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

ISSN 2095-0179

ISSN 2095-0187(Online)

CN 11-5981/TQ

邮发代号 80-969

2019 Impact Factor: 3.552

Frontiers of Chemical Science and Engineering  2022, Vol. 16 Issue (3): 333-344   https://doi.org/10.1007/s11705-021-2059-5
  本期目录
Immunological effects of nano-enabled hyperthermia for solid tumors: opportunity and challenge
Xiangsheng Liu1,2, Hui Sun1, Xueqing Wang3(), Huan Meng1()
1. CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China
2. The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
3. Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
 全文: PDF(3418 KB)   HTML
Abstract

Compared to conventional hyperthermia that is limited by low selectivity and severe side effects, nano-enabled hyperthermia yields great potentials to tackle these limitations for cancer treatment. Another major advance is the observation of immunological responses associated with nano-enabled hyperthermia, which introduces a new avenue, allowing a potential paradigm shift from the acutely effective and cytotoxicity-centric response to the next-phase discovery, i.e., long-lasting and/or systemic anti-tumor immunity. This perspective first discusses the temperature-gradient and the spatially-structured immunological landscape in solid tumors receiving nano-enabled hyperthermia. This includes the discussion about underlying mechanism such as immunogenic cell death, which initiates a profound immunological chain reaction. In order to propagate the immune activation as a viable therapeutic principle, we further discussed the tumor type-specific complexity in the immunological tumor microenvironment, including the creative design of nano-enabled combination therapy to synergize with nano-enabled hyperthermia.

Key wordsnano-enabled hyperthermia    immunogenic cell death    heterogeneous immunological landscape    tumor microenvironment
收稿日期: 2020-12-25      出版日期: 2022-02-24
Corresponding Author(s): Xueqing Wang,Huan Meng   
 引用本文:   
. [J]. Frontiers of Chemical Science and Engineering, 2022, 16(3): 333-344.
Xiangsheng Liu, Hui Sun, Xueqing Wang, Huan Meng. Immunological effects of nano-enabled hyperthermia for solid tumors: opportunity and challenge. Front. Chem. Sci. Eng., 2022, 16(3): 333-344.
 链接本文:  
https://academic.hep.com.cn/fcse/CN/10.1007/s11705-021-2059-5
https://academic.hep.com.cn/fcse/CN/Y2022/V16/I3/333
HT type Design of NP Animal model Administration route Major immunological endpoints Ref.
NIR PTT Chitosan-coated hollow CuS NPs with immunoadjuvants oligodeoxynucleotides containing the cytosine-guanine (CpG) motifs EMT6 breast cancer i.t. (intratumorally injection) The HCuSNPs-CpG-mediated photothermal immunotherapy elicits more effective systemic immune responses than immunotherapy or PTT alone, resulting in combined anticancer effects against primary treated as well as distant untreated tumors [29]
NIR PTT PEGylated SWNTs 4T1 breast cancer i.t. The PEGylated SWNTs mediated photothermal tumor destruction could release tumor-associated antigens and act as an immunological adjuvant to greatly promote maturation of DCs and production of anti-tumor cytokines. The combination of SWNT-based PTT with antiCTLA-4 therapy could modulate the adaptive immune responses especially cellular immunity for the treatment of metastatic cancer [30]
NIR PTT PEG and polyethylenimine (PEI) dual-polymer-functionalized GO (GO-PEG-PEI) carrying CpG CT26 colon cancer i.t. The GO-PEG-PEI mediated photothermal effect enhanced immunostimulation responses of CpG, owing to the photothermally induced local heating that accelerated intracellular trafficking of nanovectors [31]
NIR PTT Poly(lactic-co-glycolic) acid (PLGA) NPs encapsulating
NIR photothermal agent indocyanine green (ICG) and toll-like-receptor-7 agonist imiquimod (R837)
4T1 breast cancer
CT26 colon cancer
s.c. (subcutaneously injection), i.v. (intravenously injection) The photothermal ablation of primary tumours using PLGA-ICG-R837 NPs, generated tumour-associated antigens, which in the presence of R837-containing NPs as the adjuvant showed vaccine-like functions. In combination with anti-CTLA4, the generated immunological responses was able to attack remaining tumour cells in mice to inhibit metastasis. This strategy offered a strong immunological memory effect, which provided protection against tumour rechallenging post elimination of their initial tumours [32]
NIR PTT PEG coated plasmonic gold nanostar (GNS) MB49 bladder cancer i.t., i.v. The GNS-mediated PTT combined with anti-PD-L1 were able to achieve complete eradication of primary treated tumors and distant untreated tumors in some mice with effective long-lasting immunity against MB49 cancer cells rechallenge [33]
NIR PTT Polydopamine-coated spiky AuNPs (SGNP@PDA) CT26 colorectal cancer;
TC-1 head and neck squamous cell carcinoma
i.t. SGNP@PDA PTT combined with a sub-therapeutic chemo dose of doxorubicin (DOX), elicits robust anti-tumor responses in both cellular (CD8+ T and NK cells) and humoral compartments. Chemo-PTT eliminates residual tumor cells from locally treated tumors and exerts an abscopal effect against untreated, distant tumors, and also exhibits long-term resistance against tumor rechallenge due to the establishment of immunological memory [34]
NIR PTT MDSC membrane-coated iron oxide MNP
(MNP@MDSC)
B16-F10 melanoma i.v. MNPs@MDSC mediated PTT effects enhanced antitumor response by inducing ICD, reprogramming the tumor infiltrating macrophages, and reducing the tumor’s metabolic activity [35]
NIR PTT CpG self-crosslinked
NPs-loaded IR820-conjugated hydrogel
B16 melanoma i.t. IR820-hydrogel mediated PTT induced tumor antigens release for enhancing the immunotherapy effect. CpG NPs serve as adjuvant to improve the immune stimulation. The combined specific antitumor immunity achieved more effective systemic therapeutic effect than PTT or immunotherapy alone [36]
NIR PTT Intracellularly generated AuNPs B16F10 Melanoma
4T1 breast cancer
s.c. The AuNPs were generated intracellularly and then exocytosed as nanoparticle trapped vesicles with retained original bioinformation. After further introduction to DCs, DCs-derived immunological AuNPs induced HT under NIR irradiation and provoked strong antitumor immune responses, promoting DCs maturation, multiple cytokines secretion, and T cells activation [37]
Magnetic HT PEGylated iron NPs (FeNPs) 4T1 breast cancer
CT26 colon cancer
i.t. The combination of FeNP-based MHT with local injection of nanoadjuvant and systemic injection of anti-CTLA4 checkpoint blockade would result in systemic therapeutic responses to inhibit tumor metastasis and a robust immune memory effect to prevent tumor recurrence [38]
NIR PTT Mesoporous silica NPs decorated with AuNPs (Au@XL-MSNs) loaded with CpG-ODNs 4T1 breast cancer i.t. The photothermal effect of AuNPs enhanced cancer immunotherapy by generating a cancer antigen at the tumor site, which can be processed by tumor-infiltrated DCs and induce antigen-specific adaptive immune response [39]
NIR(II) PTT Self-assembly complex of liposome with AuNPs;
two-dimensional polypyrrole nanosheets
4T1 breast cancer i.v. PTT effect induced by NIR(II) light could trigger ICD more homogeneously and deeper than NIR(I) and red light, and more effective than oxaliplatin in solid tumors. The NIR(II) PTT provoked innate and adaptive immunity led to efficient antitumor and anti-metastasis effects when combined with checkpoint blockade therapy [40]
NIR PTT c-RGD-functionalized conjugated polymer NPs (CP NPs) 4T1 breast cancer i.t. The CP NPs mediated photothermal effect demonstrated effective activation of proinflammatory immune response, induced antitumor immunity activation and ultimately inhibited tumor growth [41]
NIR PTT M NPs coated with cancer cell membrane (M@C NPs) 4T1 breast cancer i.v. M@C NPs mediated PTT effects enhanced antitumor immune response by inducing ICD, which led to good therapeutic effect for primary and abscopal tumor when combined with immunoblocking inhibitor [42]
Tab.1  
Fig.1  
Fig.2  
Fig.3  
Fig.4  
Fig.5  
Fig.6  
1 K F Chu, D E Dupuy. Thermal ablation of tumours: biological mechanisms and advances in therapy. Nature Reviews. Cancer, 2014, 14(3): 199–208
https://doi.org/10.1038/nrc3672
2 M H Falk, R D Issels. Hyperthermia in oncology. International Journal of Hyperthermia, 2001, 17(1): 1–18
https://doi.org/10.1080/02656730150201552
3 S B Field, N M Bleehen. Hyperthermia in the treatment of cancer. Cancer Treatment Reviews, 1979, 6(2): 63–94
https://doi.org/10.1016/S0305-7372(79)80043-2
4 P Wust, B Hildebrandt, G Sreenivasa, B Rau, J Gellermann, H Riess, R Felix, P M Schlag. Hyperthermia in combined treatment of cancer. Lancet. Oncology, 2002, 3(8): 487–497
https://doi.org/10.1016/S1470-2045(02)00818-5
5 P Cherukuri, E S Glazer, S A Curley. Targeted hyperthermia using metal nanoparticles. Advanced Drug Delivery Reviews, 2010, 62(3): 339–345
https://doi.org/10.1016/j.addr.2009.11.006
6 B Thiesen, A Jordan. Clinical applications of magnetic nanoparticles for hyperthermia. International Journal of Hyperthermia, 2008, 24(6): 467–474
https://doi.org/10.1080/02656730802104757
7 J Beik, Z Abed, F S Ghoreishi, S Hosseini-Nami, S Mehrzadi, A Shakeri-Zadeh, S K Kamrava. Nanotechnology in hyperthermia cancer therapy: from fundamental principles to advanced applications. Journal of Controlled Release, 2016, 235: 205–221
https://doi.org/10.1016/j.jconrel.2016.05.062
8 X Huang, P K Jain, I H El-Sayed, M A El-Sayed. Plasmonic photothermal therapy (PPTT) using gold nanoparticles. Lasers in Medical Science, 2008, 23(3): 217–228
https://doi.org/10.1007/s10103-007-0470-x
9 D Jaque, L M Maestro, B Del Rosal, P Haro Gonzalez, A Benayas, J Plaza, E M Rodriguez, J Sole. Nanoparticles for photothermal therapies. Nanoscale, 2014, 6(16): 9494–9530
https://doi.org/10.1039/C4NR00708E
10 X Liu, N Huang, H Li, H Wang, Q Jin, J Ji. Multidentate polyethylene glycol modified gold nanorods for in vivo near-infrared photothermal cancer therapy. ACS Applied Materials & Interfaces, 2014, 6(8): 5657–5668
https://doi.org/10.1021/am5001823
11 H C Kim, E Kim, S W Jeong, T L Ha, S I Park, S G Lee, S J Lee, S W Lee. Magnetic nanoparticle-conjugated polymeric micelles for combined hyperthermia and chemotherapy. Nanoscale, 2015, 7(39): 16470–16480
https://doi.org/10.1039/C5NR04130A
12 K P Tamarov, L A Osminkina, S V Zinovyev, K A Maximova, J V Kargina, M B Gongalsky, Y Ryabchikov, A Al Kattan, A P Sviridov, M Sentis. Radio frequency radiation-induced hyperthermia using Si nanoparticle-based sensitizers for mild cancer therapy. Scientific Reports, 2014, 4(1): 1–7
13 R Kumar, A Chauhan, S K Jha, B K Kuanr. Localized cancer treatment by radio-frequency hyperthermia using magnetic nanoparticles immobilized on graphene oxide: from novel synthesis to in vitro studies. Journal of Materials Chemistry. B, Materials for Biology and Medicine, 2018, 6(33): 5385–5399
https://doi.org/10.1039/C8TB01365A
14 J Cardinal, J R Klune, E Chory, G Jeyabalan, J S Kanzius, M Nalesnik, D A Geller. Noninvasive radiofrequency ablation of cancer targeted by gold nanoparticles. Surgery, 2008, 144(2): 125–132
https://doi.org/10.1016/j.surg.2008.03.036
15 K Kaczmarek, T Hornowski, M Kubovcikova, M Timko, M Koralewski, A Jozefczak. Heating induced by therapeutic ultrasound in the presence of magnetic nanoparticles. ACS Applied Materials & Interfaces, 2018, 10(14): 11554–11564
https://doi.org/10.1021/acsami.8b02496
16 J Beik, Z Abed, A Shakeri-Zadeh, M Nourbakhsh, M B Shiran. Evaluation of the sonosensitizing properties of nano-graphene oxide in comparison with iron oxide and gold nanoparticles. Physica E, Low-Dimensional Systems and Nanostructures, 2016, 81: 308–314
https://doi.org/10.1016/j.physe.2016.03.023
17 S B Devarakonda, M R Myers, M Lanier, C Dumoulin, R K Banerjee. Assessment of gold nanoparticle-mediated-enhanced hyperthermia using MR-guided high-intensity focused ultrasound ablation procedure. Nano Letters, 2017, 17(4): 2532–2538
https://doi.org/10.1021/acs.nanolett.7b00272
18 B Hildebrandt, P Wust, O Ahlers, A Dieing, G Sreenivasa, T Kerner, R Felix, H Riess. The cellular and molecular basis of hyperthermia. Critical Reviews in Oncology/Hematology, 2002, 43(1): 33–56
https://doi.org/10.1016/S1040-8428(01)00179-2
19 J R Lepock. Cellular effects of hyperthermia: relevance to the minimum dose for thermal damage. International Journal of Hyperthermia, 2003, 19(3): 252–266
https://doi.org/10.1080/0265673031000065042
20 J L Roti. Cellular responses to hyperthermia (40 °C–46 °C): cell killing and molecular events. International Journal of Hyperthermia, 2008, 24(1): 3–15
https://doi.org/10.1080/02656730701769841
21 K D Fairchild, R M Viscardi, L Hester, I S Singh, J D Hasday. Effects of hypothermia and hyperthermia on cytokine production by cultured human mononuclear phagocytes from adults and newborns. Journal of Interferon & Cytokine Research, 2000, 20(12): 1049–1055
https://doi.org/10.1089/107999000750053708
22 A Ito, H Honda, T Kobayashi. Cancer immunotherapy based on intracellular hyperthermia using magnetite nanoparticles: a novel concept of “heat-controlled necrosis” with heat shock protein expression. Cancer Immunology, Immunotherapy, 2006, 55(3): 320–328
https://doi.org/10.1007/s00262-005-0049-y
23 A Ito, M Shinkai, H Honda, K Yoshikawa, S Saga, T Wakabayashi, J Yoshida, T Kobayashi. Heat shock protein 70 expression induces antitumor immunity during intracellular hyperthermia using magnetite nanoparticles. Cancer Immunology, Immunotherapy, 2003, 52(2): 80–88
https://doi.org/10.1007/s00262-002-0335-x
24 L Galluzzi, A Buqué, O Kepp, L Zitvogel, G Kroemer. Immunogenic cell death in cancer and infectious disease. Nature Reviews. Immunology, 2017, 17(2): 97–111
https://doi.org/10.1038/nri.2016.107
25 S Todryk, A Melcher, A Dalgleish, R G Vile. Heat shock proteins refine the danger theory. Immunology, 2000, 99(3): 334–337
https://doi.org/10.1046/j.1365-2567.2000.00002.x
26 Z Zhang, J Wang, C Chen. Gold nanorods based platforms for light-mediated theranostics. Theranostics, 2013, 3(3): 223–238
https://doi.org/10.7150/thno.5409
27 E B Dickerson, E C Dreaden, X Huang, I H El Sayed, H Chu, S Pushpanketh, J F McDonald, M A El Sayed. Gold nanorod assisted near-infrared plasmonic photothermal therapy (PPTT) of squamous cell carcinoma in mice. Cancer Letters, 2008, 269(1): 57–66
https://doi.org/10.1016/j.canlet.2008.04.026
28 K Yang, L Feng, X Shi, Z Liu. Nano-graphene in biomedicine: theranostic applications. Chemical Society Reviews, 2013, 42(2): 530–547
https://doi.org/10.1039/C2CS35342C
29 L Guo, D D Yan, D Yang, Y Li, X Wang, O Zalewski, B Yan, W Lu. Combinatorial photothermal and immuno cancer therapy using chitosan-coated hollow copper sulfide nanoparticles. ACS Nano, 2014, 8(6): 5670–5681
https://doi.org/10.1021/nn5002112
30 C Wang, L Xu, C Liang, J Xiang, R Peng, Z Liu. Immunological responses triggered by photothermal therapy with carbon nanotubes in combination with anti-CTLA-4 therapy to inhibit cancer metastasis. Advanced Materials, 2014, 26(48): 8154–8162
https://doi.org/10.1002/adma.201402996
31 Y Tao, E Ju, J Ren, X Qu. Immunostimulatory oligonucleotides-loaded cationic graphene oxide with photothermally enhanced immunogenicity for photothermal/immune cancer therapy. Biomaterials, 2014, 35(37): 9963–9971
https://doi.org/10.1016/j.biomaterials.2014.08.036
32 Q Chen, L Xu, C Liang, C Wang, R Peng, Z Liu. Photothermal therapy with immune-adjuvant nanoparticles together with checkpoint blockade for effective cancer immunotherapy. Nature Communications, 2016, 7(1): 1–13
https://doi.org/10.1038/ncomms13193
33 Y Liu, P Maccarini, G M Palmer, W Etienne, Y Zhao, C T Lee, X Ma, B A Inman, T Vo-Dinh. Synergistic immuno photothermal nanotherapy (SYMPHONY) for the treatment of unresectable and metastatic cancers. Scientific Reports, 2017, 7(1): 8606
https://doi.org/10.1038/s41598-017-09116-1
34 J Nam, S Son, L J Ochyl, R Kuai, A Schwendeman, J J Moon. Chemo-photothermal therapy combination elicits anti-tumor immunity against advanced metastatic cancer. Nature Communications, 2018, 9(1): 1074
https://doi.org/10.1038/s41467-018-03473-9
35 G T Yu, L Rao, H Wu, L L Yang, L L Bu, W W Deng, L Wu, X Nan, W F Zhang, X Z Zhao, W Liu, Z J Sun. Myeloid-derived suppressor cell membrane-coated magnetic nanoparticles for cancer theranostics by Inducing macrophage polarization and synergizing immunogenic cell death. Advanced Functional Materials, 2018, 28(37): 1801389
https://doi.org/10.1002/adfm.201801389
36 X Dong, J Liang, A Yang, Z Qian, D Kong, F Lv. Fluorescence imaging guided CpG nanoparticles-loaded IR820-hydrogel for synergistic photothermal immunotherapy. Biomaterials, 2019, 209: 111–125
https://doi.org/10.1016/j.biomaterials.2019.04.024
37 D Zhang, T Wu, X Qin, Q Qiao, L Shang, Q Song, C Yang, Z Zhang. Intracellularly generated immunological gold nanoparticles for combinatorial photothermal therapy and immunotherapy against tumor. Nano Letters, 2019, 19(9): 6635–6646
https://doi.org/10.1021/acs.nanolett.9b02903
38 Y Chao, G Chen, C Liang, J Xu, Z Dong, X Han, C Wang, Z Liu. Iron nanoparticles for low-power local magnetic hyperthermia in combination with immune checkpoint blockade for systemic antitumor therapy. Nano Letters, 2019, 19(7): 4287–4296
https://doi.org/10.1021/acs.nanolett.9b00579
39 C Ong, B G Cha, J Kim. Mesoporous silica nanoparticles doped with gold nanoparticles for combined cancer immunotherapy and photothermal therapy. ACS Applied Bio Materials, 2019, 2(8): 3630–3638
https://doi.org/10.1021/acsabm.9b00483
40 Y Ma, Y Zhang, X Li, Y Zhao, M Li, W Jiang, X Tang, J Dou, L Lu, F Wang, Y Wang. Near-infrared II phototherapy induces deep tissue immunogenic cell death and potentiates cancer immunotherapy. ACS Nano, 2019, 13(10): 11967–11980
https://doi.org/10.1021/acsnano.9b06040
41 Z Wang, B Guo, E Middha, Z Huang, Q Hu, Z Fu, B Liu. Microfluidics-prepared uniform conjugated polymer nanoparticles for photo-triggered immune microenvironment modulation and cancer therapy. ACS Applied Materials & Interfaces, 2019, 11(12): 11167–11176
https://doi.org/10.1021/acsami.8b22579
42 Y Li, X Liu, W Pan, N Li, B Tang. Photothermal therapy-induced immunogenic cell death based on natural melanin nanoparticles against breast cancer. Chemical Communications, 2020, 56(9): 1389–1392
https://doi.org/10.1039/C9CC08447A
43 L Bezu, L C Gomes-da-Silva, H Dewitte, K Breckpot, J Fucikova, R Spisek, L Galluzzi, O Kepp, G Kroemer. Combinatorial strategies for the induction of immunogenic cell death. Frontiers in Immunology, 2015, 6: 187
https://doi.org/10.3389/fimmu.2015.00187
44 X Duan, C Chan, W Lin. Nanoparticle-mediated immunogenic cell death enables and potentiates cancer immunotherapy. Angewandte Chemie International Edition, 2019, 58(3): 670–680
https://doi.org/10.1002/anie.201804882
45 A Rodallec, G Sicard, R Fanciullino, S Benzekry, B Lacarelle, G Milano, J Ciccolini. Turning cold tumors into hot tumors: harnessing the potential of tumor immunity using nanoparticles. Expert Opinion on Drug Metabolism & Toxicology, 2018, 14(11): 1139–1147
https://doi.org/10.1080/17425255.2018.1540588
46 S Wang, A Riedinger, H Li, C Fu, H Liu, L Li, T Liu, L Tan, M J Barthel, G Pugliese, F De Donato, M Scotto D’Abbusco, X Meng, L Manna, H Meng, T Pellegrino. Plasmonic copper sulfide nanocrystals exhibiting near-infrared photothermal and photodynamic therapeutic effects. ACS Nano, 2015, 9(2): 1788–1800
https://doi.org/10.1021/nn506687t
47 G Kroemer, L Galluzzi, O Kepp, L Zitvogel. Immunogenic cell death in cancer therapy. Annual Review of Immunology, 2013, 31(1): 51–72
https://doi.org/10.1146/annurev-immunol-032712-100008
48 A D Garg, A M Dudek-Peric, E Romano, P Agostinis. Immunogenic cell death. International Journal of Developmental Biology, 2015, 59(1-3): 131–140
https://doi.org/10.1387/ijdb.150061pa
49 X Liu, J Jiang, Y P Liao, I Tang, E Zheng, W Qiu, M Lin, X Wang, Y Ji, K C Mei, et al.. Combination chemo-immunotherapy for pancreatic cancer using the immunogenic effects of an irinotecan silicasome nanocarrier plus anti-PD-1. Advancement of Science, 2021, 8(6): 2002147
50 D Y Wong, W W Ong, W H Ang. Induction of immunogenic cell death by chemotherapeutic platinum complexes. Angewandte Chemie International Edition in English, 2015, 54(22): 6483–6487
https://doi.org/10.1002/anie.201500934
51 T Verfaillie, N Rubio, A D Garg, G Bultynck, R Rizzuto, J P Decuypere, J Piette, C Linehan, S Gupta, A Samali, P Agostinis. PERK is required at the ER-mitochondrial contact sites to convey apoptosis after ROS-based ER stress. Cell Death and Differentiation, 2012, 19(11): 1880–1891
https://doi.org/10.1038/cdd.2012.74
52 A D Garg, D V Krysko, T Verfaillie, A Kaczmarek, G B Ferreira, T Marysael, N Rubio, M Firczuk, C Mathieu, A J Roebroek, W Annaert, J Golab, P de Witte, P Vandenabeele, P Agostinis. A novel pathway combining calreticulin exposure and ATP secretion in immunogenic cancer cell death. EMBO Journal, 2012, 31(5): 1062–1079
https://doi.org/10.1038/emboj.2011.497
53 F Radogna, M Diederich. Stress-induced cellular responses in immunogenic cell death: implications for cancer immunotherapy. Biochemical Pharmacology, 2018, 153: 12–23
https://doi.org/10.1016/j.bcp.2018.02.006
54 D V Krysko, A D Garg, A Kaczmarek, O Krysko, P Agostinis, P Vandenabeele. Immunogenic cell death and DAMPs in cancer therapy. Nature Reviews. Cancer, 2012, 12(12): 860–875
https://doi.org/10.1038/nrc3380
55 W Li, J Yang, L Luo, M Jiang, B Qin, H Yin, C Zhu, X Yuan, J Zhang, Z Luo, Y Du, Q Li, Y Lou, Y Qiu, J You. Targeting photodynamic and photothermal therapy to the endoplasmic reticulum enhances immunogenic cancer cell death. Nature Communications, 2019, 10(1): 3349
https://doi.org/10.1038/s41467-019-11269-8
56 A Dimou, K N Syrigos, M W Saif. Overcoming the stromal barrier: technologies to optimize drug delivery in pancreatic cancer. Therapeutic Advances in Medical Oncology, 2012, 4(5): 271–279
https://doi.org/10.1177/1758834012446008
57 H Meng, Y Zhao, J Dong, M Xue, Y S Lin, Z Ji, W X Mai, H Zhang, C H Chang, C J Brinker, J I Zink, A E Nel. Two-wave nanotherapy to target the stroma and optimize gemcitabine delivery to a human pancreatic cancer model in mice. ACS Nano, 2013, 7(11): 10048–10065
https://doi.org/10.1021/nn404083m
58 L Miao, C M Lin, L Huang. Stromal barriers and strategies for the delivery of nanomedicine to desmoplastic tumors. Journal of Controlled Release, 2015, 219: 192–204
https://doi.org/10.1016/j.jconrel.2015.08.017
59 H Meng, A E Nel. Use of nano engineered approaches to overcome the stromal barrier in pancreatic cancer. Advanced Drug Delivery Reviews, 2018, 130: 50–57
https://doi.org/10.1016/j.addr.2018.06.014
60 A Rosenberg, D Mahalingam. Immunotherapy in pancreatic adenocarcinoma-overcoming barriers to response. Journal of Gastrointestinal Oncology, 2018, 9(1): 143–159
https://doi.org/10.21037/jgo.2018.01.13
61 R J Torphy, Y Zhu, R D Schulick. Immunotherapy for pancreatic cancer: barriers and breakthroughs. Annals of Gastroenterological Surgery, 2018, 2(4): 274–281
https://doi.org/10.1002/ags3.12176
62 R D Issels. Hyperthermia adds to chemotherapy. European Journal of Cancer, 2008, 44(17): 2546–2554
https://doi.org/10.1016/j.ejca.2008.07.038
63 J Nam, S Son, K S Park, W P 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
64 J Peng, Y Xiao, W Li, Q Yang, L Tan, Y Jia, Y Qu, Z Qian. Photosensitizer micelles together with IDO inhibitor enhance cancer photothermal therapy and immunotherapy. Advancement of Science, 2018, 5(5): 1700891
https://doi.org/10.1002/advs.201700891
65 Y Li, X Li, F Zhou, A Doughty, A R Hoover, R E Nordquist, W R Chen. Nanotechnology-based photoimmunological therapies for cancer. Cancer Letters, 2019, 442: 429–438
https://doi.org/10.1016/j.canlet.2018.10.044
66 X Liu, Y Zhang, Y Wang, W Zhu, G Li, X Ma, Y Zhang, S Chen, S Tiwari, K Shi, S Zhang, H M Fan, Y X Zhao, X J Liang. Comprehensive understanding of magnetic hyperthermia for improving antitumor therapeutic efficacy. Theranostics, 2020, 10(8): 3793–3815
https://doi.org/10.7150/thno.40805
67 K C Mei, Y P Liao, J Jiang, M Chiang, M Khazaieli, X Liu, X Wang, Q Liu, C H Chang, X Zhang, J Li, Y Ji, B Melano, D Telesca, T Xia, H Meng, A E Nel. Liposomal delivery of mitoxantrone and a cholesteryl indoximod prodrug provides effective chemo-immunotherapy in multiple solid tumors. ACS Nano, 2020, 14(10): 13343–13366
https://doi.org/10.1021/acsnano.0c05194
68 Z R Stephen, M Zhang. Recent progress in the synergistic combination of nanoparticle-mediated hyperthermia and immunotherapy for treatment of cancer. Advanced Healthcare Materials, 2020, 10(2): 2001415
69 S Wang, Z Sun, Y Hou. Engineering nanoparticles toward the modulation of emerging cancer immunotherapy. Advanced Healthcare Materials, 2020, 10(5): e2000845
https://doi.org/10.1002/adhm.202000845
70 K Mortezaee, A Narmani, M Salehi, H Bagheri, B Farhood, H Haghi-Aminjan, M Najafi. Synergic effects of nanoparticles-mediated hyperthermia in radiotherapy/chemotherapy of cancer. Life Sciences, 2021, 269: 119020
https://doi.org/10.1016/j.lfs.2021.119020
71 X S Liu, J H Jiang, C H Chang, Y P Liao, J L Jared, I Tang, E Zheng, W Qiu, M Lin, X Wang, et al.. Development of facile and versatile platinum drug delivering silicasome nanocarriers for efficient pancreatic cancer chemo-immunotherapy. Small, 2021, 17(14): 2005993
https://doi.org/10.1002/smll.202005993
72 J Chen, L Lin, N Yan, Y Hu, H Fang, Z Guo, P Sun, H Tian, X Chen. Macrophages loaded CpG and GNR-PEI for combination of tumor photothermal therapy and immunotherapy. Science China Materials, 2018, 61(11): 1484–1494
https://doi.org/10.1007/s40843-018-9238-6
73 S Musetti, L Huang. Nanoparticle-mediated remodeling of the tumor microenvironment to enhance immunotherapy. ACS Nano, 2018, 12(12): 11740–11755
https://doi.org/10.1021/acsnano.8b05893
74 S D Allen, X Liu, J Jiang, Y P Liao, C H Chang, A E Nel, H Meng. Immune checkpoint inhibition in syngeneic mouse cancer models by a silicasome nanocarrier delivering a GSK3 inhibitor. Biomaterials, 2021, 269: 120635
https://doi.org/10.1016/j.biomaterials.2020.120635
75 P M Chen, W Y Pan, C Y Wu, C Y Yeh, C Korupalli, P K Luo, C J Chou, W T Chia, H W Sung. Modulation of tumor microenvironment using a TLR-7/8 agonist-loaded nanoparticle system that exerts low-temperature hyperthermia and immunotherapy for in situ cancer vaccination. Biomaterials, 2020, 230: 119629
https://doi.org/10.1016/j.biomaterials.2019.119629
76 N Shobaki, Y Sato, Y Suzuki, N Okabe, H Harashima. Manipulating the function of tumor-associated macrophages by siRNA-loaded lipid nanoparticles for cancer immunotherapy. Journal of Controlled Release, 2020, 325: 235–248
https://doi.org/10.1016/j.jconrel.2020.07.001
77 P Y Teo, C Yang, L M Whilding, A C Parente-Pereira, J Maher, A J George, J L Hedrick, Y Y Yang, S Ghaem-Maghami. Ovarian cancer immunotherapy using PD-L1 siRNA targeted delivery from folic acid-functionalized polyethylenimine: strategies to enhance T cell killing. Advanced Healthcare Materials, 2015, 4(8): 1180–1189
https://doi.org/10.1002/adhm.201500089
78 E B Golden, L Apetoh. Radiotherapy and immunogenic cell death. Seminars in Radiation Oncology, 2015, 25(1): 11–17
https://doi.org/10.1016/j.semradonc.2014.07.005
79 L Galluzzi, O Kepp, G Kroemer. Immunogenic cell death in radiation therapy. OncoImmunology, 2013, 2(10): e26536
https://doi.org/10.4161/onci.26536
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed