The combination of traditional Chinese medicine (TCM) nanoparticles with modern immunotherapy presents a promising approach to enhancing the treatment of gastrointestinal tumors. TCM nanoparticles can significantly augment the efficacy of conventional immunotherapies by enhancing immune cell activation, modulating the tumor microenvironment, and overcoming immune evasion mechanisms. This review explores the integration of TCM nanoparticles with existing immunotherapy strategies such as immune checkpoint inhibitors, cancer vaccines, and cytokine therapies. It also summarizes the progress in preclinical studies and clinical trials, analyzing their efficacy and safety profiles. Current research indicates that this combinatory approach has shown substantial improvements in anti-tumor immune responses in animal models, with promising early-stage clinical trial results demonstrating favorable safety and potential therapeutic effects. However, further large-scale clinical trials are necessary to confirm its clinical efficacy and safety. Future studies should focus on optimizing treatment regimens and identifying suitable patient populations to advance the clinical application of TCM nanoparticle-based immunotherapy in gastrointestinal cancers.
T.R. de Back, S.R. van Hooff, D.W. Sommeijer, et al. Transcriptomic subtyping of gastrointestinal malignancies. Trends in Cancer, 2024, 10(9): 842−856. https://doi.org/10.1016/j.trecan.2024.06.007
P. Sharma, C. Hassan. Artificial intelligence and deep learning for upper gastrointestinal neoplasia. Gastroenterology, 2022, 162(4): 1056−1066. https://doi.org/10.1053/j.gastro.2021.11.040
R. Pittayanon, W. Khongka, S. Linlawan, et al. Hemostatic powder vs standard endoscopic treatment for gastrointestinal tumor bleeding: A multicenter randomized trial. Gastroenterology, 2023, 165(3): 762−772.e2. https://doi.org/10.1053/j.gastro.2023.05.042
X. Chong, Y. Madeti, J. Cai, et al. Recent developments in immunotherapy for gastrointestinal tract cancers. Journal of Hematology & Oncology, 2024, 17(1): 65. https://doi.org/10.1186/s13045-024-01578-x
Q. Gan, Y. Li, Y. Li, et al. Pathways and molecules for overcoming immunotolerance in metastatic gastrointestinal tumors. Frontiers in Immunology, 2024, 15: 1359914. https://doi.org/10.3389/fimmu.2024.1359914
K. Miao, W. Liu, J. Xu, et al. Harnessing the power of traditional Chinese medicine monomers and compound prescriptions to boost cancer immunotherapy. Frontiers in Immunology, 2023, 14: 1277243. https://doi.org/10.3389/fimmu.2023.1277243
G. Chen, N. Wang, R. Yang, et al. Efficacy and safety of herbal medicines intervention for cachexia associated with cancer: A systematic review and meta-analysis. Phytotherapy Research, 2023, 37(11): 5243−5278. https://doi.org/10.1002/ptr.7956
Y. Yan, J. Liu, Y. Pang, et al. Efficacy of Traditional Chinese Medicine Combined Online Group Psychotherapy (TCM-eRhab) on improving quality of life and relieving psychological burden for colorectal cancer survivors: A study protocol for a phase-II randomized controlled trial. BMC Complementary Medicine and Therapies, 2024, 24(1): 290. https://doi.org/10.1186/s12906-024-04533-y
X. Feng, Z. Li, W. Guo, et al. The effects of traditional Chinese medicine and dietary compounds on digestive cancer immunotherapy and gut microbiota modulation: A review. Frontiers in Immunology, 2023, 14: 1087755. https://doi.org/10.3389/fimmu.2023.1087755
Y.H. Jin, Y.P. Wang, Y.L. Xie, et al. Research on the development methodology for clinical practice guidelines for organic integration of traditional Chinese and Western medicine. Military Medical Research, 2023, 10(1): 45. https://doi.org/10.1186/s40779-023-00481-9
R. Sun, J. Dai, M. Ling, et al. Delivery of triptolide: A combination of traditional Chinese medicine and nanomedicine. Journal of Nanobiotechnology, 2022, 20(1): 194. https://doi.org/10.1186/s12951-022-01389-7
L. Gervaso, D. Ciardiello, R.A. Oliveira, et al. Immunotherapy in the neoadjuvant treatment of gastrointestinal tumors: Is the time ripe. Journal for ImmunoTherapy of Cancer, 2024, 12(5): e008027. https://doi.org/10.1136/jitc-2023-008027
X. Zhu, J. Xue, H. Jiang, et al. CAR-NK cells for gastrointestinal cancer immunotherapy: From bench to bedside. Molecular Cancer, 2024, 23(1): 237. https://doi.org/10.1186/s12943-024-02151-3
S. Ai, Y. Li, H. Zheng, et al. Collision of herbal medicine and nanotechnology: A bibliometric analysis of herbal nanoparticles from 2004 to 2023. Journal of Nanobiotechnology, 2024, 22(1): 140. https://doi.org/10.1186/s12951-024-02426-3
M. Zheng, K. Liu, L. Li, et al. Traditional Chinese medicine inspired dual-drugs loaded inhalable nano-therapeutics alleviated idiopathic pulmonary fibrosis by targeting early inflammation and late fibrosis. J Nanobiotechnology, 2024, 22(1): 14. https://doi.org/10.1186/s12951-023-02251-0
D. Wei, H. Yang, Y. Zhang, et al. Nano-traditional Chinese medicine: A promising strategy and its recent advances. Journal of Materials Chemistry B, 2022, 10(16): 2973−2994. https://doi.org/10.1039/D2TB00225F
Q. Xiang, J. Wang, K. Tao, et al. Optimization of phenolic-enriched extracts from olive leaves via ball milling-assisted extraction using response surface methodology. Molecules, 2024, 29(15): 3658. https://doi.org/10.3390/molecules29153658
J. Wang, Y. Zhao, B. Zhai, et al. Phloretin transfersomes for transdermal delivery: Design, optimization, and in vivo evaluation. Molecules, 2023, 28(19): 6790. https://doi.org/10.3390/molecules28196790
L. Ke, X. Duan, J. Cui, et al. Research progress on the extraction technology and activity study of Epimedium polysaccharides. Carbohydr Polym, 2023, 306: 120602. https://doi.org/10.1016/j.carbpol.2023.120602
G. You, T. Feng, G. Zhang, et al. Preparation, optimization, characterization and in vitro release of baicalein-solubilizing glycyrrhizic acid nano-micelles. International Journal of Pharmaceutics, 2021, 601: 120546. https://doi.org/10.1016/j.ijpharm.2021.120546
R. Al-Shdefat, M. Hailat, O.Y. Alshogran, et al. Ribociclib hybrid lipid-polymer nanoparticle preparation and characterization for cancer treatment. Polymers, 2023, 15(13): 2844. https://doi.org/10.3390/polym15132844
M. Khang, J.H. Lee, T. Lee, et al. Intrathecal delivery of nanoparticle PARP inhibitor to the cerebrospinal fluid for the treatment of metastatic medulloblastoma. Science Translational Medicine, 2023, 15(720): eadi1617. https://doi.org/10.1126/scitranslmed.adi1617
A.L. Luss, D.V. Bagrov, A.V. Yagolovich, et al. Toxicity evaluation and controlled-release of curcumin-loaded amphiphilic poly-N-vinylpyrrolidone nanoparticles: in vitro and in vivo models. Pharmaceutics, 2023, 16(1): 8. https://doi.org/10.3390/pharmaceutics16010008
C.R. Liu, B.X. Xu, D.J. McClements, et al. Properties of curcumin-loaded zein-tea saponin nanoparticles prepared by antisolvent co-precipitation and precipitation. Food Chemistry, 2022, 391: 133224. https://doi.org/10.1016/j.foodchem.2022.133224
Z.H. Zhang, X.J. Li, S.Y. Sang, et al. Preparation, properties and interaction of curcumin loaded zein/HP-β-CD nanoparticles based on electrostatic interactions by antisolvent co-precipitation. Food Chemistry, 2023, 403: 134344. https://doi.org/10.1016/j.foodchem.2022.134344
J. Jing, K.R. Tupally, G.R. Kokil, et al. Development of a hybrid peptide dendrimer micellar carrier system and its application in the reformulation of a hydrophobic therapeutic agent derived from traditional Chinese medicine. RSC Adv, 2019, 9(5): 2458−2463. https://doi.org/10.1039/C8RA09606F
W.R. Lee, T.H. Huang, S. Hu, et al. Laser-assisted nanoparticle delivery to promote skin absorption and penetration depth of retinoic acid with the aim for treating photoaging. International Journal of Pharmaceutics, 2022, 627: 122162. https://doi.org/10.1016/j.ijpharm.2022.122162
D.V. Bhalani, B. Nutan, A. Kumar, et al. Bioavailability enhancement techniques for poorly aqueous soluble drugs and therapeutics. Biomedicines, 2022, 10(9): 2055. https://doi.org/10.3390/biomedicines10092055
I.D. Boateng. Polyprenols in Ginkgo biloba; a review of their chemistry (synthesis of polyprenols and their derivatives), extraction, purification, and bioactivities. Food Chemistry, 2023, 418: 136006. https://doi.org/10.1016/j.foodchem.2023.136006
A. Gul, Fozia, A. Shaheen, et al. Green synthesis, characterization, enzyme inhibition, antimicrobial potential, and cytotoxic activity of plant mediated silver nanoparticle using Ricinus communis leaf and root extracts. Biomolecules, 2021, 11(2): 206. https://doi.org/10.3390/biom11020206
A. Balčiūnaitienė, P. Štreimikytė, V. Puzerytė, et al. Antimicrobial activities against opportunistic pathogenic bacteria using green synthesized silver nanoparticles in plant and lichen enzyme-assisted extracts. Plants, 2022, 11(14): 1833. https://doi.org/10.3390/plants11141833
V. Sakaray, Y.S. Rao, N.V. Naidu. Green synthesis of silver nanoparticles by Acalypha indica plant extract and their approach towards multifunctional applications. Nano Biomedicine and Engineering, 2024, 16(4): 665−676. https://doi.org/10.26599/NBE.2024.9290064
W.J. Feng, Z.Q. Wang, T. Zhang, et al. Biomimetic synthesis of maltodextrin-derived dendritic nanoparticle and its structural characterizations. Carbohydrate Polymers, 2023, 312: 120816. https://doi.org/10.1016/j.carbpol.2023.120816
X.Y. Xiao, Y. Zhang, K.D.Sun, et al. Enzymatic and ultrasound assisted β-cyclodextrin extraction of active ingredients from Forsythia suspensa and their antioxidant and anti-inflammatory activities. Ultrasonics Sonochemistry, 2024, 108: 106944. https://doi.org/10.1016/j.ultsonch.2024.106944
J. Jeevanandam, S.F. Kiew, S. Boakye-Ansah, et al. Green approaches for the synthesis of metal and metal oxide nanoparticles using microbial and plant extracts. Nanoscale, 2022, 14(7): 2534−2571. https://doi.org/10.1039/D1NR08144F
H.G. Lazcano-Ramírez, J.J.O. Garza-García, J.A. Hernández-Díaz, et al. Antifungal activity of selenium nanoparticles obtained by plant-mediated synthesis. Antibiotics, 2023, 12(1): 115. https://doi.org/10.3390/antibiotics12010115
M.A. Khan, D. Singh, A. Arif, et al. Protective effect of green synthesized Selenium Nanoparticles against Doxorubicin induced multiple adverse effects in Swiss albino mice. Life Sciences, 2022, 305: 120792. https://doi.org/10.1016/j.lfs.2022.120792
Y. Liu, J. Zhang, Y. Tu, et al. Potential-independent intracellular drug delivery and mitochondrial targeting. ACS Nano, 2022, 16(1): 1409−1420. https://doi.org/10.1021/acsnano.1c09456
M.M. Agwa, H. Elmotasem, R.I. Moustafa, et al. Advent in proteins, nucleic acids, and biological cell membranes functionalized nanocarriers to accomplish active or homologous tumor targeting for smart amalgamated chemotherapy/photo-therapy: A review. International Journal of Biological Macromolecules, 2023, 253: 127460. https://doi.org/10.1016/j.ijbiomac.2023.127460
A. Bandyopadhyay, T. Das, S. Nandy, et al. Ligand-based active targeting strategies for cancer theranostics. Naunyn-Schmiedeberg’s Archives of Pharmacology, 2023, 396(12): 3417−3441. https://doi.org/10.1007/s00210-023-02612-4
A.S. Postovalova, Y.A. Tishchenko, M.S. Istomina, et al. Comparison of passive targeted delivery of inorganic and organic nanocarriers among different types of tumors. Nanomedicine: Nanotechnology, Biology and Medicine, 2024, 59: 102753. https://doi.org/10.1016/j.nano.2024.102753
N. AlSawaftah, W.G. Pitt, G.A. Husseini. Dual-targeting and stimuli-triggered liposomal drug delivery in cancer treatment. ACS Pharmacology & Translational Science, 2021, 4(3): 1028−1049. https://doi.org/10.1021/acsptsci.1c00066
M.H. Mohd-Zahid, S.N. Zulkifli, C.A. Che Abdullah, et al. Gold nanoparticles conjugated with anti-CD133 monoclonal antibody and 5-fluorouracil chemotherapeutic agent as nanocarriers for cancer cell targeting. RSC Advance, 2021, 11(26): 16131−16141. https://doi.org/10.1039/D1RA01093J
S. Wang, Y. Liu, Q. Sun, et al. Triple cross-linked dynamic responsive hydrogel loaded with selenium nanoparticles for modulating the inflammatory microenvironment via PI3K/Akt/NF-κB and MAPK signaling pathways. Advanced Science, 2023, 10(31): e2303167. https://doi.org/10.1002/advs.202303167
P. Dosta, M.Z. Dion, M. Prado, et al. Matrix metalloproteinase- and pH-sensitive nanoparticle system enhances drug retention and penetration in glioblastoma. ACS Nano, 2024, 18(22): 14145−14160. https://doi.org/10.1021/acsnano.3c03409
Y.S. Choi, H. Cho, W.G. Choi, et al. Beyond hydrophilic polymers in amphiphilic polymer-based self-assembled NanoCarriers: Small hydrophilic carboxylate-capped disulfide drug delivery system and its multifunctionality and multispatial targetability. Biomaterials, 2022, 280: 121307. https://doi.org/10.1016/j.biomaterials.2021.121307
M.F. Wu, Q. Wang, Y.Y. Peng, et al. Enhancing targeted therapy in hepatocellular carcinoma through a pH-responsive delivery system: Folic acid-modified polydopamine-paclitaxel-loaded poly(3-hydroxybutyrate- co-3-hydroxyvalerate) nanoparticles. Molecular Pharmaceutics, 2024, 21(2): 581−595. https://doi.org/10.1021/acs.molpharmaceut.3c00710
Q. Zhang, G. Kuang, H. Wang, et al. Multi-bioinspired MOF delivery systems from microfluidics for tumor multimodal therapy. Advanced Science, 2023, 10(33): e2303818. https://doi.org/10.1002/advs.202303818
S.J. Wang, S.K. Dougan, M. Dougan. Immune mechanisms of toxicity from checkpoint inhibitors. Trends in Cancer, 2023, 9(7): 543−553. https://doi.org/10.1016/j.trecan.2023.04.002
M. Oladejo, W. Paulishak, L. Wood. Synergistic potential of immune checkpoint inhibitors and therapeutic cancer vaccines. Seminars in Cancer Biology, 2023, 88: 81−95. https://doi.org/10.1016/j.semcancer.2022.12.003
D.T. Le, T.W. Kim, E. Van Cutsem, et al. Phase II open-label study of pembrolizumab in treatment-refractory, microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: KEYNOTE-164. Journal of Clinical Oncology, 2020, 38(1): 11−19. https://doi.org/10.1200/JCO.19.02107
H.J. Lenz, E. Van Cutsem, M. Luisa Limon, et al. First-line nivolumab plus low-dose ipilimumab for microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer: The phase II CheckMate 142 study. Journal of Clinical Oncology, 2022, 40(2): 161−170. https://doi.org/10.1200/JCO.21.01015
M.D. Hellmann, L. Paz-Ares, R.B. Caro, et al. Nivolumab plus ipilimumab in advanced non-small-cell lung cancer. The New England Journal of Medicine, 2019, 381(21): 2020−2031. https://doi.org/10.1056/NEJMoa1910231
M. Lei, N.O. Siemers, D. Pandya, et al. Analyses of PD-L1 and inflammatory gene expression association with efficacy of nivolumab ± ipilimumab in gastric cancer/gastroesophageal junction cancer. Clinical Cancer Research, 2021, 27(14): 3926−3935. https://doi.org/10.1158/1078-0432.CCR-20-2790
S.Y. Rha, D.Y. Oh, P. Yañez, et al. Pembrolizumab plus chemotherapy versus placebo plus chemotherapy for HER2-negative advanced gastric cancer (KEYNOTE-859): A multicentre, randomised, double-blind, phase 3 trial. The Lancet Oncology, 2023, 24(11): 1181−1195. https://doi.org/10.1016/S1470-2045(23)00515-6
M. Tampaki, E. Ionas, E. Hadziyannis, et al. Association of TIM-3 with BCLC stage, serum PD-L1 detection, and response to transarterial chemoembolization in patients with hepatocellular carcinoma. Cancers, 2020, 12(1): E212. https://doi.org/10.3390/cancers12010212
M. de Miguel, E. Calvo. Clinical challenges of immune checkpoint inhibitors. Cancer Cell, 2020, 38(3): 326−333. https://doi.org/10.1016/j.ccell.2020.07.004
P. Sharma, S. Goswami, D. Raychaudhuri, et al. Immune checkpoint therapy-current perspectives and future directions. Cell, 2023, 186(8): 1652−1669. https://doi.org/10.1016/j.cell.2023.03.006
S. Mir, A. Venugopalan, J.L. Zhang, et al. Persistence of activated anti-mesothelin hYP218 chimeric antigen receptor T cells in the tumour is associated with efficacy in gastric and colorectal carcinomas. Clinical and Translational Medicine, 2024, 14(11): e70057. https://doi.org/10.1002/ctm2.70057
N. Puebla-Osorio N.W., Fowlkes, H.B. Barsoumian, et al. Enhanced tumor control and survival in preclinical models with adoptive cell therapy preceded by low-dose radiotherapy. Frontiers in Oncology, 2024, 14: 1407143. https://doi.org/10.3389/fonc.2024.1407143
W. Lam, R. Hu, S.H. Liu, et al. YIV-906 enhances nuclear factor of activated T-cells (NFAT) activity of T cells and promotes immune checkpoint blockade antibody action and CAR T-cell activity. Frontiers in Pharmacology, 2022, 13: 1095186. https://doi.org/10.3389/fphar.2022.1095186
A. Guerreiro, I. Compañón, F.S. Lazaris, et al. Non-natural MUC1 glycopeptide homogeneous cancer vaccine with enhanced immunogenicity and therapeutic activity. Angewandte Chemie, 2024, 63(49): e202411009. https://doi.org/10.1002/anie.202411009
K.P. Fabian, M.R. Padget, R. Fujii, et al. Differential combination immunotherapy requirements for inflamed (warm) tumors versus T cell excluded (cool) tumors: Engage, expand, enable, and evolve. Journal for Immunotherapy of Cancer, 2021, 9(2): e001691. https://doi.org/10.1136/jitc-2020-001691
L. Huang, Y. Rong, X. Tang, et al. Engineered exosomes as an in situ DC-primed vaccine to boost antitumor immunity in breast cancer. Molecular Cancer, 2022, 21(1): 45. https://doi.org/10.1186/s12943-022-01515-x
Y. Liu, J. Pagacz, D.J. Wolfgeher, et al. Senescent cancer cell vaccines induce cytotoxic T cell responses targeting primary tumors and disseminated tumor cells. Journal for Immunotherapy of Cancer, 2023, 11(2): e005862. https://doi.org/10.1136/jitc-2022-005862
G. Deng, L. Zhou, B. Wang, et al. Targeting cathepsin B by cycloastragenol enhances antitumor immunity of CD8 T cells via inhibiting MHC-I degradation. J Immunother Cancer, 2022, 10(10): e004874. https://doi.org/10.1136/jitc-2022-004874
H. Liu, Z.Y. Wang, Y.C. Zhou, et al. Immunomodulation of Chinese Herbal Medicines on NK cell populations for cancer therapy: A systematic review. Journal of Ethnopharmacology, 2021, 268: 113561. https://doi.org/10.1016/j.jep.2020.113561
Q. Zeng, Y. Zhang, W. Zhang, et al. Baicalein suppresses the proliferation and invasiveness of colorectal cancer cells by inhibiting Snail-induced epithelial-mesenchymal transition. Molecular Medicine Reports, 2020, 21(6): 2544−2552. https://doi.org/10.3892/mmr.2020.11051
S. Xiaodan, C. Ying. Role of ginsenoside Rh2 in tumor therapy and tumor microenvironment immunomodulation. Biomed Pharmacother, 2022, 156: 113912. https://doi.org/10.1016/j.biopha.2022.113912
J. Qian, Y. Jiang, H. Hu. Ginsenosides: An immunomodulator for the treatment of colorectal cancer. Frontiers in Pharmacology, 2024, 15: 1408993. https://doi.org/10.3389/fphar.2024.1408993
X.H. Wu, J.L. Xia, Z.Q. Wang, et al. Feiyanning downregulating CXCLs/CXCR2 axis to suppress TANs infiltration in the prevention of lung cancer metastasis. Journal of Ethnopharmacology, 2022, 295: 115277. https://doi.org/10.1016/j.jep.2022.115277
H. Liu, R. Deng, C.W. Zhu, et al. Rosmarinic acid in combination with ginsenoside Rg1 suppresses colon cancer metastasis via co-inhition of COX-2 and PD1/PD-L1 signaling axis. Acta Pharmacologica Sinica, 2024, 45: 193−208. https://doi.org/10.1038/s41401-023-01158-8
Y.F. Jiang, Y.E. Hu, Y. Yang, et al. Tong-Xie-Yao-Fang promotes dendritic cells maturation and retards tumor growth in colorectal cancer mice with chronic restraint stress. Journal of Ethnopharmacology, 2024, 319: 117069. https://doi.org/10.1016/j.jep.2023.117069
J. Li, S.P. Fan, H.X. Li, et al. Evaluation of efficacy, safety and underlying mechanism on Traditional Chinese medicine as synergistic agents for cancer immunotherapy: A preclinical systematic review and meta-analysis. Journal of Ethnopharmacology, 2025, 338: 119035. https://doi.org/10.1016/j.jep.2024.119035
M.T. Wang, F. Yang, J.W. Kong, et al. Traditional Chinese medicine enhances the effectiveness of immune checkpoint inhibitors in tumor treatment: A mechanism discussion. Journal of Ethnopharmacology, 2025, 338: 118955. https://doi.org/10.1016/j.jep.2024.118955
M. De Martino, J.C. Rathmell, L. Galluzzi, et al. Cancer cell metabolism and antitumour immunity. Nature Reviews Immunology, 2024, 24: 654−669. https://doi.org/10.1038/s41577-024-01026-4
L. Maiorino, J. Daßler-Plenker, L. Sun, et al. Innate immunity and cancer pathophysiology. Annual Review of Pathology: Mechanisms of Disease, 2022, 17: 425−457. https://doi.org/10.1146/annurev-pathmechdis-032221-115501
A. Del Prete, V. Salvi, A. Soriani, et al. Dendritic cell subsets in cancer immunity and tumor antigen sensing. Cellular & Molecular Immunology, 2023, 20: 432−447. https://doi.org/10.1038/s41423-023-00990-6
G. Oliveira, C.J. Wu. Dynamics and specificities of T cells in cancer immunotherapy. Nature Reviews Cancer, 2023, 23(5): 295−316. https://doi.org/10.1038/s41568-023-00560-y
M. Tsoumakidou. The advent of immune stimulating CAFs in cancer. Nature Reviews Cancer, 2023, 23: 258−269. https://doi.org/10.1038/s41568-023-00549-7
D. Galati, S. Zanotta. Dendritic cell and cancer therapy. International Journal of Molecular Sciences, 2023, 24(4): 4253. https://doi.org/10.3390/ijms24044253
X. Wu, H. Yang, X. Chen, et al. Nano-herb medicine and PDT induced synergistic immunotherapy for colon cancer treatment. Biomaterials, 2021, 269: 120654. https://doi.org/10.1016/j.biomaterials.2021.120654
F. Huang, Q. Zhang, J. Xiao, et al. Cancer cell membrane-coated gambogic acid nanoparticles for effective anticancer vaccination by activating dendritic cells. International Journal of Nanomedicine, 2023, 18: 2261−2273. https://doi.org/10.2147/IJN.S408521
Q. Mao, J. Min, R. Zeng, et al. Self-assembled traditional Chinese nanomedicine modulating tumor immunosuppressive microenvironment for colorectal cancer immunotherapy. Theranostics, 2022, 12(14): 6088−6105. https://doi.org/10.7150/thno.72509
Y. Lv, M.Y. Li, L. Weng, et al. Ginseng-derived nanoparticles reprogram macrophages to regulate arginase-1 release for ameliorating T cell exhaustion in tumor microenvironment. Journal of Experimental & Clinical Cancer Research, 2023, 42(1): 322. https://doi.org/10.1186/s13046-023-02888-7
X. Ye, Y. Liu, L. Wei, et al. Monocyte/macrophage-mediated transport of dual-drug ZIF nanoplatforms synergized with programmed cell death protein-1 inhibitor against microsatellite-stable colorectal cancer. Advanced Science, 2024, 11(38): e2405886. https://doi.org/10.1002/advs.202405886
S. Kumagai, S. Koyama, K. Itahashi, et al. Lactic acid promotes PD-1 expression in regulatory T cells in highly glycolytic tumor microenvironments. Cancer Cell, 2022, 40(2): 201−218.e9. https://doi.org/10.1016/j.ccell.2022.01.001
S. Kumagai, S. Koyama, K. Itahashi, et al. Lactic acid supports an immunosuppressive environment and reduces ICB response. Cancer Discovery, 2022, 12(4): OF4. https://doi.org/10.1158/2159-8290.CD-RW2022-020
J. Ye, J.M. Baer, D.V. Faget, et al. Senescent CAFs mediate immunosuppression and drive breast cancer progression. Cancer Discovery, 2024, 14(7): 1302−1323. https://doi.org/10.1158/2159-8290.CD-23-0426
C. Ma, C. Yang, A. Peng, et al. Pan-cancer spatially resolved single-cell analysis reveals the crosstalk between cancer-associated fibroblasts and tumor microenvironment. Molecular Cancer, 2023, 22(1): 170. https://doi.org/10.1186/s12943-023-01876-x
J.X. Feng, M.J. Xu, J.H. Wang, et al. Sequential delivery of nanoformulated α-mangostin and triptolide overcomes permeation obstacles and improves therapeutic effects in pancreatic cancer. Biomaterials, 2020, 241: 119907. https://doi.org/10.1016/j.biomaterials.2020.119907
J. Peng, J. Zhou, R. Sun, et al. Dual-targeting of artesunate and chloroquine to tumor cells and tumor-associated macrophages by a biomimetic PLGA nanoparticle for colorectal cancer treatment. Int J Biol Macromol, 2023, 244: 125163. https://doi.org/10.1016/j.ijbiomac.2023.125163
S. Han, S. Bi, T. Guo, et al. Nano co-delivery of Plumbagin and Dihydrotanshinone I reverses immunosuppressive TME of liver cancer. Journal of Controlled Release, 2022, 348: 250−263. https://doi.org/10.1016/j.jconrel.2022.05.057
Z. Yu, J. Guo, M. Hu, et al. Icaritin exacerbates mitophagy and synergizes with doxorubicin to induce immunogenic cell death in hepatocellular carcinoma. ACS Nano, 2020, 14(4): 4816−4828. https://doi.org/10.1021/acsnano.0c00708
L. Li, Y. Zou, L. Wang, et al. Nanodelivery of scutellarin induces immunogenic cell death for treating hepatocellular carcinoma. International Journal of Pharmaceutics, 2023, 642: 123114. https://doi.org/10.1016/j.ijpharm.2023.123114
Y. Cao, Z. Chen, L. Sun, et al. Herb polysaccharide-based drug delivery system: Fabrication, properties, and applications for immunotherapy. Pharmaceutics, 2022, 14(8): 1703. https://doi.org/10.3390/pharmaceutics14081703
C. Yang, H. Ming, B.W. Li, et al. A pH and glutathione-responsive carbon monoxide-driven nano-herb delivery system for enhanced immunotherapy in colorectal cancer. Journal of Controlled Release, 2024, 376: 659−677. https://doi.org/10.1016/j.jconrel.2024.10.043
Y.X. Xiong, N. Li, M.M. Han, et al. Rhodiola rosea polysaccharides-based nanoparticles loaded with DOX boosts chemo-immunotherapy for triple-negative breast cancer by re-educating Tumor-associated macrophages. International Journal of Biological Macromolecules, 2023, 239: 124110. https://doi.org/10.1016/j.ijbiomac.2023.124110
X. Lang, X. Wang, M. Han, et al. Nanoparticle-mediated synergistic chemoimmunotherapy for cancer treatment. International Journal of Nanomedicine, 2024, 19: 4533−4568. https://doi.org/10.2147/IJN.S455213
B. Zhao, H. Lin, X. Jiang, et al. Exosome-like nanoparticles derived from fruits, vegetables, and herbs: Innovative strategies of therapeutic and drug delivery. Theranostics, 2024, 14(12): 4598−4621. https://doi.org/10.7150/thno.97096
Q. Yi, Z. Xu, A. Thakur, et al. Current understanding of plant-derived exosome-like nanoparticles in regulating the inflammatory response and immune system microenvironment. Pharmacological Research, 2023, 190: 106733. https://doi.org/10.1016/j.phrs.2023.106733
X.D. Wang, J. Fu, C. Jiang, et al. Specific and long-term luminescent monitoring of hydrogen peroxide in tumor metastasis. Advanced Materials, 2023, 35(20): 2210948. https://doi.org/10.1002/adma.202210948
S.M. Wei, Y.H. Wang, Z.S. Tang, et al. A novel green synthesis of silver nanoparticles by the residues of Chinese herbal medicine and their biological activities. RSC Advances, 2021, 11(3): 1411−1419. https://doi.org/10.1039/D0RA08287B