice | SMMC-7721 and H22 | [113] | |
MDSC | FA-chitosan/mIP-10 nanoparticles | 315.5 | sustained local IP-10 expression reduced the number of MDSCs, and attracted CXCR3+CD8+ T cells to the tumor | Female C57BL/6 mice | Hepa1-6 | [117] |
T cell | FA modified TMC co-delivery DOX and IL-2 | 198.1 ± 1.4 | improve the amounts of infiltrated cytotoxic T lymphocytes cells. | Female Kunming mice | SMMC-7721 and A549 | [121] |
poly(d,l-lactide-co-glycolide) nanoparticle, by loading IL-12 and modifying with CD8 and Glypican-3 antibodies o | 145−172 | target T cells and deliver IL-12 to T cells for effective activation and proliferation. | NA | HepG-2 | [123] | |
2DG-encapsulated PLGA nanoparticles | 120 | activated CD8+ T-cell chemotaxis in the tumor microenvironment via the decreased production of lactate in tumors, the increased IFN-γ production and glucose uptake in CD8+ T cells, and production of CXCL9/CXCL10/CXCL11 in both the tumors and CD8+ T cells | nude mice with xenograft tumors | The Huh7, HepG2, B16F10, BxPC3, OS-RC-2, and HT29 cells | [129] | |
TAM | AMD3100 modified lipid-coated PLGA nanoparticles with sorafenib-containing | 150−200 | suppressed the infiltration of TAMs | Male C3H/HeNCrNarl mice | HCA-1 and JHH-7 | [132] |
a nanoliposome-loaded C6-ceramide | NA | reduces not only TAM frequency but also its suppressive function and increased the activity of CD8+ T cells | Male C57BL/6 mice | TAg-transformed B6/WT-19 cells | [133] | |
mannose-modified IMD-0354 loaded cationic lipid-based nanoparticles coated with polymer O-carboxymethyl-chitosan | 129.4 ± 6.8 | TAM re-polarization | C57BL/6 mice | Hepa1-6 | [135] | |
MNPs-MPLA-siRNA | 40−400 | inhibiting the activity of c-MYC oncogene to reduce the pro-tumoral response from M2 macrophages. | NA | Hep-G2 | [139] |
4. Conclusions and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
References
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Llovet, J.M.; Kelley, R.K.; Villanueva, A.; Singal, A.G.; Pikarsky, E.; Roayaie, S.; Lencioni, R.; Koike, K.; Zucman-Rossi, J.; Finn, R.S. Hepatocellular carcinoma. Nat. Rev. Dis. Primers 2021, 7, 6. [Google Scholar] [CrossRef] [PubMed]
- Pinato, D.J.; Sharma, R.; Allara, E.; Yen, C.; Arizumi, T.; Kubota, K.; Bettinger, D.; Jang, J.W.; Smirne, C.; Kim, Y.W.; et al. The ALBI grade provides objective hepatic reserve estimation across each BCLC stage of hepatocellular carcinoma. J. Hepatol. 2017, 66, 338–346. [Google Scholar] [CrossRef] [PubMed]
- Roayaie, S.; Obeidat, K.; Sposito, C.; Mariani, L.; Bhoori, S.; Pellegrinelli, A.; Labow, D.; Llovet, J.M.; Schwartz, M.; Mazzaferro, V. Resection of hepatocellular cancer ≤2 cm: Results from two Western centers. Hepatology 2013, 57, 1426–1435. [Google Scholar] [CrossRef]
- Roxburgh, P.; Evans, T.R.J. Systemic therapy of hepatocellular carcinoma: Are we making progress? Adv. Ther. 2008, 25, 1089–1104. [Google Scholar] [CrossRef]
- Jin, H.; Qin, S.; He, J.; Xiao, J.; Li, Q.; Mao, Y.; Zhao, L. New insights into checkpoint inhibitor immunotherapy and its combined therapies in hepatocellular carcinoma: From mechanisms to clinical trials. Int. J. Biol. Sci. 2022, 18, 2775–2794. [Google Scholar] [CrossRef]
- Al-Salama, Z.T.; Syed, Y.Y.; Scott, L.J. Lenvatinib: A Review in Hepatocellular Carcinoma. Drugs 2019, 79, 665–674. [Google Scholar] [CrossRef]
- Thomas, M. Molecular targeted therapy for hepatocellular carcinoma. J. Gastroenterol. 2009, 44, 136–141. [Google Scholar] [CrossRef]
- Tang, W.; Chen, Z.; Zhang, W.; Cheng, Y.; Zhang, B.; Wu, F.; Wang, Q.; Wang, S.; Rong, D.; Reiter, F.P.; et al. The mechanisms of sorafenib resistance in hepatocellular carcinoma: Theoretical basis and therapeutic aspects. Signal Transduct. Target. Ther. 2020, 5, 87. [Google Scholar] [CrossRef]
- Van den Bulk, J.; Verdegaal, E.M.; de Miranda, N.F. Cancer immunotherapy: Broadening the scope of targetable tumours. Open Biol. 2018, 8, 180037. [Google Scholar] [CrossRef]
- Hoos, A.; Britten, C. The immuno-oncology framework: Enabling a new era of cancer therapy. Oncoimmunology 2012, 1, 334–339. [Google Scholar] [CrossRef]
- Marrero, J.A.; Kulik, L.M.; Sirlin, C.B.; Zhu, A.X.; Finn, R.S.; Abecassis, M.M.; Roberts, L.R.; Heimbach, J.K. Diagnosis, Staging, and Management of Hepatocellular Carcinoma: 2018 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology 2018, 68, 723–750. [Google Scholar] [CrossRef]
- Yau, T.; Hsu, C.; Kim, T.Y.; Choo, S.P.; Kang, Y.K.; Hou, M.M.; Numata, K.; Yeo, W.; Chopra, A.; Ikeda, M.; et al. Nivolumab in advanced hepatocellular carcinoma: Sorafenib-experienced Asian cohort analysis. J. Hepatol. 2019, 71, 543–552. [Google Scholar] [CrossRef]
- Pan, C.; Liu, H.; Robins, E.; Song, W.; Liu, D.; Li, Z.; Zheng, L. Next-generation immuno-oncology agents: Current momentum shifts in cancer immunotherapy. J. Hematol. Oncol. 2020, 13, 29. [Google Scholar] [CrossRef]
- Yan, Y.; Zheng, L.; Du, Q.; Yan, B.; Geller, D.A. Interferon regulatory factor 1 (IRF-1) and IRF-2 regulate PD-L1 expression in hepatocellular carcinoma (HCC) cells. Cancer Immunol. Immunother. CII 2020, 69, 1891–1903. [Google Scholar] [CrossRef]
- Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef]
- El-Khoueiry, A.B.; Sangro, B.; Yau, T.; Crocenzi, T.S.; Kudo, M.; Hsu, C.; Kim, T.-Y.; Choo, S.-P.; Trojan, J.; Welling, T.H.R.; et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): An open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017, 389, 2492–2502. [Google Scholar] [CrossRef]
- Sheng, H.; Huang, Y.; Xiao, Y.; Zhu, Z.; Shen, M.; Zhou, P.; Guo, Z.; Wang, J.; Wang, H.; Dai, W.; et al. ATR inhibitor AZD6738 enhances the antitumor activity of radiotherapy and immune checkpoint inhibitors by potentiating the tumor immune microenvironment in hepatocellular carcinoma. J. Immunother. Cancer 2020, 8, e000340. [Google Scholar] [CrossRef]
- Wu, Y.; Kuang, D.M.; Pan, W.D.; Wan, Y.L.; Lao, X.M.; Wang, D.; Li, X.F.; Zheng, L. Monocyte/macrophage-elicited natural killer cell dysfunction in hepatocellular carcinoma is mediated by CD48/2B4 interactions. Hepatology 2013, 57, 1107–1116. [Google Scholar] [CrossRef]
- Hato, T.; Goyal, L.; Greten, T.F.; Duda, D.G.; Zhu, A.X. Immune checkpoint blockade in hepatocellular carcinoma: Current progress and future directions. Hepatology 2014, 60, 1776–1782. [Google Scholar] [CrossRef]
- Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef]
- Eggert, T.; Greten, T.F. Tumor regulation of the tissue environment in the liver. Pharm. Ther. 2017, 173, 47–57. [Google Scholar] [CrossRef]
- Sun, K.; Yu, J.; Hu, J.; Chen, J.; Song, J.; Chen, Z.; Cai, Z.; Lu, Z.; Zhang, L.; Wang, Z. Salicylic acid-based hypoxia-responsive chemodynamic nanomedicines boost antitumor immunotherapy by modulating immunosuppressive tumor microenvironment. Acta Biomater. 2022, 148, 230–243. [Google Scholar] [CrossRef]
- Dai, Q.; Wilhelm, S.; Ding, D.; Syed, A.M.; Sindhwani, S.; Zhang, Y.; Chen, Y.Y.; MacMillan, P.; Chan, W.C.W. Quantifying the Ligand-Coated Nanoparticle Delivery to Cancer Cells in Solid Tumors. ACS Nano 2018, 12, 8423–8435. [Google Scholar] [CrossRef]
- Golombek, S.K.; May, J.-N.; Theek, B.; Appold, L.; Drude, N.; Kiessling, F.; Lammers, T. Tumor targeting via EPR: Strategies to enhance patient responses. Adv. Drug Deliv. Rev. 2018, 130, 17–38. [Google Scholar] [CrossRef]
- Zhao, P.; Wang, Y.; Kang, X.; Wu, A.; Yin, W.; Tang, Y.; Wang, J.; Zhang, M.; Duan, Y.; Huang, Y. Dual-targeting biomimetic delivery for anti-glioma activity via remodeling the tumor microenvironment and directing macrophage-mediated immunotherapy. Chem. Sci. 2018, 9, 2674–2689. [Google Scholar] [CrossRef]
- Wan, J.L.; Wang, B.; Wu, M.L.; Li, J.; Gong, R.M.; Song, L.N.; Zhang, H.S.; Zhu, G.Q.; Chen, S.P.; Cai, J.L.; et al. MTDH antisense oligonucleotides reshape the immunosuppressive tumor microenvironment to sensitize Hepatocellular Carcinoma to immune checkpoint blockade therapy. Cancer Lett. 2022, 541, 215750. [Google Scholar] [CrossRef]
- Wang, Y.; Wang, Z.; Jia, F.; Xu, Q.; Shu, Z.; Deng, J.; Li, A.; Yu, M.; Yu, Z. CXCR4-guided liposomes regulating hypoxic and immunosuppressive microenvironment for sorafenib-resistant tumor treatment. Bioact. Mater. 2022, 17, 147–161. [Google Scholar] [CrossRef]
- Xiao, Y.; Chen, J.; Zhou, H.; Zeng, X.; Ruan, Z.; Pu, Z.; Jiang, X.; Matsui, A.; Zhu, L.; Amoozgar, Z.; et al. Combining p53 mRNA nanotherapy with immune checkpoint blockade reprograms the immune microenvironment for effective cancer therapy. Nat. Commun. 2022, 13, 758. [Google Scholar] [CrossRef]
- Zhang, J.; Shan, W.F.; Jin, T.T.; Wu, G.Q.; Xiong, X.X.; Jin, H.Y.; Zhu, S.M. Propofol exerts anti-hepatocellular carcinoma by microvesicle-mediated transfer of miR-142-3p from macrophage to cancer cells. J. Transl. Med. 2014, 12, 279. [Google Scholar] [CrossRef]
- Bejarano, L.; Jordāo, M.J.C.; Joyce, J.A. Therapeutic Targeting of the Tumor Microenvironment. Cancer Discov. 2021, 11, 933–959. [Google Scholar] [CrossRef] [PubMed]
- Petitprez, F.; Meylan, M.; de Reyniès, A.; Sautès-Fridman, C.; Fridman, W.H. The Tumor Microenvironment in the Response to Immune Checkpoint Blockade Therapies. Front. Immunol. 2020, 11, 784. [Google Scholar] [CrossRef] [PubMed]
- Jain, R.K. Normalizing tumor microenvironment to treat cancer: Bench to bedside to biomarkers. J. Clin. Oncol. 2013, 31, 2205–2218. [Google Scholar] [CrossRef] [PubMed]
- Wilson, W.R.; Hay, M.P. Targeting hypoxia in cancer therapy. Nat. Rev. Cancer 2011, 11, 393–410. [Google Scholar] [CrossRef]
- Facciabene, A.; Peng, X.; Hagemann, I.S.; Balint, K.; Barchetti, A.; Wang, L.-P.; Gimotty, P.A.; Gilks, C.B.; Lal, P.; Zhang, L.; et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and Treg cells. Nature 2011, 475, 226–230. [Google Scholar] [CrossRef]
- Carmeliet, P.; Jain, R.K. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat. Rev. Drug Discov. 2011, 10, 417–427. [Google Scholar] [CrossRef]
- Schaaf, M.B.; Garg, A.D.; Agostinis, P. Defining the role of the tumor vasculature in antitumor immunity and immunotherapy. Cell Death Dis. 2018, 9, 115. [Google Scholar] [CrossRef]
- Poon, R.T.-P.; Fan, S.-T.; Wong, J. Clinical Implications of Circulating Angiogenic Factors in Cancer Patients. J. Clin. Oncol. 2001, 19, 1207–1225. [Google Scholar] [CrossRef]
- Kalluri, R. The biology and function of fibroblasts in cancer. Nat. Rev. Cancer 2016, 16, 582–598. [Google Scholar] [CrossRef]
- Yin, Z.; Dong, C.; Jiang, K.; Xu, Z.; Li, R.; Guo, K.; Shao, S.; Wang, L. Heterogeneity of cancer-associated fibroblasts and roles in the progression, prognosis, and therapy of hepatocellular carcinoma. J. Hematol. Oncol. 2019, 12, 101. [Google Scholar] [CrossRef]
- Kubo, N.; Araki, K.; Kuwano, H.; Shirabe, K. Cancer-associated fibroblasts in hepatocellular carcinoma. World J. Gastroenterol. 2016, 22, 6841–6850. [Google Scholar] [CrossRef]
- Fu, R.; Zhang, Y.-W.; Li, H.-M.; Lv, W.-C.; Zhao, L.; Guo, Q.-L.; Lu, T.; Weiss, S.J.; Li, Z.-Y.; Wu, Z.-Q. LW106, a novel indoleamine 2,3-dioxygenase 1 inhibitor, suppresses tumour progression by limiting stroma-immune crosstalk and cancer stem cell enrichment in tumour micro-environment. Br. J. Pharm. 2018, 175, 3034–3049. [Google Scholar] [CrossRef]
- Najafi, M.; Farhood, B.; Mortezaee, K. Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J. Cell. Biochem. 2019, 120, 2782–2790. [Google Scholar] [CrossRef]
- Carloni, V.; Luong, T.V.; Rombouts, K. Hepatic stellate cells and extracellular matrix in hepatocellular carcinoma: More complicated than ever. Liver Int. 2014, 34, 834–843. [Google Scholar] [CrossRef]
- Kumar, V.; Patel, S.; Tcyganov, E.; Gabrilovich, D.I. The Nature of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. Trends Immunol. 2016, 37, 208–220. [Google Scholar] [CrossRef]
- Hao, X.; Sun, G.; Zhang, Y.; Kong, X.; Rong, D.; Song, J.; Tang, W.; Wang, X. Targeting Immune Cells in the Tumor Microenvironment of HCC: New Opportunities and Challenges. Front. Cell Dev. Biol. 2021, 9, 775462. [Google Scholar] [CrossRef]
- Venook, A.P.; Papandreou, C.; Furuse, J.; de Guevara, L.L. The incidence and epidemiology of hepatocellular carcinoma: A global and regional perspective. Oncologist 2010, 15 (Suppl. S4), 5–13. [Google Scholar] [CrossRef]
- Pallett, L.J.; Gill, U.S.; Quaglia, A.; Sinclair, L.V.; Jover-Cobos, M.; Schurich, A.; Singh, K.P.; Thomas, N.; Das, A.; Chen, A.; et al. Metabolic regulation of hepatitis B immunopathology by myeloid-derived suppressor cells. Nat. Med. 2015, 21, 591–600. [Google Scholar] [CrossRef]
- Zhang, X.; Fu, X.; Li, T.; Yan, H. The prognostic value of myeloid derived suppressor cell level in hepatocellular carcinoma: A systematic review and meta-analysis. PLoS ONE 2019, 14, e0225327. [Google Scholar] [CrossRef]
- Rodríguez, P.C.; Ochoa, A.C. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: Mechanisms and therapeutic perspectives. Immunol. Rev. 2008, 222, 180–191. [Google Scholar] [CrossRef]
- Wang, Y.; Zhang, T.; Sun, M.; Ji, X.; Xie, M.; Huang, W.; Xia, L. Therapeutic Values of Myeloid-Derived Suppressor Cells in Hepatocellular Carcinoma: Facts and Hopes. Cancers 2021, 13, 5127. [Google Scholar] [CrossRef] [PubMed]
- Sakaguchi, S.; Yamaguchi, T.; Nomura, T.; Ono, M. Regulatory T Cells and Immune Tolerance. Cell 2008, 133, 775–787. [Google Scholar] [CrossRef] [PubMed]
- Eisenstein, E.M.; Williams, C.B. The Treg/Th17 Cell Balance: A New Paradigm for Autoimmunity. Pediatr. Res. 2009, 65, 26–31. [Google Scholar] [CrossRef] [PubMed]
- Fu, J.; Xu, D.; Liu, Z.; Shi, M.; Zhao, P.; Fu, B.; Zhang, Z.; Yang, H.; Zhang, H.; Zhou, C.; et al. Increased regulatory T cells correlate with CD8 T-cell impairment and poor survival in hepatocellular carcinoma patients. Gastroenterology 2007, 132, 2328–2339. [Google Scholar] [CrossRef]
- Vignali, D.A.A.; Collison, L.W.; Workman, C.J. How regulatory T cells work. Nat. Rev. Immunol. 2008, 8, 523–532. [Google Scholar] [CrossRef]
- Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef]
- Cassetta, L.; Pollard, J.W. Tumor-associated macrophages. Curr. Biol. 2020, 30, R246–R248. [Google Scholar] [CrossRef]
- Yang, J.; Zhang, J.-X.; Wang, H.; Wang, G.-L.; Hu, Q.-G.; Zheng, Q.-C. Hepatocellular carcinoma and macrophage interaction induced tumor immunosuppression via Treg requires TLR4 signaling. World J. Gastroenterol. 2012, 18, 2938–2947. [Google Scholar] [CrossRef]
- Ju, C.; Tacke, F. Hepatic macrophages in homeostasis and liver diseases: From pathogenesis to novel therapeutic strategies. Cell Mol. Immunol. 2016, 13, 316–327. [Google Scholar] [CrossRef]
- Yin, C.; Han, Q.; Xu, D.; Zheng, B.; Zhao, X.; Zhang, J. SALL4-mediated upregulation of exosomal miR-146a-5p drives T-cell exhaustion by M2 tumor-associated macrophages in HCC. Oncoimmunology 2019, 8, 1601479. [Google Scholar] [CrossRef]
- Hou, P.-p.; Luo, L.-j.; Chen, H.-z.; Chen, Q.-t.; Bian, X.-l.; Wu, S.-f.; Zhou, J.-x.; Zhao, W.-x.; Liu, J.-m.; Wang, X.-m.; et al. Ectosomal PKM2 Promotes HCC by Inducing Macrophage Differentiation and Remodeling the Tumor Microenvironment. Mol. Cell 2020, 78, 1192–1206.e1110. [Google Scholar] [CrossRef]
- Chen, J.; Lin, Z.; Liu, L.; Zhang, R.; Geng, Y.; Fan, M.; Zhu, W.; Lu, M.; Lu, L.; Jia, H.; et al. GOLM1 exacerbates CD8(+) T cell suppression in hepatocellular carcinoma by promoting exosomal PD-L1 transport into tumor-associated macrophages. Signal Transduct. Target. Ther. 2021, 6, 397. [Google Scholar] [CrossRef]
- LaGory, E.L.; Giaccia, A.J. The ever-expanding role of HIF in tumour and stromal biology. Nat. Cell Biol. 2016, 18, 356–365. [Google Scholar] [CrossRef]
- Dysthe, M.; Parihar, R. Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. Adv. Exp. Med. Biol. 2020, 1224, 117–140. [Google Scholar] [CrossRef]
- Cho, H.; Seo, Y.; Loke, K.M.; Kim, S.W.; Oh, S.M.; Kim, J.H.; Soh, J.; Kim, H.S.; Lee, H.; Kim, J.; et al. Cancer-Stimulated CAFs Enhance Monocyte Differentiation and Protumoral TAM Activation via IL6 and GM-CSF Secretion. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2018, 24, 5407–5421. [Google Scholar] [CrossRef]
- Ranganathan, R.; Madanmohan, S.; Kesavan, A.; Baskar, G.; Krishnamoorthy, Y.R.; Santosham, R.; Ponraju, D.; Rayala, S.K.; Venkatraman, G. Nanomedicine: Towards development of patient-friendly drug-delivery systems for oncological applications. Int. J. Nanomed. 2012, 7, 1043–1060. [Google Scholar] [CrossRef]
- Turato, C.; Balasso, A.; Carloni, V.; Tiribelli, C.; Mastrotto, F.; Mazzocca, A.; Pontisso, P. New molecular targets for functionalized nanosized drug delivery systems in personalized therapy for hepatocellular carcinoma. J. Control. Release Off. J. Control. Release Soc. 2017, 268, 184–197. [Google Scholar] [CrossRef]
- Martin, D.N.; Uprichard, S.L. Identification of transferrin receptor 1 as a hepatitis C virus entry factor. Proc. Natl. Acad. Sci. USA 2013, 110, 10777–10782. [Google Scholar] [CrossRef]
- Pramanik, A.; Laha, D.; Pramanik, P.; Karmakar, P. A novel drug “copper acetylacetonate” loaded in folic acid-tagged chitosan nanoparticle for efficient cancer cell targeting. J. Drug Target. 2014, 22, 23–33. [Google Scholar] [CrossRef]
- Wang, J.; Qian, Y.; Xu, L.; Shao, Y.; Zhang, H.; Shi, F.; Chen, J.; Cui, S.; Chen, X.; Zhu, D.; et al. Hyaluronic acid-shelled, peptide drug conjugate-cored nanomedicine for the treatment of hepatocellular carcinoma. Mater. Sci. Eng. C 2020, 117, 111261. [Google Scholar] [CrossRef]
- Maghsoudinia, F.; Tavakoli, M.B.; Samani, R.K.; Hejazi, S.H.; Sobhani, T.; Mehradnia, F.; Mehrgardi, M.A. Folic acid-functionalized gadolinium-loaded phase transition nanodroplets for dual-modal ultrasound/magnetic resonance imaging of hepatocellular carcinoma. Talanta 2021, 228, 122245. [Google Scholar] [CrossRef] [PubMed]
- Zhan, X.; Guan, Y.-Q. Design of magnetic nanoparticles for hepatocellular carcinoma treatment using the control mechanisms of the cell internal nucleus and external membrane. J. Mater. Chem. B 2015, 3, 4191–4204. [Google Scholar] [CrossRef] [PubMed]
- Kumari, P.; Ghosh, B.; Biswas, S. Nanocarriers for cancer-targeted drug delivery. J. Drug Target. 2016, 24, 179–191. [Google Scholar] [CrossRef] [PubMed]
- Shafei, A.; El-Bakly, W.; Sobhy, A.; Wagdy, O.; Reda, A.; Aboelenin, O.; Marzouk, A.; El Habak, K.; Mostafa, R.; Ali, M.A.; et al. A review on the efficacy and toxicity of different doxorubicin nanoparticles for targeted therapy in metastatic breast cancer. Biomed. Pharmacother. 2017, 95, 1209–1218. [Google Scholar] [CrossRef] [PubMed]
- Pramanik, A.; Xu, Z.; Shamsuddin, S.H.; Khaled, Y.S.; Ingram, N.; Maisey, T.; Tomlinson, D.; Coletta, P.L.; Jayne, D.; Hughes, T.A.; et al. Affimer Tagged Cubosomes: Targeting of Carcinoembryonic Antigen Expressing Colorectal Cancer Cells Using In Vitro and In Vivo Models. ACS Appl. Mater. Interfaces 2022, 14, 11078–11091. [Google Scholar] [CrossRef] [PubMed]
- Wang, Q.; Sun, Y.; Zhang, Z.; Duan, Y. Targeted polymeric therapeutic nanoparticles: Design and interactions with hepatocellular carcinoma. Biomaterials 2015, 56, 229–240. [Google Scholar] [CrossRef]
- Lou, J.; Zhang, L.; Zheng, G. Advancing Cancer Immunotherapies with Nanotechnology. Adv. Ther. 2019, 2, 1800128. [Google Scholar] [CrossRef]
- Wu, S.; Zhang, D.; Yu, J.; Dou, J.; Li, X.; Mu, M.; Liang, P. Chemotherapeutic Nanoparticle-Based Liposomes Enhance the Efficiency of Mild Microwave Ablation in Hepatocellular Carcinoma Therapy. Front. Pharmacol. 2020, 11, 85. [Google Scholar] [CrossRef]
- Cen, D.; Ge, Q.; Xie, C.; Zheng, Q.; Guo, J.; Zhang, Y.; Wang, Y.; Li, X.; Gu, Z.; Cai, X. ZnS@BSA Nanoclusters Potentiate Efficacy of Cancer Immunotherapy. Adv. Mater. 2021, 33, 2104037. [Google Scholar] [CrossRef]
- Liao, J.; Jia, Y.; Wu, Y.; Shi, K.; Yang, D.; Li, P.; Qian, Z. Physical-, chemical-, and biological-responsive nanomedicine for cancer therapy. WIREs Nanomed. Nanobiotechnol. 2020, 12, e1581. [Google Scholar] [CrossRef]
- Barenholz, Y. Doxil®—The first FDA-approved nano-drug: Lessons learned. J. Control. Release 2012, 160, 117–134. [Google Scholar] [CrossRef]
- Shi, J.; Kantoff, P.W.; Wooster, R.; Farokhzad, O.C. Cancer nanomedicine: Progress, challenges and opportunities. Nat. Rev. Cancer 2017, 17, 20–37. [Google Scholar] [CrossRef]
- Azzi, S.; Hebda, J.K.; Gavard, J. Vascular permeability and drug delivery in cancers. Front. Oncol. 2013, 3, 211. [Google Scholar] [CrossRef]
- Berretta, M.; Rinaldi, L.; Di Benedetto, F.; Lleshi, A.; De Re, V.; Facchini, G.; De Paoli, P.; Di Francia, R. Angiogenesis Inhibitors for the Treatment of Hepatocellular Carcinoma. Front. Pharmacol. 2016, 7, 428. [Google Scholar] [CrossRef]
- Taketomi, A. Clinical trials of antiangiogenic therapy for hepatocellular carcinoma. Int. J. Clin. Oncol. 2016, 21, 213–218. [Google Scholar] [CrossRef]
- Zhang, Y.N.; Poon, W.; Tavares, A.J.; McGilvray, I.D.; Chan, W.C.W. Nanoparticle-liver interactions: Cellular uptake and hepatobiliary elimination. J. Control. Release Off. J. Control. Release Soc. 2016, 240, 332–348. [Google Scholar] [CrossRef]
- Hao, Q.; Wang, Z.; Zhao, W.; Wen, L.; Wang, W.; Lu, S.; Xing, D.; Zhan, M.; Hu, X. Dual-Responsive Polyprodrug Nanoparticles with Cascade-Enhanced Magnetic Resonance Signals for Deep-Penetration Drug Release in Tumor Therapy. ACS Appl. Mater. Interfaces 2020, 12, 49489–49501. [Google Scholar] [CrossRef]
- Bazak, R.; Houri, M.; Achy, S.E.; Hussein, W.; Refaat, T. Passive targeting of nanoparticles to cancer: A comprehensive review of the literature. Mol. Clin. Oncol. 2014, 2, 904–908. [Google Scholar] [CrossRef]
- Li, Z.; Ye, L.; Liu, J.; Lian, D.; Li, X. Sorafenib-Loaded Nanoparticles Based on Biodegradable Dendritic Polymers for Enhanced Therapy of Hepatocellular Carcinoma. Int. J. Nanomed. 2020, 15, 1469–1480. [Google Scholar] [CrossRef]
- Kanasty, R.; Dorkin, J.R.; Vegas, A.; Anderson, D. Delivery materials for siRNA therapeutics. Nat. Mater. 2013, 12, 967–977. [Google Scholar] [CrossRef]
- Han, L.; Tang, C.; Yin, C. Oral delivery of shRNA and siRNA via multifunctional polymeric nanoparticles for synergistic cancer therapy. Biomaterials 2014, 35, 4589–4600. [Google Scholar] [CrossRef]
- Zheng, G.; Zhao, R.; Xu, A.; Shen, Z.; Chen, X.; Shao, J. Co-delivery of sorafenib and siVEGF based on mesoporous silica nanoparticles for ASGPR mediated targeted HCC therapy. Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci. 2018, 111, 492–502. [Google Scholar] [CrossRef]
- Chase, D.M.; Chaplin, D.J.; Monk, B.J. The development and use of vascular targeted therapy in ovarian cancer. Gynecol. Oncol. 2017, 145, 393–406. [Google Scholar] [CrossRef]
- Wang, Y.; Chen, H.; Liu, Y.; Wu, J.; Zhou, P.; Wang, Y.; Li, R.; Yang, X.; Zhang, N. pH-sensitive pullulan-based nanoparticle carrier of methotrexate and combretastatin A4 for the combination therapy against hepatocellular carcinoma. Biomaterials 2013, 34, 7181–7190. [Google Scholar] [CrossRef] [PubMed]
- Inglis, D.J.; Lavranos, T.C.; Beaumont, D.M.; Leske, A.F.; Brown, C.K.; Hall, A.J.; Kremmidiotis, G. The vascular disrupting agent BNC105 potentiates the efficacy of VEGF and mTOR inhibitors in renal and breast cancer. Cancer Biol. Ther. 2014, 15, 1552–1560. [Google Scholar] [CrossRef] [PubMed]
- Bao, X.; Shen, N.; Lou, Y.; Yu, H.; Wang, Y.; Liu, L.; Tang, Z.; Chen, X. Enhanced anti-PD-1 therapy in hepatocellular carcinoma by tumor vascular disruption and normalization dependent on combretastatin A4 nanoparticles and DC101. Theranostics 2021, 11, 5955–5969. [Google Scholar] [CrossRef] [PubMed]
- Chang, C.-C.; Dinh, T.K.; Lee, Y.-A.; Wang, F.-N.; Sung, Y.-C.; Yu, P.-L.; Chiu, S.-C.; Shih, Y.-C.; Wu, C.-Y.; Huang, Y.-D.; et al. Nanoparticle Delivery of MnO2 and Antiangiogenic Therapy to Overcome Hypoxia-Driven Tumor Escape and Suppress Hepatocellular Carcinoma. ACS Appl. Mater. Interfaces 2020, 12, 44407–44419. [Google Scholar] [CrossRef] [PubMed]
- Karimi, M.; Ghasemi, A.; Sahandi Zangabad, P.; Rahighi, R.; Moosavi Basri, S.M.; Mirshekari, H.; Amiri, M.; Shafaei Pishabad, Z.; Aslani, A.; Bozorgomid, M.; et al. Smart micro/nanoparticles in stimulus-responsive drug/gene delivery systems. Chem. Soc. Rev. 2016, 45, 1457–1501. [Google Scholar] [CrossRef]
- Zhang, J.; Li, J.; Shi, Z.; Yang, Y.; Xie, X.; Lee, S.M.; Wang, Y.; Leong, K.W.; Chen, M. pH-sensitive polymeric nanoparticles for co-delivery of doxorubicin and curcumin to treat cancer via enhanced pro-apoptotic and anti-angiogenic activities. Acta Biomater. 2017, 58, 349–364. [Google Scholar] [CrossRef]
- Arbiser, J.L.; Klauber, N.; Rohan, R.; van Leeuwen, R.; Huang, M.T.; Fisher, C.; Flynn, E.; Byers, H.R. Curcumin is an in vivo inhibitor of angiogenesis. Mol. Med. 1998, 4, 376–383. [Google Scholar] [CrossRef]
- Sadzuka, Y.; Nagamine, M.; Toyooka, T.; Ibuki, Y.; Sonobe, T. Beneficial effects of curcumin on antitumor activity and adverse reactions of doxorubicin. Int. J. Pharm. 2012, 432, 42–49. [Google Scholar] [CrossRef]
- Xiong, Y.-Q.; Sun, H.-C.; Zhang, W.; Zhu, X.-D.; Zhuang, P.-Y.; Zhang, J.-B.; Wang, L.; Wu, W.-z.; Qin, L.-X.; Tang, Z.-Y. Human Hepatocellular Carcinoma Tumor–derived Endothelial Cells Manifest Increased Angiogenesis Capability and Drug Resistance Compared with Normal Endothelial Cells. Clin. Cancer Res. 2009, 15, 4838–4846. [Google Scholar] [CrossRef]
- Chaudhuri, O.; Koshy, S.T.; Branco da Cunha, C.; Shin, J.W.; Verbeke, C.S.; Allison, K.H.; Mooney, D.J. Extracellular matrix stiffness and composition jointly regulate the induction of malignant phenotypes in mammary epithelium. Nat. Mater. 2014, 13, 970–978. [Google Scholar] [CrossRef]
- Bourboulia, D.; Stetler-Stevenson, W.G. Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs): Positive and negative regulators in tumor cell adhesion. Semin. Cancer Biol. 2010, 20, 161–168. [Google Scholar] [CrossRef]
- Liang, S.; Hu, J.; Xie, Y.; Zhou, Q.; Zhu, Y.; Yang, X. A polyethylenimine-modified carboxyl-poly(styrene/acrylamide) copolymer nanosphere for co-delivering of CpG and TGF-β receptor I inhibitor with remarkable additive tumor regression effect against liver cancer in mice. Int. J. Nanomed. 2016, 11, 6753–6762. [Google Scholar] [CrossRef]
- Affo, S.; Yu, L.-X.; Schwabe, R.F. The Role of Cancer-Associated Fibroblasts and Fibrosis in Liver Cancer. Annu. Rev. Pathol. 2017, 12, 153–186. [Google Scholar] [CrossRef]
- Greupink, R.; Bakker, H.I.; Reker-Smit, C.; van Loenen-Weemaes, A.M.; Kok, R.J.; Meijer, D.K.; Beljaars, L.; Poelstra, K. Studies on the targeted delivery of the antifibrogenic compound mycophenolic acid to the hepatic stellate cell. J. Hepatol. 2005, 43, 884–892. [Google Scholar] [CrossRef]
- Yang, Z.; Zhang, L.; Zhu, H.; Zhou, K.; Wang, H.; Wang, Y.; Su, R.; Guo, D.; Zhou, L.; Xu, X.; et al. Nanoparticle formulation of mycophenolate mofetil achieves enhanced efficacy against hepatocellular carcinoma by targeting tumour-associated fibroblast. J. Cell Mol. Med. 2021, 25, 3511–3523. [Google Scholar] [CrossRef]
- Arii, S.; Mise, M.; Harada, T.; Furutani, M.; Ishigami, S.; Niwano, M.; Mizumoto, M.; Fukumoto, M.; Imamura, M. Overexpression of matrix metalloproteinase 9 gene in hepatocellular carcinoma with invasive potential. Hepatology 1996, 24, 316–322. [Google Scholar] [CrossRef]
- Cui, N.; Hu, M.; Khalil, R.A. Biochemical and Biological Attributes of Matrix Metalloproteinases. Prog. Mol. Biol. Transl. Sci. 2017, 147, 1–73. [Google Scholar] [CrossRef]
- Ji, Y.; Xiao, Y.; Xu, L.; He, J.; Qian, C.; Li, W.; Wu, L.; Chen, R.; Wang, J.; Hu, R.; et al. Drug-Bearing Supramolecular MMP Inhibitor Nanofibers for Inhibition of Metastasis and Growth of Liver Cancer. Adv. Sci. 2018, 5, 1700867. [Google Scholar] [CrossRef]
- Yeow, Y.L.; Wu, J.; Wang, X.; Winteringham, L.; Feindel, K.W.; Tirnitz-Parker, J.E.E.; Leedman, P.J.; Ganss, R.; Hamzah, J. ECM Depletion Is Required to Improve the Intratumoral Uptake of Iron Oxide Nanoparticles in Poorly Perfused Hepatocellular Carcinoma. Front. Oncol. 2022, 12, 837234. [Google Scholar] [CrossRef]
- Luo, J.; Gong, T.; Ma, L. Chondroitin-modified lipid nanoparticles target the Golgi to degrade extracellular matrix for liver cancer management. Carbohydr. Polym. 2020, 249, 116887. [Google Scholar] [CrossRef]
- Suzuki, E.; Kapoor, V.; Jassar, A.S.; Kaiser, L.R.; Albelda, S.M. Gemcitabine Selectively Eliminates Splenic Gr-1+/CD11b+ Myeloid Suppressor Cells in Tumor-Bearing Animals and Enhances Antitumor Immune Activity. Clin. Cancer Res. 2005, 11, 6713–6721. [Google Scholar] [CrossRef]
- Plebanek, M.P.; Bhaumik, D.; Bryce, P.J.; Thaxton, C.S. Scavenger Receptor Type B1 and Lipoprotein Nanoparticle Inhibit Myeloid-Derived Suppressor Cells. Mol. Cancer 2018, 17, 686–697. [Google Scholar] [CrossRef]
- Lai, C.; Yu, X.; Zhuo, H.; Zhou, N.; Xie, Y.; He, J.; Peng, Y.; Xie, X.; Luo, G.; Zhou, S.; et al. Anti-tumor immune response of folate-conjugated chitosan nanoparticles containing the IP-10 gene in mice with hepatocellular carcinoma. J. Biomed. Nanotechnol. 2014, 10, 3576–3589. [Google Scholar] [CrossRef]
- Hu, Z.; Chen, J.; Zhou, S.; Yang, N.; Duan, S.; Zhang, Z.; Su, J.; He, J.; Zhang, Z.; Lu, X.; et al. Mouse IP-10 Gene Delivered by Folate-modified Chitosan Nanoparticles and Dendritic/tumor Cells Fusion Vaccine Effectively Inhibit the Growth of Hepatocellular Carcinoma in Mice. Theranostics 2017, 7, 1942–1952. [Google Scholar] [CrossRef]
- Wang, Z.; He, L.; Li, W.; Xu, C.; Zhang, J.; Wang, D.; Dou, K.; Zhuang, R.; Jin, B.; Zhang, W.; et al. GDF15 induces immunosuppression via CD48 on regulatory T cells in hepatocellular carcinoma. J. Immunother. Cancer 2021, 9, e002787. [Google Scholar] [CrossRef]
- Rosenberg, S.A. IL-2: The first effective immunotherapy for human cancer. J. Immunol. 2014, 192, 5451–5458. [Google Scholar] [CrossRef]
- Tahvildari, M.; Dana, R. Low-Dose IL-2 Therapy in Transplantation, Autoimmunity, and Inflammatory Diseases. J. Immunol. 2019, 203, 2749–2755. [Google Scholar] [CrossRef]
- Wu, J.; Tang, C.; Yin, C. Co-delivery of doxorubicin and interleukin-2 via chitosan based nanoparticles for enhanced antitumor efficacy. Acta Biomater. 2017, 47, 81–90. [Google Scholar] [CrossRef] [PubMed]
- Huang, K.-W.; Hsu, F.-F.; Qiu, J.T.; Chern, G.-J.; Lee, Y.-A.; Chang, C.-C.; Huang, Y.-T.; Sung, Y.-C.; Chiang, C.-C.; Huang, R.-L.; et al. Highly efficient and tumor-selective nanoparticles for dual-targeted immunogene therapy against cancer. Sci. Adv. 2020, 6, eaax5032. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Lin, W.; Chen, H.; Xu, Z.; Ye, Y.; Chen, M. Dual-target IL-12-containing nanoparticles enhance T cell functions for cancer immunotherapy. Cell. Immunol. 2020, 349, 104042. [Google Scholar] [CrossRef] [PubMed]
- Tennant, D.A.; Durán, R.V.; Gottlieb, E. Targeting metabolic transformation for cancer therapy. Nat. Rev. Cancer 2010, 10, 267–277. [Google Scholar] [CrossRef]
- Warburg, O. On the facultative anaerobiosis of cancer cells and its use in chemotherapy. Munch. Med. Wochenschr. (1950) 1961, 103, 2504–2506. [Google Scholar]
- Dietl, K.; Renner, K.; Dettmer, K.; Timischl, B.; Eberhart, K.; Dorn, C.; Hellerbrand, C.; Kastenberger, M.; Kunz-Schughart, L.A.; Oefner, P.J.; et al. Lactic Acid and Acidification Inhibit TNF Secretion and Glycolysis of Human Monocytes. J. Immunol. 2010, 184, 1200. [Google Scholar] [CrossRef]
- Brand, A.; Singer, K.; Koehl, G.E.; Kolitzus, M.; Schoenhammer, G.; Thiel, A.; Matos, C.; Bruss, C.; Klobuch, S.; Peter, K.; et al. LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells. Cell Metab. 2016, 24, 657–671. [Google Scholar] [CrossRef]
- Raez, L.E.; Papadopoulos, K.; Ricart, A.D.; Chiorean, E.G.; Dipaola, R.S.; Stein, M.N.; Rocha Lima, C.M.; Schlesselman, J.J.; Tolba, K.; Langmuir, V.K.; et al. A phase I dose-escalation trial of 2-deoxy-D-glucose alone or combined with docetaxel in patients with advanced solid tumors. Cancer Chemother. Pharmacol. 2013, 71, 523–530. [Google Scholar] [CrossRef]
- Sasaki, K.; Nishina, S.; Yamauchi, A.; Fukuda, K.; Hara, Y.; Yamamura, M.; Egashira, K.; Hino, K. Nanoparticle-Mediated Delivery of 2-Deoxy-D-Glucose Induces Antitumor Immunity and Cytotoxicity in Liver Tumors in Mice. Cell. Mol. Gastroenterol. Hepatol. 2021, 11, 739–762. [Google Scholar] [CrossRef]
- Chen, Y.; Huang, Y.; Reiberger, T.; Duyverman, A.M.; Huang, P.; Samuel, R.; Hiddingh, L.; Roberge, S.; Koppel, C.; Lauwers, G.Y.; et al. Differential effects of sorafenib on liver versus tumor fibrosis mediated by stromal-derived factor 1 alpha/C-X-C receptor type 4 axis and myeloid differentiation antigen–positive myeloid cell infiltration in mice. Hepatology 2014, 59, 1435–1447. [Google Scholar] [CrossRef]
- Duda, D.G.; Kozin, S.V.; Kirkpatrick, N.D.; Xu, L.; Fukumura, D.; Jain, R.K. CXCL12 (SDF1alpha)-CXCR4/CXCR7 pathway inhibition: An emerging sensitizer for anticancer therapies? Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2011, 17, 2074–2080. [Google Scholar] [CrossRef]
- Gao, D.-Y.; Lin, T.-T.; Sung, Y.-C.; Liu, Y.C.; Chiang, W.-H.; Chang, C.-C.; Liu, J.-Y.; Chen, Y. CXCR4-targeted lipid-coated PLGA nanoparticles deliver sorafenib and overcome acquired drug resistance in liver cancer. Biomaterials 2015, 67, 194–203. [Google Scholar] [CrossRef]
- Li, G.; Liu, D.; Kimchi, E.T.; Kaifi, J.T.; Qi, X.; Manjunath, Y.; Liu, X.; Deering, T.; Avella, D.M.; Fox, T.; et al. Nanoliposome C6-Ceramide Increases the Anti-tumor Immune Response and Slows Growth of Liver Tumors in Mice. Gastroenterology 2018, 154, 1024–1036.e1029. [Google Scholar] [CrossRef]
- Ngambenjawong, C.; Gustafson, H.H.; Pun, S.H. Progress in tumor-associated macrophage (TAM)-targeted therapeutics. Adv. Drug Deliv. Rev. 2017, 114, 206–221. [Google Scholar] [CrossRef]
- Wang, T.; Zhang, J.; Hou, T.; Yin, X.; Zhang, N. Selective targeting of tumor cells and tumor associated macrophages separately by twin-like core–shell nanoparticles for enhanced tumor-localized chemoimmunotherapy. Nanoscale 2019, 11, 13934–13946. [Google Scholar] [CrossRef]
- Kaps, L.; Leber, N.; Klefenz, A.; Choteschovsky, N.; Zentel, R.; Nuhn, L.; Schuppan, D. In Vivo siRNA Delivery to Immunosuppressive Liver Macrophages by α-Mannosyl-Functionalized Cationic Nanohydrogel Particles. Cells 2020, 9, 1905. [Google Scholar] [CrossRef]
- Boutilier, A.J.; Elsawa, S.F. Macrophage Polarization States in the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 6995. [Google Scholar] [CrossRef]
- Dai, X.; Ruan, J.; Guo, Y.; Sun, Z.; Liu, J.; Bao, X.; Zhang, H.; Li, Q.; Ye, C.; Wang, X.; et al. Enhanced radiotherapy efficacy and induced anti-tumor immunity in HCC by improving hypoxia microenvironment using oxygen microcapsules. Chem. Eng. J. 2021, 422, 130109. [Google Scholar] [CrossRef]
- Comparetti, E.J.; Lins, P.M.P.; Quitiba, J.; Zucolotto, V. Cancer cell membrane-derived nanoparticles block the expression of immune checkpoint proteins on cancer cells and coordinate modulatory activity on immunosuppressive macrophages. J. Biomed. Mater. Res. Part A 2022, 110, 1499–1511. [Google Scholar] [CrossRef]
- Li, Y.; Ayala-Orozco, C.; Rauta, P.R.; Krishnan, S. The application of nanotechnology in enhancing immunotherapy for cancer treatment: Current effects and perspective. Nanoscale 2019, 11, 17157–17178. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Lu, Y.; Feng, N.; Du, Y.; Yu, R. Nanoparticle-Based Therapeutics to Overcome Obstacles in the Tumor Microenvironment of Hepatocellular Carcinoma. Nanomaterials 2022, 12, 2832. https://doi.org/10.3390/nano12162832
Lu Y, Feng N, Du Y, Yu R. Nanoparticle-Based Therapeutics to Overcome Obstacles in the Tumor Microenvironment of Hepatocellular Carcinoma. Nanomaterials. 2022; 12(16):2832. https://doi.org/10.3390/nano12162832
Chicago/Turabian StyleLu, Yuanfei, Na Feng, Yongzhong Du, and Risheng Yu. 2022. "Nanoparticle-Based Therapeutics to Overcome Obstacles in the Tumor Microenvironment of Hepatocellular Carcinoma" Nanomaterials 12, no. 16: 2832. https://doi.org/10.3390/nano12162832