Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2024

Open Access 01-12-2024 | Cancer Immunotherapy | Review

Nanoparticles in tumor microenvironment remodeling and cancer immunotherapy

Authors: Qiang Lu, Dongquan Kou, Shenghan Lou, Milad Ashrafizadeh, Amir Reza Aref, Israel Canadas, Yu Tian, Xiaojia Niu, Yuzhuo Wang, Pedram Torabian, Lingzhi Wang, Gautam Sethi, Vinay Tergaonkar, Franklin Tay, Zhennan Yuan, Peng Han

Published in: Journal of Hematology & Oncology | Issue 1/2024

Login to get access

Abstract

Cancer immunotherapy and vaccine development have significantly improved the fight against cancers. Despite these advancements, challenges remain, particularly in the clinical delivery of immunomodulatory compounds. The tumor microenvironment (TME), comprising macrophages, fibroblasts, and immune cells, plays a crucial role in immune response modulation. Nanoparticles, engineered to reshape the TME, have shown promising results in enhancing immunotherapy by facilitating targeted delivery and immune modulation. These nanoparticles can suppress fibroblast activation, promote M1 macrophage polarization, aid dendritic cell maturation, and encourage T cell infiltration. Biomimetic nanoparticles further enhance immunotherapy by increasing the internalization of immunomodulatory agents in immune cells such as dendritic cells. Moreover, exosomes, whether naturally secreted by cells in the body or bioengineered, have been explored to regulate the TME and immune-related cells to affect cancer immunotherapy. Stimuli-responsive nanocarriers, activated by pH, redox, and light conditions, exhibit the potential to accelerate immunotherapy. The co-application of nanoparticles with immune checkpoint inhibitors is an emerging strategy to boost anti-tumor immunity. With their ability to induce long-term immunity, nanoarchitectures are promising structures in vaccine development. This review underscores the critical role of nanoparticles in overcoming current challenges and driving the advancement of cancer immunotherapy and TME modification.
Literature
2.
go back to reference Kennedy LB, Salama AKS. A review of cancer immunotherapy toxicity. CA Cancer J Clin. 2020;70(2):86–104.PubMedCrossRef Kennedy LB, Salama AKS. A review of cancer immunotherapy toxicity. CA Cancer J Clin. 2020;70(2):86–104.PubMedCrossRef
3.
go back to reference Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov. 2015;14(8):561–84.PubMedCrossRef Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov. 2015;14(8):561–84.PubMedCrossRef
4.
go back to reference Webster RM. The immune checkpoint inhibitors: where are we now? Nat Rev Drug Discov. 2014;13(12):883–4.PubMedCrossRef Webster RM. The immune checkpoint inhibitors: where are we now? Nat Rev Drug Discov. 2014;13(12):883–4.PubMedCrossRef
7.
go back to reference Liotta F, et al. Toll-like receptors 3 and 4 are expressed by human bone marrow-derived mesenchymal stem cells and can inhibit their T-cell modulatory activity by impairing notch signaling. Stem Cells. 2008;26(1):279–89.PubMedCrossRef Liotta F, et al. Toll-like receptors 3 and 4 are expressed by human bone marrow-derived mesenchymal stem cells and can inhibit their T-cell modulatory activity by impairing notch signaling. Stem Cells. 2008;26(1):279–89.PubMedCrossRef
8.
go back to reference Majeti R, et al. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 2009;138(2):286–99.PubMedPubMedCentralCrossRef Majeti R, et al. CD47 is an adverse prognostic factor and therapeutic antibody target on human acute myeloid leukemia stem cells. Cell. 2009;138(2):286–99.PubMedPubMedCentralCrossRef
10.
11.
go back to reference Barkal AA, et al. Engagement of MHC class I by the inhibitory receptor LILRB1 suppresses macrophages and is a target of cancer immunotherapy. Nat Immunol. 2018;19(1):76–84.PubMedCrossRef Barkal AA, et al. Engagement of MHC class I by the inhibitory receptor LILRB1 suppresses macrophages and is a target of cancer immunotherapy. Nat Immunol. 2018;19(1):76–84.PubMedCrossRef
15.
go back to reference Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39(1):1–10.PubMedCrossRef Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013;39(1):1–10.PubMedCrossRef
16.
go back to reference Nakayama M. Antigen Presentation by MHC-Dressed cells. Front Immunol. 2014;5:672.PubMed Nakayama M. Antigen Presentation by MHC-Dressed cells. Front Immunol. 2014;5:672.PubMed
18.
go back to reference Broz ML, et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell. 2014;26(5):638–52.PubMedPubMedCentralCrossRef Broz ML, et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell. 2014;26(5):638–52.PubMedPubMedCentralCrossRef
21.
go back to reference Sánchez-Paulete AR, et al. Cancer Immunotherapy with Immunomodulatory Anti-CD137 and Anti-PD-1 monoclonal antibodies requires BATF3-Dependent dendritic cells. Cancer Discov. 2016;6(1):71–9.PubMedCrossRef Sánchez-Paulete AR, et al. Cancer Immunotherapy with Immunomodulatory Anti-CD137 and Anti-PD-1 monoclonal antibodies requires BATF3-Dependent dendritic cells. Cancer Discov. 2016;6(1):71–9.PubMedCrossRef
22.
go back to reference Hildner K, et al. Batf3 deficiency reveals a critical role for CD8alpha + dendritic cells in cytotoxic T cell immunity. Science. 2008;322(5904):1097–100.PubMedPubMedCentralCrossRef Hildner K, et al. Batf3 deficiency reveals a critical role for CD8alpha + dendritic cells in cytotoxic T cell immunity. Science. 2008;322(5904):1097–100.PubMedPubMedCentralCrossRef
23.
go back to reference Roberts EW, et al. Critical role for CD103(+)/CD141(+) dendritic cells bearing CCR7 for Tumor Antigen Trafficking and priming of T cell immunity in Melanoma. Cancer Cell. 2016;30(2):324–36.PubMedPubMedCentralCrossRef Roberts EW, et al. Critical role for CD103(+)/CD141(+) dendritic cells bearing CCR7 for Tumor Antigen Trafficking and priming of T cell immunity in Melanoma. Cancer Cell. 2016;30(2):324–36.PubMedPubMedCentralCrossRef
24.
go back to reference Salmon H, et al. Expansion and activation of CD103(+) dendritic cell progenitors at the Tumor Site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity. 2016;44(4):924–38.PubMedPubMedCentralCrossRef Salmon H, et al. Expansion and activation of CD103(+) dendritic cell progenitors at the Tumor Site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity. 2016;44(4):924–38.PubMedPubMedCentralCrossRef
25.
go back to reference de Pulido M. TIM-3 regulates CD103(+) dendritic cell function and response to chemotherapy in breast Cancer. Cancer Cell. 2018;33(1):60–74.e6.CrossRef de Pulido M. TIM-3 regulates CD103(+) dendritic cell function and response to chemotherapy in breast Cancer. Cancer Cell. 2018;33(1):60–74.e6.CrossRef
26.
go back to reference Garris CS, et al. Successful Anti-PD-1 Cancer Immunotherapy requires T cell-dendritic cell crosstalk involving the cytokines IFN-γ and IL-12. Immunity. 2018;49(6):1148–e11617.PubMedPubMedCentralCrossRef Garris CS, et al. Successful Anti-PD-1 Cancer Immunotherapy requires T cell-dendritic cell crosstalk involving the cytokines IFN-γ and IL-12. Immunity. 2018;49(6):1148–e11617.PubMedPubMedCentralCrossRef
27.
go back to reference Spranger S, et al. Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell. 2017;31(5):711–e7234.PubMedPubMedCentralCrossRef Spranger S, et al. Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell. 2017;31(5):711–e7234.PubMedPubMedCentralCrossRef
28.
go back to reference Chow MT, et al. Intratumoral Activity of the CXCR3 chemokine system is required for the efficacy of Anti-PD-1 therapy. Immunity. 2019;50(6):1498–e15125.PubMedPubMedCentralCrossRef Chow MT, et al. Intratumoral Activity of the CXCR3 chemokine system is required for the efficacy of Anti-PD-1 therapy. Immunity. 2019;50(6):1498–e15125.PubMedPubMedCentralCrossRef
29.
go back to reference Xia Y, et al. Engineering macrophages for Cancer Immunotherapy and Drug Delivery. Adv Mater. 2020;32(40):e2002054.PubMedCrossRef Xia Y, et al. Engineering macrophages for Cancer Immunotherapy and Drug Delivery. Adv Mater. 2020;32(40):e2002054.PubMedCrossRef
30.
go back to reference Gao J, Liang Y, Wang L. Shap Polarization Tumor-Associated Macrophages Cancer Immunotherapy Front Immunol. 2022;13:888713. Gao J, Liang Y, Wang L. Shap Polarization Tumor-Associated Macrophages Cancer Immunotherapy Front Immunol. 2022;13:888713.
31.
32.
go back to reference Li L, Ma SR, Yu ZL. Targeting the lipid metabolic reprogramming of tumor-associated macrophages: a novel insight into cancer immunotherapy. Cell Oncol (Dordr); 2023. Li L, Ma SR, Yu ZL. Targeting the lipid metabolic reprogramming of tumor-associated macrophages: a novel insight into cancer immunotherapy. Cell Oncol (Dordr); 2023.
33.
34.
go back to reference Zhang H, et al. Define cancer-associated fibroblasts (CAFs) in the tumor microenvironment: new opportunities in cancer immunotherapy and advances in clinical trials. Mol Cancer. 2023;22(1):159.PubMedPubMedCentralCrossRef Zhang H, et al. Define cancer-associated fibroblasts (CAFs) in the tumor microenvironment: new opportunities in cancer immunotherapy and advances in clinical trials. Mol Cancer. 2023;22(1):159.PubMedPubMedCentralCrossRef
35.
36.
go back to reference Thiery J. Modulation of the antitumor immune response by cancer-associated fibroblasts: mechanisms and targeting strategies to hamper their immunosuppressive functions. Explor Target Antitumor Ther. 2022;3(5):598–629.PubMedPubMedCentralCrossRef Thiery J. Modulation of the antitumor immune response by cancer-associated fibroblasts: mechanisms and targeting strategies to hamper their immunosuppressive functions. Explor Target Antitumor Ther. 2022;3(5):598–629.PubMedPubMedCentralCrossRef
38.
go back to reference Han S et al. Tumor Microenvironment Regulation and Cancer Targeting Therapy based on nanoparticles. J Funct Biomater, 2023. 14(3). Han S et al. Tumor Microenvironment Regulation and Cancer Targeting Therapy based on nanoparticles. J Funct Biomater, 2023. 14(3).
39.
go back to reference Li Z, Yin P. Tumor microenvironment diversity and plasticity in cancer multidrug resistance. Biochim Biophys Acta Rev Cancer. 2023;1878(6):188997.PubMedCrossRef Li Z, Yin P. Tumor microenvironment diversity and plasticity in cancer multidrug resistance. Biochim Biophys Acta Rev Cancer. 2023;1878(6):188997.PubMedCrossRef
44.
46.
48.
go back to reference Bockorny B, Grossman JE, Hidalgo M. Facts and hopes in Immunotherapy of Pancreatic Cancer. Clin Cancer Res. 2022;28(21):4606–17.PubMedCrossRef Bockorny B, Grossman JE, Hidalgo M. Facts and hopes in Immunotherapy of Pancreatic Cancer. Clin Cancer Res. 2022;28(21):4606–17.PubMedCrossRef
49.
go back to reference Yang M, et al. The application of nanoparticles in cancer immunotherapy: targeting tumor microenvironment. Bioact Mater. 2021;6(7):1973–87.PubMed Yang M, et al. The application of nanoparticles in cancer immunotherapy: targeting tumor microenvironment. Bioact Mater. 2021;6(7):1973–87.PubMed
50.
go back to reference Overchuk M, Zheng G. Overcoming obstacles in the tumor microenvironment: recent advancements in nanoparticle delivery for cancer theranostics. Biomaterials. 2018;156:217–37.PubMedCrossRef Overchuk M, Zheng G. Overcoming obstacles in the tumor microenvironment: recent advancements in nanoparticle delivery for cancer theranostics. Biomaterials. 2018;156:217–37.PubMedCrossRef
51.
go back to reference Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res. 1986;46(12 Pt 1):6387–92.PubMed Matsumura Y, Maeda H. A new concept for macromolecular therapeutics in cancer chemotherapy: mechanism of tumoritropic accumulation of proteins and the antitumor agent smancs. Cancer Res. 1986;46(12 Pt 1):6387–92.PubMed
52.
go back to reference Prabhakar U, et al. Challenges and key considerations of the enhanced permeability and retention effect for nanomedicine drug delivery in oncology. Cancer Res. 2013;73(8):2412–7.PubMedPubMedCentralCrossRef Prabhakar U, et al. Challenges and key considerations of the enhanced permeability and retention effect for nanomedicine drug delivery in oncology. Cancer Res. 2013;73(8):2412–7.PubMedPubMedCentralCrossRef
53.
go back to reference Yang S, Gao H. Nanoparticles for modulating tumor microenvironment to improve drug delivery and tumor therapy. Pharmacol Res. 2017;126:97–108.PubMedCrossRef Yang S, Gao H. Nanoparticles for modulating tumor microenvironment to improve drug delivery and tumor therapy. Pharmacol Res. 2017;126:97–108.PubMedCrossRef
54.
go back to reference Chen Q, et al. Remodeling the Tumor Microenvironment with Emerging Nanotherapeutics. Trends Pharmacol Sci. 2018;39(1):59–74.PubMedCrossRef Chen Q, et al. Remodeling the Tumor Microenvironment with Emerging Nanotherapeutics. Trends Pharmacol Sci. 2018;39(1):59–74.PubMedCrossRef
55.
go back to reference Li M, et al. Nanoparticles designed to regulate tumor microenvironment for cancer therapy. Life Sci. 2018;201:37–44.PubMedCrossRef Li M, et al. Nanoparticles designed to regulate tumor microenvironment for cancer therapy. Life Sci. 2018;201:37–44.PubMedCrossRef
56.
go back to reference Guo B, et al. Cuproptosis Induced by ROS responsive nanoparticles with Elesclomol and Copper Combined with αPD-L1 for enhanced Cancer Immunotherapy. Adv Mater. 2023;35(22):e2212267.PubMedCrossRef Guo B, et al. Cuproptosis Induced by ROS responsive nanoparticles with Elesclomol and Copper Combined with αPD-L1 for enhanced Cancer Immunotherapy. Adv Mater. 2023;35(22):e2212267.PubMedCrossRef
57.
go back to reference Wang-Bishop L, et al. STING-activating nanoparticles normalize the vascular-immune interface to potentiate cancer immunotherapy. Sci Immunol. 2023;8(83):eadd1153.PubMedCrossRef Wang-Bishop L, et al. STING-activating nanoparticles normalize the vascular-immune interface to potentiate cancer immunotherapy. Sci Immunol. 2023;8(83):eadd1153.PubMedCrossRef
58.
go back to reference Ding B, et al. ZIF-8 nanoparticles evoke pyroptosis for high-efficiency Cancer Immunotherapy. Angew Chem Int Ed Engl. 2023;62(10):e202215307.PubMedCrossRef Ding B, et al. ZIF-8 nanoparticles evoke pyroptosis for high-efficiency Cancer Immunotherapy. Angew Chem Int Ed Engl. 2023;62(10):e202215307.PubMedCrossRef
59.
go back to reference Liu J et al. Ultrathin clay nanoparticles-mediated mutual reinforcement of ferroptosis and Cancer immunotherapy. Adv Mater, 2023: p. e2309562. Liu J et al. Ultrathin clay nanoparticles-mediated mutual reinforcement of ferroptosis and Cancer immunotherapy. Adv Mater, 2023: p. e2309562.
60.
go back to reference Huang A, et al. Engineered apoptosis-bioinspired nanoparticles initiate Immune Cascade for Cancer Immunotherapy of Malignant ascites. ACS Appl Mater Interfaces. 2023;15(8):10371–82.PubMedCrossRef Huang A, et al. Engineered apoptosis-bioinspired nanoparticles initiate Immune Cascade for Cancer Immunotherapy of Malignant ascites. ACS Appl Mater Interfaces. 2023;15(8):10371–82.PubMedCrossRef
61.
go back to reference Liu D et al. Tumor Microenvironment-Responsive nanoparticles amplifying STING Signaling Pathway for Cancer Immunotherapy. Adv Mater, 2023: p. e2304845. Liu D et al. Tumor Microenvironment-Responsive nanoparticles amplifying STING Signaling Pathway for Cancer Immunotherapy. Adv Mater, 2023: p. e2304845.
62.
go back to reference Sun Y, et al. Sialic acid-targeted cyclodextrin-based nanoparticles deliver CSF-1R siRNA and reprogram tumour-associated macrophages for immunotherapy of prostate cancer. Eur J Pharm Sci. 2023;185:106427.PubMedCrossRef Sun Y, et al. Sialic acid-targeted cyclodextrin-based nanoparticles deliver CSF-1R siRNA and reprogram tumour-associated macrophages for immunotherapy of prostate cancer. Eur J Pharm Sci. 2023;185:106427.PubMedCrossRef
63.
go back to reference Liu C, et al. Microfluidic sonication to assemble Exosome membrane-coated nanoparticles for Immune evasion-mediated targeting. Nano Lett. 2019;19(11):7836–44.PubMedCrossRef Liu C, et al. Microfluidic sonication to assemble Exosome membrane-coated nanoparticles for Immune evasion-mediated targeting. Nano Lett. 2019;19(11):7836–44.PubMedCrossRef
64.
go back to reference Li M et al. Signaling pathways in macrophages: molecular mechanisms and therapeutic targets MedComm (2020), 2023;4(5):e349. Li M et al. Signaling pathways in macrophages: molecular mechanisms and therapeutic targets MedComm (2020), 2023;4(5):e349.
65.
go back to reference Ginhoux F, Guilliams M. Tissue-Resident Macrophage Ontogeny and Homeostasis. Immunity. 2016;44(3):439–49.PubMedCrossRef Ginhoux F, Guilliams M. Tissue-Resident Macrophage Ontogeny and Homeostasis. Immunity. 2016;44(3):439–49.PubMedCrossRef
68.
go back to reference Schulz C, et al. A lineage of myeloid cells independent of myb and hematopoietic stem cells. Science. 2012;336(6077):86–90.PubMedCrossRef Schulz C, et al. A lineage of myeloid cells independent of myb and hematopoietic stem cells. Science. 2012;336(6077):86–90.PubMedCrossRef
69.
go back to reference Sieweke MH, Allen JE. Beyond stem cells: self-renewal of differentiated macrophages. Science. 2013;342(6161):1242974.PubMedCrossRef Sieweke MH, Allen JE. Beyond stem cells: self-renewal of differentiated macrophages. Science. 2013;342(6161):1242974.PubMedCrossRef
70.
go back to reference Zhan C et al. Antitumor therapy for breast cancer: focus on tumor-associated macrophages and nanosized drug delivery systems. Cancer Med, 2023. Zhan C et al. Antitumor therapy for breast cancer: focus on tumor-associated macrophages and nanosized drug delivery systems. Cancer Med, 2023.
72.
go back to reference Vitale I, et al. Macrophages and metabolism in the Tumor Microenvironment. Cell Metab. 2019;30(1):36–50.PubMedCrossRef Vitale I, et al. Macrophages and metabolism in the Tumor Microenvironment. Cell Metab. 2019;30(1):36–50.PubMedCrossRef
73.
go back to reference Wang J, et al. Crosstalk between cancer and immune cells: role of tumor-associated macrophages in the tumor microenvironment. Cancer Med. 2019;8(10):4709–21.PubMedPubMedCentralCrossRef Wang J, et al. Crosstalk between cancer and immune cells: role of tumor-associated macrophages in the tumor microenvironment. Cancer Med. 2019;8(10):4709–21.PubMedPubMedCentralCrossRef
74.
75.
go back to reference Munir MT et al. Tumor-Associated macrophages as multifaceted regulators of breast tumor growth. Int J Mol Sci, 2021. 22(12). Munir MT et al. Tumor-Associated macrophages as multifaceted regulators of breast tumor growth. Int J Mol Sci, 2021. 22(12).
76.
go back to reference Qiu SQ, et al. Tumor-associated macrophages in breast cancer: innocent bystander or important player? Cancer Treat Rev. 2018;70:178–89.PubMedCrossRef Qiu SQ, et al. Tumor-associated macrophages in breast cancer: innocent bystander or important player? Cancer Treat Rev. 2018;70:178–89.PubMedCrossRef
77.
go back to reference Wang C, et al. Macrophage balance fraction determines the degree of immunosuppression and metastatic ability of breast cancer. Int Immunopharmacol. 2021;97:107682.PubMedCrossRef Wang C, et al. Macrophage balance fraction determines the degree of immunosuppression and metastatic ability of breast cancer. Int Immunopharmacol. 2021;97:107682.PubMedCrossRef
78.
go back to reference Steenbrugge J, et al. Anti-inflammatory signaling by mammary tumor cells mediates prometastatic macrophage polarization in an innovative intraductal mouse model for triple-negative breast cancer. J Exp Clin Cancer Res. 2018;37(1):191.PubMedPubMedCentralCrossRef Steenbrugge J, et al. Anti-inflammatory signaling by mammary tumor cells mediates prometastatic macrophage polarization in an innovative intraductal mouse model for triple-negative breast cancer. J Exp Clin Cancer Res. 2018;37(1):191.PubMedPubMedCentralCrossRef
79.
go back to reference Shu Y, Cheng P. Targeting tumor-associated macrophages for cancer immunotherapy Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, 2020;1874(2):188434. Shu Y, Cheng P. Targeting tumor-associated macrophages for cancer immunotherapy Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, 2020;1874(2):188434.
80.
go back to reference Nascimento C, et al. Reprogramming of tumor-associated macrophages by polyaniline-coated iron oxide nanoparticles applied to treatment of breast cancer. Int J Pharm. 2023;636:122866.PubMedCrossRef Nascimento C, et al. Reprogramming of tumor-associated macrophages by polyaniline-coated iron oxide nanoparticles applied to treatment of breast cancer. Int J Pharm. 2023;636:122866.PubMedCrossRef
81.
go back to reference Shang Y, et al. Bioengineered Nanospores selectively blocking LC3-Associated phagocytosis in Tumor-Associated macrophages Potentiate Antitumor Immunity. ACS Nano. 2023;17(11):10872–87.PubMedCrossRef Shang Y, et al. Bioengineered Nanospores selectively blocking LC3-Associated phagocytosis in Tumor-Associated macrophages Potentiate Antitumor Immunity. ACS Nano. 2023;17(11):10872–87.PubMedCrossRef
82.
go back to reference Liu Y, et al. Multifunctional nanoparticles inhibit tumor and tumor-associated macrophages for triple-negative breast cancer therapy. J Colloid Interface Sci. 2024;657:598–610.PubMedCrossRef Liu Y, et al. Multifunctional nanoparticles inhibit tumor and tumor-associated macrophages for triple-negative breast cancer therapy. J Colloid Interface Sci. 2024;657:598–610.PubMedCrossRef
83.
go back to reference Liang DS, et al. Targeted delivery of pexidartinib to tumor-associated macrophages via legumain-sensitive dual-coating nanoparticles for cancer immunotherapy. Colloids Surf B Biointerfaces. 2023;226:113283.PubMedCrossRef Liang DS, et al. Targeted delivery of pexidartinib to tumor-associated macrophages via legumain-sensitive dual-coating nanoparticles for cancer immunotherapy. Colloids Surf B Biointerfaces. 2023;226:113283.PubMedCrossRef
85.
go back to reference Gu J, et al. The role of PKM2 nuclear translocation in the constant activation of the NF-κB signaling pathway in cancer-associated fibroblasts. Cell Death Dis. 2021;12(4):291.PubMedPubMedCentralCrossRef Gu J, et al. The role of PKM2 nuclear translocation in the constant activation of the NF-κB signaling pathway in cancer-associated fibroblasts. Cell Death Dis. 2021;12(4):291.PubMedPubMedCentralCrossRef
86.
go back to reference Bu L, et al. Biological heterogeneity and versatility of cancer-associated fibroblasts in the tumor microenvironment. Oncogene. 2019;38(25):4887–901.PubMedCrossRef Bu L, et al. Biological heterogeneity and versatility of cancer-associated fibroblasts in the tumor microenvironment. Oncogene. 2019;38(25):4887–901.PubMedCrossRef
87.
88.
go back to reference Kobayashi H, et al. Cancer-associated fibroblasts in gastrointestinal cancer. Nat Rev Gastroenterol Hepatol. 2019;16(5):282–95.PubMedCrossRef Kobayashi H, et al. Cancer-associated fibroblasts in gastrointestinal cancer. Nat Rev Gastroenterol Hepatol. 2019;16(5):282–95.PubMedCrossRef
90.
91.
go back to reference Biffi G, et al. IL1-Induced JAK/STAT signaling is antagonized by TGFβ to shape CAF heterogeneity in pancreatic ductal adenocarcinoma. Cancer Discov. 2019;9(2):282–301.PubMedCrossRef Biffi G, et al. IL1-Induced JAK/STAT signaling is antagonized by TGFβ to shape CAF heterogeneity in pancreatic ductal adenocarcinoma. Cancer Discov. 2019;9(2):282–301.PubMedCrossRef
92.
go back to reference Elyada E, et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals Antigen-Presenting Cancer-Associated fibroblasts. Cancer Discov. 2019;9(8):1102–23.PubMedPubMedCentralCrossRef Elyada E, et al. Cross-species single-cell analysis of pancreatic ductal adenocarcinoma reveals Antigen-Presenting Cancer-Associated fibroblasts. Cancer Discov. 2019;9(8):1102–23.PubMedPubMedCentralCrossRef
93.
go back to reference Fang Z, et al. Signaling pathways in cancer-associated fibroblasts: recent advances and future perspectives. Cancer Commun. 2023;43(1):3–41.CrossRef Fang Z, et al. Signaling pathways in cancer-associated fibroblasts: recent advances and future perspectives. Cancer Commun. 2023;43(1):3–41.CrossRef
94.
go back to reference Ai W, et al. Modulation of cancer-associated fibroblasts by nanodelivery system to enhance efficacy of tumor therapy. Nanomed (Lond). 2023;18(15):1025–39.CrossRef Ai W, et al. Modulation of cancer-associated fibroblasts by nanodelivery system to enhance efficacy of tumor therapy. Nanomed (Lond). 2023;18(15):1025–39.CrossRef
95.
go back to reference Geng S, et al. Safe engineering of cancer-associated fibroblasts enhances checkpoint blockade immunotherapy. J Control Release. 2023;356:272–87.PubMedCrossRef Geng S, et al. Safe engineering of cancer-associated fibroblasts enhances checkpoint blockade immunotherapy. J Control Release. 2023;356:272–87.PubMedCrossRef
96.
go back to reference Jian C et al. Biomimetic Nanoplatform for Dual-Targeted Clearance of Activated and Senescent Cancer-Associated Fibroblasts to Improve Radiation Resistance in Breast Cancer Small, 2024: p. e2309279. Jian C et al. Biomimetic Nanoplatform for Dual-Targeted Clearance of Activated and Senescent Cancer-Associated Fibroblasts to Improve Radiation Resistance in Breast Cancer Small, 2024: p. e2309279.
97.
go back to reference Hu M, et al. Salvianolic acid B-loaded polydopamine-modified hollow mesoporous organic silica nanoparticles for treatment of breast cancer metastasis via suppressing cancer-associated fibroblasts. Eur J Pharm Sci. 2024;192:106641.PubMedCrossRef Hu M, et al. Salvianolic acid B-loaded polydopamine-modified hollow mesoporous organic silica nanoparticles for treatment of breast cancer metastasis via suppressing cancer-associated fibroblasts. Eur J Pharm Sci. 2024;192:106641.PubMedCrossRef
99.
go back to reference Wang J, et al. The clinical significance of tumor-infiltrating neutrophils and neutrophil-to-CD8 + lymphocyte ratio in patients with resectable esophageal squamous cell carcinoma. J Transl Med. 2014;12:7.PubMedPubMedCentralCrossRef Wang J, et al. The clinical significance of tumor-infiltrating neutrophils and neutrophil-to-CD8 + lymphocyte ratio in patients with resectable esophageal squamous cell carcinoma. J Transl Med. 2014;12:7.PubMedPubMedCentralCrossRef
100.
go back to reference Deniset JF, Kubes P. Recent Adv Underst Neutrophils F1000Res. 2016;5:2912. Deniset JF, Kubes P. Recent Adv Underst Neutrophils F1000Res. 2016;5:2912.
103.
go back to reference Jin W, et al. Tumor-infiltrating NETs predict postsurgical survival in patients with pancreatic ductal adenocarcinoma. Ann Surg Oncol. 2019;26(2):635–43.PubMedCrossRef Jin W, et al. Tumor-infiltrating NETs predict postsurgical survival in patients with pancreatic ductal adenocarcinoma. Ann Surg Oncol. 2019;26(2):635–43.PubMedCrossRef
104.
go back to reference Fridlender ZG, Albelda SM. Tumor-associated neutrophils: friend or foe? Carcinogenesis. 2012;33(5):949–55.PubMedCrossRef Fridlender ZG, Albelda SM. Tumor-associated neutrophils: friend or foe? Carcinogenesis. 2012;33(5):949–55.PubMedCrossRef
106.
go back to reference He M, et al. Peritumoral stromal neutrophils are essential for c-Met-elicited metastasis in human hepatocellular carcinoma. Oncoimmunology. 2016;5(10):e1219828.PubMedPubMedCentralCrossRef He M, et al. Peritumoral stromal neutrophils are essential for c-Met-elicited metastasis in human hepatocellular carcinoma. Oncoimmunology. 2016;5(10):e1219828.PubMedPubMedCentralCrossRef
107.
go back to reference Schauer C, et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat Med. 2014;20(5):511–7.PubMedCrossRef Schauer C, et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat Med. 2014;20(5):511–7.PubMedCrossRef
108.
go back to reference Ralph SJ, Reynolds MJ. Intratumoral pro-oxidants promote cancer immunotherapy by recruiting and reprogramming neutrophils to eliminate tumors. Cancer Immunol Immunother. 2023;72(3):527–42.PubMedCrossRef Ralph SJ, Reynolds MJ. Intratumoral pro-oxidants promote cancer immunotherapy by recruiting and reprogramming neutrophils to eliminate tumors. Cancer Immunol Immunother. 2023;72(3):527–42.PubMedCrossRef
109.
go back to reference Benguigui M et al. Interferon-stimulated neutrophils as a predictor of immunotherapy response. Cancer Cell, 2024. Benguigui M et al. Interferon-stimulated neutrophils as a predictor of immunotherapy response. Cancer Cell, 2024.
110.
go back to reference Huntington ND, Cursons J, Rautela J. The cancer–natural killer cell immunity cycle. Nat Rev Cancer. 2020;20(8):437–54.PubMedCrossRef Huntington ND, Cursons J, Rautela J. The cancer–natural killer cell immunity cycle. Nat Rev Cancer. 2020;20(8):437–54.PubMedCrossRef
111.
go back to reference Huntington ND, Vosshenrich CA, Di Santo JP. Developmental pathways that generate natural-killer-cell diversity in mice and humans. Nat Rev Immunol. 2007;7(9):703–14.PubMedCrossRef Huntington ND, Vosshenrich CA, Di Santo JP. Developmental pathways that generate natural-killer-cell diversity in mice and humans. Nat Rev Immunol. 2007;7(9):703–14.PubMedCrossRef
112.
go back to reference Pfefferle A, et al. Intra-lineage plasticity and functional reprogramming maintain natural killer cell repertoire diversity. Cell Rep. 2019;29(8):2284–e22944.PubMedCrossRef Pfefferle A, et al. Intra-lineage plasticity and functional reprogramming maintain natural killer cell repertoire diversity. Cell Rep. 2019;29(8):2284–e22944.PubMedCrossRef
113.
114.
go back to reference Gil M, Kim KE. Interleukin-18 is a prognostic biomarker correlated with CD8 + T cell and natural killer cell infiltration in skin cutaneous melanoma Journal of clinical medicine, 2019;8(11):1993. Gil M, Kim KE. Interleukin-18 is a prognostic biomarker correlated with CD8 + T cell and natural killer cell infiltration in skin cutaneous melanoma Journal of clinical medicine, 2019;8(11):1993.
115.
go back to reference Cursons J, et al. A gene signature Predicting Natural Killer Cell Infiltration and Improved Survival in Melanoma patients. Cancer Immunol Res. 2019;7(7):1162–74.PubMedCrossRef Cursons J, et al. A gene signature Predicting Natural Killer Cell Infiltration and Improved Survival in Melanoma patients. Cancer Immunol Res. 2019;7(7):1162–74.PubMedCrossRef
116.
go back to reference Lee H, et al. Integrated molecular and immunophenotypic analysis of NK cells in anti-PD-1 treated metastatic melanoma patients. Oncoimmunology. 2019;8(2):e1537581.PubMedCrossRef Lee H, et al. Integrated molecular and immunophenotypic analysis of NK cells in anti-PD-1 treated metastatic melanoma patients. Oncoimmunology. 2019;8(2):e1537581.PubMedCrossRef
117.
go back to reference Li B et al. Natural killer cell and stroma abundance are independently prognostic and predict gastric cancer chemotherapy benefit. JCI Insight, 2020. 5(9). Li B et al. Natural killer cell and stroma abundance are independently prognostic and predict gastric cancer chemotherapy benefit. JCI Insight, 2020. 5(9).
118.
go back to reference Ran GH, et al. Natural killer cell homing and trafficking in tissues and tumors: from biology to application. Signal Transduct Target Therapy. 2022;7(1):205.CrossRef Ran GH, et al. Natural killer cell homing and trafficking in tissues and tumors: from biology to application. Signal Transduct Target Therapy. 2022;7(1):205.CrossRef
119.
go back to reference Scoville SD, Freud AG, Caligiuri MA. Model Hum Nat Killer Cell Dev Era Innate Lymphoid Cells Front Immunol. 2017;8:360. Scoville SD, Freud AG, Caligiuri MA. Model Hum Nat Killer Cell Dev Era Innate Lymphoid Cells Front Immunol. 2017;8:360.
120.
go back to reference Stokic-Trtica V, Diefenbach A, Klose CSN. NK Cell Dev Times Innate Lymphoid Cell Divers Front Immunol. 2020;11:813. Stokic-Trtica V, Diefenbach A, Klose CSN. NK Cell Dev Times Innate Lymphoid Cell Divers Front Immunol. 2020;11:813.
123.
go back to reference Björkström NK, Ljunggren HG, Michaëlsson J. Emerging insights into natural killer cells in human peripheral tissues. Nat Rev Immunol. 2016;16(5):310–20.PubMedCrossRef Björkström NK, Ljunggren HG, Michaëlsson J. Emerging insights into natural killer cells in human peripheral tissues. Nat Rev Immunol. 2016;16(5):310–20.PubMedCrossRef
124.
go back to reference Yu J, Freud AG, Caligiuri MA. Location and cellular stages of natural killer cell development. Trends Immunol. 2013;34(12):573–82.PubMedCrossRef Yu J, Freud AG, Caligiuri MA. Location and cellular stages of natural killer cell development. Trends Immunol. 2013;34(12):573–82.PubMedCrossRef
125.
126.
127.
go back to reference Hammer Q, Rückert T, Romagnani C. Natural killer cell specificity for viral infections. Nat Immunol. 2018;19(8):800–8.PubMedCrossRef Hammer Q, Rückert T, Romagnani C. Natural killer cell specificity for viral infections. Nat Immunol. 2018;19(8):800–8.PubMedCrossRef
128.
go back to reference Björkström NK, Strunz B, Ljunggren HG. Natural killer cells in antiviral immunity. Nat Rev Immunol. 2022;22(2):112–23.PubMedCrossRef Björkström NK, Strunz B, Ljunggren HG. Natural killer cells in antiviral immunity. Nat Rev Immunol. 2022;22(2):112–23.PubMedCrossRef
130.
go back to reference Shimasaki N, Jain A, Campana D. NK cells for cancer immunotherapy. Nat Rev Drug Discov. 2020;19(3):200–18.PubMedCrossRef Shimasaki N, Jain A, Campana D. NK cells for cancer immunotherapy. Nat Rev Drug Discov. 2020;19(3):200–18.PubMedCrossRef
131.
go back to reference Maskalenko NA, Zhigarev D, Campbell KS. Harnessing natural killer cells for cancer immunotherapy: dispatching the first responders. Nat Rev Drug Discov. 2022;21(8):559–77.PubMedPubMedCentralCrossRef Maskalenko NA, Zhigarev D, Campbell KS. Harnessing natural killer cells for cancer immunotherapy: dispatching the first responders. Nat Rev Drug Discov. 2022;21(8):559–77.PubMedPubMedCentralCrossRef
132.
go back to reference Pockley AG, Vaupel P, Multhoff G. NK cell-based therapeutics for lung cancer. Expert Opin Biol Ther. 2020;20(1):23–33.PubMedCrossRef Pockley AG, Vaupel P, Multhoff G. NK cell-based therapeutics for lung cancer. Expert Opin Biol Ther. 2020;20(1):23–33.PubMedCrossRef
133.
go back to reference Muntasell A, et al. NK Cell infiltrates and HLA Class I expression in primary HER2(+) breast Cancer Predict and Uncouple Pathological Response and Disease-free survival. Clin Cancer Res. 2019;25(5):1535–45.PubMedCrossRef Muntasell A, et al. NK Cell infiltrates and HLA Class I expression in primary HER2(+) breast Cancer Predict and Uncouple Pathological Response and Disease-free survival. Clin Cancer Res. 2019;25(5):1535–45.PubMedCrossRef
134.
go back to reference Liu P, Chen L, Zhang H. Natural killer cells in Liver Disease and Hepatocellular Carcinoma and the NK cell-based immunotherapy. J Immunol Res. 2018;2018:p1206737.CrossRef Liu P, Chen L, Zhang H. Natural killer cells in Liver Disease and Hepatocellular Carcinoma and the NK cell-based immunotherapy. J Immunol Res. 2018;2018:p1206737.CrossRef
135.
go back to reference Ali TH, et al. Enrichment of CD56(dim)KIR + CD57 + highly cytotoxic NK cells in tumour-infiltrated lymph nodes of melanoma patients. Nat Commun. 2014;5:5639.PubMedCrossRef Ali TH, et al. Enrichment of CD56(dim)KIR + CD57 + highly cytotoxic NK cells in tumour-infiltrated lymph nodes of melanoma patients. Nat Commun. 2014;5:5639.PubMedCrossRef
136.
go back to reference Terrén I et al. NK Cell-based immunotherapy in renal cell carcinoma. Cancers (Basel), 2020. 12(2). Terrén I et al. NK Cell-based immunotherapy in renal cell carcinoma. Cancers (Basel), 2020. 12(2).
137.
go back to reference Lim SA, et al. Lipid metabolism in T cell signaling and function. Nat Chem Biol. 2022;18(5):470–81.PubMedCrossRef Lim SA, et al. Lipid metabolism in T cell signaling and function. Nat Chem Biol. 2022;18(5):470–81.PubMedCrossRef
138.
go back to reference Ando M, et al. Memory T cell, exhaustion, and tumor immunity. Immunological Med. 2020;43(1):1–9.CrossRef Ando M, et al. Memory T cell, exhaustion, and tumor immunity. Immunological Med. 2020;43(1):1–9.CrossRef
139.
go back to reference Franco F, et al. Metabolic and epigenetic regulation of T-cell exhaustion. Nat Metabolism. 2020;2(10):1001–12.CrossRef Franco F, et al. Metabolic and epigenetic regulation of T-cell exhaustion. Nat Metabolism. 2020;2(10):1001–12.CrossRef
140.
go back to reference Douka S, et al. Lipid nanoparticle-mediated messenger RNA delivery for ex vivo engineering of natural killer cells. J Control Release. 2023;361:455–69.PubMedCrossRef Douka S, et al. Lipid nanoparticle-mediated messenger RNA delivery for ex vivo engineering of natural killer cells. J Control Release. 2023;361:455–69.PubMedCrossRef
142.
go back to reference Kim KS, et al. Cationic nanoparticle-mediated activation of natural killer cells for Effective Cancer Immunotherapy. ACS Appl Mater Interfaces. 2020;12(51):56731–40.PubMedCrossRef Kim KS, et al. Cationic nanoparticle-mediated activation of natural killer cells for Effective Cancer Immunotherapy. ACS Appl Mater Interfaces. 2020;12(51):56731–40.PubMedCrossRef
143.
go back to reference Sun X, et al. Acid-switchable nanoparticles induce self-adaptive aggregation for enhancing antitumor immunity of natural killer cells. Acta Pharm Sin B. 2023;13(7):3093–105.PubMedPubMedCentralCrossRef Sun X, et al. Acid-switchable nanoparticles induce self-adaptive aggregation for enhancing antitumor immunity of natural killer cells. Acta Pharm Sin B. 2023;13(7):3093–105.PubMedPubMedCentralCrossRef
145.
go back to reference Félétou M. Integrated Systems Physiology: from molecule to function to Disease, in The endothelium: part 1: multiple functions of the endothelial cells—focus on endothelium-derived Vasoactive mediators. 2011, Morgan & Claypool Life Sciences copyright © 2011 by Morgan & Claypool Life Sciences Publishers.: San Rafael (CA). Félétou M. Integrated Systems Physiology: from molecule to function to Disease, in The endothelium: part 1: multiple functions of the endothelial cells—focus on endothelium-derived Vasoactive mediators. 2011, Morgan & Claypool Life Sciences copyright © 2011 by Morgan & Claypool Life Sciences Publishers.: San Rafael (CA).
146.
go back to reference Stoltz JF, et al. Introduction to endothelial cell biology. Clin Hemorheol Microcirc. 2007;37(1–2):5–8.PubMed Stoltz JF, et al. Introduction to endothelial cell biology. Clin Hemorheol Microcirc. 2007;37(1–2):5–8.PubMed
147.
go back to reference Martin FA, Murphy RP, Cummins PM. Thrombomodulin and the vascular endothelium: insights into functional, regulatory, and therapeutic aspects. Am J Physiol Heart Circ Physiol. 2013;304(12):H1585–97.PubMedPubMedCentralCrossRef Martin FA, Murphy RP, Cummins PM. Thrombomodulin and the vascular endothelium: insights into functional, regulatory, and therapeutic aspects. Am J Physiol Heart Circ Physiol. 2013;304(12):H1585–97.PubMedPubMedCentralCrossRef
148.
149.
go back to reference Mohan Rao LV, Esmon CT, Pendurthi UR. Endothelial cell protein C receptor: a multiliganded and multifunctional receptor. Blood. 2014;124(10):1553–62.PubMedPubMedCentralCrossRef Mohan Rao LV, Esmon CT, Pendurthi UR. Endothelial cell protein C receptor: a multiliganded and multifunctional receptor. Blood. 2014;124(10):1553–62.PubMedPubMedCentralCrossRef
150.
go back to reference Bremnes RM, et al. The role of tumor stroma in cancer progression and prognosis: emphasis on carcinoma-associated fibroblasts and non-small cell lung cancer. J Thorac Oncol. 2011;6(1):209–17.PubMedCrossRef Bremnes RM, et al. The role of tumor stroma in cancer progression and prognosis: emphasis on carcinoma-associated fibroblasts and non-small cell lung cancer. J Thorac Oncol. 2011;6(1):209–17.PubMedCrossRef
151.
go back to reference Pietras K, Ostman A. Hallmarks of cancer: interactions with the tumor stroma. Exp Cell Res. 2010;316(8):1324–31.PubMedCrossRef Pietras K, Ostman A. Hallmarks of cancer: interactions with the tumor stroma. Exp Cell Res. 2010;316(8):1324–31.PubMedCrossRef
152.
go back to reference McAllister SS, Weinberg RA. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat Cell Biol. 2014;16(8):717–27.PubMedPubMedCentralCrossRef McAllister SS, Weinberg RA. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat Cell Biol. 2014;16(8):717–27.PubMedPubMedCentralCrossRef
154.
go back to reference Khorana AA, et al. Thromboembolism is a leading cause of death in cancer patients receiving outpatient chemotherapy. J Thromb Haemost. 2007;5(3):632–4.PubMedCrossRef Khorana AA, et al. Thromboembolism is a leading cause of death in cancer patients receiving outpatient chemotherapy. J Thromb Haemost. 2007;5(3):632–4.PubMedCrossRef
156.
go back to reference Preuss SF, Grieshober D, Augustin HG. Systemic Reprogramming Endothelial cell Signal Metastasis cachexia Physiol. 2023;38(4):189–202. Preuss SF, Grieshober D, Augustin HG. Systemic Reprogramming Endothelial cell Signal Metastasis cachexia Physiol. 2023;38(4):189–202.
158.
go back to reference Rouget C. Memoire sur les development, la structure et la proprietes physiologiques des capillaires sanguines et lymphatiques. Arch Physiol, 1873: p. 603–63. Rouget C. Memoire sur les development, la structure et la proprietes physiologiques des capillaires sanguines et lymphatiques. Arch Physiol, 1873: p. 603–63.
159.
go back to reference Zimmermann KW. Der Feinere Bau Der Blutcapillaren. Z für Anatomie und Entwicklungsgeschichte. 1923;68:29–109.CrossRef Zimmermann KW. Der Feinere Bau Der Blutcapillaren. Z für Anatomie und Entwicklungsgeschichte. 1923;68:29–109.CrossRef
160.
go back to reference Schrimpf C, et al. The role of pericyte detachment in vascular rarefaction. J Vasc Res. 2014;51(4):247–58.PubMedCrossRef Schrimpf C, et al. The role of pericyte detachment in vascular rarefaction. J Vasc Res. 2014;51(4):247–58.PubMedCrossRef
161.
go back to reference Hirschi KK, D’Amore PA. Pericytes in the microvasculature. Cardiovascular Res. 1996;32(4):687–98.CrossRef Hirschi KK, D’Amore PA. Pericytes in the microvasculature. Cardiovascular Res. 1996;32(4):687–98.CrossRef
162.
go back to reference Díaz-Flores L, et al. Microvascular pericytes, a review of their morphological and functional characteristics. Histology and histopathology; 1991. Díaz-Flores L, et al. Microvascular pericytes, a review of their morphological and functional characteristics. Histology and histopathology; 1991.
164.
go back to reference Birbrair A, et al. Pericytes at the intersection between tissue regeneration and pathology. Clin Sci. 2015;128(2):81–93.CrossRef Birbrair A, et al. Pericytes at the intersection between tissue regeneration and pathology. Clin Sci. 2015;128(2):81–93.CrossRef
165.
go back to reference Picoli CC et al. Pericytes cross-talks within the tumor microenvironment Biochimica et Biophysica Acta (BBA)-Reviews on Cancer, 2021;1876(2):188608. Picoli CC et al. Pericytes cross-talks within the tumor microenvironment Biochimica et Biophysica Acta (BBA)-Reviews on Cancer, 2021;1876(2):188608.
166.
go back to reference Jiang Z et al. Pericytes in the tumor microenvironment. Cancer Lett, 2023: p. 216074. Jiang Z et al. Pericytes in the tumor microenvironment. Cancer Lett, 2023: p. 216074.
167.
go back to reference Chen Z, Xu XH, Hu J. Role of pericytes in angiogenesis: focus on cancer angiogenesis and anti-angiogenic therapy. Neoplasma. 2016;63(2):173–82.PubMed Chen Z, Xu XH, Hu J. Role of pericytes in angiogenesis: focus on cancer angiogenesis and anti-angiogenic therapy. Neoplasma. 2016;63(2):173–82.PubMed
168.
go back to reference Hong CL, et al. CD248 regulates wnt signaling in Pericytes to Promote Angiogenesis and Tumor Growth in Lung Cancer. Cancer Res. 2022;82(20):3734–50.PubMedCrossRef Hong CL, et al. CD248 regulates wnt signaling in Pericytes to Promote Angiogenesis and Tumor Growth in Lung Cancer. Cancer Res. 2022;82(20):3734–50.PubMedCrossRef
169.
go back to reference Meng YM, et al. Hexokinase 2-driven glycolysis in pericytes activates their contractility leading to tumor blood vessel abnormalities. Nat Commun. 2021;12(1):6011.PubMedPubMedCentralCrossRef Meng YM, et al. Hexokinase 2-driven glycolysis in pericytes activates their contractility leading to tumor blood vessel abnormalities. Nat Commun. 2021;12(1):6011.PubMedPubMedCentralCrossRef
170.
go back to reference Dasgupta S, et al. RGS5-TGFβ-Smad2/3 axis switches pro- to anti-apoptotic signaling in tumor-residing pericytes, assisting tumor growth. Cell Death Differ. 2021;28(11):3052–76.PubMedPubMedCentralCrossRef Dasgupta S, et al. RGS5-TGFβ-Smad2/3 axis switches pro- to anti-apoptotic signaling in tumor-residing pericytes, assisting tumor growth. Cell Death Differ. 2021;28(11):3052–76.PubMedPubMedCentralCrossRef
171.
go back to reference Pillay J, et al. Immune suppression by neutrophils and granulocytic myeloid-derived suppressor cells: similarities and differences. Cell Mol Life Sci. 2013;70(20):3813–27.PubMedPubMedCentralCrossRef Pillay J, et al. Immune suppression by neutrophils and granulocytic myeloid-derived suppressor cells: similarities and differences. Cell Mol Life Sci. 2013;70(20):3813–27.PubMedPubMedCentralCrossRef
173.
go back to reference Rodriguez PC, et al. Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res. 2009;69(4):1553–60.PubMedPubMedCentralCrossRef Rodriguez PC, et al. Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res. 2009;69(4):1553–60.PubMedPubMedCentralCrossRef
174.
go back to reference Youn JI, et al. Characterization of the nature of granulocytic myeloid-derived suppressor cells in tumor-bearing mice. J Leukoc Biol. 2012;91(1):167–81.PubMedPubMedCentralCrossRef Youn JI, et al. Characterization of the nature of granulocytic myeloid-derived suppressor cells in tumor-bearing mice. J Leukoc Biol. 2012;91(1):167–81.PubMedPubMedCentralCrossRef
175.
176.
178.
go back to reference Condamine T, et al. Regulation of tumor metastasis by myeloid-derived suppressor cells. Annu Rev Med. 2015;66:97–110.PubMedCrossRef Condamine T, et al. Regulation of tumor metastasis by myeloid-derived suppressor cells. Annu Rev Med. 2015;66:97–110.PubMedCrossRef
179.
go back to reference Condamine T et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci Immunol, 2016. 1(2). Condamine T et al. Lectin-type oxidized LDL receptor-1 distinguishes population of human polymorphonuclear myeloid-derived suppressor cells in cancer patients. Sci Immunol, 2016. 1(2).
180.
go back to reference Corzo CA, et al. HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med. 2010;207(11):2439–53.PubMedPubMedCentralCrossRef Corzo CA, et al. HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med. 2010;207(11):2439–53.PubMedPubMedCentralCrossRef
181.
go back to reference Hossain F, et al. Inhibition of fatty acid oxidation modulates immunosuppressive functions of myeloid-derived suppressor cells and enhances Cancer therapies. Cancer Immunol Res. 2015;3(11):1236–47.PubMedPubMedCentralCrossRef Hossain F, et al. Inhibition of fatty acid oxidation modulates immunosuppressive functions of myeloid-derived suppressor cells and enhances Cancer therapies. Cancer Immunol Res. 2015;3(11):1236–47.PubMedPubMedCentralCrossRef
182.
go back to reference Zhou J, et al. Hepatoma-intrinsic CCRK inhibition diminishes myeloid-derived suppressor cell immunosuppression and enhances immune-checkpoint blockade efficacy. Gut. 2018;67(5):931–44.PubMedCrossRef Zhou J, et al. Hepatoma-intrinsic CCRK inhibition diminishes myeloid-derived suppressor cell immunosuppression and enhances immune-checkpoint blockade efficacy. Gut. 2018;67(5):931–44.PubMedCrossRef
183.
go back to reference Wu C, et al. Repolarization of myeloid derived suppressor cells via magnetic nanoparticles to promote radiotherapy for glioma treatment. Nanomedicine. 2019;16:126–37.PubMedCrossRef Wu C, et al. Repolarization of myeloid derived suppressor cells via magnetic nanoparticles to promote radiotherapy for glioma treatment. Nanomedicine. 2019;16:126–37.PubMedCrossRef
184.
go back to reference Ding M, et al. Augmenting immunogenic cell death and alleviating myeloid-derived suppressor cells by Sono-Activatable Semiconducting Polymer Nanopartners for Immunotherapy. Adv Mater. 2023;35(33):e2302508.PubMedCrossRef Ding M, et al. Augmenting immunogenic cell death and alleviating myeloid-derived suppressor cells by Sono-Activatable Semiconducting Polymer Nanopartners for Immunotherapy. Adv Mater. 2023;35(33):e2302508.PubMedCrossRef
185.
go back to reference Zuo H, et al. Circumventing myeloid-derived suppressor cell-mediated immunosuppression using an oxygen-generated and -economized nanoplatform. ACS Appl Mater Interfaces. 2020;12(50):55723–36.PubMedCrossRef Zuo H, et al. Circumventing myeloid-derived suppressor cell-mediated immunosuppression using an oxygen-generated and -economized nanoplatform. ACS Appl Mater Interfaces. 2020;12(50):55723–36.PubMedCrossRef
186.
go back to reference Habanjar O et al. Crosstalk of inflammatory cytokines within the breast Tumor Microenvironment. Int J Mol Sci, 2023. 24(4). Habanjar O et al. Crosstalk of inflammatory cytokines within the breast Tumor Microenvironment. Int J Mol Sci, 2023. 24(4).
189.
go back to reference Bachelerie F, et al. International Union of Basic and Clinical Pharmacology. [corrected]. LXXXIX. Update on the extended family of chemokine receptors and introducing a new nomenclature for atypical chemokine receptors. Pharmacol Rev. 2014;66(1):1–79.PubMedPubMedCentralCrossRef Bachelerie F, et al. International Union of Basic and Clinical Pharmacology. [corrected]. LXXXIX. Update on the extended family of chemokine receptors and introducing a new nomenclature for atypical chemokine receptors. Pharmacol Rev. 2014;66(1):1–79.PubMedPubMedCentralCrossRef
190.
go back to reference Mélik-Parsadaniantz S, Rostène W. Chemokines and neuromodulation. J Neuroimmunol. 2008;198(1–2):62–8.PubMedCrossRef Mélik-Parsadaniantz S, Rostène W. Chemokines and neuromodulation. J Neuroimmunol. 2008;198(1–2):62–8.PubMedCrossRef
193.
go back to reference Akdis M et al. Interleukins, from 1 to 37, and interferon-γ: receptors, functions, and roles in diseases J Allergy Clin Immunol, 2011;127(3):701 – 21.e1-70. Akdis M et al. Interleukins, from 1 to 37, and interferon-γ: receptors, functions, and roles in diseases J Allergy Clin Immunol, 2011;127(3):701 – 21.e1-70.
194.
go back to reference Shin E, Koo JS. The role of Adipokines and bone marrow adipocytes in breast Cancer bone metastasis. Int J Mol Sci, 2020. 21(14). Shin E, Koo JS. The role of Adipokines and bone marrow adipocytes in breast Cancer bone metastasis. Int J Mol Sci, 2020. 21(14).
195.
go back to reference Blüher M, Mantzoros CS. From leptin to other adipokines in health and disease: facts and expectations at the beginning of the 21st century. Metabolism. 2015;64(1):131–45.PubMedCrossRef Blüher M, Mantzoros CS. From leptin to other adipokines in health and disease: facts and expectations at the beginning of the 21st century. Metabolism. 2015;64(1):131–45.PubMedCrossRef
197.
go back to reference Barchetta I, et al. Sick fat: the good and the bad of old and new circulating markers of adipose tissue inflammation. J Endocrinol Invest. 2019;42(11):1257–72.PubMedCrossRef Barchetta I, et al. Sick fat: the good and the bad of old and new circulating markers of adipose tissue inflammation. J Endocrinol Invest. 2019;42(11):1257–72.PubMedCrossRef
198.
go back to reference Nehme R et al. Targeting adiponectin in breast Cancer. Biomedicines, 2022. 10(11). Nehme R et al. Targeting adiponectin in breast Cancer. Biomedicines, 2022. 10(11).
199.
200.
go back to reference Ferrer I, et al. Transforming growth factor-alpha (TGF-alpha) and epidermal growth factor-receptor (EGF-R) immunoreactivity in normal and pathologic brain. Prog Neurobiol. 1996;49(2):99–123.PubMedCrossRef Ferrer I, et al. Transforming growth factor-alpha (TGF-alpha) and epidermal growth factor-receptor (EGF-R) immunoreactivity in normal and pathologic brain. Prog Neurobiol. 1996;49(2):99–123.PubMedCrossRef
201.
202.
go back to reference Isaacs A, Lindenmann J. Virus interference. I. The interferon. Proc R Soc Lond B Biol Sci. 1957;147(927):258–67.PubMedCrossRef Isaacs A, Lindenmann J. Virus interference. I. The interferon. Proc R Soc Lond B Biol Sci. 1957;147(927):258–67.PubMedCrossRef
203.
go back to reference Schroder K, et al. Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol. 2004;75(2):163–89.PubMedCrossRef Schroder K, et al. Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol. 2004;75(2):163–89.PubMedCrossRef
204.
go back to reference Peng S, et al. Tumor-microenvironment-responsive nanomedicine for enhanced Cancer Immunotherapy. Adv Sci (Weinh). 2022;9(1):e2103836.PubMedCrossRef Peng S, et al. Tumor-microenvironment-responsive nanomedicine for enhanced Cancer Immunotherapy. Adv Sci (Weinh). 2022;9(1):e2103836.PubMedCrossRef
206.
go back to reference Hu J, Zhang G, Liu S. Enzyme-responsive polymeric assemblies, nanoparticles and hydrogels. Chem Soc Rev. 2012;41(18):5933–49.PubMedCrossRef Hu J, Zhang G, Liu S. Enzyme-responsive polymeric assemblies, nanoparticles and hydrogels. Chem Soc Rev. 2012;41(18):5933–49.PubMedCrossRef
207.
go back to reference Shahriari M, et al. Enzyme responsive drug delivery systems in cancer treatment. J Control Release. 2019;308:172–89.PubMedCrossRef Shahriari M, et al. Enzyme responsive drug delivery systems in cancer treatment. J Control Release. 2019;308:172–89.PubMedCrossRef
208.
go back to reference Qiu N, et al. Esterase-activated charge-reversal polymer for fibroblast-exempt Cancer Gene Therapy. Adv Mater. 2016;28(48):10613–22.PubMedCrossRef Qiu N, et al. Esterase-activated charge-reversal polymer for fibroblast-exempt Cancer Gene Therapy. Adv Mater. 2016;28(48):10613–22.PubMedCrossRef
209.
210.
go back to reference Li H, et al. The relationship between MMP-2 and MMP-9 expression levels with breast cancer incidence and prognosis. Oncol Lett. 2017;14(5):5865–70.PubMedPubMedCentral Li H, et al. The relationship between MMP-2 and MMP-9 expression levels with breast cancer incidence and prognosis. Oncol Lett. 2017;14(5):5865–70.PubMedPubMedCentral
211.
go back to reference Pham HT, Block NL, Lokeshwar VB. Tumor-derived hyaluronidase: a diagnostic urine marker for high-grade bladder cancer. Cancer Res. 1997;57(4):778–83.PubMed Pham HT, Block NL, Lokeshwar VB. Tumor-derived hyaluronidase: a diagnostic urine marker for high-grade bladder cancer. Cancer Res. 1997;57(4):778–83.PubMed
212.
go back to reference Moreira AM et al. The Extracellular Matrix: an accomplice in gastric Cancer Development and Progression. Cells, 2020. 9(2). Moreira AM et al. The Extracellular Matrix: an accomplice in gastric Cancer Development and Progression. Cells, 2020. 9(2).
213.
214.
go back to reference Mohan V, Das A, Sagi I. Emerging roles of ECM remodeling processes in cancer. Semin Cancer Biol. 2020;62:192–200.PubMedCrossRef Mohan V, Das A, Sagi I. Emerging roles of ECM remodeling processes in cancer. Semin Cancer Biol. 2020;62:192–200.PubMedCrossRef
215.
go back to reference Sung JY, Cheong JH. The Matrisome is Associated with metabolic reprograming in stem-like phenotypes of gastric Cancer. Cancers (Basel), 2022. 14(6). Sung JY, Cheong JH. The Matrisome is Associated with metabolic reprograming in stem-like phenotypes of gastric Cancer. Cancers (Basel), 2022. 14(6).
216.
217.
go back to reference Jang M, et al. Increased extracellular matrix density disrupts E-cadherin/β-catenin complex in gastric cancer cells. Biomater Sci. 2018;6(10):2704–13.PubMedCrossRef Jang M, et al. Increased extracellular matrix density disrupts E-cadherin/β-catenin complex in gastric cancer cells. Biomater Sci. 2018;6(10):2704–13.PubMedCrossRef
218.
go back to reference Acerbi I, et al. Human breast cancer invasion and aggression correlates with ECM stiffening and immune cell infiltration. Integr Biol (Camb). 2015;7(10):1120–34.PubMedCrossRef Acerbi I, et al. Human breast cancer invasion and aggression correlates with ECM stiffening and immune cell infiltration. Integr Biol (Camb). 2015;7(10):1120–34.PubMedCrossRef
222.
223.
go back to reference Saito G, Swanson JA, Lee K-D. Drug delivery strategy utilizing conjugation via reversible disulfide linkages: role and site of cellular reducing activities. Adv Drug Deliv Rev. 2003;55(2):199–215.PubMedCrossRef Saito G, Swanson JA, Lee K-D. Drug delivery strategy utilizing conjugation via reversible disulfide linkages: role and site of cellular reducing activities. Adv Drug Deliv Rev. 2003;55(2):199–215.PubMedCrossRef
224.
225.
go back to reference Naughton D. Drug targeting to hypoxic tissue using self-inactivating bioreductive delivery systems. Adv Drug Deliv Rev. 2001;53(2):229–33.PubMedCrossRef Naughton D. Drug targeting to hypoxic tissue using self-inactivating bioreductive delivery systems. Adv Drug Deliv Rev. 2001;53(2):229–33.PubMedCrossRef
227.
go back to reference Li Q, et al. PRDM1/BLIMP1 induces cancer immune evasion by modulating the USP22-SPI1-PD-L1 axis in hepatocellular carcinoma cells. Nat Commun. 2022;13(1):7677.PubMedPubMedCentralCrossRef Li Q, et al. PRDM1/BLIMP1 induces cancer immune evasion by modulating the USP22-SPI1-PD-L1 axis in hepatocellular carcinoma cells. Nat Commun. 2022;13(1):7677.PubMedPubMedCentralCrossRef
228.
go back to reference Yang H, et al. RNF31 represses cell progression and immune evasion via YAP/PD-L1 suppression in triple negative breast Cancer. J Exp Clin Cancer Res. 2022;41(1):364.PubMedPubMedCentralCrossRef Yang H, et al. RNF31 represses cell progression and immune evasion via YAP/PD-L1 suppression in triple negative breast Cancer. J Exp Clin Cancer Res. 2022;41(1):364.PubMedPubMedCentralCrossRef
230.
go back to reference Wang JX et al. Lactic acid and an acidic Tumor Microenvironment suppress Anticancer Immunity. Int J Mol Sci, 2020. 21(21). Wang JX et al. Lactic acid and an acidic Tumor Microenvironment suppress Anticancer Immunity. Int J Mol Sci, 2020. 21(21).
232.
go back to reference Zhang B et al. Tumor CEMIP drives immune evasion of colorectal cancer via MHC-I internalization and degradation. J Immunother Cancer, 2023. 11(1). Zhang B et al. Tumor CEMIP drives immune evasion of colorectal cancer via MHC-I internalization and degradation. J Immunother Cancer, 2023. 11(1).
233.
go back to reference Quah HS, et al. Single cell analysis in head and neck cancer reveals potential immune evasion mechanisms during early metastasis. Nat Commun. 2023;14(1):1680.PubMedPubMedCentralCrossRef Quah HS, et al. Single cell analysis in head and neck cancer reveals potential immune evasion mechanisms during early metastasis. Nat Commun. 2023;14(1):1680.PubMedPubMedCentralCrossRef
234.
go back to reference Bergholz JS, et al. PI3Kβ controls immune evasion in PTEN-deficient breast tumours. Nature. 2023;617(7959):139–46.PubMedCrossRef Bergholz JS, et al. PI3Kβ controls immune evasion in PTEN-deficient breast tumours. Nature. 2023;617(7959):139–46.PubMedCrossRef
235.
go back to reference Barriga FM, et al. MACHETE identifies interferon-encompassing chromosome 9p21.3 deletions as mediators of immune evasion and metastasis. Nat Cancer. 2022;3(11):1367–85.PubMedPubMedCentralCrossRef Barriga FM, et al. MACHETE identifies interferon-encompassing chromosome 9p21.3 deletions as mediators of immune evasion and metastasis. Nat Cancer. 2022;3(11):1367–85.PubMedPubMedCentralCrossRef
237.
go back to reference Lidström T, et al. Extracellular galectin 4 drives Immune Evasion and promotes T-cell apoptosis in pancreatic Cancer. Cancer Immunol Res. 2023;11(1):72–92.PubMedCrossRef Lidström T, et al. Extracellular galectin 4 drives Immune Evasion and promotes T-cell apoptosis in pancreatic Cancer. Cancer Immunol Res. 2023;11(1):72–92.PubMedCrossRef
238.
go back to reference Vinay DS, et al. Immune evasion in cancer: mechanistic basis and therapeutic strategies. Sem Cancer Biol. 2015;35:S185–98.CrossRef Vinay DS, et al. Immune evasion in cancer: mechanistic basis and therapeutic strategies. Sem Cancer Biol. 2015;35:S185–98.CrossRef
239.
go back to reference Polyak K, et al. Breast tumor heterogeneity: causes and consequences. Breast Cancer Res. 2009;11:1–1.CrossRef Polyak K, et al. Breast tumor heterogeneity: causes and consequences. Breast Cancer Res. 2009;11:1–1.CrossRef
240.
241.
go back to reference Nicolson GL. Generation of phenotypic diversity and progression in metastatic tumor cells. Cancer Metastasis Rev. 1984;3:25–42.PubMedCrossRef Nicolson GL. Generation of phenotypic diversity and progression in metastatic tumor cells. Cancer Metastasis Rev. 1984;3:25–42.PubMedCrossRef
242.
go back to reference Merlo LM, et al. Cancer as an evolutionary and ecological process. Nat Rev Cancer. 2006;6(12):924–35.PubMedCrossRef Merlo LM, et al. Cancer as an evolutionary and ecological process. Nat Rev Cancer. 2006;6(12):924–35.PubMedCrossRef
243.
go back to reference Sceneay J, et al. Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G + immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche. Cancer Res. 2012;72(16):3906–11.PubMedCrossRef Sceneay J, et al. Primary tumor hypoxia recruits CD11b+/Ly6Cmed/Ly6G + immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche. Cancer Res. 2012;72(16):3906–11.PubMedCrossRef
244.
go back to reference Gabay C, Kushner I. Acute-phase proteins and other systemic responses to inflammation. N Engl J Med. 1999;340(6):448–54.PubMedCrossRef Gabay C, Kushner I. Acute-phase proteins and other systemic responses to inflammation. N Engl J Med. 1999;340(6):448–54.PubMedCrossRef
245.
go back to reference Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004;22:329–60.PubMedCrossRef Dunn GP, Old LJ, Schreiber RD. The three Es of cancer immunoediting. Annu Rev Immunol. 2004;22:329–60.PubMedCrossRef
246.
go back to reference Lauritzsen GF, et al. Clonal deletion of thymocytes as a tumor escape mechanism. Int J Cancer. 1998;78(2):216–22.PubMedCrossRef Lauritzsen GF, et al. Clonal deletion of thymocytes as a tumor escape mechanism. Int J Cancer. 1998;78(2):216–22.PubMedCrossRef
247.
249.
go back to reference Yang K, et al. Zinc cyclic di-AMP nanoparticles target and suppress tumours via endothelial STING activation and tumour-associated macrophage reinvigoration. Nat Nanotechnol. 2022;17(12):1322–31.PubMedCrossRef Yang K, et al. Zinc cyclic di-AMP nanoparticles target and suppress tumours via endothelial STING activation and tumour-associated macrophage reinvigoration. Nat Nanotechnol. 2022;17(12):1322–31.PubMedCrossRef
250.
252.
go back to reference Jiang Q, et al. Platelet membrane-camouflaged magnetic nanoparticles for ferroptosis-enhanced Cancer Immunotherapy. Small. 2020;16(22):e2001704.PubMedCrossRef Jiang Q, et al. Platelet membrane-camouflaged magnetic nanoparticles for ferroptosis-enhanced Cancer Immunotherapy. Small. 2020;16(22):e2001704.PubMedCrossRef
253.
go back to reference Melamed JR, et al. Ionizable lipid nanoparticles deliver mRNA to pancreatic β cells via macrophage-mediated gene transfer. Sci Adv. 2023;9(4):eade1444.PubMedPubMedCentralCrossRef Melamed JR, et al. Ionizable lipid nanoparticles deliver mRNA to pancreatic β cells via macrophage-mediated gene transfer. Sci Adv. 2023;9(4):eade1444.PubMedPubMedCentralCrossRef
254.
go back to reference Chen C, et al. Tumor-Associated-Macrophage-membrane-coated nanoparticles for improved photodynamic immunotherapy. Nano Lett. 2021;21(13):5522–31.PubMedCrossRef Chen C, et al. Tumor-Associated-Macrophage-membrane-coated nanoparticles for improved photodynamic immunotherapy. Nano Lett. 2021;21(13):5522–31.PubMedCrossRef
255.
go back to reference Ramesh A, et al. CSF1R- and SHP2-Inhibitor-loaded nanoparticles enhance cytotoxic activity and phagocytosis in Tumor-Associated macrophages. Adv Mater. 2019;31(51):e1904364.PubMedCrossRef Ramesh A, et al. CSF1R- and SHP2-Inhibitor-loaded nanoparticles enhance cytotoxic activity and phagocytosis in Tumor-Associated macrophages. Adv Mater. 2019;31(51):e1904364.PubMedCrossRef
256.
go back to reference Zanganeh S, et al. Iron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissues. Nat Nanotechnol. 2016;11(11):986–94.PubMedPubMedCentralCrossRef Zanganeh S, et al. Iron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissues. Nat Nanotechnol. 2016;11(11):986–94.PubMedPubMedCentralCrossRef
257.
go back to reference Chim LK, et al. Tumor-associated macrophages induce inflammation and drug resistance in a mechanically tunable engineered model of osteosarcoma. Biomaterials. 2023;296:122076.PubMedCrossRef Chim LK, et al. Tumor-associated macrophages induce inflammation and drug resistance in a mechanically tunable engineered model of osteosarcoma. Biomaterials. 2023;296:122076.PubMedCrossRef
258.
go back to reference Rohila D, et al. Syk Inhibition reprograms Tumor-Associated macrophages and overcomes Gemcitabine-Induced Immunosuppression in Pancreatic Ductal Adenocarcinoma. Cancer Res. 2023;83(16):2675–89.PubMedPubMedCentralCrossRef Rohila D, et al. Syk Inhibition reprograms Tumor-Associated macrophages and overcomes Gemcitabine-Induced Immunosuppression in Pancreatic Ductal Adenocarcinoma. Cancer Res. 2023;83(16):2675–89.PubMedPubMedCentralCrossRef
259.
go back to reference Xie R, et al. Furin-instructed aggregated gold nanoparticles for re-educating tumor associated macrophages and overcoming breast cancer chemoresistance. Biomaterials. 2021;275:120891.PubMedCrossRef Xie R, et al. Furin-instructed aggregated gold nanoparticles for re-educating tumor associated macrophages and overcoming breast cancer chemoresistance. Biomaterials. 2021;275:120891.PubMedCrossRef
260.
go back to reference Shen W, et al. Metabolic homeostasis-regulated nanoparticles for antibody-independent Cancer Radio-Immunotherapy. Adv Mater. 2022;34(51):e2207343.PubMedCrossRef Shen W, et al. Metabolic homeostasis-regulated nanoparticles for antibody-independent Cancer Radio-Immunotherapy. Adv Mater. 2022;34(51):e2207343.PubMedCrossRef
261.
go back to reference Sun M, et al. Fibroblast activation Protein-α responsive peptide assembling Prodrug nanoparticles for Remodeling the Immunosuppressive Microenvironment and Boosting Cancer Immunotherapy. Small. 2022;18(9):e2106296.PubMedCrossRef Sun M, et al. Fibroblast activation Protein-α responsive peptide assembling Prodrug nanoparticles for Remodeling the Immunosuppressive Microenvironment and Boosting Cancer Immunotherapy. Small. 2022;18(9):e2106296.PubMedCrossRef
262.
go back to reference Gao C, et al. Injectable Immunotherapeutic Hydrogel containing RNA-Loaded lipid nanoparticles reshapes Tumor Microenvironment for Pancreatic Cancer Therapy. Nano Lett. 2022;22(22):8801–9.PubMedCrossRef Gao C, et al. Injectable Immunotherapeutic Hydrogel containing RNA-Loaded lipid nanoparticles reshapes Tumor Microenvironment for Pancreatic Cancer Therapy. Nano Lett. 2022;22(22):8801–9.PubMedCrossRef
263.
go back to reference Yoon J, et al. Macrophage-reprogramming upconverting nanoparticles for enhanced TAM-mediated antitumor therapy of hypoxic breast cancer. J Control Release. 2023;360:482–95.PubMedCrossRef Yoon J, et al. Macrophage-reprogramming upconverting nanoparticles for enhanced TAM-mediated antitumor therapy of hypoxic breast cancer. J Control Release. 2023;360:482–95.PubMedCrossRef
264.
go back to reference Rong L, et al. Iron chelated melanin-like nanoparticles for tumor-associated macrophage repolarization and cancer therapy. Biomaterials. 2019;225:119515.PubMedCrossRef Rong L, et al. Iron chelated melanin-like nanoparticles for tumor-associated macrophage repolarization and cancer therapy. Biomaterials. 2019;225:119515.PubMedCrossRef
265.
go back to reference Ramesh A, et al. Dual inhibition of CSF1R and MAPK pathways using supramolecular nanoparticles enhances macrophage immunotherapy. Biomaterials. 2020;227:119559.PubMedCrossRef Ramesh A, et al. Dual inhibition of CSF1R and MAPK pathways using supramolecular nanoparticles enhances macrophage immunotherapy. Biomaterials. 2020;227:119559.PubMedCrossRef
266.
go back to reference He Y, et al. Targeted MIP-3β plasmid nanoparticles induce dendritic cell maturation and inhibit M2 macrophage polarisation to suppress cancer growth. Biomaterials. 2020;249:120046.PubMedCrossRef He Y, et al. Targeted MIP-3β plasmid nanoparticles induce dendritic cell maturation and inhibit M2 macrophage polarisation to suppress cancer growth. Biomaterials. 2020;249:120046.PubMedCrossRef
267.
go back to reference Su WP, et al. Polyaniline-based glyco-condensation on au nanoparticles enhances Immunotherapy in Lung Cancer. ACS Appl Mater Interfaces. 2022;14(21):24144–59.PubMedCrossRef Su WP, et al. Polyaniline-based glyco-condensation on au nanoparticles enhances Immunotherapy in Lung Cancer. ACS Appl Mater Interfaces. 2022;14(21):24144–59.PubMedCrossRef
268.
go back to reference Yang S, et al. CaCO(3)-Encapsulated au nanoparticles modulate macrophages toward M1-like phenotype. ACS Appl Bio Mater. 2021;4(4):3214–23.PubMedCrossRef Yang S, et al. CaCO(3)-Encapsulated au nanoparticles modulate macrophages toward M1-like phenotype. ACS Appl Bio Mater. 2021;4(4):3214–23.PubMedCrossRef
269.
go back to reference Wan J, et al. Biodegradable NIR-II Pseudo Conjugate Polymeric nanoparticles amplify photodynamic immunotherapy via alleviation of Tumor Hypoxia and Tumor-Associated Macrophage Reprogramming. Adv Mater. 2023;35(31):e2209799.PubMedCrossRef Wan J, et al. Biodegradable NIR-II Pseudo Conjugate Polymeric nanoparticles amplify photodynamic immunotherapy via alleviation of Tumor Hypoxia and Tumor-Associated Macrophage Reprogramming. Adv Mater. 2023;35(31):e2209799.PubMedCrossRef
270.
go back to reference Li L, et al. Functional Gadofullerene nanoparticles trigger Robust Cancer Immunotherapy based on rebuilding an immunosuppressive Tumor Microenvironment. Nano Lett. 2020;20(6):4487–96.PubMedCrossRef Li L, et al. Functional Gadofullerene nanoparticles trigger Robust Cancer Immunotherapy based on rebuilding an immunosuppressive Tumor Microenvironment. Nano Lett. 2020;20(6):4487–96.PubMedCrossRef
271.
go back to reference Guo Q, et al. Dandelion-Like Tailorable nanoparticles for Tumor Microenvironment Modulation. Adv Sci (Weinh). 2019;6(21):1901430.PubMedCrossRef Guo Q, et al. Dandelion-Like Tailorable nanoparticles for Tumor Microenvironment Modulation. Adv Sci (Weinh). 2019;6(21):1901430.PubMedCrossRef
272.
go back to reference Liu Y, et al. Targeting Tumor-Associated macrophages by MMP2-Sensitive apoptotic body-mimicking nanoparticles. ACS Appl Mater Interfaces. 2020;12(47):52402–14.PubMedPubMedCentralCrossRef Liu Y, et al. Targeting Tumor-Associated macrophages by MMP2-Sensitive apoptotic body-mimicking nanoparticles. ACS Appl Mater Interfaces. 2020;12(47):52402–14.PubMedPubMedCentralCrossRef
273.
go back to reference Parayath NN, Parikh A, Amiji MM. Repolarization of Tumor-Associated macrophages in a genetically Engineered Nonsmall Cell Lung Cancer Model by Intraperitoneal Administration of Hyaluronic Acid-based nanoparticles encapsulating MicroRNA-125b. Nano Lett. 2018;18(6):3571–9.PubMedCrossRef Parayath NN, Parikh A, Amiji MM. Repolarization of Tumor-Associated macrophages in a genetically Engineered Nonsmall Cell Lung Cancer Model by Intraperitoneal Administration of Hyaluronic Acid-based nanoparticles encapsulating MicroRNA-125b. Nano Lett. 2018;18(6):3571–9.PubMedCrossRef
274.
go back to reference Xu L, et al. Engineering the Intestinal Lymphatic Transport of oral nanoparticles to Educate macrophages for Cancer Combined Immunotherapy. ACS Nano. 2023;17(12):11817–37.PubMedCrossRef Xu L, et al. Engineering the Intestinal Lymphatic Transport of oral nanoparticles to Educate macrophages for Cancer Combined Immunotherapy. ACS Nano. 2023;17(12):11817–37.PubMedCrossRef
275.
go back to reference Zhang Y, et al. Gold nanoparticles inhibit activation of cancer-associated fibroblasts by disrupting communication from tumor and microenvironmental cells. Bioact Mater. 2021;6(2):326–32.PubMed Zhang Y, et al. Gold nanoparticles inhibit activation of cancer-associated fibroblasts by disrupting communication from tumor and microenvironmental cells. Bioact Mater. 2021;6(2):326–32.PubMed
276.
go back to reference Kovács D, et al. Core-shell nanoparticles suppress metastasis and modify the tumour-supportive activity of cancer-associated fibroblasts. J Nanobiotechnol. 2020;18(1):18.CrossRef Kovács D, et al. Core-shell nanoparticles suppress metastasis and modify the tumour-supportive activity of cancer-associated fibroblasts. J Nanobiotechnol. 2020;18(1):18.CrossRef
277.
go back to reference Lopez S, et al. Magneto-mechanical destruction of cancer-associated fibroblasts using ultra-small iron oxide nanoparticles and low frequency rotating magnetic fields. Nanoscale Adv. 2022;4(2):421–36.PubMedCrossRef Lopez S, et al. Magneto-mechanical destruction of cancer-associated fibroblasts using ultra-small iron oxide nanoparticles and low frequency rotating magnetic fields. Nanoscale Adv. 2022;4(2):421–36.PubMedCrossRef
278.
go back to reference Li L, et al. Photosensitizer-encapsulated ferritins mediate photodynamic therapy against Cancer-Associated fibroblasts and improve Tumor Accumulation of nanoparticles. Mol Pharm. 2018;15(8):3595–9.PubMedPubMedCentralCrossRef Li L, et al. Photosensitizer-encapsulated ferritins mediate photodynamic therapy against Cancer-Associated fibroblasts and improve Tumor Accumulation of nanoparticles. Mol Pharm. 2018;15(8):3595–9.PubMedPubMedCentralCrossRef
279.
go back to reference Jin J, et al. Human Cancer Cell membrane-coated biomimetic nanoparticles reduce fibroblast-mediated Invasion and Metastasis and induce T-Cells. ACS Appl Mater Interfaces. 2019;11(8):7850–61.PubMedPubMedCentralCrossRef Jin J, et al. Human Cancer Cell membrane-coated biomimetic nanoparticles reduce fibroblast-mediated Invasion and Metastasis and induce T-Cells. ACS Appl Mater Interfaces. 2019;11(8):7850–61.PubMedPubMedCentralCrossRef
280.
go back to reference Zhou S et al. FAP-Targeted photodynamic therapy mediated by Ferritin nanoparticles elicits an Immune response against Cancer cells and Cancer Associated fibroblasts. Adv Funct Mater, 2021. 31(7). Zhou S et al. FAP-Targeted photodynamic therapy mediated by Ferritin nanoparticles elicits an Immune response against Cancer cells and Cancer Associated fibroblasts. Adv Funct Mater, 2021. 31(7).
281.
go back to reference Yu Q, et al. Targeting cancer-associated fibroblasts by dual-responsive lipid-albumin nanoparticles to enhance drug perfusion for pancreatic tumor therapy. J Control Release. 2020;321:564–75.PubMedCrossRef Yu Q, et al. Targeting cancer-associated fibroblasts by dual-responsive lipid-albumin nanoparticles to enhance drug perfusion for pancreatic tumor therapy. J Control Release. 2020;321:564–75.PubMedCrossRef
282.
283.
284.
go back to reference Xiong J, et al. Cancer-Erythrocyte Hybrid membrane-camouflaged magnetic nanoparticles with enhanced photothermal-immunotherapy for ovarian Cancer. ACS Nano. 2021;15(12):19756–70.PubMedCrossRef Xiong J, et al. Cancer-Erythrocyte Hybrid membrane-camouflaged magnetic nanoparticles with enhanced photothermal-immunotherapy for ovarian Cancer. ACS Nano. 2021;15(12):19756–70.PubMedCrossRef
285.
286.
go back to reference Billingsley MM, et al. Orthogonal Design of experiments for optimization of lipid nanoparticles for mRNA Engineering of CAR T cells. Nano Lett. 2022;22(1):533–42.PubMedCrossRef Billingsley MM, et al. Orthogonal Design of experiments for optimization of lipid nanoparticles for mRNA Engineering of CAR T cells. Nano Lett. 2022;22(1):533–42.PubMedCrossRef
287.
go back to reference Lin YX et al. Reactivation of the tumor suppressor PTEN by mRNA nanoparticles enhances antitumor immunity in preclinical models Sci Transl Med, 2021. 13(599). Lin YX et al. Reactivation of the tumor suppressor PTEN by mRNA nanoparticles enhances antitumor immunity in preclinical models Sci Transl Med, 2021. 13(599).
288.
go back to reference Jiang Y, et al. Engineered cell-membrane-coated nanoparticles directly Present Tumor antigens to promote anticancer immunity. Adv Mater. 2020;32(30):e2001808.PubMedPubMedCentralCrossRef Jiang Y, et al. Engineered cell-membrane-coated nanoparticles directly Present Tumor antigens to promote anticancer immunity. Adv Mater. 2020;32(30):e2001808.PubMedPubMedCentralCrossRef
289.
go back to reference Li Z, et al. Immunogenic cell death augmented by Manganese Zinc Sulfide nanoparticles for metastatic Melanoma Immunotherapy. ACS Nano. 2022;16(9):15471–83.PubMedCrossRef Li Z, et al. Immunogenic cell death augmented by Manganese Zinc Sulfide nanoparticles for metastatic Melanoma Immunotherapy. ACS Nano. 2022;16(9):15471–83.PubMedCrossRef
290.
go back to reference Cao L, et al. Activating cGAS-STING pathway with ROS-responsive nanoparticles delivering a hybrid prodrug for enhanced chemo-immunotherapy. Biomaterials. 2022;290:121856.PubMedCrossRef Cao L, et al. Activating cGAS-STING pathway with ROS-responsive nanoparticles delivering a hybrid prodrug for enhanced chemo-immunotherapy. Biomaterials. 2022;290:121856.PubMedCrossRef
291.
go back to reference Chen C, et al. Asynchronous blockade of PD-L1 and CD155 by polymeric nanoparticles inhibits triple-negative breast cancer progression and metastasis. Biomaterials. 2021;275:p120988.CrossRef Chen C, et al. Asynchronous blockade of PD-L1 and CD155 by polymeric nanoparticles inhibits triple-negative breast cancer progression and metastasis. Biomaterials. 2021;275:p120988.CrossRef
292.
go back to reference Tang S, et al. MnO(2)-melittin nanoparticles serve as an effective anti-tumor immunotherapy by enhancing systemic immune response. Biomaterials. 2022;288:121706.PubMedCrossRef Tang S, et al. MnO(2)-melittin nanoparticles serve as an effective anti-tumor immunotherapy by enhancing systemic immune response. Biomaterials. 2022;288:121706.PubMedCrossRef
293.
go back to reference Sun F, et al. Regulating glucose metabolism with Prodrug nanoparticles for promoting photoimmunotherapy of pancreatic Cancer. Adv Sci (Weinh). 2021;8(4):2002746.PubMedCrossRef Sun F, et al. Regulating glucose metabolism with Prodrug nanoparticles for promoting photoimmunotherapy of pancreatic Cancer. Adv Sci (Weinh). 2021;8(4):2002746.PubMedCrossRef
294.
go back to reference Chou PY, et al. Glycosylation of OVA antigen-loaded PLGA nanoparticles enhances DC-targeting for cancer vaccination. J Control Release. 2022;351:970–88.PubMedCrossRef Chou PY, et al. Glycosylation of OVA antigen-loaded PLGA nanoparticles enhances DC-targeting for cancer vaccination. J Control Release. 2022;351:970–88.PubMedCrossRef
295.
go back to reference Son S, et al. Induction of T-helper-17-cell-mediated anti-tumour immunity by pathogen-mimicking polymer nanoparticles. Nat Biomed Eng. 2023;7(1):72–84.PubMedCrossRef Son S, et al. Induction of T-helper-17-cell-mediated anti-tumour immunity by pathogen-mimicking polymer nanoparticles. Nat Biomed Eng. 2023;7(1):72–84.PubMedCrossRef
296.
go back to reference Kim KS, et al. Ovalbumin and poly(i:c) encapsulated dendritic cell-targeted nanoparticles for Immune activation in the small intestinal lymphatic system. Adv Healthc Mater. 2022;11(21):e2200909.PubMedPubMedCentralCrossRef Kim KS, et al. Ovalbumin and poly(i:c) encapsulated dendritic cell-targeted nanoparticles for Immune activation in the small intestinal lymphatic system. Adv Healthc Mater. 2022;11(21):e2200909.PubMedPubMedCentralCrossRef
297.
go back to reference Li S, et al. Pre-induced ICD membrane-coated carrier-free nanoparticles for the personalized Lung Cancer Immunotherapy. Small Methods. 2023;7(5):e2201569.PubMedCrossRef Li S, et al. Pre-induced ICD membrane-coated carrier-free nanoparticles for the personalized Lung Cancer Immunotherapy. Small Methods. 2023;7(5):e2201569.PubMedCrossRef
298.
go back to reference Zhang Z, et al. Antitumor Activity of Anti-mir-21 delivered through lipid nanoparticles. Adv Healthc Mater. 2023;12(6):e2202412.PubMedCrossRef Zhang Z, et al. Antitumor Activity of Anti-mir-21 delivered through lipid nanoparticles. Adv Healthc Mater. 2023;12(6):e2202412.PubMedCrossRef
299.
300.
go back to reference Ou W, et al. Combination of NIR therapy and regulatory T cell modulation using layer-by-layer hybrid nanoparticles for effective cancer photoimmunotherapy. Theranostics. 2018;8(17):4574–90.PubMedPubMedCentralCrossRef Ou W, et al. Combination of NIR therapy and regulatory T cell modulation using layer-by-layer hybrid nanoparticles for effective cancer photoimmunotherapy. Theranostics. 2018;8(17):4574–90.PubMedPubMedCentralCrossRef
301.
go back to reference Zang X, et al. Polymeric indoximod based prodrug nanoparticles with doxorubicin entrapment for inducing immunogenic cell death and improving the immunotherapy of breast cancer. J Mater Chem B. 2022;10(12):2019–27.PubMedCrossRef Zang X, et al. Polymeric indoximod based prodrug nanoparticles with doxorubicin entrapment for inducing immunogenic cell death and improving the immunotherapy of breast cancer. J Mater Chem B. 2022;10(12):2019–27.PubMedCrossRef
302.
go back to reference Yang T, et al. Dual polymeric prodrug co-assembled nanoparticles with precise ratiometric co-delivery of cisplatin and metformin for lung cancer chemoimmunotherapy. Biomater Sci. 2020;8(20):5698–714.PubMedCrossRef Yang T, et al. Dual polymeric prodrug co-assembled nanoparticles with precise ratiometric co-delivery of cisplatin and metformin for lung cancer chemoimmunotherapy. Biomater Sci. 2020;8(20):5698–714.PubMedCrossRef
303.
go back to reference 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.PubMedCrossRef 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.PubMedCrossRef
304.
go back to reference He Y, et al. Remodeling tumor immunosuppression with molecularly imprinted nanoparticles to enhance immunogenic cell death for cancer immunotherapy. J Control Release. 2023;362:44–57.PubMedCrossRef He Y, et al. Remodeling tumor immunosuppression with molecularly imprinted nanoparticles to enhance immunogenic cell death for cancer immunotherapy. J Control Release. 2023;362:44–57.PubMedCrossRef
305.
go back to reference Wan WJ, et al. Coadministration of iRGD peptide with ROS-sensitive nanoparticles co-delivering siFGL1 and siPD-L1 enhanced tumor immunotherapy. Acta Biomater. 2021;136:473–84.PubMedCrossRef Wan WJ, et al. Coadministration of iRGD peptide with ROS-sensitive nanoparticles co-delivering siFGL1 and siPD-L1 enhanced tumor immunotherapy. Acta Biomater. 2021;136:473–84.PubMedCrossRef
306.
go back to reference Wang W, et al. The development of Chiral nanoparticles to target NK cells and CD8(+) T cells for Cancer Immunotherapy. Adv Mater. 2022;34(16):e2109354.PubMedCrossRef Wang W, et al. The development of Chiral nanoparticles to target NK cells and CD8(+) T cells for Cancer Immunotherapy. Adv Mater. 2022;34(16):e2109354.PubMedCrossRef
307.
go back to reference Wang B, et al. Liposomes co-loaded with ursolic acid and ginsenoside Rg3 in the treatment of hepatocellular carcinoma. Acta Biochim Pol. 2021;68(4):711–5.PubMed Wang B, et al. Liposomes co-loaded with ursolic acid and ginsenoside Rg3 in the treatment of hepatocellular carcinoma. Acta Biochim Pol. 2021;68(4):711–5.PubMed
308.
go back to reference Yao H, et al. Cisplatin nanoparticles promote Intratumoral CD8(+) T cell priming via Antigen Presentation and T cell receptor crosstalk. Nano Lett. 2022;22(8):3328–39.PubMedCrossRef Yao H, et al. Cisplatin nanoparticles promote Intratumoral CD8(+) T cell priming via Antigen Presentation and T cell receptor crosstalk. Nano Lett. 2022;22(8):3328–39.PubMedCrossRef
309.
go back to reference Munakata L, et al. Lipid nanoparticles of Type-A CpG D35 suppress tumor growth by changing tumor immune-microenvironment and activate CD8 T cells in mice. J Control Release. 2019;313:106–19.PubMedCrossRef Munakata L, et al. Lipid nanoparticles of Type-A CpG D35 suppress tumor growth by changing tumor immune-microenvironment and activate CD8 T cells in mice. J Control Release. 2019;313:106–19.PubMedCrossRef
310.
go back to reference Chang HC, et al. Targeting and specific activation of Antigen-presenting cells by endogenous Antigen-Loaded nanoparticles elicits tumor-specific immunity. Adv Sci (Weinh). 2020;7(1):1900069.PubMedCrossRef Chang HC, et al. Targeting and specific activation of Antigen-presenting cells by endogenous Antigen-Loaded nanoparticles elicits tumor-specific immunity. Adv Sci (Weinh). 2020;7(1):1900069.PubMedCrossRef
311.
go back to reference Smith R, et al. Cationic nanoparticles enhance T cell tumor infiltration and antitumor immune responses to a melanoma vaccine. Sci Adv. 2022;8(29):eabk3150.PubMedPubMedCentralCrossRef Smith R, et al. Cationic nanoparticles enhance T cell tumor infiltration and antitumor immune responses to a melanoma vaccine. Sci Adv. 2022;8(29):eabk3150.PubMedPubMedCentralCrossRef
312.
go back to reference Gao Y, et al. Engineering platelets with PDL1 antibodies and Iron oxide nanoparticles for Postsurgical Cancer Immunotherapy. ACS Appl Bio Mater. 2023;6(1):257–66.PubMedCrossRef Gao Y, et al. Engineering platelets with PDL1 antibodies and Iron oxide nanoparticles for Postsurgical Cancer Immunotherapy. ACS Appl Bio Mater. 2023;6(1):257–66.PubMedCrossRef
313.
go back to reference Patel RB, et al. Development of an in situ Cancer vaccine via Combinational Radiation and bacterial-membrane-coated nanoparticles. Adv Mater. 2019;31(43):e1902626.PubMedPubMedCentralCrossRef Patel RB, et al. Development of an in situ Cancer vaccine via Combinational Radiation and bacterial-membrane-coated nanoparticles. Adv Mater. 2019;31(43):e1902626.PubMedPubMedCentralCrossRef
314.
go back to reference Choi J, et al. Visible-light-triggered Prodrug nanoparticles combine chemotherapy and photodynamic therapy to Potentiate Checkpoint Blockade Cancer Immunotherapy. ACS Nano. 2021;15(7):12086–98.PubMedCrossRef Choi J, et al. Visible-light-triggered Prodrug nanoparticles combine chemotherapy and photodynamic therapy to Potentiate Checkpoint Blockade Cancer Immunotherapy. ACS Nano. 2021;15(7):12086–98.PubMedCrossRef
315.
go back to reference Ding B, et al. Biodegradable Upconversion nanoparticles induce pyroptosis for Cancer Immunotherapy. Nano Lett. 2021;21(19):8281–9.PubMedCrossRef Ding B, et al. Biodegradable Upconversion nanoparticles induce pyroptosis for Cancer Immunotherapy. Nano Lett. 2021;21(19):8281–9.PubMedCrossRef
316.
go back to reference Zhang L, et al. Glutathione-responsive nanoparticles of Camptothecin Prodrug for Cancer Therapy. Adv Sci (Weinh). 2023;10(3):e2205246.PubMedCrossRef Zhang L, et al. Glutathione-responsive nanoparticles of Camptothecin Prodrug for Cancer Therapy. Adv Sci (Weinh). 2023;10(3):e2205246.PubMedCrossRef
317.
go back to reference Zhang Y, et al. Gemcitabine nanoparticles promote antitumor immunity against melanoma. Biomaterials. 2019;189:48–59.PubMedCrossRef Zhang Y, et al. Gemcitabine nanoparticles promote antitumor immunity against melanoma. Biomaterials. 2019;189:48–59.PubMedCrossRef
318.
go back to reference Gowd V, et al. Advancement of cancer immunotherapy using nanoparticles-based nanomedicine. Sem Cancer Biol. 2022;86:624–44.CrossRef Gowd V, et al. Advancement of cancer immunotherapy using nanoparticles-based nanomedicine. Sem Cancer Biol. 2022;86:624–44.CrossRef
319.
go back to reference Casazza A, et al. Tumor stroma: a complexity dictated by the hypoxic tumor microenvironment. Oncogene. 2014;33(14):1743–54.PubMedCrossRef Casazza A, et al. Tumor stroma: a complexity dictated by the hypoxic tumor microenvironment. Oncogene. 2014;33(14):1743–54.PubMedCrossRef
320.
go back to reference Yu JL, et al. Effect of p53 status on tumor response to antiangiogenic therapy. Science. 2002;295(5559):1526–8.PubMedCrossRef Yu JL, et al. Effect of p53 status on tumor response to antiangiogenic therapy. Science. 2002;295(5559):1526–8.PubMedCrossRef
321.
322.
go back to reference Facciabene A, et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and Treg cells. Nature. 2011;475(7355):226–30.PubMedCrossRef Facciabene A, et al. Tumour hypoxia promotes tolerance and angiogenesis via CCL28 and Treg cells. Nature. 2011;475(7355):226–30.PubMedCrossRef
323.
go back to reference Doedens AL, et al. Macrophage expression of hypoxia-inducible factor-1α suppresses T-cell function and promotes tumor progression. Cancer Res. 2010;70(19):7465–75.PubMedPubMedCentralCrossRef Doedens AL, et al. Macrophage expression of hypoxia-inducible factor-1α suppresses T-cell function and promotes tumor progression. Cancer Res. 2010;70(19):7465–75.PubMedPubMedCentralCrossRef
324.
go back to reference Abou Khouzam R, et al. Hypoxia as a potential inducer of immune tolerance, tumor plasticity and a driver of tumor mutational burden: impact on cancer immunotherapy. Semin Cancer Biol. 2023;97:104–23.PubMedCrossRef Abou Khouzam R, et al. Hypoxia as a potential inducer of immune tolerance, tumor plasticity and a driver of tumor mutational burden: impact on cancer immunotherapy. Semin Cancer Biol. 2023;97:104–23.PubMedCrossRef
325.
go back to reference Finisguerra V et al. Metformin improves cancer immunotherapy by directly rescuing tumor-infiltrating CD8 T lymphocytes from hypoxia-induced immunosuppression. J Immunother Cancer, 2023. 11(5). Finisguerra V et al. Metformin improves cancer immunotherapy by directly rescuing tumor-infiltrating CD8 T lymphocytes from hypoxia-induced immunosuppression. J Immunother Cancer, 2023. 11(5).
326.
go back to reference Yan H, et al. Exercise sensitizes PD-1/PD-L1 immunotherapy as a hypoxia modulator in the tumor microenvironment of melanoma. Front Immunol. 2023;14:1265914.PubMedPubMedCentralCrossRef Yan H, et al. Exercise sensitizes PD-1/PD-L1 immunotherapy as a hypoxia modulator in the tumor microenvironment of melanoma. Front Immunol. 2023;14:1265914.PubMedPubMedCentralCrossRef
327.
go back to reference Barsoum IB, et al. A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res. 2014;74(3):665–74.PubMedCrossRef Barsoum IB, et al. A mechanism of hypoxia-mediated escape from adaptive immunity in cancer cells. Cancer Res. 2014;74(3):665–74.PubMedCrossRef
328.
go back to reference Silva VL. Exploiting the cancer niche: tumor-associated macrophages and hypoxia as promising synergistic targets for nano-based therapy. J Controlled Release. 2017;253:82–96.CrossRef Silva VL. Exploiting the cancer niche: tumor-associated macrophages and hypoxia as promising synergistic targets for nano-based therapy. J Controlled Release. 2017;253:82–96.CrossRef
329.
go back to reference Jiang T et al. TAM-Hijacked Immunoreaction Rescued by Hypoxia-Pathway-Intervened Strategy for Enhanced Metastatic Cancer Immunotherapy Small, 2023: p. e2305728. Jiang T et al. TAM-Hijacked Immunoreaction Rescued by Hypoxia-Pathway-Intervened Strategy for Enhanced Metastatic Cancer Immunotherapy Small, 2023: p. e2305728.
330.
go back to reference Qu S et al. Light-enhanced hypoxia-responsive gene editing for Hypoxia-resistant photodynamic and Immunotherapy. Adv Healthc Mater, 2023: p. e2302615. Qu S et al. Light-enhanced hypoxia-responsive gene editing for Hypoxia-resistant photodynamic and Immunotherapy. Adv Healthc Mater, 2023: p. e2302615.
331.
go back to reference Sun T et al. Metal-Organic Framework-Mediated Synergistic Hypoxia-Activated Chemo-Immunotherapy Induced by High Intensity Focused Ultrasound for Enhanced Cancer Theranostics Small, 2023: p. e2306338. Sun T et al. Metal-Organic Framework-Mediated Synergistic Hypoxia-Activated Chemo-Immunotherapy Induced by High Intensity Focused Ultrasound for Enhanced Cancer Theranostics Small, 2023: p. e2306338.
332.
go back to reference Luo W et al. Strengthening the Combinational Immunotherapy from modulating the Tumor Inflammatory Environment via Hypoxia-Responsive Nanogels. Adv Healthc Mater, 2023: p. e2302865. Luo W et al. Strengthening the Combinational Immunotherapy from modulating the Tumor Inflammatory Environment via Hypoxia-Responsive Nanogels. Adv Healthc Mater, 2023: p. e2302865.
333.
go back to reference Wang M, et al. Controllable hypoxia-activated chemotherapy as a dual enhancer for synergistic cancer photodynamic immunotherapy. Biomaterials. 2023;301:122257.PubMedCrossRef Wang M, et al. Controllable hypoxia-activated chemotherapy as a dual enhancer for synergistic cancer photodynamic immunotherapy. Biomaterials. 2023;301:122257.PubMedCrossRef
334.
go back to reference Luo G, et al. Multifunctional calcium-manganese Nanomodulator provides Antitumor Treatment and Improved Immunotherapy via Reprogramming of the Tumor Microenvironment. ACS Nano. 2023;17(16):15449–65.PubMedPubMedCentralCrossRef Luo G, et al. Multifunctional calcium-manganese Nanomodulator provides Antitumor Treatment and Improved Immunotherapy via Reprogramming of the Tumor Microenvironment. ACS Nano. 2023;17(16):15449–65.PubMedPubMedCentralCrossRef
335.
go back to reference Kang X, et al. A photo-triggered self-accelerated nanoplatform for multifunctional image-guided combination cancer immunotherapy. Nat Commun. 2023;14(1):5216.PubMedPubMedCentralCrossRef Kang X, et al. A photo-triggered self-accelerated nanoplatform for multifunctional image-guided combination cancer immunotherapy. Nat Commun. 2023;14(1):5216.PubMedPubMedCentralCrossRef
336.
go back to reference Gao C, et al. Conjugation of macrophage-mimetic Microalgae and Liposome for Antitumor Sonodynamic Immunotherapy via Hypoxia Alleviation and Autophagy Inhibition. ACS Nano. 2023;17(4):4034–49.PubMedCrossRef Gao C, et al. Conjugation of macrophage-mimetic Microalgae and Liposome for Antitumor Sonodynamic Immunotherapy via Hypoxia Alleviation and Autophagy Inhibition. ACS Nano. 2023;17(4):4034–49.PubMedCrossRef
337.
go back to reference Qin Y, et al. Autophagy and cancer drug resistance in dialogue: pre-clinical and clinical evidence. Cancer Lett. 2023;570:216307.PubMedCrossRef Qin Y, et al. Autophagy and cancer drug resistance in dialogue: pre-clinical and clinical evidence. Cancer Lett. 2023;570:216307.PubMedCrossRef
338.
go back to reference Ashrafizadeh M, et al. A bioinformatics analysis, pre-clinical and clinical conception of autophagy in pancreatic cancer: complexity and simplicity in crosstalk. Pharmacol Res. 2023;194:106822.PubMedCrossRef Ashrafizadeh M, et al. A bioinformatics analysis, pre-clinical and clinical conception of autophagy in pancreatic cancer: complexity and simplicity in crosstalk. Pharmacol Res. 2023;194:106822.PubMedCrossRef
339.
go back to reference Wang S, et al. Metabolic intervention Liposome boosted Lung Cancer Radio-Immunotherapy via Hypoxia Amelioration and PD-L1 Restraint. Adv Sci (Weinh). 2023;10(18):e2207608.PubMedCrossRef Wang S, et al. Metabolic intervention Liposome boosted Lung Cancer Radio-Immunotherapy via Hypoxia Amelioration and PD-L1 Restraint. Adv Sci (Weinh). 2023;10(18):e2207608.PubMedCrossRef
340.
go back to reference Liu Y et al. An Injectable puerarin depot can potentiate chimeric antigen receptor natural killer cell immunotherapy against targeted solid tumors by reversing tumor immunosuppression Small, 2024: p. e2307521. Liu Y et al. An Injectable puerarin depot can potentiate chimeric antigen receptor natural killer cell immunotherapy against targeted solid tumors by reversing tumor immunosuppression Small, 2024: p. e2307521.
341.
go back to reference Jiang X, et al. Mitochondrial disruption Nanosystem simultaneously depressed programmed death Ligand-1 and transforming growth Factor-β to overcome photodynamic immunotherapy resistance. ACS Nano; 2024. Jiang X, et al. Mitochondrial disruption Nanosystem simultaneously depressed programmed death Ligand-1 and transforming growth Factor-β to overcome photodynamic immunotherapy resistance. ACS Nano; 2024.
342.
go back to reference Li M, et al. Sono-activatable semiconducting polymer nanoreshapers multiply remodel tumor microenvironment for potent immunotherapy of orthotopic pancreatic cancer. Adv Sci (Weinh). 2023;10(35):e2305150.PubMedCrossRef Li M, et al. Sono-activatable semiconducting polymer nanoreshapers multiply remodel tumor microenvironment for potent immunotherapy of orthotopic pancreatic cancer. Adv Sci (Weinh). 2023;10(35):e2305150.PubMedCrossRef
343.
go back to reference Jang H et al. Nanoparticles targeting innate immune cells in tumor microenvironment. Int J Mol Sci, 2021. 22(18). Jang H et al. Nanoparticles targeting innate immune cells in tumor microenvironment. Int J Mol Sci, 2021. 22(18).
344.
go back to reference Bronte V, Zanovello P. Regulation of immune responses by L-arginine metabolism. Nat Rev Immunol. 2005;5(8):641–54.PubMedCrossRef Bronte V, Zanovello P. Regulation of immune responses by L-arginine metabolism. Nat Rev Immunol. 2005;5(8):641–54.PubMedCrossRef
346.
go back to reference Rodríguez PC, Ochoa AC. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol Rev. 2008;222:180–91.PubMedPubMedCentralCrossRef Rodríguez PC, Ochoa AC. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol Rev. 2008;222:180–91.PubMedPubMedCentralCrossRef
347.
go back to reference Rodriguez PC, et al. Regulation of T cell receptor CD3zeta chain expression by L-arginine. J Biol Chem. 2002;277(24):21123–9.PubMedCrossRef Rodriguez PC, et al. Regulation of T cell receptor CD3zeta chain expression by L-arginine. J Biol Chem. 2002;277(24):21123–9.PubMedCrossRef
349.
go back to reference Kusmartsev S, et al. Antigen-specific inhibition of CD8 + T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J Immunol. 2004;172(2):989–99.PubMedCrossRef Kusmartsev S, et al. Antigen-specific inhibition of CD8 + T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J Immunol. 2004;172(2):989–99.PubMedCrossRef
350.
go back to reference Kusmartsev S, Nagaraj S, Gabrilovich DI. Tumor-associated CD8 + T cell tolerance induced by bone marrow-derived immature myeloid cells. J Immunol. 2005;175(7):4583–92.PubMedCrossRef Kusmartsev S, Nagaraj S, Gabrilovich DI. Tumor-associated CD8 + T cell tolerance induced by bone marrow-derived immature myeloid cells. J Immunol. 2005;175(7):4583–92.PubMedCrossRef
351.
go back to reference Huang B, et al. Gr-1 + CD115 + immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res. 2006;66(2):1123–31.PubMedCrossRef Huang B, et al. Gr-1 + CD115 + immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer Res. 2006;66(2):1123–31.PubMedCrossRef
352.
go back to reference Sawant DV, et al. Adaptive plasticity of IL-10(+) and IL-35(+) T(reg) cells cooperatively promotes tumor T cell exhaustion. Nat Immunol. 2019;20(6):724–35.PubMedPubMedCentralCrossRef Sawant DV, et al. Adaptive plasticity of IL-10(+) and IL-35(+) T(reg) cells cooperatively promotes tumor T cell exhaustion. Nat Immunol. 2019;20(6):724–35.PubMedPubMedCentralCrossRef
353.
354.
go back to reference Liu H, et al. A novel DNA aptamer for dual targeting of Polymorphonuclear myeloid-derived suppressor cells and Tumor cells. Theranostics. 2018;8(1):31–44.PubMedPubMedCentralCrossRef Liu H, et al. A novel DNA aptamer for dual targeting of Polymorphonuclear myeloid-derived suppressor cells and Tumor cells. Theranostics. 2018;8(1):31–44.PubMedPubMedCentralCrossRef
355.
go back to reference Song X, et al. CD11b+/Gr-1 + immature myeloid cells mediate suppression of T cells in mice bearing tumors of IL-1beta-secreting cells. J Immunol. 2005;175(12):8200–8.PubMedCrossRef Song X, et al. CD11b+/Gr-1 + immature myeloid cells mediate suppression of T cells in mice bearing tumors of IL-1beta-secreting cells. J Immunol. 2005;175(12):8200–8.PubMedCrossRef
356.
go back to reference Bunt SK, et al. Inflammation induces myeloid-derived suppressor cells that facilitate tumor progression. J Immunol. 2006;176(1):284–90.PubMedCrossRef Bunt SK, et al. Inflammation induces myeloid-derived suppressor cells that facilitate tumor progression. J Immunol. 2006;176(1):284–90.PubMedCrossRef
357.
go back to reference Jiang M, et al. Interleukin-6 trans-signaling pathway promotes immunosuppressive myeloid-derived suppressor cells via Suppression of Suppressor of Cytokine Signaling 3 in breast Cancer. Front Immunol. 2017;8:1840.PubMedPubMedCentralCrossRef Jiang M, et al. Interleukin-6 trans-signaling pathway promotes immunosuppressive myeloid-derived suppressor cells via Suppression of Suppressor of Cytokine Signaling 3 in breast Cancer. Front Immunol. 2017;8:1840.PubMedPubMedCentralCrossRef
358.
go back to reference Sinha P, et al. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res. 2007;67(9):4507–13.PubMedCrossRef Sinha P, et al. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res. 2007;67(9):4507–13.PubMedCrossRef
359.
go back to reference Gabrilovich DI, et al. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med. 1996;2(10):1096–103.PubMedCrossRef Gabrilovich DI, et al. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med. 1996;2(10):1096–103.PubMedCrossRef
360.
go back to reference Groth C, et al. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br J Cancer. 2019;120(1):16–25.PubMedCrossRef Groth C, et al. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br J Cancer. 2019;120(1):16–25.PubMedCrossRef
361.
go back to reference Kong M, et al. Biodegradable Hollow Mesoporous silica nanoparticles for regulating Tumor Microenvironment and Enhancing Antitumor Efficiency. Theranostics. 2017;7(13):3276–92.PubMedPubMedCentralCrossRef Kong M, et al. Biodegradable Hollow Mesoporous silica nanoparticles for regulating Tumor Microenvironment and Enhancing Antitumor Efficiency. Theranostics. 2017;7(13):3276–92.PubMedPubMedCentralCrossRef
362.
363.
go back to reference Zolnik BS, et al. Minireview: nanoparticles and the immune system. Endocrinology. 2010;151(2):458–65.PubMedCrossRef Zolnik BS, et al. Minireview: nanoparticles and the immune system. Endocrinology. 2010;151(2):458–65.PubMedCrossRef
364.
go back to reference Dwivedi PD, et al. Impact of nanoparticles on the immune system. J Biomed Nanotechnol. 2011;7(1):193–4.PubMedCrossRef Dwivedi PD, et al. Impact of nanoparticles on the immune system. J Biomed Nanotechnol. 2011;7(1):193–4.PubMedCrossRef
365.
go back to reference Falo L Jr, et al. Targeting antigen into the phagocytic pathway in vivo induces protective tumour immunity. Nat Med. 1995;1(7):649–53.PubMedCrossRef Falo L Jr, et al. Targeting antigen into the phagocytic pathway in vivo induces protective tumour immunity. Nat Med. 1995;1(7):649–53.PubMedCrossRef
366.
go back to reference Fifis T, et al. Size-dependent immunogenicity: therapeutic and protective properties of nano-vaccines against tumors. J Immunol. 2004;173(5):3148–54.PubMedCrossRef Fifis T, et al. Size-dependent immunogenicity: therapeutic and protective properties of nano-vaccines against tumors. J Immunol. 2004;173(5):3148–54.PubMedCrossRef
367.
go back to reference Uto T, et al. Targeting of antigen to dendritic cells with poly (γ-glutamic acid) nanoparticles induces antigen-specific humoral and cellular immunity. J Immunol. 2007;178(5):2979–86.PubMedCrossRef Uto T, et al. Targeting of antigen to dendritic cells with poly (γ-glutamic acid) nanoparticles induces antigen-specific humoral and cellular immunity. J Immunol. 2007;178(5):2979–86.PubMedCrossRef
368.
go back to reference Cho N-H, et al. A multifunctional core–shell nanoparticle for dendritic cell-based cancer immunotherapy. Nat Nanotechnol. 2011;6(10):675–82.PubMedCrossRef Cho N-H, et al. A multifunctional core–shell nanoparticle for dendritic cell-based cancer immunotherapy. Nat Nanotechnol. 2011;6(10):675–82.PubMedCrossRef
369.
go back to reference Bourquin C, et al. Targeting CpG oligonucleotides to the lymph node by nanoparticles elicits efficient antitumoral immunity. J Immunol. 2008;181(5):2990–8.PubMedCrossRef Bourquin C, et al. Targeting CpG oligonucleotides to the lymph node by nanoparticles elicits efficient antitumoral immunity. J Immunol. 2008;181(5):2990–8.PubMedCrossRef
370.
go back to reference Zwiorek K, et al. Delivery by cationic gelatin nanoparticles strongly increases the immunostimulatory effects of CpG oligonucleotides. Pharm Res. 2008;25:551–62.PubMedCrossRef Zwiorek K, et al. Delivery by cationic gelatin nanoparticles strongly increases the immunostimulatory effects of CpG oligonucleotides. Pharm Res. 2008;25:551–62.PubMedCrossRef
371.
go back to reference Romero P, et al. Ex vivo staining of metastatic lymph nodes by class I major histocompatibility complex tetramers reveals high numbers of antigen-experienced tumor-specific cytolytic T lymphocytes. J Exp Med. 1998;188(9):1641–50.PubMedPubMedCentralCrossRef Romero P, et al. Ex vivo staining of metastatic lymph nodes by class I major histocompatibility complex tetramers reveals high numbers of antigen-experienced tumor-specific cytolytic T lymphocytes. J Exp Med. 1998;188(9):1641–50.PubMedPubMedCentralCrossRef
372.
go back to reference Cochran AJ, et al. Sentinel lymph nodes show profound downregulation of antigen-presenting cells of the paracortex: implications for tumor biology and treatment. Mod Pathol. 2001;14(6):604–8.PubMedCrossRef Cochran AJ, et al. Sentinel lymph nodes show profound downregulation of antigen-presenting cells of the paracortex: implications for tumor biology and treatment. Mod Pathol. 2001;14(6):604–8.PubMedCrossRef
373.
go back to reference Curiel TJ, et al. Blockade of B7-H1 improves myeloid dendritic cell–mediated antitumor immunity. Nat Med. 2003;9(5):562–7.PubMedCrossRef Curiel TJ, et al. Blockade of B7-H1 improves myeloid dendritic cell–mediated antitumor immunity. Nat Med. 2003;9(5):562–7.PubMedCrossRef
374.
go back to reference Munn DH, et al. Expression of indoleamine 2, 3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J Clin Investig. 2004;114(2):280–90.PubMedPubMedCentralCrossRef Munn DH, et al. Expression of indoleamine 2, 3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J Clin Investig. 2004;114(2):280–90.PubMedPubMedCentralCrossRef
375.
go back to reference Gabrilovich D. Mechanisms and functional significance of tumour-induced dendritic-cell defects. Nat Rev Immunol. 2004;4(12):941–52.PubMedCrossRef Gabrilovich D. Mechanisms and functional significance of tumour-induced dendritic-cell defects. Nat Rev Immunol. 2004;4(12):941–52.PubMedCrossRef
376.
go back to reference Pinzon-Charry A, Maxwell T, López JA. Dendritic cell dysfunction in cancer: a mechanism for immunosuppression. Immunol Cell Biol. 2005;83(5):451–61.PubMedCrossRef Pinzon-Charry A, Maxwell T, López JA. Dendritic cell dysfunction in cancer: a mechanism for immunosuppression. Immunol Cell Biol. 2005;83(5):451–61.PubMedCrossRef
377.
go back to reference Vicari AP, et al. Reversal of tumor-induced dendritic cell paralysis by CpG immunostimulatory oligonucleotide and anti–interleukin 10 receptor antibody. J Exp Med. 2002;196(4):541–9.PubMedPubMedCentralCrossRef Vicari AP, et al. Reversal of tumor-induced dendritic cell paralysis by CpG immunostimulatory oligonucleotide and anti–interleukin 10 receptor antibody. J Exp Med. 2002;196(4):541–9.PubMedPubMedCentralCrossRef
378.
go back to reference van Mierlo GJ, et al. Activation of dendritic cells that cross-present tumor-derived antigen licenses CD8 + CTL to cause tumor eradication. J Immunol. 2004;173(11):6753–9.PubMedCrossRef van Mierlo GJ, et al. Activation of dendritic cells that cross-present tumor-derived antigen licenses CD8 + CTL to cause tumor eradication. J Immunol. 2004;173(11):6753–9.PubMedCrossRef
379.
go back to reference Munn DH, Mellor AL. The tumor-draining lymph node as an immune‐privileged site. Immunol Rev. 2006;213(1):146–58.PubMedCrossRef Munn DH, Mellor AL. The tumor-draining lymph node as an immune‐privileged site. Immunol Rev. 2006;213(1):146–58.PubMedCrossRef
380.
go back to reference Jeanbart L, et al. Enhancing efficacy of anticancer vaccines by targeted delivery to tumor-draining lymph nodes. Cancer Immunol Res. 2014;2(5):436–47.PubMedCrossRef Jeanbart L, et al. Enhancing efficacy of anticancer vaccines by targeted delivery to tumor-draining lymph nodes. Cancer Immunol Res. 2014;2(5):436–47.PubMedCrossRef
381.
go back to reference Nam J, et al. Cancer nanomedicine for combination cancer immunotherapy. Nat Reviews Mater. 2019;4(6):398–414.CrossRef Nam J, et al. Cancer nanomedicine for combination cancer immunotherapy. Nat Reviews Mater. 2019;4(6):398–414.CrossRef
382.
go back to reference Shi J, et al. Cancer nanomedicine: progress, challenges and opportunities. Nat Rev Cancer. 2017;17(1):20–37.PubMedCrossRef Shi J, et al. Cancer nanomedicine: progress, challenges and opportunities. Nat Rev Cancer. 2017;17(1):20–37.PubMedCrossRef
383.
go back to reference Ojha T, et al. Pharmacological and physical vessel modulation strategies to improve EPR-mediated drug targeting to tumors. Adv Drug Deliv Rev. 2017;119:44–60.PubMedPubMedCentralCrossRef Ojha T, et al. Pharmacological and physical vessel modulation strategies to improve EPR-mediated drug targeting to tumors. Adv Drug Deliv Rev. 2017;119:44–60.PubMedPubMedCentralCrossRef
384.
go back to reference Wilhelm S, et al. Analysis of nanoparticle delivery to tumours. Nat Reviews Mater. 2016;1(5):1–12.CrossRef Wilhelm S, et al. Analysis of nanoparticle delivery to tumours. Nat Reviews Mater. 2016;1(5):1–12.CrossRef
385.
go back to reference Siegler EL, Kim YJ, Wang P. Nanomedicine targeting the tumor microenvironment: therapeutic strategies to inhibit angiogenesis, remodel matrix, and modulate immune responses. J Cell Immunotherapy. 2016;2(2):69–78.CrossRef Siegler EL, Kim YJ, Wang P. Nanomedicine targeting the tumor microenvironment: therapeutic strategies to inhibit angiogenesis, remodel matrix, and modulate immune responses. J Cell Immunotherapy. 2016;2(2):69–78.CrossRef
386.
go back to reference Shi K, Haynes M, Huang L. Nanovaccines for remodeling the suppressive tumor microenvironment: new horizons in cancer immunotherapy. Front Chem Sci Eng. 2017;11:676–84.CrossRef Shi K, Haynes M, Huang L. Nanovaccines for remodeling the suppressive tumor microenvironment: new horizons in cancer immunotherapy. Front Chem Sci Eng. 2017;11:676–84.CrossRef
387.
go back to reference Marabelle A, et al. Intratumoral immunotherapy: using the tumor as the remedy. Ann Oncol. 2017;28:xii33–43.PubMedCrossRef Marabelle A, et al. Intratumoral immunotherapy: using the tumor as the remedy. Ann Oncol. 2017;28:xii33–43.PubMedCrossRef
388.
go back to reference Aznar MA, et al. Intratumoral delivery of immunotherapy—act locally, think globally. J Immunol. 2017;198(1):31–9.PubMedCrossRef Aznar MA, et al. Intratumoral delivery of immunotherapy—act locally, think globally. J Immunol. 2017;198(1):31–9.PubMedCrossRef
389.
go back to reference Ishihara J, et al. Matrix-binding checkpoint immunotherapies enhance antitumor efficacy and reduce adverse events. Sci Transl Med. 2017;9(415):eaan0401.PubMedCrossRef Ishihara J, et al. Matrix-binding checkpoint immunotherapies enhance antitumor efficacy and reduce adverse events. Sci Transl Med. 2017;9(415):eaan0401.PubMedCrossRef
390.
go back to reference Raavé R, van Kuppevelt TH, Daamen WF. Chemotherapeutic drug delivery by tumoral extracellular matrix targeting. J Controlled Release. 2018;274:1–8.CrossRef Raavé R, van Kuppevelt TH, Daamen WF. Chemotherapeutic drug delivery by tumoral extracellular matrix targeting. J Controlled Release. 2018;274:1–8.CrossRef
391.
go back to reference Yu S et al. Reinforcing the immunogenic cell death to enhance cancer immunotherapy efficacy Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, 2023;1878(5):188946. Yu S et al. Reinforcing the immunogenic cell death to enhance cancer immunotherapy efficacy Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, 2023;1878(5):188946.
393.
go back to reference Petroni G, et al. Immunomodulation by targeted anticancer agents. Cancer Cell. 2021;39(3):310–45.PubMedCrossRef Petroni G, et al. Immunomodulation by targeted anticancer agents. Cancer Cell. 2021;39(3):310–45.PubMedCrossRef
394.
go back to reference Huang Z, et al. KCl nanoparticles as potential inducer of immunogenic cell death for Cancer Immunotherapy. ACS Appl Bio Mater. 2023;6(6):2404–14.PubMedCrossRef Huang Z, et al. KCl nanoparticles as potential inducer of immunogenic cell death for Cancer Immunotherapy. ACS Appl Bio Mater. 2023;6(6):2404–14.PubMedCrossRef
395.
go back to reference Félix-Piña P et al. Antitumoral and Immunogenic Capacity of β-D-Glucose-reduced silver nanoparticles in breast Cancer. Int J Mol Sci, 2023. 24(10). Félix-Piña P et al. Antitumoral and Immunogenic Capacity of β-D-Glucose-reduced silver nanoparticles in breast Cancer. Int J Mol Sci, 2023. 24(10).
396.
go back to reference Liu Q, et al. Exploiting immunostimulatory mechanisms of immunogenic cell death to develop membrane-encapsulated nanoparticles as a potent tumor vaccine. J Nanobiotechnol. 2023;21(1):326.CrossRef Liu Q, et al. Exploiting immunostimulatory mechanisms of immunogenic cell death to develop membrane-encapsulated nanoparticles as a potent tumor vaccine. J Nanobiotechnol. 2023;21(1):326.CrossRef
397.
go back to reference Xie B, et al. Supramolecularly Engineered Conjugate of Bacteria and cell membrane-coated magnetic nanoparticles for enhanced ferroptosis and immunotherapy of tumors. Adv Sci (Weinh). 2023;10(34):e2304407.PubMedCrossRef Xie B, et al. Supramolecularly Engineered Conjugate of Bacteria and cell membrane-coated magnetic nanoparticles for enhanced ferroptosis and immunotherapy of tumors. Adv Sci (Weinh). 2023;10(34):e2304407.PubMedCrossRef
398.
go back to reference Lu Y, et al. Self-driven bioactive hybrids co-deliver doxorubicin and indocyanine green nanoparticles for chemo/photothermal therapy of breast cancer. Biomed Pharmacother. 2023;169:115846.PubMedCrossRef Lu Y, et al. Self-driven bioactive hybrids co-deliver doxorubicin and indocyanine green nanoparticles for chemo/photothermal therapy of breast cancer. Biomed Pharmacother. 2023;169:115846.PubMedCrossRef
399.
go back to reference Wang H et al. Near-Infrared Light-Driven Nanoparticles for Cancer Photoimmunotherapy by Synergizing Immune Cell Death and Epigenetic Regulation Small, 2023: p. e2309202. Wang H et al. Near-Infrared Light-Driven Nanoparticles for Cancer Photoimmunotherapy by Synergizing Immune Cell Death and Epigenetic Regulation Small, 2023: p. e2309202.
400.
go back to reference Wu Q et al. Photothermal ferrotherapy - induced immunogenic cell death via iron-based ternary chalcogenide nanoparticles against triple-negative breast cancer Small, 2023: p. e2306766. Wu Q et al. Photothermal ferrotherapy - induced immunogenic cell death via iron-based ternary chalcogenide nanoparticles against triple-negative breast cancer Small, 2023: p. e2306766.
401.
go back to reference Song S, et al. All-in-one glycol chitosan nanoparticles for co-delivery of doxorubicin and anti-PD-L1 peptide in cancer immunotherapy. Bioact Mater. 2023;28:358–75.PubMedPubMedCentral Song S, et al. All-in-one glycol chitosan nanoparticles for co-delivery of doxorubicin and anti-PD-L1 peptide in cancer immunotherapy. Bioact Mater. 2023;28:358–75.PubMedPubMedCentral
402.
go back to reference Wang R, et al. Hydroxyapatite nanoparticles promote TLR4 agonist-mediated anti-tumor immunity through synergically enhanced macrophage polarization. Acta Biomater. 2023;164:626–40.PubMedCrossRef Wang R, et al. Hydroxyapatite nanoparticles promote TLR4 agonist-mediated anti-tumor immunity through synergically enhanced macrophage polarization. Acta Biomater. 2023;164:626–40.PubMedCrossRef
403.
go back to reference Wang Y, et al. Docetaxel-loaded pH/ROS dual-responsive nanoparticles with self-supplied ROS for inhibiting metastasis and enhancing immunotherapy of breast cancer. J Nanobiotechnol. 2023;21(1):286.CrossRef Wang Y, et al. Docetaxel-loaded pH/ROS dual-responsive nanoparticles with self-supplied ROS for inhibiting metastasis and enhancing immunotherapy of breast cancer. J Nanobiotechnol. 2023;21(1):286.CrossRef
404.
go back to reference Yang J, et al. Self-assembled albumin nanoparticles induce pyroptosis for photodynamic/photothermal/immuno synergistic therapies in triple-negative breast cancer. Front Immunol. 2023;14:1173487.PubMedPubMedCentralCrossRef Yang J, et al. Self-assembled albumin nanoparticles induce pyroptosis for photodynamic/photothermal/immuno synergistic therapies in triple-negative breast cancer. Front Immunol. 2023;14:1173487.PubMedPubMedCentralCrossRef
405.
go back to reference Xu X, et al. Guanidine-modified nanoparticles as robust BTZ delivery carriers and activators of immune responses. J Control Release. 2023;357:310–8.PubMedCrossRef Xu X, et al. Guanidine-modified nanoparticles as robust BTZ delivery carriers and activators of immune responses. J Control Release. 2023;357:310–8.PubMedCrossRef
406.
go back to reference Chen W, et al. PEI-Based nanoparticles for Tumor Immunotherapy via in situ Antigen-capture triggered by Photothermal Therapy. ACS Appl Mater Interfaces. 2023;15(48):55433–46.PubMedCrossRef Chen W, et al. PEI-Based nanoparticles for Tumor Immunotherapy via in situ Antigen-capture triggered by Photothermal Therapy. ACS Appl Mater Interfaces. 2023;15(48):55433–46.PubMedCrossRef
407.
go back to reference Li Y, et al. Antigen-capturing dendritic-cell-targeting nanoparticles for enhanced Tumor Immunotherapy based on Photothermal-Therapy-Induced in situ vaccination. Adv Healthc Mater. 2023;12(22):e2202871.PubMedCrossRef Li Y, et al. Antigen-capturing dendritic-cell-targeting nanoparticles for enhanced Tumor Immunotherapy based on Photothermal-Therapy-Induced in situ vaccination. Adv Healthc Mater. 2023;12(22):e2202871.PubMedCrossRef
408.
go back to reference Zou C, et al. cRGD-modified nanoparticles of multi-bioactive agent conjugate with pH-sensitive linkers and PD-L1 antagonist for integrative collaborative treatment of breast cancer. Nanoscale Horiz. 2023;8(7):870–86.PubMedCrossRef Zou C, et al. cRGD-modified nanoparticles of multi-bioactive agent conjugate with pH-sensitive linkers and PD-L1 antagonist for integrative collaborative treatment of breast cancer. Nanoscale Horiz. 2023;8(7):870–86.PubMedCrossRef
409.
go back to reference Chen S, et al. Tumor Acidic Microenvironment-Responsive Promodulator Iron Oxide nanoparticles for Photothermal-enhanced Chemodynamic Immunotherapy of Cancer. ACS Biomater Sci Eng. 2023;9(2):773–83.PubMedCrossRef Chen S, et al. Tumor Acidic Microenvironment-Responsive Promodulator Iron Oxide nanoparticles for Photothermal-enhanced Chemodynamic Immunotherapy of Cancer. ACS Biomater Sci Eng. 2023;9(2):773–83.PubMedCrossRef
410.
go back to reference Ma W, et al. Biomimetic nanoerythrosome-coated Aptamer-DNA Tetrahedron/Maytansine conjugates: pH-Responsive and targeted cytotoxicity for HER2-Positive breast Cancer. Adv Mater. 2022;34(46):e2109609.PubMedCrossRef Ma W, et al. Biomimetic nanoerythrosome-coated Aptamer-DNA Tetrahedron/Maytansine conjugates: pH-Responsive and targeted cytotoxicity for HER2-Positive breast Cancer. Adv Mater. 2022;34(46):e2109609.PubMedCrossRef
411.
go back to reference Zheng X, et al. Biomimetic co-assembled nanodrug of doxorubicin and berberine suppresses chemotherapy-exacerbated breast cancer metastasis. Biomaterials. 2021;271:120716.PubMedCrossRef Zheng X, et al. Biomimetic co-assembled nanodrug of doxorubicin and berberine suppresses chemotherapy-exacerbated breast cancer metastasis. Biomaterials. 2021;271:120716.PubMedCrossRef
412.
go back to reference Liu XL, et al. Biomimetic liposomal nanoplatinum for targeted Cancer Chemophototherapy. Adv Sci (Weinh). 2021;8(8):2003679.PubMedCrossRef Liu XL, et al. Biomimetic liposomal nanoplatinum for targeted Cancer Chemophototherapy. Adv Sci (Weinh). 2021;8(8):2003679.PubMedCrossRef
413.
go back to reference Zhao Y, et al. Biomimetic manganese-based theranostic nanoplatform for cancer multimodal imaging and twofold immunotherapy. Bioact Mater. 2023;19:237–50.PubMed Zhao Y, et al. Biomimetic manganese-based theranostic nanoplatform for cancer multimodal imaging and twofold immunotherapy. Bioact Mater. 2023;19:237–50.PubMed
414.
go back to reference Zhao P, et al. Programming cell pyroptosis with biomimetic nanoparticles for solid tumor immunotherapy. Biomaterials. 2020;254:120142.PubMedCrossRef Zhao P, et al. Programming cell pyroptosis with biomimetic nanoparticles for solid tumor immunotherapy. Biomaterials. 2020;254:120142.PubMedCrossRef
415.
go back to reference Ying K, et al. Macrophage membrane-biomimetic adhesive polycaprolactone nanocamptothecin for improving cancer-targeting efficiency and impairing metastasis. Bioact Mater. 2023;20:449–62.PubMed Ying K, et al. Macrophage membrane-biomimetic adhesive polycaprolactone nanocamptothecin for improving cancer-targeting efficiency and impairing metastasis. Bioact Mater. 2023;20:449–62.PubMed
416.
go back to reference Miao Y, et al. Cell membrane-camouflaged nanocarriers with Biomimetic Deformability of erythrocytes for Ultralong circulation and enhanced Cancer Therapy. ACS Nano. 2022;16(4):6527–40.PubMedCrossRef Miao Y, et al. Cell membrane-camouflaged nanocarriers with Biomimetic Deformability of erythrocytes for Ultralong circulation and enhanced Cancer Therapy. ACS Nano. 2022;16(4):6527–40.PubMedCrossRef
417.
go back to reference Guo Y, et al. Chemotherapy mediated by Biomimetic Polymeric Nanoparticles Potentiates Enhanced Tumor Immunotherapy via Amplification of Endoplasmic Reticulum Stress and mitochondrial dysfunction. Adv Mater. 2022;34(47):e2206861.PubMedCrossRef Guo Y, et al. Chemotherapy mediated by Biomimetic Polymeric Nanoparticles Potentiates Enhanced Tumor Immunotherapy via Amplification of Endoplasmic Reticulum Stress and mitochondrial dysfunction. Adv Mater. 2022;34(47):e2206861.PubMedCrossRef
418.
go back to reference Wu P, et al. Engineered EGCG-Containing Biomimetic nanoassemblies as effective delivery platform for enhanced Cancer Therapy. Adv Sci (Weinh). 2022;9(15):e2105894.PubMedCrossRef Wu P, et al. Engineered EGCG-Containing Biomimetic nanoassemblies as effective delivery platform for enhanced Cancer Therapy. Adv Sci (Weinh). 2022;9(15):e2105894.PubMedCrossRef
420.
go back to reference Molina M, et al. Stimuli-responsive nanogel composites and their application in nanomedicine. Chem Soc Rev. 2015;44(17):6161–86.PubMedCrossRef Molina M, et al. Stimuli-responsive nanogel composites and their application in nanomedicine. Chem Soc Rev. 2015;44(17):6161–86.PubMedCrossRef
421.
go back to reference Chuard N, et al. Strain-promoted thiol-mediated Cellular Uptake of Giant substrates: liposomes and polymersomes. Angew Chem Int Ed Engl. 2017;56(11):2947–50.PubMedCrossRef Chuard N, et al. Strain-promoted thiol-mediated Cellular Uptake of Giant substrates: liposomes and polymersomes. Angew Chem Int Ed Engl. 2017;56(11):2947–50.PubMedCrossRef
422.
go back to reference Wang H, et al. A cooperative dimensional strategy for enhanced nucleus-targeted delivery of anticancer drugs. Adv Funct Mater. 2017;27(24):1700339.CrossRef Wang H, et al. A cooperative dimensional strategy for enhanced nucleus-targeted delivery of anticancer drugs. Adv Funct Mater. 2017;27(24):1700339.CrossRef
423.
424.
go back to reference Sun CY, et al. Tumor acidity-sensitive polymeric vector for active targeted siRNA delivery. J Am Chem Soc. 2015;137(48):15217–24.PubMedCrossRef Sun CY, et al. Tumor acidity-sensitive polymeric vector for active targeted siRNA delivery. J Am Chem Soc. 2015;137(48):15217–24.PubMedCrossRef
425.
go back to reference Shang L, et al. Enhancing cancer chemo-immunotherapy by biomimetic nanogel with tumor targeting capacity and rapid drug-releasing in tumor microenvironment. Acta Pharm Sin B. 2022;12(5):2550–67.PubMedCrossRef Shang L, et al. Enhancing cancer chemo-immunotherapy by biomimetic nanogel with tumor targeting capacity and rapid drug-releasing in tumor microenvironment. Acta Pharm Sin B. 2022;12(5):2550–67.PubMedCrossRef
426.
go back to reference Wan L, et al. Biomimetic, pH-Responsive nanoplatforms for Cancer Multimodal Imaging and Photothermal Immunotherapy. ACS Appl Mater Interfaces. 2023;15(1):1784–97.PubMedCrossRef Wan L, et al. Biomimetic, pH-Responsive nanoplatforms for Cancer Multimodal Imaging and Photothermal Immunotherapy. ACS Appl Mater Interfaces. 2023;15(1):1784–97.PubMedCrossRef
427.
go back to reference Yang C, et al. Biomimetic nanovaccines potentiating dendritic cell internalization via CXCR4-Mediated macropinocytosis. Adv Healthc Mater. 2023;12(5):e2202064.PubMedCrossRef Yang C, et al. Biomimetic nanovaccines potentiating dendritic cell internalization via CXCR4-Mediated macropinocytosis. Adv Healthc Mater. 2023;12(5):e2202064.PubMedCrossRef
428.
go back to reference Wei X et al. Biomimetic nano-immunoactivator via ionic metabolic modulation for strengthened nir-ii photothermal immunotherapy small, 2023: p. e2304370. Wei X et al. Biomimetic nano-immunoactivator via ionic metabolic modulation for strengthened nir-ii photothermal immunotherapy small, 2023: p. e2304370.
429.
go back to reference Wang Y, et al. Engineering Endogenous Tumor-Associated macrophage-targeted biomimetic Nano-RBC to Reprogram Tumor Immunosuppressive Microenvironment for enhanced chemo-immunotherapy. Adv Mater. 2021;33(39):e2103497.PubMedCrossRef Wang Y, et al. Engineering Endogenous Tumor-Associated macrophage-targeted biomimetic Nano-RBC to Reprogram Tumor Immunosuppressive Microenvironment for enhanced chemo-immunotherapy. Adv Mater. 2021;33(39):e2103497.PubMedCrossRef
430.
go back to reference Zhao H, et al. Biomimetic Decoy inhibits Tumor Growth and Lung Metastasis by reversing the drawbacks of Sonodynamic Therapy. Adv Healthc Mater. 2020;9(1):e1901335.PubMedCrossRef Zhao H, et al. Biomimetic Decoy inhibits Tumor Growth and Lung Metastasis by reversing the drawbacks of Sonodynamic Therapy. Adv Healthc Mater. 2020;9(1):e1901335.PubMedCrossRef
431.
go back to reference Feng C, et al. Biomimetic and bioinspired nano-platforms for cancer vaccine development. Explor (Beijing). 2023;3(3):20210263. Feng C, et al. Biomimetic and bioinspired nano-platforms for cancer vaccine development. Explor (Beijing). 2023;3(3):20210263.
432.
go back to reference Fang RH, et al. Cell Membrane Coat Nanatechnol Adv Mater. 2018;30(23):e1706759. Fang RH, et al. Cell Membrane Coat Nanatechnol Adv Mater. 2018;30(23):e1706759.
434.
go back to reference Zheng S, Guan XY. Ferroptosis: promising approach for cancer and cancer immunotherapy. Cancer Lett. 2023;561:216152.PubMedCrossRef Zheng S, Guan XY. Ferroptosis: promising approach for cancer and cancer immunotherapy. Cancer Lett. 2023;561:216152.PubMedCrossRef
435.
436.
go back to reference Xu H, et al. Ferroptosis in the tumor microenvironment: perspectives for immunotherapy. Trends Mol Med. 2021;27(9):856–67.PubMedCrossRef Xu H, et al. Ferroptosis in the tumor microenvironment: perspectives for immunotherapy. Trends Mol Med. 2021;27(9):856–67.PubMedCrossRef
438.
go back to reference Lei Y et al. Autophagy in cancer immunotherapy cells, 2022. 11(19). Lei Y et al. Autophagy in cancer immunotherapy cells, 2022. 11(19).
440.
441.
go back to reference Hamidi M, et al. Applications of carrier erythrocytes in delivery of biopharmaceuticals. J Control Release. 2007;118(2):145–60.PubMedCrossRef Hamidi M, et al. Applications of carrier erythrocytes in delivery of biopharmaceuticals. J Control Release. 2007;118(2):145–60.PubMedCrossRef
442.
go back to reference Banz A, et al. In situ targeting of dendritic cells by antigen-loaded red blood cells: a novel approach to cancer immunotherapy. Vaccine. 2010;28(17):2965–72.PubMedCrossRef Banz A, et al. In situ targeting of dendritic cells by antigen-loaded red blood cells: a novel approach to cancer immunotherapy. Vaccine. 2010;28(17):2965–72.PubMedCrossRef
443.
go back to reference Hu CMJ, Fang RH, Zhang L. Erythrocyte-inspired delivery systems. Adv Healthc Mater. 2012;1(5):537–47.PubMedCrossRef Hu CMJ, Fang RH, Zhang L. Erythrocyte-inspired delivery systems. Adv Healthc Mater. 2012;1(5):537–47.PubMedCrossRef
444.
go back to reference Guo Y, et al. Erythrocyte membrane-enveloped polymeric nanoparticles as Nanovaccine for induction of Antitumor immunity against Melanoma. ACS Nano. 2015;9(7):6918–33.PubMedCrossRef Guo Y, et al. Erythrocyte membrane-enveloped polymeric nanoparticles as Nanovaccine for induction of Antitumor immunity against Melanoma. ACS Nano. 2015;9(7):6918–33.PubMedCrossRef
447.
go back to reference Bahmani B et al. Intratumoral immunotherapy using platelet-cloaked nanoparticles enhances antitumor immunity in solid tumors Nat Commun, 2021;12(1):1999. Bahmani B et al. Intratumoral immunotherapy using platelet-cloaked nanoparticles enhances antitumor immunity in solid tumors Nat Commun, 2021;12(1):1999.
448.
go back to reference Mai X, et al. Integration of immunogenic activation and immunosuppressive reversion using mitochondrial-respiration-inhibited platelet-mimicking nanoparticles. Biomaterials. 2020;232:119699.PubMedCrossRef Mai X, et al. Integration of immunogenic activation and immunosuppressive reversion using mitochondrial-respiration-inhibited platelet-mimicking nanoparticles. Biomaterials. 2020;232:119699.PubMedCrossRef
449.
go back to reference Wang Q et al. Biomimetic Nanophotosensitizer amplifies immunogenic pyroptosis and triggers synergistic Cancer Therapy. Adv Healthc Mater, 2023: p. e2301641. Wang Q et al. Biomimetic Nanophotosensitizer amplifies immunogenic pyroptosis and triggers synergistic Cancer Therapy. Adv Healthc Mater, 2023: p. e2301641.
450.
go back to reference Gong P et al. Immunocyte membrane-coated nanoparticles for Cancer Immunotherapy. Cancers (Basel), 2020. 13(1). Gong P et al. Immunocyte membrane-coated nanoparticles for Cancer Immunotherapy. Cancers (Basel), 2020. 13(1).
451.
go back to reference Sun K, et al. Saikosaponin D loaded macrophage membrane-biomimetic nanoparticles target angiogenic signaling for breast cancer therapy. Appl Mater Today. 2020;18:100505.CrossRef Sun K, et al. Saikosaponin D loaded macrophage membrane-biomimetic nanoparticles target angiogenic signaling for breast cancer therapy. Appl Mater Today. 2020;18:100505.CrossRef
452.
go back to reference Chen LJ, et al. Macrophage membrane coated persistent luminescence nanoparticle@MOF-derived mesoporous carbon core-shell nanocomposites for autofluorescence-free imaging-guided chemotherapy. J Mater Chem B. 2020;8(35):8071–83.PubMedCrossRef Chen LJ, et al. Macrophage membrane coated persistent luminescence nanoparticle@MOF-derived mesoporous carbon core-shell nanocomposites for autofluorescence-free imaging-guided chemotherapy. J Mater Chem B. 2020;8(35):8071–83.PubMedCrossRef
453.
go back to reference Cao H, et al. Liposomes coated with isolated macrophage membrane can target lung metastasis of breast Cancer. ACS Nano. 2016;10(8):7738–48.PubMedCrossRef Cao H, et al. Liposomes coated with isolated macrophage membrane can target lung metastasis of breast Cancer. ACS Nano. 2016;10(8):7738–48.PubMedCrossRef
454.
go back to reference Meng QF, et al. Macrophage membrane-coated iron oxide nanoparticles for enhanced photothermal tumor therapy. Nanotechnology. 2018;29(13):134004.PubMedCrossRef Meng QF, et al. Macrophage membrane-coated iron oxide nanoparticles for enhanced photothermal tumor therapy. Nanotechnology. 2018;29(13):134004.PubMedCrossRef
455.
go back to reference Zhao H, et al. C-C Chemokine Ligand 2 (CCL2) recruits macrophage-membrane-camouflaged Hollow Bismuth Selenide nanoparticles to facilitate Photothermal Sensitivity and inhibit lung metastasis of breast Cancer. ACS Appl Mater Interfaces. 2018;10(37):31124–35.PubMedCrossRef Zhao H, et al. C-C Chemokine Ligand 2 (CCL2) recruits macrophage-membrane-camouflaged Hollow Bismuth Selenide nanoparticles to facilitate Photothermal Sensitivity and inhibit lung metastasis of breast Cancer. ACS Appl Mater Interfaces. 2018;10(37):31124–35.PubMedCrossRef
456.
go back to reference Liang B, et al. Biomimetic theranostic strategy for anti-metastasis therapy of breast cancer via the macrophage membrane camouflaged superparticles. Mater Sci Eng C Mater Biol Appl. 2020;115:111097.PubMedCrossRef Liang B, et al. Biomimetic theranostic strategy for anti-metastasis therapy of breast cancer via the macrophage membrane camouflaged superparticles. Mater Sci Eng C Mater Biol Appl. 2020;115:111097.PubMedCrossRef
457.
go back to reference Xuan M, et al. Macrophage cell membrane camouflaged mesoporous silica nanocapsules for in vivo Cancer therapy. Adv Healthc Mater. 2015;4(11):1645–52.PubMedCrossRef Xuan M, et al. Macrophage cell membrane camouflaged mesoporous silica nanocapsules for in vivo Cancer therapy. Adv Healthc Mater. 2015;4(11):1645–52.PubMedCrossRef
458.
go back to reference Rao L, et al. Effective cancer targeting and imaging using macrophage membrane-camouflaged upconversion nanoparticles. J Biomed Mater Res A. 2017;105(2):521–30.PubMedCrossRef Rao L, et al. Effective cancer targeting and imaging using macrophage membrane-camouflaged upconversion nanoparticles. J Biomed Mater Res A. 2017;105(2):521–30.PubMedCrossRef
459.
go back to reference Cao X, et al. Paclitaxel-loaded macrophage membrane camouflaged Albumin nanoparticles for targeted Cancer Therapy. Int J Nanomed. 2020;15:1915–28.CrossRef Cao X, et al. Paclitaxel-loaded macrophage membrane camouflaged Albumin nanoparticles for targeted Cancer Therapy. Int J Nanomed. 2020;15:1915–28.CrossRef
460.
go back to reference Ma J, et al. Copresentation of Tumor antigens and Costimulatory molecules via Biomimetic nanoparticles for Effective Cancer Immunotherapy. Nano Lett. 2020;20(6):4084–94.PubMedCrossRef Ma J, et al. Copresentation of Tumor antigens and Costimulatory molecules via Biomimetic nanoparticles for Effective Cancer Immunotherapy. Nano Lett. 2020;20(6):4084–94.PubMedCrossRef
461.
go back to reference Li L, et al. Cholesterol removal improves performance of a model biomimetic system to co-deliver a photothermal agent and a STING agonist for cancer immunotherapy. Nat Commun. 2023;14(1):5111.PubMedPubMedCentralCrossRef Li L, et al. Cholesterol removal improves performance of a model biomimetic system to co-deliver a photothermal agent and a STING agonist for cancer immunotherapy. Nat Commun. 2023;14(1):5111.PubMedPubMedCentralCrossRef
462.
go back to reference Xie W, et al. Biomimetic nanoplatform loading type I Aggregation-Induced Emission Photosensitizer and glutamine blockade to regulate nutrient partitioning for enhancing Antitumor Immunotherapy. ACS Nano. 2022;16(7):10742–53.PubMedCrossRef Xie W, et al. Biomimetic nanoplatform loading type I Aggregation-Induced Emission Photosensitizer and glutamine blockade to regulate nutrient partitioning for enhancing Antitumor Immunotherapy. ACS Nano. 2022;16(7):10742–53.PubMedCrossRef
463.
go back to reference Zuo L, et al. Biomimetic nanovesicle with Mitochondria-Synthesized Sonosensitizer and Mitophagy Inhibition for Cancer Sono-Immunotherapy. Nano Lett. 2023;23(7):3005–13.PubMedCrossRef Zuo L, et al. Biomimetic nanovesicle with Mitochondria-Synthesized Sonosensitizer and Mitophagy Inhibition for Cancer Sono-Immunotherapy. Nano Lett. 2023;23(7):3005–13.PubMedCrossRef
464.
go back to reference Pan P, et al. A heterogenic membrane-based biomimetic hybrid nanoplatform for combining radiotherapy and immunotherapy against breast cancer. Biomaterials. 2022;289:121810.PubMedCrossRef Pan P, et al. A heterogenic membrane-based biomimetic hybrid nanoplatform for combining radiotherapy and immunotherapy against breast cancer. Biomaterials. 2022;289:121810.PubMedCrossRef
465.
go back to reference Yang C, et al. Leveraging β-Adrenergic receptor signaling blockade for Improved Cancer Immunotherapy through Biomimetic Nanovaccine. Small. 2023;19(14):e2207029.PubMedCrossRef Yang C, et al. Leveraging β-Adrenergic receptor signaling blockade for Improved Cancer Immunotherapy through Biomimetic Nanovaccine. Small. 2023;19(14):e2207029.PubMedCrossRef
466.
go back to reference Wang X, et al. Intelligent Biomimetic Nanoplatform for systemic treatment of metastatic triple-negative breast Cancer via enhanced EGFR-Targeted therapy and immunotherapy. ACS Appl Mater Interfaces; 2022. Wang X, et al. Intelligent Biomimetic Nanoplatform for systemic treatment of metastatic triple-negative breast Cancer via enhanced EGFR-Targeted therapy and immunotherapy. ACS Appl Mater Interfaces; 2022.
467.
go back to reference Li J, et al. Multifunctional biomimetic nanovaccines based on Photothermal and weak-immunostimulatory Nanoparticulate cores for the immunotherapy of solid tumors. Adv Mater. 2022;34(9):e2108012.PubMedCrossRef Li J, et al. Multifunctional biomimetic nanovaccines based on Photothermal and weak-immunostimulatory Nanoparticulate cores for the immunotherapy of solid tumors. Adv Mater. 2022;34(9):e2108012.PubMedCrossRef
468.
go back to reference Xu X, et al. A Biomimetic Aggregation-Induced Emission Photosensitizer with Antigen-presenting and hitchhiking function for lipid droplet targeted photodynamic immunotherapy. Adv Mater. 2021;33(33):e2102322.PubMedCrossRef Xu X, et al. A Biomimetic Aggregation-Induced Emission Photosensitizer with Antigen-presenting and hitchhiking function for lipid droplet targeted photodynamic immunotherapy. Adv Mater. 2021;33(33):e2102322.PubMedCrossRef
469.
go back to reference Fang X, et al. Biomimetic Anti-PD-1 peptide-loaded 2D FePSe(3) nanosheets for efficient photothermal and enhanced Immune Therapy with Multimodal MR/PA/Thermal Imaging. Adv Sci (Weinh). 2021;8(2):2003041.PubMedCrossRef Fang X, et al. Biomimetic Anti-PD-1 peptide-loaded 2D FePSe(3) nanosheets for efficient photothermal and enhanced Immune Therapy with Multimodal MR/PA/Thermal Imaging. Adv Sci (Weinh). 2021;8(2):2003041.PubMedCrossRef
471.
go back to reference Zhang F, et al. Janus nanocarrier-based co-delivery of doxorubicin and berberine weakens chemotherapy-exacerbated hepatocellular carcinoma recurrence. Acta Biomater. 2019;100:352–64.PubMedCrossRef Zhang F, et al. Janus nanocarrier-based co-delivery of doxorubicin and berberine weakens chemotherapy-exacerbated hepatocellular carcinoma recurrence. Acta Biomater. 2019;100:352–64.PubMedCrossRef
472.
473.
go back to reference Moradi-Chaleshtori M, et al. Overexpression of pigment epithelium-derived factor in breast cancer cell-derived exosomes induces M1 polarization in macrophages. Immunol Lett. 2022;248:31–6.PubMedCrossRef Moradi-Chaleshtori M, et al. Overexpression of pigment epithelium-derived factor in breast cancer cell-derived exosomes induces M1 polarization in macrophages. Immunol Lett. 2022;248:31–6.PubMedCrossRef
474.
go back to reference Wang Y et al. G-MDSC-derived exosomes mediate the differentiation of M-MDSC into M2 macrophages promoting colitis-to-cancer transition. J Immunother Cancer, 2023. 11(6). Wang Y et al. G-MDSC-derived exosomes mediate the differentiation of M-MDSC into M2 macrophages promoting colitis-to-cancer transition. J Immunother Cancer, 2023. 11(6).
475.
go back to reference Zheng N, et al. M2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance. Oncoimmunology. 2023;12(1):2210959.PubMedPubMedCentralCrossRef Zheng N, et al. M2 macrophage-derived exosomes suppress tumor intrinsic immunogenicity to confer immunotherapy resistance. Oncoimmunology. 2023;12(1):2210959.PubMedPubMedCentralCrossRef
476.
go back to reference Chen J, et al. PDL1-positive exosomes suppress antitumor immunity by inducing tumor-specific CD8(+) T cell exhaustion during metastasis. Cancer Sci. 2021;112(9):3437–54.PubMedPubMedCentralCrossRef Chen J, et al. PDL1-positive exosomes suppress antitumor immunity by inducing tumor-specific CD8(+) T cell exhaustion during metastasis. Cancer Sci. 2021;112(9):3437–54.PubMedPubMedCentralCrossRef
477.
go back to reference Wei Y, et al. An RNA-RNA crosstalk network involving HMGB1 and RICTOR facilitates hepatocellular carcinoma tumorigenesis by promoting glutamine metabolism and impedes immunotherapy by PD-L1 + exosomes activity. Signal Transduct Target Ther. 2021;6(1):421.PubMedPubMedCentralCrossRef Wei Y, et al. An RNA-RNA crosstalk network involving HMGB1 and RICTOR facilitates hepatocellular carcinoma tumorigenesis by promoting glutamine metabolism and impedes immunotherapy by PD-L1 + exosomes activity. Signal Transduct Target Ther. 2021;6(1):421.PubMedPubMedCentralCrossRef
478.
go back to reference Wang C, et al. Tumor Cell-associated exosomes robustly elicit anti-tumor Immune responses through modulating dendritic cell vaccines in Lung Tumor. Int J Biol Sci. 2020;16(4):633–43.PubMedPubMedCentralCrossRef Wang C, et al. Tumor Cell-associated exosomes robustly elicit anti-tumor Immune responses through modulating dendritic cell vaccines in Lung Tumor. Int J Biol Sci. 2020;16(4):633–43.PubMedPubMedCentralCrossRef
479.
go back to reference Wang S, et al. Macrophage-tumor chimeric exosomes accumulate in lymph node and tumor to activate the immune response and the tumor microenvironment. Sci Transl Med. 2021;13(615):eabb6981.PubMedCrossRef Wang S, et al. Macrophage-tumor chimeric exosomes accumulate in lymph node and tumor to activate the immune response and the tumor microenvironment. Sci Transl Med. 2021;13(615):eabb6981.PubMedCrossRef
480.
go back to reference Lu Z, et al. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol. 2017;67(4):739–48.PubMedCrossRef Lu Z, et al. Dendritic cell-derived exosomes elicit tumor regression in autochthonous hepatocellular carcinoma mouse models. J Hepatol. 2017;67(4):739–48.PubMedCrossRef
481.
go back to reference Zhou W, et al. Exosomes derived from immunogenically dying tumor cells as a versatile tool for vaccination against pancreatic cancer. Biomaterials. 2022;280:121306.PubMedCrossRef Zhou W, et al. Exosomes derived from immunogenically dying tumor cells as a versatile tool for vaccination against pancreatic cancer. Biomaterials. 2022;280:121306.PubMedCrossRef
482.
483.
go back to reference Morishita M, et al. Exosome-based tumor antigens-adjuvant co-delivery utilizing genetically engineered tumor cell-derived exosomes with immunostimulatory CpG DNA. Biomaterials. 2016;111:55–65.PubMedCrossRef Morishita M, et al. Exosome-based tumor antigens-adjuvant co-delivery utilizing genetically engineered tumor cell-derived exosomes with immunostimulatory CpG DNA. Biomaterials. 2016;111:55–65.PubMedCrossRef
484.
go back to reference Gehrmann U, et al. Synergistic induction of adaptive antitumor immunity by codelivery of antigen with α-galactosylceramide on exosomes. Cancer Res. 2013;73(13):3865–76.PubMedCrossRef Gehrmann U, et al. Synergistic induction of adaptive antitumor immunity by codelivery of antigen with α-galactosylceramide on exosomes. Cancer Res. 2013;73(13):3865–76.PubMedCrossRef
485.
go back to reference Kim GB et al. Xenogenization of tumor cells by fusogenic exosomes in tumor microenvironment ignites and propagates antitumor immunity. Sci Adv, 2020. 6(27). Kim GB et al. Xenogenization of tumor cells by fusogenic exosomes in tumor microenvironment ignites and propagates antitumor immunity. Sci Adv, 2020. 6(27).
486.
go back to reference Zhou W, et al. Pancreatic cancer-targeting exosomes for enhancing immunotherapy and reprogramming tumor microenvironment. Biomaterials. 2021;268:120546.PubMedCrossRef Zhou W, et al. Pancreatic cancer-targeting exosomes for enhancing immunotherapy and reprogramming tumor microenvironment. Biomaterials. 2021;268:120546.PubMedCrossRef
487.
go back to reference Gunassekaran GR, et al. M1 macrophage exosomes engineered to foster M1 polarization and target the IL-4 receptor inhibit tumor growth by reprogramming tumor-associated macrophages into M1-like macrophages. Biomaterials. 2021;278:121137.PubMedCrossRef Gunassekaran GR, et al. M1 macrophage exosomes engineered to foster M1 polarization and target the IL-4 receptor inhibit tumor growth by reprogramming tumor-associated macrophages into M1-like macrophages. Biomaterials. 2021;278:121137.PubMedCrossRef
488.
go back to reference Cheng L, et al. Gene-engineered exosomes-thermosensitive liposomes hybrid nanovesicles by the blockade of CD47 signal for combined photothermal therapy and cancer immunotherapy. Biomaterials. 2021;275:120964.PubMedCrossRef Cheng L, et al. Gene-engineered exosomes-thermosensitive liposomes hybrid nanovesicles by the blockade of CD47 signal for combined photothermal therapy and cancer immunotherapy. Biomaterials. 2021;275:120964.PubMedCrossRef
489.
go back to reference Li J, et al. Dendritic cell derived exosomes loaded neoantigens for personalized cancer immunotherapies. J Control Release. 2023;353:423–33.PubMedCrossRef Li J, et al. Dendritic cell derived exosomes loaded neoantigens for personalized cancer immunotherapies. J Control Release. 2023;353:423–33.PubMedCrossRef
490.
go back to reference Xu J, et al. Enhanced penetrative siRNA delivery by a nanodiamond drug delivery platform against hepatocellular carcinoma 3D models. Nanoscale. 2021;13(38):16131–45.PubMedCrossRef Xu J, et al. Enhanced penetrative siRNA delivery by a nanodiamond drug delivery platform against hepatocellular carcinoma 3D models. Nanoscale. 2021;13(38):16131–45.PubMedCrossRef
491.
go back to reference Jiang H, et al. M1 macrophage-derived exosomes and their key molecule lncRNA HOTTIP suppress head and neck squamous cell carcinoma progression by upregulating the TLR5/NF-κB pathway. Cell Death Dis. 2022;13(2):183.PubMedPubMedCentralCrossRef Jiang H, et al. M1 macrophage-derived exosomes and their key molecule lncRNA HOTTIP suppress head and neck squamous cell carcinoma progression by upregulating the TLR5/NF-κB pathway. Cell Death Dis. 2022;13(2):183.PubMedPubMedCentralCrossRef
492.
go back to reference Zhou WJ, et al. CD45RO(-)CD8(+) T cell-derived exosomes restrict estrogen-driven endometrial cancer development via the ERβ/miR-765/PLP2/Notch axis. Theranostics. 2021;11(11):5330–45.PubMedPubMedCentralCrossRef Zhou WJ, et al. CD45RO(-)CD8(+) T cell-derived exosomes restrict estrogen-driven endometrial cancer development via the ERβ/miR-765/PLP2/Notch axis. Theranostics. 2021;11(11):5330–45.PubMedPubMedCentralCrossRef
493.
go back to reference Cheng L, et al. Multifunctional hybrid exosomes enhanced cancer chemo-immunotherapy by activating the STING pathway. Biomaterials. 2023;301:122259.PubMedCrossRef Cheng L, et al. Multifunctional hybrid exosomes enhanced cancer chemo-immunotherapy by activating the STING pathway. Biomaterials. 2023;301:122259.PubMedCrossRef
494.
go back to reference Wang X et al. Exosomes derived from γδ-T cells synergize with radiotherapy and preserve antitumor activities against nasopharyngeal carcinoma in immunosuppressive microenvironment. J Immunother Cancer, 2022. 10(2). Wang X et al. Exosomes derived from γδ-T cells synergize with radiotherapy and preserve antitumor activities against nasopharyngeal carcinoma in immunosuppressive microenvironment. J Immunother Cancer, 2022. 10(2).
495.
go back to reference Ji P, et al. Smart exosomes with lymph node homing and immune-amplifying capacities for enhanced immunotherapy of metastatic breast cancer. Mol Ther Nucleic Acids. 2021;26:987–96.PubMedPubMedCentralCrossRef Ji P, et al. Smart exosomes with lymph node homing and immune-amplifying capacities for enhanced immunotherapy of metastatic breast cancer. Mol Ther Nucleic Acids. 2021;26:987–96.PubMedPubMedCentralCrossRef
496.
go back to reference Fan M, et al. A CAR T-inspiring platform based on antibody-engineered exosomes from antigen-feeding dendritic cells for precise solid tumor therapy. Biomaterials. 2022;282:121424.PubMedCrossRef Fan M, et al. A CAR T-inspiring platform based on antibody-engineered exosomes from antigen-feeding dendritic cells for precise solid tumor therapy. Biomaterials. 2022;282:121424.PubMedCrossRef
497.
go back to reference Li Y, et al. A nanotherapeutic system for gastric cancer suppression by synergistic chemotherapy and immunotherapy based on iPSCs and DCs exosomes. Cancer Immunol Immunother. 2023;72(6):1673–83.PubMedCrossRef Li Y, et al. A nanotherapeutic system for gastric cancer suppression by synergistic chemotherapy and immunotherapy based on iPSCs and DCs exosomes. Cancer Immunol Immunother. 2023;72(6):1673–83.PubMedCrossRef
498.
go back to reference Huang H, et al. Synergistic strategy with hyperthermia therapy based immunotherapy and engineered exosomes-liposomes targeted chemotherapy prevents tumor recurrence and metastasis in advanced breast cancer. Bioeng Transl Med. 2022;7(2):e10284.PubMedCrossRef Huang H, et al. Synergistic strategy with hyperthermia therapy based immunotherapy and engineered exosomes-liposomes targeted chemotherapy prevents tumor recurrence and metastasis in advanced breast cancer. Bioeng Transl Med. 2022;7(2):e10284.PubMedCrossRef
499.
go back to reference Pakravan N, Abbasi A, Hassan ZM. Immunotherapy using oxygenated water and tumor-derived exosomes potentiates antitumor immune response and attenuates malignancy tendency in mice model of breast cancer oxid med cell longev, 2021;2021:5529484. Pakravan N, Abbasi A, Hassan ZM. Immunotherapy using oxygenated water and tumor-derived exosomes potentiates antitumor immune response and attenuates malignancy tendency in mice model of breast cancer oxid med cell longev, 2021;2021:5529484.
500.
go back to reference Javeed N, et al. Immunosuppressive CD14(+)HLA-DR(lo/neg) monocytes are elevated in pancreatic cancer and primed by tumor-derived exosomes. Oncoimmunology. 2017;6(1):e1252013.PubMedCrossRef Javeed N, et al. Immunosuppressive CD14(+)HLA-DR(lo/neg) monocytes are elevated in pancreatic cancer and primed by tumor-derived exosomes. Oncoimmunology. 2017;6(1):e1252013.PubMedCrossRef
501.
go back to reference Zhao Y, et al. Docetaxel-loaded M1 macrophage-derived exosomes for a safe and efficient chemoimmunotherapy of breast cancer. J Nanobiotechnol. 2022;20(1):359.CrossRef Zhao Y, et al. Docetaxel-loaded M1 macrophage-derived exosomes for a safe and efficient chemoimmunotherapy of breast cancer. J Nanobiotechnol. 2022;20(1):359.CrossRef
502.
go back to reference Lin J, et al. Dendritic cell-derived exosomes Driven Drug Co-delivery Biomimetic Nanosystem for Effective Combination of Malignant Melanoma Immunotherapy and Gene Therapy. Drug Des Devel Ther. 2023;17:2087–106.PubMedPubMedCentralCrossRef Lin J, et al. Dendritic cell-derived exosomes Driven Drug Co-delivery Biomimetic Nanosystem for Effective Combination of Malignant Melanoma Immunotherapy and Gene Therapy. Drug Des Devel Ther. 2023;17:2087–106.PubMedPubMedCentralCrossRef
503.
go back to reference Wang D, et al. Sonodynamical reversion of immunosuppressive microenvironment in prostate cancer via engineered exosomes. Drug Deliv. 2022;29(1):702–13.PubMedPubMedCentralCrossRef Wang D, et al. Sonodynamical reversion of immunosuppressive microenvironment in prostate cancer via engineered exosomes. Drug Deliv. 2022;29(1):702–13.PubMedPubMedCentralCrossRef
504.
go back to reference Zhang Y et al. pH-Responsive STING-Activating DNA nanovaccines for Cancer Immunotherapy. Adv Ther (Weinh), 2020. 3(9). Zhang Y et al. pH-Responsive STING-Activating DNA nanovaccines for Cancer Immunotherapy. Adv Ther (Weinh), 2020. 3(9).
505.
go back to reference Yao D, et al. A self-cascaded unimolecular prodrug for pH-responsive chemotherapy and tumor-detained photodynamic-immunotherapy of triple-negative breast cancer. Biomaterials. 2023;292:121920.PubMedCrossRef Yao D, et al. A self-cascaded unimolecular prodrug for pH-responsive chemotherapy and tumor-detained photodynamic-immunotherapy of triple-negative breast cancer. Biomaterials. 2023;292:121920.PubMedCrossRef
506.
go back to reference Liu Y, et al. Dual pH-responsive multifunctional nanoparticles for targeted treatment of breast cancer by combining immunotherapy and chemotherapy. Acta Biomater. 2018;66:310–24.PubMedCrossRef Liu Y, et al. Dual pH-responsive multifunctional nanoparticles for targeted treatment of breast cancer by combining immunotherapy and chemotherapy. Acta Biomater. 2018;66:310–24.PubMedCrossRef
507.
go back to reference Lim B, Kim KS, Na K. pH-Responsive zinc Ion regulating Immunomodulatory nanoparticles for Effective Cancer Immunotherapy. Biomacromolecules. 2023;24(9):4263–73.PubMedCrossRef Lim B, Kim KS, Na K. pH-Responsive zinc Ion regulating Immunomodulatory nanoparticles for Effective Cancer Immunotherapy. Biomacromolecules. 2023;24(9):4263–73.PubMedCrossRef
508.
go back to reference Duong HTT, et al. Smart vaccine delivery based on microneedle arrays decorated with ultra-ph-responsive copolymers for cancer immunotherapy. Biomaterials. 2018;185:13–24.PubMedCrossRef Duong HTT, et al. Smart vaccine delivery based on microneedle arrays decorated with ultra-ph-responsive copolymers for cancer immunotherapy. Biomaterials. 2018;185:13–24.PubMedCrossRef
509.
go back to reference Gu J, et al. Injectable pH-responsive hydrogel for combinatorial chemoimmunotherapy tailored to the tumor microenvironment. J Nanobiotechnol. 2022;20(1):372.CrossRef Gu J, et al. Injectable pH-responsive hydrogel for combinatorial chemoimmunotherapy tailored to the tumor microenvironment. J Nanobiotechnol. 2022;20(1):372.CrossRef
510.
go back to reference Gong C, et al. Regulating the immunosuppressive tumor microenvironment to enhance breast cancer immunotherapy using pH-responsive hybrid membrane-coated nanoparticles. J Nanobiotechnol. 2021;19(1):58.CrossRef Gong C, et al. Regulating the immunosuppressive tumor microenvironment to enhance breast cancer immunotherapy using pH-responsive hybrid membrane-coated nanoparticles. J Nanobiotechnol. 2021;19(1):58.CrossRef
511.
go back to reference Ding Y, et al. Construction of pH-Sensitive nanovaccines encapsulating tumor cell lysates and Immune adjuvants for breast Cancer therapy. Small. 2023;19(37):e2301420.PubMedCrossRef Ding Y, et al. Construction of pH-Sensitive nanovaccines encapsulating tumor cell lysates and Immune adjuvants for breast Cancer therapy. Small. 2023;19(37):e2301420.PubMedCrossRef
512.
go back to reference Jiang M, et al. Sequentially pH-Responsive drug-delivery Nanosystem for Tumor Immunogenic Cell Death and cooperating with Immune checkpoint blockade for efficient Cancer Chemoimmunotherapy. ACS Appl Mater Interfaces. 2021;13(37):43963–74.PubMedCrossRef Jiang M, et al. Sequentially pH-Responsive drug-delivery Nanosystem for Tumor Immunogenic Cell Death and cooperating with Immune checkpoint blockade for efficient Cancer Chemoimmunotherapy. ACS Appl Mater Interfaces. 2021;13(37):43963–74.PubMedCrossRef
513.
go back to reference Yang G, et al. Smart Nanoreactors for pH-Responsive Tumor Homing, Mitochondria-Targeting, and enhanced photodynamic-immunotherapy of Cancer. Nano Lett. 2018;18(4):2475–84.PubMedCrossRef Yang G, et al. Smart Nanoreactors for pH-Responsive Tumor Homing, Mitochondria-Targeting, and enhanced photodynamic-immunotherapy of Cancer. Nano Lett. 2018;18(4):2475–84.PubMedCrossRef
514.
go back to reference Sun X, et al. Binary regulation of the tumor microenvironment by a pH-responsive reversible shielding nanoplatform for improved tumor chemo-immunotherapy. Acta Biomater. 2022;138:505–17.PubMedCrossRef Sun X, et al. Binary regulation of the tumor microenvironment by a pH-responsive reversible shielding nanoplatform for improved tumor chemo-immunotherapy. Acta Biomater. 2022;138:505–17.PubMedCrossRef
515.
go back to reference Lv X, et al. Multi-signaling pathway activation by pH responsive manganese particles for enhanced vaccination. J Control Release. 2023;357:109–19.PubMedCrossRef Lv X, et al. Multi-signaling pathway activation by pH responsive manganese particles for enhanced vaccination. J Control Release. 2023;357:109–19.PubMedCrossRef
516.
go back to reference Wagner J, et al. Mesoporous silica nanoparticles as pH-Responsive carrier for the Immune-Activating Drug Resiquimod enhance the local Immune response in mice. ACS Nano. 2021;15(3):4450–66.PubMedCrossRef Wagner J, et al. Mesoporous silica nanoparticles as pH-Responsive carrier for the Immune-Activating Drug Resiquimod enhance the local Immune response in mice. ACS Nano. 2021;15(3):4450–66.PubMedCrossRef
517.
go back to reference Zhao M, et al. Immune/Hypoxic Tumor Microenvironment Regulation-enhanced photodynamic treatment realized by pH-Responsive phase Transition-Targeting Nanobubbles. ACS Appl Mater Interfaces. 2021;13(28):32763–79.PubMedCrossRef Zhao M, et al. Immune/Hypoxic Tumor Microenvironment Regulation-enhanced photodynamic treatment realized by pH-Responsive phase Transition-Targeting Nanobubbles. ACS Appl Mater Interfaces. 2021;13(28):32763–79.PubMedCrossRef
518.
go back to reference Yuba E, et al. The application of pH-sensitive polymer-lipids to antigen delivery for cancer immunotherapy. Biomaterials. 2013;34(22):5711–21.PubMedCrossRef Yuba E, et al. The application of pH-sensitive polymer-lipids to antigen delivery for cancer immunotherapy. Biomaterials. 2013;34(22):5711–21.PubMedCrossRef
519.
go back to reference Yoshizaki Y, et al. Potentiation of pH-sensitive polymer-modified liposomes with cationic lipid inclusion as antigen delivery carriers for cancer immunotherapy. Biomaterials. 2014;35(28):8186–96.PubMedCrossRef Yoshizaki Y, et al. Potentiation of pH-sensitive polymer-modified liposomes with cationic lipid inclusion as antigen delivery carriers for cancer immunotherapy. Biomaterials. 2014;35(28):8186–96.PubMedCrossRef
520.
go back to reference Yuba E, et al. A liposome-based antigen delivery system using pH-sensitive fusogenic polymers for cancer immunotherapy. Biomaterials. 2013;34(12):3042–52.PubMedCrossRef Yuba E, et al. A liposome-based antigen delivery system using pH-sensitive fusogenic polymers for cancer immunotherapy. Biomaterials. 2013;34(12):3042–52.PubMedCrossRef
521.
go back to reference Yoshizaki Y, et al. pH-sensitive polymer-modified liposome-based immunity-inducing system: effects of inclusion of cationic lipid and CpG-DNA. Biomaterials. 2017;141:272–83.PubMedCrossRef Yoshizaki Y, et al. pH-sensitive polymer-modified liposome-based immunity-inducing system: effects of inclusion of cationic lipid and CpG-DNA. Biomaterials. 2017;141:272–83.PubMedCrossRef
522.
go back to reference Zhang J, et al. pH-sensitive tumor-tropism hybrid membrane-coated nanoparticles for reprogramming the tumor microenvironment and boosting the antitumor immunity. Acta Biomater. 2023;166:470–84.PubMedCrossRef Zhang J, et al. pH-sensitive tumor-tropism hybrid membrane-coated nanoparticles for reprogramming the tumor microenvironment and boosting the antitumor immunity. Acta Biomater. 2023;166:470–84.PubMedCrossRef
523.
go back to reference Yuba E, et al. Bioactive polysaccharide-based pH-sensitive polymers for cytoplasmic delivery of antigen and activation of antigen-specific immunity. Biomaterials. 2017;120:32–45.PubMedCrossRef Yuba E, et al. Bioactive polysaccharide-based pH-sensitive polymers for cytoplasmic delivery of antigen and activation of antigen-specific immunity. Biomaterials. 2017;120:32–45.PubMedCrossRef
524.
go back to reference Kocabas BB, et al. Dual-adjuvant effect of pH-sensitive liposomes loaded with STING and TLR9 agonists regress tumor development by enhancing Th1 immune response. J Control Release. 2020;328:587–95.PubMedCrossRef Kocabas BB, et al. Dual-adjuvant effect of pH-sensitive liposomes loaded with STING and TLR9 agonists regress tumor development by enhancing Th1 immune response. J Control Release. 2020;328:587–95.PubMedCrossRef
525.
go back to reference Zhang W, et al. Multifunctional redox-responsive nanoplatform with Dual Activation of Macrophages and T Cells for Antitumor Immunotherapy. ACS Nano. 2023;17(15):14424–41.PubMedCrossRef Zhang W, et al. Multifunctional redox-responsive nanoplatform with Dual Activation of Macrophages and T Cells for Antitumor Immunotherapy. ACS Nano. 2023;17(15):14424–41.PubMedCrossRef
526.
go back to reference Yan P, et al. A redox-responsive Nanovaccine combined with A2A receptor antagonist for Cancer Immunotherapy. Adv Healthc Mater. 2021;10(21):e2101222.PubMedCrossRef Yan P, et al. A redox-responsive Nanovaccine combined with A2A receptor antagonist for Cancer Immunotherapy. Adv Healthc Mater. 2021;10(21):e2101222.PubMedCrossRef
527.
go back to reference Lv M, et al. Redox-responsive hyperbranched poly(amido amine) and polymer dots as a vaccine delivery system for cancer immunotherapy. J Mater Chem B. 2017;5(48):9532–45.PubMedCrossRef Lv M, et al. Redox-responsive hyperbranched poly(amido amine) and polymer dots as a vaccine delivery system for cancer immunotherapy. J Mater Chem B. 2017;5(48):9532–45.PubMedCrossRef
528.
go back to reference Jiang D, et al. Cascade Cytosol Delivery of Dual-Sensitive Micelle-tailored vaccine for Enhancing Cancer Immunotherapy. ACS Appl Mater Interfaces. 2018;10(44):37797–811.PubMedCrossRef Jiang D, et al. Cascade Cytosol Delivery of Dual-Sensitive Micelle-tailored vaccine for Enhancing Cancer Immunotherapy. ACS Appl Mater Interfaces. 2018;10(44):37797–811.PubMedCrossRef
529.
go back to reference Chen Q, et al. Cancer cell membrane-coated nanoparticles for bimodal imaging-guided photothermal therapy and docetaxel-enhanced immunotherapy against cancer. J Nanobiotechnol. 2021;19(1):449.CrossRef Chen Q, et al. Cancer cell membrane-coated nanoparticles for bimodal imaging-guided photothermal therapy and docetaxel-enhanced immunotherapy against cancer. J Nanobiotechnol. 2021;19(1):449.CrossRef
530.
go back to reference Chen L, et al. Tumor-targeted drug and CpG delivery system for Phototherapy and Docetaxel-enhanced immunotherapy with polarization toward M1-Type macrophages on Triple negative breast cancers. Adv Mater. 2019;31(52):e1904997.PubMedCrossRef Chen L, et al. Tumor-targeted drug and CpG delivery system for Phototherapy and Docetaxel-enhanced immunotherapy with polarization toward M1-Type macrophages on Triple negative breast cancers. Adv Mater. 2019;31(52):e1904997.PubMedCrossRef
531.
go back to reference Sun Y, et al. An enzyme-responsive and transformable PD-L1 blocking peptide-photosensitizer conjugate enables efficient photothermal immunotherapy for breast cancer. Bioact Mater. 2023;22:47–59.PubMed Sun Y, et al. An enzyme-responsive and transformable PD-L1 blocking peptide-photosensitizer conjugate enables efficient photothermal immunotherapy for breast cancer. Bioact Mater. 2023;22:47–59.PubMed
532.
go back to reference Ma S, et al. Boosting cancer immunotherapy by biomineralized nanovaccine with ferroptosis-inducing and photothermal properties. Biomater Sci. 2023;11(2):518–32.PubMedCrossRef Ma S, et al. Boosting cancer immunotherapy by biomineralized nanovaccine with ferroptosis-inducing and photothermal properties. Biomater Sci. 2023;11(2):518–32.PubMedCrossRef
533.
go back to reference Xiao Y, et al. Microenvironment-responsive prodrug-induced pyroptosis boosts cancer immunotherapy. Adv Sci (Weinh). 2021;8(24):e2101840.PubMedCrossRef Xiao Y, et al. Microenvironment-responsive prodrug-induced pyroptosis boosts cancer immunotherapy. Adv Sci (Weinh). 2021;8(24):e2101840.PubMedCrossRef
534.
go back to reference Wu X, et al. Nano-herb medicine and PDT induced synergistic immunotherapy for colon cancer treatment. Biomaterials. 2021;269:120654.PubMedCrossRef Wu X, et al. Nano-herb medicine and PDT induced synergistic immunotherapy for colon cancer treatment. Biomaterials. 2021;269:120654.PubMedCrossRef
535.
go back to reference Ding D, et al. Multifunctional Nanodrug mediates synergistic photodynamic therapy and MDSCs-Targeting immunotherapy of Colon cancer. Adv Sci (Weinh). 2021;8(14):e2100712.PubMedCrossRef Ding D, et al. Multifunctional Nanodrug mediates synergistic photodynamic therapy and MDSCs-Targeting immunotherapy of Colon cancer. Adv Sci (Weinh). 2021;8(14):e2100712.PubMedCrossRef
536.
go back to reference Zhou H, et al. A tumor-microenvironment-activatable Molecular Pro-theranostic Agent for Photodynamic and Immunotherapy of Cancer. Adv Mater. 2023;35(30):e2211485.PubMedCrossRef Zhou H, et al. A tumor-microenvironment-activatable Molecular Pro-theranostic Agent for Photodynamic and Immunotherapy of Cancer. Adv Mater. 2023;35(30):e2211485.PubMedCrossRef
537.
go back to reference Yu Y, et al. Polymeric PD-L1 blockade nanoparticles for cancer photothermal-immunotherapy. Biomaterials. 2022;280:121312.PubMedCrossRef Yu Y, et al. Polymeric PD-L1 blockade nanoparticles for cancer photothermal-immunotherapy. Biomaterials. 2022;280:121312.PubMedCrossRef
538.
go back to reference Liang X, et al. Photothermal cancer immunotherapy by erythrocyte membrane-coated black phosphorus formulation. J Control Release. 2019;296:150–61.PubMedCrossRef Liang X, et al. Photothermal cancer immunotherapy by erythrocyte membrane-coated black phosphorus formulation. J Control Release. 2019;296:150–61.PubMedCrossRef
539.
go back to reference Zhang R, et al. Chemodynamic/Photothermal synergistic Cancer immunotherapy based on yeast microcapsule-derived Au/Pt nanoparticles. ACS Appl Mater Interfaces. 2023;15(20):24134–48.PubMedCrossRef Zhang R, et al. Chemodynamic/Photothermal synergistic Cancer immunotherapy based on yeast microcapsule-derived Au/Pt nanoparticles. ACS Appl Mater Interfaces. 2023;15(20):24134–48.PubMedCrossRef
540.
go back to reference Wang M, et al. Synergistic interventional photothermal therapy and immunotherapy using an iron oxide nanoplatform for the treatment of pancreatic cancer. Acta Biomater. 2022;138:453–62.PubMedCrossRef Wang M, et al. Synergistic interventional photothermal therapy and immunotherapy using an iron oxide nanoplatform for the treatment of pancreatic cancer. Acta Biomater. 2022;138:453–62.PubMedCrossRef
541.
go back to reference Debele TA, Yeh C-F, Su W-P. Cancer immunotherapy and application of nanoparticles in cancers immunotherapy as the delivery of immunotherapeutic agents and as the immunomodulators. Cancers. 2020;12(12):3773.PubMedPubMedCentralCrossRef Debele TA, Yeh C-F, Su W-P. Cancer immunotherapy and application of nanoparticles in cancers immunotherapy as the delivery of immunotherapeutic agents and as the immunomodulators. Cancers. 2020;12(12):3773.PubMedPubMedCentralCrossRef
542.
go back to reference Zhang C, et al. Checkpoint Nano-PROTACs for Activatable Cancer Photo-Immunotherapy. Adv Mater. 2023;35(6):e2208553.PubMedCrossRef Zhang C, et al. Checkpoint Nano-PROTACs for Activatable Cancer Photo-Immunotherapy. Adv Mater. 2023;35(6):e2208553.PubMedCrossRef
543.
go back to reference Qiu W, et al. Immunomodulatory-photodynamic nanostimulators for invoking pyroptosis to augment Tumor Immunotherapy. Adv Healthc Mater. 2022;11(21):e2201233.PubMedCrossRef Qiu W, et al. Immunomodulatory-photodynamic nanostimulators for invoking pyroptosis to augment Tumor Immunotherapy. Adv Healthc Mater. 2022;11(21):e2201233.PubMedCrossRef
544.
go back to reference Zhou JY, et al. Ru(II)-modified TiO(2) nanoparticles for hypoxia-adaptive photo-immunotherapy of oral squamous cell carcinoma. Biomaterials. 2022;289:121757.PubMedCrossRef Zhou JY, et al. Ru(II)-modified TiO(2) nanoparticles for hypoxia-adaptive photo-immunotherapy of oral squamous cell carcinoma. Biomaterials. 2022;289:121757.PubMedCrossRef
545.
go back to reference Duan Y, et al. Immune Modulator and Low-Temperature PTT-Induced Synergistic Immunotherapy for Cancer Treatment. ACS Appl Bio Mater. 2021;4(2):1524–35.PubMedCrossRef Duan Y, et al. Immune Modulator and Low-Temperature PTT-Induced Synergistic Immunotherapy for Cancer Treatment. ACS Appl Bio Mater. 2021;4(2):1524–35.PubMedCrossRef
546.
go back to reference Yan T, et al. Synergistic photothermal cancer immunotherapy by Cas9 ribonucleoprotein-based copper sulfide nanotherapeutic platform targeting PTPN2. Biomaterials. 2021;279:121233.PubMedCrossRef Yan T, et al. Synergistic photothermal cancer immunotherapy by Cas9 ribonucleoprotein-based copper sulfide nanotherapeutic platform targeting PTPN2. Biomaterials. 2021;279:121233.PubMedCrossRef
547.
go back to reference Li J, et al. Second Near-Infrared Photothermal Semiconducting polymer nanoadjuvant for enhanced Cancer Immunotherapy. Adv Mater. 2021;33(4):e2003458.PubMedCrossRef Li J, et al. Second Near-Infrared Photothermal Semiconducting polymer nanoadjuvant for enhanced Cancer Immunotherapy. Adv Mater. 2021;33(4):e2003458.PubMedCrossRef
548.
go back to reference Jana D et al. A defect-Engineered Nanozyme for targeted NIR-II Photothermal Immunotherapy of Cancer. Adv Mater, 2022: p. e2206401. Jana D et al. A defect-Engineered Nanozyme for targeted NIR-II Photothermal Immunotherapy of Cancer. Adv Mater, 2022: p. e2206401.
549.
go back to reference Hwang J, et al. Escherichia coli Mimetic Gold nanorod-mediated photo- and immunotherapy for Treating Cancer and its metastasis. ACS Nano. 2022;16(5):8472–83.PubMedCrossRef Hwang J, et al. Escherichia coli Mimetic Gold nanorod-mediated photo- and immunotherapy for Treating Cancer and its metastasis. ACS Nano. 2022;16(5):8472–83.PubMedCrossRef
550.
go back to reference Tang Y, et al. Pyroptosis-mediated synergistic photodynamic and photothermal immunotherapy enabled by a tumor-membrane-targeted photosensitive dimer. Adv Mater. 2023;35(25):e2300232.PubMedCrossRef Tang Y, et al. Pyroptosis-mediated synergistic photodynamic and photothermal immunotherapy enabled by a tumor-membrane-targeted photosensitive dimer. Adv Mater. 2023;35(25):e2300232.PubMedCrossRef
551.
go back to reference Xu M, et al. Activatable Immunoprotease Nanorestimulator for Second Near-Infrared Photothermal Immunotherapy of Cancer. ACS Nano. 2023;17(9):8183–94.PubMedCrossRef Xu M, et al. Activatable Immunoprotease Nanorestimulator for Second Near-Infrared Photothermal Immunotherapy of Cancer. ACS Nano. 2023;17(9):8183–94.PubMedCrossRef
552.
go back to reference Wang M, et al. NIR-Triggered Phototherapy and Immunotherapy via an Antigen-capturing nanoplatform for metastatic Cancer treatment. Adv Sci (Weinh). 2019;6(10):1802157.PubMedCrossRef Wang M, et al. NIR-Triggered Phototherapy and Immunotherapy via an Antigen-capturing nanoplatform for metastatic Cancer treatment. Adv Sci (Weinh). 2019;6(10):1802157.PubMedCrossRef
553.
go back to reference Ye X, et al. Surgical Tumor-Derived Personalized Photothermal Vaccine Formulation for Cancer Immunotherapy. ACS Nano. 2019;13(3):2956–68.PubMedCrossRef Ye X, et al. Surgical Tumor-Derived Personalized Photothermal Vaccine Formulation for Cancer Immunotherapy. ACS Nano. 2019;13(3):2956–68.PubMedCrossRef
554.
go back to reference Wang S, et al. Photothermal therapy mediated by gold nanocages composed of anti-PDL1 and galunisertib for improved synergistic immunotherapy in colorectal cancer. Acta Biomater. 2021;134:621–32.PubMedCrossRef Wang S, et al. Photothermal therapy mediated by gold nanocages composed of anti-PDL1 and galunisertib for improved synergistic immunotherapy in colorectal cancer. Acta Biomater. 2021;134:621–32.PubMedCrossRef
555.
go back to reference Mikelez-Alonso I, Aires A, Cortajarena AL. Cancer nano-immunotherapy from the injection to the target: the role of protein corona. Int J Mol Sci. 2020;21(2):519.PubMedPubMedCentralCrossRef Mikelez-Alonso I, Aires A, Cortajarena AL. Cancer nano-immunotherapy from the injection to the target: the role of protein corona. Int J Mol Sci. 2020;21(2):519.PubMedPubMedCentralCrossRef
556.
go back to reference Yetisgin AA et al. Therapeutic nanoparticles and their targeted delivery applications. Molecules, 2020. 25(9). Yetisgin AA et al. Therapeutic nanoparticles and their targeted delivery applications. Molecules, 2020. 25(9).
558.
go back to reference Kranz LM, et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature. 2016;534(7607):396–401.PubMedCrossRef Kranz LM, et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature. 2016;534(7607):396–401.PubMedCrossRef
559.
go back to reference Beg MS, et al. Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Invest New Drugs. 2017;35(2):180–8.PubMedCrossRef Beg MS, et al. Phase I study of MRX34, a liposomal miR-34a mimic, administered twice weekly in patients with advanced solid tumors. Invest New Drugs. 2017;35(2):180–8.PubMedCrossRef
560.
go back to reference Burris III et al. H.A., A phase 1, open-label, multicenter study to assess the safety, tolerability, and immunogenicity of mRNA-4157 alone in subjects with resected solid tumors and in combination with pembrolizumab in subjects with unresectable solid tumors (Keynote-603). 2019, American Society of Clinical Oncology. Burris III et al. H.A., A phase 1, open-label, multicenter study to assess the safety, tolerability, and immunogenicity of mRNA-4157 alone in subjects with resected solid tumors and in combination with pembrolizumab in subjects with unresectable solid tumors (Keynote-603). 2019, American Society of Clinical Oncology.
561.
go back to reference Pfannenstiel LW, et al. Paclitaxel enhances early dendritic cell maturation and function through TLR4 signaling in mice. Cell Immunol. 2010;263(1):79–87.PubMedPubMedCentralCrossRef Pfannenstiel LW, et al. Paclitaxel enhances early dendritic cell maturation and function through TLR4 signaling in mice. Cell Immunol. 2010;263(1):79–87.PubMedPubMedCentralCrossRef
562.
go back to reference Graziani SR, et al. Phase II study of paclitaxel associated with lipid core nanoparticles (LDE) as third-line treatment of patients with epithelial ovarian carcinoma. Med Oncol. 2017;34:1–7.CrossRef Graziani SR, et al. Phase II study of paclitaxel associated with lipid core nanoparticles (LDE) as third-line treatment of patients with epithelial ovarian carcinoma. Med Oncol. 2017;34:1–7.CrossRef
563.
go back to reference Shen C, et al. Phase I study of NBTXR3 activated by radiotherapy in patients with advanced cancers treated with an anti-PD-1 therapy. American Society of Clinical Oncology; 2020. Shen C, et al. Phase I study of NBTXR3 activated by radiotherapy in patients with advanced cancers treated with an anti-PD-1 therapy. American Society of Clinical Oncology; 2020.
564.
go back to reference Li S, et al. Polymer nanoparticles as adjuvants in cancer immunotherapy. Nano Res. 2018;11:5769–86.CrossRef Li S, et al. Polymer nanoparticles as adjuvants in cancer immunotherapy. Nano Res. 2018;11:5769–86.CrossRef
567.
go back to reference Chen P-G, et al. Chitosan nanoparticles based nanovaccines for cancer immunotherapy. Pure Appl Chem. 2017;89(7):931–9.CrossRef Chen P-G, et al. Chitosan nanoparticles based nanovaccines for cancer immunotherapy. Pure Appl Chem. 2017;89(7):931–9.CrossRef
568.
go back to reference Shi GN, et al. Enhanced antitumor immunity by targeting dendritic cells with tumor cell lysate-loaded chitosan nanoparticles vaccine. Biomaterials. 2017;113:191–202.PubMedCrossRef Shi GN, et al. Enhanced antitumor immunity by targeting dendritic cells with tumor cell lysate-loaded chitosan nanoparticles vaccine. Biomaterials. 2017;113:191–202.PubMedCrossRef
569.
go back to reference Yang R, et al. Cancer Cell membrane-coated adjuvant nanoparticles with mannose modification for effective anticancer vaccination. ACS Nano. 2018;12(6):5121–9.PubMedCrossRef Yang R, et al. Cancer Cell membrane-coated adjuvant nanoparticles with mannose modification for effective anticancer vaccination. ACS Nano. 2018;12(6):5121–9.PubMedCrossRef
570.
go back to reference Kim H, et al. Polymeric nanoparticles encapsulating novel TLR7/8 agonists as immunostimulatory adjuvants for enhanced cancer immunotherapy. Biomaterials. 2018;164:38–53.PubMedCrossRef Kim H, et al. Polymeric nanoparticles encapsulating novel TLR7/8 agonists as immunostimulatory adjuvants for enhanced cancer immunotherapy. Biomaterials. 2018;164:38–53.PubMedCrossRef
571.
572.
go back to reference Klinguer-Hamour C, et al. DDA adjuvant induces a mixed Th1/Th2 immune response when associated with BBG2Na, a respiratory syncytial virus potential vaccine. Vaccine. 2002;20(21–22):2743–51.PubMedCrossRef Klinguer-Hamour C, et al. DDA adjuvant induces a mixed Th1/Th2 immune response when associated with BBG2Na, a respiratory syncytial virus potential vaccine. Vaccine. 2002;20(21–22):2743–51.PubMedCrossRef
573.
go back to reference Christensen D, et al. Cationic liposomes as vaccine adjuvants. Expert Rev Vaccines. 2011;10(4):513–21.PubMedCrossRef Christensen D, et al. Cationic liposomes as vaccine adjuvants. Expert Rev Vaccines. 2011;10(4):513–21.PubMedCrossRef
574.
go back to reference Korsholm KS, et al. The adjuvant mechanism of cationic dimethyldioctadecylammonium liposomes. Immunology. 2007;121(2):216–26.PubMedCrossRef Korsholm KS, et al. The adjuvant mechanism of cationic dimethyldioctadecylammonium liposomes. Immunology. 2007;121(2):216–26.PubMedCrossRef
575.
go back to reference Zamani P, et al. Nanoliposomes as the adjuvant delivery systems in cancer immunotherapy. J Cell Physiol. 2018;233(7):5189–99.PubMedCrossRef Zamani P, et al. Nanoliposomes as the adjuvant delivery systems in cancer immunotherapy. J Cell Physiol. 2018;233(7):5189–99.PubMedCrossRef
576.
go back to reference Yuba E, et al. Dextran derivative-based pH-sensitive liposomes for cancer immunotherapy. Biomaterials. 2014;35(9):3091–101.PubMedCrossRef Yuba E, et al. Dextran derivative-based pH-sensitive liposomes for cancer immunotherapy. Biomaterials. 2014;35(9):3091–101.PubMedCrossRef
577.
go back to reference Zeng Q, et al. Tailoring polymeric hybrid micelles with lymph node targeting ability to improve the potency of cancer vaccines. Biomaterials. 2017;122:105–13.PubMedCrossRef Zeng Q, et al. Tailoring polymeric hybrid micelles with lymph node targeting ability to improve the potency of cancer vaccines. Biomaterials. 2017;122:105–13.PubMedCrossRef
578.
go back to reference de Faria PC, et al. Oxidized multiwalled carbon nanotubes as antigen delivery system to promote superior CD8(+) T cell response and protection against cancer. Nano Lett. 2014;14(9):5458–70.PubMedCrossRef de Faria PC, et al. Oxidized multiwalled carbon nanotubes as antigen delivery system to promote superior CD8(+) T cell response and protection against cancer. Nano Lett. 2014;14(9):5458–70.PubMedCrossRef
579.
go back to reference Hassan HA, et al. Dual stimulation of antigen presenting cells using carbon nanotube-based vaccine delivery system for cancer immunotherapy. Biomaterials. 2016;104:310–22.PubMedPubMedCentralCrossRef Hassan HA, et al. Dual stimulation of antigen presenting cells using carbon nanotube-based vaccine delivery system for cancer immunotherapy. Biomaterials. 2016;104:310–22.PubMedPubMedCentralCrossRef
580.
go back to reference Zhou Q, et al. Different-sized gold nanoparticle Activator/Antigen increases dendritic cells Accumulation in Liver-Draining Lymph Nodes and CD8 + T cell responses. ACS Nano. 2016;10(2):2678–92.PubMedCrossRef Zhou Q, et al. Different-sized gold nanoparticle Activator/Antigen increases dendritic cells Accumulation in Liver-Draining Lymph Nodes and CD8 + T cell responses. ACS Nano. 2016;10(2):2678–92.PubMedCrossRef
581.
go back to reference Fallarini S, et al. Factors affecting T cell responses induced by fully synthetic glyco-gold-nanoparticles. Nanoscale. 2013;5(1):390–400.PubMedCrossRef Fallarini S, et al. Factors affecting T cell responses induced by fully synthetic glyco-gold-nanoparticles. Nanoscale. 2013;5(1):390–400.PubMedCrossRef
582.
go back to reference Dykman LA, et al. Gold nanoparticles as an adjuvant: influence of size, shape, and technique of combination with CpG on antibody production. Int Immunopharmacol. 2018;54:163–8.PubMedCrossRef Dykman LA, et al. Gold nanoparticles as an adjuvant: influence of size, shape, and technique of combination with CpG on antibody production. Int Immunopharmacol. 2018;54:163–8.PubMedCrossRef
583.
584.
585.
go back to reference Luo J, et al. Self-assembled CpG oligodeoxynucleotides conjugated hollow gold nanospheres to enhance cancer-associated immunostimulation. Colloids Surf B Biointerfaces. 2019;175:248–55.PubMedCrossRef Luo J, et al. Self-assembled CpG oligodeoxynucleotides conjugated hollow gold nanospheres to enhance cancer-associated immunostimulation. Colloids Surf B Biointerfaces. 2019;175:248–55.PubMedCrossRef
586.
go back to reference Ow H, et al. Bright and stable core-shell fluorescent silica nanoparticles. Nano Lett. 2005;5(1):113–7.PubMedCrossRef Ow H, et al. Bright and stable core-shell fluorescent silica nanoparticles. Nano Lett. 2005;5(1):113–7.PubMedCrossRef
587.
go back to reference Benezra M, et al. Multimodal silica nanoparticles are effective cancer-targeted probes in a model of human melanoma. J Clin Invest. 2011;121(7):2768–80.PubMedPubMedCentralCrossRef Benezra M, et al. Multimodal silica nanoparticles are effective cancer-targeted probes in a model of human melanoma. J Clin Invest. 2011;121(7):2768–80.PubMedPubMedCentralCrossRef
588.
589.
go back to reference Mahony D, et al. Mesoporous silica nanoparticles act as a self-adjuvant for ovalbumin model antigen in mice. Small. 2013;9(18):3138–46.PubMedCrossRef Mahony D, et al. Mesoporous silica nanoparticles act as a self-adjuvant for ovalbumin model antigen in mice. Small. 2013;9(18):3138–46.PubMedCrossRef
590.
go back to reference Selvaraja VK, Gudipudi DK. Fundamentals to clinical application of nanoparticles in cancer immunotherapy and radiotherapy. Ecancermedicalscience. 2020;14:1095.PubMedPubMedCentral Selvaraja VK, Gudipudi DK. Fundamentals to clinical application of nanoparticles in cancer immunotherapy and radiotherapy. Ecancermedicalscience. 2020;14:1095.PubMedPubMedCentral
591.
go back to reference Saeed M, et al. Engineering nanoparticles to reprogram the Tumor Immune Microenvironment for Improved Cancer Immunotherapy. Theranostics. 2019;9(26):7981–8000.PubMedPubMedCentralCrossRef Saeed M, et al. Engineering nanoparticles to reprogram the Tumor Immune Microenvironment for Improved Cancer Immunotherapy. Theranostics. 2019;9(26):7981–8000.PubMedPubMedCentralCrossRef
592.
go back to reference Libutti SK, et al. Phase I and pharmacokinetic studies of CYT-6091, a novel PEGylated colloidal gold-rhTNF nanomedicine. Clin Cancer Res. 2010;16(24):6139–49.PubMedPubMedCentralCrossRef Libutti SK, et al. Phase I and pharmacokinetic studies of CYT-6091, a novel PEGylated colloidal gold-rhTNF nanomedicine. Clin Cancer Res. 2010;16(24):6139–49.PubMedPubMedCentralCrossRef
593.
go back to reference Wang S, Sun Z, Hou Y. Engineering nanoparticles toward the modulation of emerging cancer immunotherapy. Adv Healthc Mater. 2021;10(5):2000845.CrossRef Wang S, Sun Z, Hou Y. Engineering nanoparticles toward the modulation of emerging cancer immunotherapy. Adv Healthc Mater. 2021;10(5):2000845.CrossRef
595.
go back to reference Prasad S, et al. Polymer nanoparticles containing tumor lysates as antigen delivery vehicles for dendritic cell–based antitumor immunotherapy. Nanomed Nanotechnol Biol Med. 2011;7(1):1–10.CrossRef Prasad S, et al. Polymer nanoparticles containing tumor lysates as antigen delivery vehicles for dendritic cell–based antitumor immunotherapy. Nanomed Nanotechnol Biol Med. 2011;7(1):1–10.CrossRef
596.
go back to reference Higashisaka K, et al. Nano-safety research: examining the associations among the biological effects of nanoparticles and their physicochemical properties and kinetics. Biol Pharm Bull. 2017;40(3):243–8.PubMedCrossRef Higashisaka K, et al. Nano-safety research: examining the associations among the biological effects of nanoparticles and their physicochemical properties and kinetics. Biol Pharm Bull. 2017;40(3):243–8.PubMedCrossRef
597.
go back to reference Niu L, et al. Cytokine-mediated disruption of lymphocyte trafficking, hemopoiesis, and induction of lymphopenia, anemia, and thrombocytopenia in anti-CD137-treated mice. J Immunol. 2007;178(7):4194–213.PubMedCrossRef Niu L, et al. Cytokine-mediated disruption of lymphocyte trafficking, hemopoiesis, and induction of lymphopenia, anemia, and thrombocytopenia in anti-CD137-treated mice. J Immunol. 2007;178(7):4194–213.PubMedCrossRef
598.
go back to reference Leonard JP, et al. Effects of single-dose interleukin-12 exposure on interleukin-12-associated toxicity and interferon-gamma production. Blood. 1997;90(7):2541–8.PubMed Leonard JP, et al. Effects of single-dose interleukin-12 exposure on interleukin-12-associated toxicity and interferon-gamma production. Blood. 1997;90(7):2541–8.PubMed
599.
go back to reference Di Giacomo AM, Biagioli M, Maio M. The emerging toxicity profiles of anti-CTLA-4 antibodies across clinical indications. Semin Oncol. 2010;37(5):499–507.PubMedCrossRef Di Giacomo AM, Biagioli M, Maio M. The emerging toxicity profiles of anti-CTLA-4 antibodies across clinical indications. Semin Oncol. 2010;37(5):499–507.PubMedCrossRef
600.
go back to reference Xia Q et al. Size-and cell type-dependent cellular uptake, cytotoxicity and in vivo distribution of gold nanoparticles. Int J Nanomed, 2019: p. 6957–70. Xia Q et al. Size-and cell type-dependent cellular uptake, cytotoxicity and in vivo distribution of gold nanoparticles. Int J Nanomed, 2019: p. 6957–70.
601.
go back to reference Bekić M, et al. The effect of stabilisation agents on the immunomodulatory properties of gold nanoparticles obtained by ultrasonic spray pyrolysis. Materials. 2019;12(24):4121.PubMedPubMedCentralCrossRef Bekić M, et al. The effect of stabilisation agents on the immunomodulatory properties of gold nanoparticles obtained by ultrasonic spray pyrolysis. Materials. 2019;12(24):4121.PubMedPubMedCentralCrossRef
602.
go back to reference Zhong Y, et al. In vivo molecular imaging for immunotherapy using ultra-bright near-infrared-IIb rare-earth nanoparticles. Nat Biotechnol. 2019;37(11):1322–31.PubMedPubMedCentralCrossRef Zhong Y, et al. In vivo molecular imaging for immunotherapy using ultra-bright near-infrared-IIb rare-earth nanoparticles. Nat Biotechnol. 2019;37(11):1322–31.PubMedPubMedCentralCrossRef
605.
go back to reference Kwong B, et al. Localized immunotherapy via liposome-anchored Anti-CD137 + IL-2 prevents lethal toxicity and elicits local and systemic antitumor immunity. Cancer Res. 2013;73(5):1547–58.PubMedPubMedCentralCrossRef Kwong B, et al. Localized immunotherapy via liposome-anchored Anti-CD137 + IL-2 prevents lethal toxicity and elicits local and systemic antitumor immunity. Cancer Res. 2013;73(5):1547–58.PubMedPubMedCentralCrossRef
606.
go back to reference Kwong B, Liu H, Irvine DJ. Induction of potent anti-tumor responses while eliminating systemic side effects via liposome-anchored combinatorial immunotherapy. Biomaterials. 2011;32(22):5134–47.PubMedPubMedCentralCrossRef Kwong B, Liu H, Irvine DJ. Induction of potent anti-tumor responses while eliminating systemic side effects via liposome-anchored combinatorial immunotherapy. Biomaterials. 2011;32(22):5134–47.PubMedPubMedCentralCrossRef
608.
go back to reference Wu TY-H et al. Rational design of small molecules as vaccine adjuvants. Sci Transl Med, 2014. 6(263): p. 263ra160-263ra160. Wu TY-H et al. Rational design of small molecules as vaccine adjuvants. Sci Transl Med, 2014. 6(263): p. 263ra160-263ra160.
609.
610.
go back to reference Manolova V, et al. Nanoparticles target distinct dendritic cell populations according to their size. Eur J Immunol. 2008;38(5):1404–13.PubMedCrossRef Manolova V, et al. Nanoparticles target distinct dendritic cell populations according to their size. Eur J Immunol. 2008;38(5):1404–13.PubMedCrossRef
611.
go back to reference Reddy ST, et al. Exploiting lymphatic transport and complement activation in nanoparticle vaccines. Nat Biotechnol. 2007;25(10):1159–64.PubMedCrossRef Reddy ST, et al. Exploiting lymphatic transport and complement activation in nanoparticle vaccines. Nat Biotechnol. 2007;25(10):1159–64.PubMedCrossRef
612.
go back to reference Tkach AV, et al. Direct effects of carbon nanotubes on dendritic cells induce immune suppression upon pulmonary exposure. ACS Nano. 2011;5(7):5755–62.PubMedPubMedCentralCrossRef Tkach AV, et al. Direct effects of carbon nanotubes on dendritic cells induce immune suppression upon pulmonary exposure. ACS Nano. 2011;5(7):5755–62.PubMedPubMedCentralCrossRef
613.
go back to reference Ryan JJ, et al. Fullerene nanomaterials inhibit the allergic response. J Immunol. 2007;179(1):665–72.PubMedCrossRef Ryan JJ, et al. Fullerene nanomaterials inhibit the allergic response. J Immunol. 2007;179(1):665–72.PubMedCrossRef
Metadata
Title
Nanoparticles in tumor microenvironment remodeling and cancer immunotherapy
Authors
Qiang Lu
Dongquan Kou
Shenghan Lou
Milad Ashrafizadeh
Amir Reza Aref
Israel Canadas
Yu Tian
Xiaojia Niu
Yuzhuo Wang
Pedram Torabian
Lingzhi Wang
Gautam Sethi
Vinay Tergaonkar
Franklin Tay
Zhennan Yuan
Peng Han
Publication date
01-12-2024
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2024
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-024-01535-8

Other articles of this Issue 1/2024

Journal of Hematology & Oncology 1/2024 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine