Understanding the Novel Approach of Nanoferroptosis for Cancer Therapy
Corresponding Author: Fei Gao
Nano-Micro Letters,
Vol. 16 (2024), Article Number: 188
Abstract
As a new form of regulated cell death, ferroptosis has unraveled the unsolicited theory of intrinsic apoptosis resistance by cancer cells. The molecular mechanism of ferroptosis depends on the induction of oxidative stress through excessive reactive oxygen species accumulation and glutathione depletion to damage the structural integrity of cells. Due to their high loading and structural tunability, nanocarriers can escort the delivery of ferro-therapeutics to the desired site through enhanced permeation or retention effect or by active targeting. This review shed light on the necessity of iron in cancer cell growth and the fascinating features of ferroptosis in regulating the cell cycle and metastasis. Additionally, we discussed the effect of ferroptosis-mediated therapy using nanoplatforms and their chemical basis in overcoming the barriers to cancer therapy.
Highlights:
1 Nanoferroptosis: a novel cell death process using various nanoparticles.
2 Therapeutic potential of nanoferroptosis inducers in cancer.
3 Synergistic approach of nanoferroptosis with immunotherapy, sonodynamic and photodynamic therapy.
Keywords
Download Citation
Endnote/Zotero/Mendeley (RIS)BibTeX
- D.-W. Zheng, Q. Lei, J.-Y. Zhu, J.-X. Fan, C.-X. Li et al., Switching apoptosis to ferroptosis: metal-organic network for high-efficiency anticancer therapy. Nano Lett. 17, 284–291 (2017). https://doi.org/10.1021/acs.nanolett.6b04060
- M.J. Davis, B.H. Ha, E.C. Holman, R. Halaban, J. Schlessinger et al., RAC1P29S is a spontaneously activating cancer-associated GTPase. Proc. Natl. Acad. Sci. U.S.A. 110, 912–917 (2013). https://doi.org/10.1073/pnas.1220895110
- S.J. Dixon, K.M. Lemberg, M.R. Lamprecht, R. Skouta, E.M. Zaitsev et al., Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012). https://doi.org/10.1016/j.cell.2012.03.042
- C. Cerella, C. Grandjenette, M. Dicato, M. Diederich, Roles of apoptosis and cellular senescence in cancer and aging. Curr. Drug Targets 17, 405–415 (2016). https://doi.org/10.2174/1389450116666150202155915
- A. Sheikh, N.A. Alhakamy, S. Md, P. Kesharwani, Recent progress of RGD modified liposomes as multistage rocket against cancer. Front. Pharmacol. 12, 803304 (2022). https://doi.org/10.3389/fphar.2021.803304
- M. Ishibashi, H. Tamura, M. Sunakawa, A. Kondo-Onodera, N. Okuyama et al., Myeloma drug resistance induced by binding of myeloma B7–H1 (PD-L1) to PD-1. Cancer Immunol. Res. 4, 779–788 (2016). https://doi.org/10.1158/2326-6066.CIR-15-0296
- Z. Su, Z. Yang, L. Xie, J.P. DeWitt, Y. Chen, Cancer therapy in the necroptosis era. Cell Death Differ. 23, 748–756 (2016). https://doi.org/10.1038/cdd.2016.8
- W.S. Yang, B.R. Stockwell, Ferroptosis: death by lipid peroxidation. Trends Cell Biol. 26, 165–176 (2016). https://doi.org/10.1016/j.tcb.2015.10.014
- C. Nathan, A. Cunningham-Bussel, Beyond oxidative stress: an immunologist’s guide to reactive oxygen species. Nat. Rev. Immunol. 13, 349–361 (2013). https://doi.org/10.1038/nri3423
- W.S. Yang, R. SriRamaratnam, M.E. Welsch, K. Shimada, R. Skouta et al., Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014). https://doi.org/10.1016/j.cell.2013.12.010
- L. Jiang, N. Kon, T. Li, S.-J. Wang, T. Su et al., Ferroptosis as a p53-mediated activity during tumour suppression. Nature 520, 57–62 (2015). https://doi.org/10.1038/nature14344
- A. Manohar, J. Park, D.D. Geleta, C. Krishnamoorthi, R. Thangam et al., Synthesis and characterization of ZnO nanops for photocatalysis, antibacterial and cytotoxicity in kidney cancer (A498) cell lines. J. Alloys Compd. 874, 159868 (2021). https://doi.org/10.1016/j.jallcom.2021.159868
- H. Deng, J. Zhang, Y. Yang, J. Yang, Y. Wei et al., Chemodynamic and photothermal combination therapy based on dual-modified metal-organic framework for inducing tumor ferroptosis/pyroptosis. ACS Appl. Mater. Interfaces 14, 24089–24101 (2022). https://doi.org/10.1021/acsami.2c00574
- L. Yu, Y. Xu, Z. Pu, H. Kang, M. Li et al., Photocatalytic superoxide radical generator that induces pyroptosis in cancer cells. J. Am. Chem. Soc. 144, 11326–11337 (2022). https://doi.org/10.1021/jacs.2c03256
- Y. Du, J. Yang, F. He, X. Zhao, J. Zhou et al., Revealing the mutually enhanced mechanism of necroptosis and immunotherapy induced by defect engineering and piezoelectric effect. Adv. Mater. 36, e2304322 (2024). https://doi.org/10.1002/adma.202304322
- P. Zheng, B. Ding, G. Zhu, C. Li, J. Lin, Biodegradable Ca2+ nanomodulators activate pyroptosis through mitochondrial Ca2+ overload for cancer immunotherapy. Angew. Chem. Int. Ed. 61, e202204904 (2022). https://doi.org/10.1002/anie.202204904
- S.-J. Wang, D. Li, Y. Ou, L. Jiang, Y. Chen et al., Acetylation is crucial for p53-mediated ferroptosis and tumor suppression. Cell Rep. 17, 366–373 (2016). https://doi.org/10.1016/j.celrep.2016.09.022
- Y. Xie, W. Hou, X. Song, Y. Yu, J. Huang et al., Ferroptosis: process and function. Cell Death Differ. 23, 369–379 (2016). https://doi.org/10.1038/cdd.2015.158
- S. Mukherjee, S. Mukherjee, M.A.S. Abourehab, A. Sahebkar, P. Kesharwani, Exploring dendrimer-based drug delivery systems and their potential applications in cancer immunotherapy. Eur. Polym. J. 177, 111471 (2022). https://doi.org/10.1016/j.eurpolymj.2022.111471
- Y. Du, R. Zhang, J. Yang, S. Liu, J. Zhou et al., A “closed-loop” therapeutic strategy based on mutually reinforced ferroptosis and immunotherapy. Adv. Funct. Mater. 32, 2111784 (2022). https://doi.org/10.1002/adfm.202111784
- K. Zhang, Z. Ma, S. Li, Y. Wu, J. Zhang et al., Disruption of dual homeostasis by a metal-organic framework nanoreactor for ferroptosis-based immunotherapy of tumor. Biomaterials 284, 121502 (2022). https://doi.org/10.1016/j.biomaterials.2022.121502
- H. Tang, D. Chen, C. Li, C. Zheng, X. Wu et al., Dual GSH-exhausting sorafenib loaded manganese-silica nanodrugs for inducing the ferroptosis of hepatocellular carcinoma cells. Int. J. Pharm. 572, 118782 (2019). https://doi.org/10.1016/j.ijpharm.2019.118782
- P. Kesharwani, R.K. Tekade, V. Gajbhiye, K. Jain, N.K. Jain, Cancer targeting potential of some ligand-anchored poly(propylene imine) dendrimers: a comparison. Nanomed. Nanotechnol. Biol. Med. 7, 295–304 (2011). https://doi.org/10.1016/j.nano.2010.10.010
- A. Sheikh, S. Md, N.A. Alhakamy, P. Kesharwani, Recent development of aptamer conjugated chitosan nanops as cancer therapeutics. Int. J. Pharm. 620, 121751 (2022). https://doi.org/10.1016/j.ijpharm.2022.121751
- A. Sheikh, P. Kesharwani, An insight into aptamer engineered dendrimer for cancer therapy. Eur. Polym. J. 159, 110746 (2021). https://doi.org/10.1016/j.eurpolymj.2021.110746
- P. Kesharwani, A. Sheikh, M.A.S. Abourehab, R. Salve, V. Gajbhiye, A combinatorial delivery of survivin targeted siRNA using cancer selective nanops for triple negative breast cancer therapy. J. Drug Deliv. Sci. Technol. 80, 104164 (2023). https://doi.org/10.1016/j.jddst.2023.104164
- P. Kesharwani, K. Jain, N.K. Jain, Dendrimer as nanocarrier for drug delivery. Prog. Polym. Sci. 39, 268–307 (2014). https://doi.org/10.1016/j.progpolymsci.2013.07.005
- P. Kesharwani, V. Gajbhiye, N.K. Jain, A review of nanocarriers for the delivery of small interfering RNA. Biomaterials 33, 7138–7150 (2012). https://doi.org/10.1016/j.biomaterials.2012.06.068
- P. Kesharwani, A.K. Iyer, Recent advances in dendrimer-based nanovectors for tumor-targeted drug and gene delivery. Drug Discov. Today 20, 536–547 (2015). https://doi.org/10.1016/j.drudis.2014.12.012
- R.A. Bapat, T.V. Chaubal, C.P. Joshi, P.R. Bapat, H. Choudhury et al., An overview of application of silver nanops for biomaterials in dentistry. Mater. Sci. Eng. C Mater. Biol. Appl. 91, 881–898 (2018). https://doi.org/10.1016/j.msec.2018.05.069
- P. Kesharwani, S. Banerjee, U. Gupta, M.C.I. Mohd Amin, S. Padhye et al., PAMAM dendrimers as promising nanocarriers for RNAi therapeutics. Mater. Today 18, 565–572 (2015). https://doi.org/10.1016/j.mattod.2015.06.003
- A.K. Sharma, A. Gothwal, P. Kesharwani, H. Alsaab, A.K. Iyer et al., Dendrimer nanoarchitectures for cancer diagnosis and anticancer drug delivery. Drug Discov. Today 22, 314–326 (2017). https://doi.org/10.1016/j.drudis.2016.09.013
- P. Kesharwani, B. Gorain, S.Y. Low, S.A. Tan, E.C.S. Ling et al., Nanotechnology based approaches for anti-diabetic drugs delivery. Diabetes Res. Clin. Pract. 136, 52–77 (2018). https://doi.org/10.1016/j.diabres.2017.11.018
- P. Kesharwani, A. Gothwal, A.K. Iyer, K. Jain, M.K. Chourasia et al., Dendrimer nanohybrid carrier systems: an expanding horizon for targeted drug and gene delivery. Drug Discov. Today 23, 300–314 (2018). https://doi.org/10.1016/j.drudis.2017.06.009
- P. Kesharwani, H. Choudhury, J.G. Meher, M. Pandey, B. Gorain, Dendrimer-entrapped gold nanops as promising nanocarriers for anticancer therapeutics and imaging. Prog. Mater. Sci. 103, 484–508 (2019). https://doi.org/10.1016/j.pmatsci.2019.03.003
- P. Kesharwani, R. Ghanghoria, N.K. Jain, Carbon nanotube exploration in cancer cell lines. Drug Discov. Today 17, 1023–1030 (2012). https://doi.org/10.1016/j.drudis.2012.05.003
- P. Kesharwani, V. Mishra, N.K. Jain, Validating the anticancer potential of carbon nanotube-based therapeutics through cell line testing. Drug Discov. Today 20, 1049–1060 (2015). https://doi.org/10.1016/j.drudis.2015.05.004
- M. Yuan, M. Kermanian, T. Agarwal, Z. Yang, S. Yousefiasl et al., Defect engineering in biomedical sciences. Adv. Mater. 35, e2304176 (2023). https://doi.org/10.1002/adma.202304176
- R. Jiang, X. Li, D. Hu, M. Zhu, D. Zhou et al., Rapamycin-reinforced ferroptosis assisted by a lysosome-controlled disintegratable micelle in autophagy-dependent/independent manners. Appl. Mater. Today 23, 101066 (2021). https://doi.org/10.1016/j.apmt.2021.101066
- G. Zhu, H. Chi, M. Liu, Y. Yin, H. Diao et al., Multifunctional “ball-rod” Janus nanops boosting Fenton reaction for ferroptosis therapy of non-small cell lung cancer. J. Colloid Interface Sci. 621, 12–23 (2022). https://doi.org/10.1016/j.jcis.2022.04.021
- H. Zheng, J. Jiang, S. Xu, W. Liu, Q. Xie et al., Nanop-induced ferroptosis: detection methods, mechanisms and applications. Nanoscale 13, 2266–2285 (2021). https://doi.org/10.1039/d0nr08478f
- H. Yu, J. Yan, Z. Li, L. Yang, F. Ju et al., Recent trends in emerging strategies for ferroptosis-based cancer therapy. Nanoscale Adv. 5, 1271–1290 (2023). https://doi.org/10.1039/d2na00719c
- C. Zhang, X. Liu, S. Jin, Y. Chen, R. Guo, Ferroptosis in cancer therapy: a novel approach to reversing drug resistance. Mol. Cancer 21, 47 (2022). https://doi.org/10.1186/s12943-022-01530-y
- C. Qiao, H. Wang, Q. Guan, M. Wei, Z. Li, Ferroptosis-based nano delivery systems targeted therapy for colorectal cancer: insights and future perspectives. Asian J. Pharm. Sci. 17, 613–629 (2022). https://doi.org/10.1016/j.ajps.2022.09.002
- K. Liu, L. Huang, S. Qi, S. Liu, W. Xie et al., Ferroptosis: the entanglement between traditional drugs and nanodrugs in tumor therapy. Adv. Healthc. Mater. 12, 2203085 (2023). https://doi.org/10.1002/adhm.202203085
- G. Lei, L. Zhuang, B. Gan, Targeting ferroptosis as a vulnerability in cancer. Nat. Rev. Cancer 22, 381–396 (2022). https://doi.org/10.1038/s41568-022-00459-0
- N. Kang, S. Son, S. Min, H. Hong, C. Kim et al., Stimuli-responsive ferroptosis for cancer therapy. Chem. Soc. Rev. 52, 3955–3972 (2023). https://doi.org/10.1039/d3cs00001j
- X. Jiang, B.R. Stockwell, M. Conrad, Ferroptosis: mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 22, 266–282 (2021). https://doi.org/10.1038/s41580-020-00324-8
- M.J. Ko, W. Yoo, S. Min, Y. Zhang, J. Joo et al., Photonic control of image-guided ferroptosis cancer nanomedicine. Coord. Chem. Rev. 500, 215532 (2024). https://doi.org/10.1016/J.CCR.2023.215532
- M. Jung, C. Mertens, E. Tomat, B. Brüne, Iron as a central player and promising target in cancer progression. Int. J. Mol. Sci. 20, 273 (2019). https://doi.org/10.3390/ijms20020273
- S.V. Torti, F.M. Torti, Iron and cancer: more ore to be mined. Nat. Rev. Cancer 13, 342–355 (2013). https://doi.org/10.1038/nrc3495
- T.A. Rouault, N. Maio, Biogenesis and functions of mammalian iron-sulfur proteins in the regulation of iron homeostasis and pivotal metabolic pathways. J. Biol. Chem. 292, 12744–12753 (2017). https://doi.org/10.1074/jbc.R117.789537
- A. Leftin, N. Ben-Chetrit, F. Klemm, J.A. Joyce, J.A. Koutcher, Iron imaging reveals tumor and metastasis macrophage hemosiderin deposits in breast cancer. PLoS ONE 12, e0184765 (2017). https://doi.org/10.1371/journal.pone.0184765
- S. Dev, J.L. Babitt, Overview of iron metabolism in health and disease. Hemodial. Int. 21, S6–S20 (2017). https://doi.org/10.1111/hdi.12542
- A.R. Bogdan, M. Miyazawa, K. Hashimoto, Y. Tsuji, Regulators of iron homeostasis: new players in metabolism, cell death, and disease. Trends Biochem. Sci. 41, 274–286 (2016). https://doi.org/10.1016/j.tibs.2015.11.012
- N. Wilkinson, K. Pantopoulos, The IRP/IRE system in vivo: insights from mouse models. Front. Pharmacol. 5, 176 (2014). https://doi.org/10.3389/fphar.2014.00176
- C. Raggi, E. Gammella, M. Correnti, P. Buratti, E. Forti et al., Dysregulation of iron metabolism in cholangiocarcinoma stem-like cells. Sci. Rep. 7, 17667 (2017). https://doi.org/10.1038/s41598-017-17804-1
- Z. Rychtarcikova, S. Lettlova, V. Tomkova, V. Korenkova, L. Langerova et al., Tumor-initiating cells of breast and prostate origin show alterations in the expression of genes related to iron metabolism. Oncotarget 8, 6376–6398 (2017). https://doi.org/10.18632/oncotarget.14093
- T.B. Toh, J.J. Lim, E.K. Chow, Epigenetics in cancer stem cells. Mol. Cancer 16, 29 (2017). https://doi.org/10.1186/s12943-017-0596-9
- E.N. Wainwright, P. Scaffidi, Epigenetics and cancer stem cells: unleashing, hijacking, and restricting cellular plasticity. Trends Cancer 3, 372–386 (2017). https://doi.org/10.1016/j.trecan.2017.04.004
- J.P.F. Angeli, M. Schneider, B. Proneth, Y.Y. Tyurina, V.A. Tyurin et al., Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat. Cell Biol. 16, 1180–1191 (2014). https://doi.org/10.1038/ncb3064
- Y. Mou, J. Wang, J. Wu, D. He, C. Zhang et al., Ferroptosis, a new form of cell death: opportunities and challenges in cancer. J. Hematol. Oncol. 12, 34 (2019). https://doi.org/10.1186/s13045-019-0720-y
- B. Lu, X.B. Chen, M.D. Ying, Q.J. He, J. Cao et al., The role of ferroptosis in cancer development and treatment response. Front. Pharmacol. 8, 992 (2018). https://doi.org/10.3389/fphar.2017.00992
- Y. Chen, Z. Yao, P. Liu, Q. Hu, Y. Huang et al., A self-assembly nano-prodrug for triple-negative breast cancer combined treatment by ferroptosis therapy and chemotherapy. Acta Biomater. 159, 275–288 (2023). https://doi.org/10.1016/j.actbio.2023.01.050
- K. Kazan, S. Kalaipandian, Ferroptosis: yet another way to die. Trends Plant Sci. 24, 479–481 (2019). https://doi.org/10.1016/j.tplants.2019.03.005
- J.P. Friedmann Angeli, D.V. Krysko, M. Conrad, Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat. Rev. Cancer 19, 405–414 (2019). https://doi.org/10.1038/s41568-019-0149-1
- M. Conlon, S.J. Dixon, Ferroptosis-like death in plant cells. Mol. Cell. Oncol. 4, e1302906 (2017). https://doi.org/10.1080/23723556.2017.1302906
- X. Lang, M.D. Green, W. Wang, J. Yu, J.E. Choi et al., Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov. 9, 1673–1685 (2019). https://doi.org/10.1158/2159-8290.CD-19-0338
- F. Kuang, J. Liu, D. Tang, R. Kang, Oxidative damage and antioxidant defense in ferroptosis. Front. Cell Dev. Biol. 8, 586578 (2020). https://doi.org/10.3389/fcell.2020.586578
- F. Ursini, M. Maiorino, C. Gregolin, The selenoenzyme phospholipid hydroperoxide glutathione peroxidase. Biochim. Biophys. Acta BBA Gen. Subj. 839, 62–70 (1985). https://doi.org/10.1016/0304-4165(85)90182-5
- W.S. Yang, K.J. Kim, M.M. Gaschler, M. Patel, M.S. Shchepinov et al., Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc. Natl. Acad. Sci. U.S.A. 113, E4966–E4975 (2016). https://doi.org/10.1073/pnas.1603244113
- X. Sui, R. Zhang, S. Liu, T. Duan, L. Zhai et al., RSL3 drives ferroptosis through GPX4 inactivation and ROS production in colorectal cancer. Front. Pharmacol. 9, 1371 (2018). https://doi.org/10.3389/fphar.2018.01371
- J. Yang, J. Mo, J. Dai, C. Ye, W. Cen et al., Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis. 12, 1079 (2021). https://doi.org/10.1038/s41419-021-04367-3
- B. Han, Y. Liu, Q. Zhang, L. Liang, Propofol decreases cisplatin resistance of non-small cell lung cancer by inducing GPX4-mediated ferroptosis through the miR-744-5p/miR-615-3p axis. J. Proteomics 274, 104777 (2023). https://doi.org/10.1016/j.jprot.2022.104777
- S. Cai, Z. Ding, X. Liu, J. Zeng, Trabectedin induces ferroptosis via regulation of HIF-1α/IRP1/TFR1 and Keap1/Nrf2/GPX4 axis in non-small cell lung cancer cells. Chem. Biol. Interact. 369, 110262 (2023). https://doi.org/10.1016/j.cbi.2022.110262
- W. Zhang, B. Jiang, Y. Liu, L. Xu, M. Wan, Bufotalin induces ferroptosis in non-small cell lung cancer cells by facilitating the ubiquitination and degradation of GPX4. Free Radic. Biol. Med. 180, 75–84 (2022). https://doi.org/10.1016/j.freeradbiomed.2022.01.009
- R. Yu, Y. Zhou, S. Shi, X. Wang, S. Huang et al., Icariside II induces ferroptosis in renal cell carcinoma cells by regulating the miR-324-3p/GPX4 axis. Phytomedicine 102, 154182 (2022). https://doi.org/10.1016/j.phymed.2022.154182
- M. Hashemzaei, A.K. Barani, M. Iranshahi, R. Rezaee, K. Tsarouhas et al., Effects of resveratrol on carbon monoxide-induced cardiotoxicity in rats. Environ. Toxicol. Pharmacol. 46, 110–115 (2016). https://doi.org/10.1016/j.etap.2016.07.010
- T.M. Bahr, R.D. Christensen, D.M. Ward, F. Meng, L.K. Jackson et al., Ferritin in serum and urine: a pilot study. Blood Cells Mol. Dis. 76, 59–62 (2019). https://doi.org/10.1016/j.bcmd.2019.02.001
- Y. Qin, Y. Qiao, D. Wang, C. Tang, G. Yan, Ferritinophagy and ferroptosis in cardiovascular disease: mechanisms and potential applications. Biomed. Pharmacother. 141, 111872 (2021). https://doi.org/10.1016/j.biopha.2021.111872
- K. Shimada, R. Skouta, A. Kaplan, W.S. Yang, M. Hayano et al., Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat. Chem. Biol. 12, 497–503 (2016). https://doi.org/10.1038/nchembio.2079
- Z. Wu, Y. Geng, X. Lu, Y. Shi, G. Wu et al., Chaperone-mediated autophagy is involved in the execution of ferroptosis. Proc. Natl. Acad. Sci. U.S.A. 116, 2996–3005 (2019). https://doi.org/10.1073/pnas.1819728116
- M. Conrad, D.A. Pratt, The chemical basis of ferroptosis. Nat. Chem. Biol. 15, 1137–1147 (2019). https://doi.org/10.1038/s41589-019-0408-1
- B.R. Stockwell, Ferroptosis turns 10: emerging mechanisms, physiological functions, and therapeutic applications. Cell 185, 2401–2421 (2022). https://doi.org/10.1016/j.cell.2022.06.003
- Y. Zou, H. Li, E.T. Graham, A.A. Deik, J.K. Eaton et al., Cytochrome P450 oxidoreductase contributes tophospholipid peroxidation in ferroptosis. Nat. Chem. Biol. 16, 302–309 (2020). https://doi.org/10.1038/s41589-020-0472-6
- V.E. Kagan, G. Mao, F. Qu, J.P.F. Angeli, S. Doll et al., Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat. Chem. Biol. 13, 81–90 (2017). https://doi.org/10.1038/nchembio.2238
- R. Shintoku, Y. Takigawa, K. Yamada, C. Kubota, Y. Yoshimoto et al., Lipoxygenase-mediated generation of lipid peroxides enhances ferroptosis induced by erastin and RSL3. Cancer Sci. 108, 2187–2194 (2017). https://doi.org/10.1111/cas.13380
- L. Tirinato, C. Liberale, S. Di Franco, P. Candeloro, A. Benfante et al., Lipid droplets: a new player in colorectal cancer stem cells unveiled by spectroscopic imaging. Stem Cells 33, 35–44 (2015). https://doi.org/10.1002/stem.1837
- M. Visweswaran, F. Arfuso, S. Warrier, A. Dharmarajan, Aberrant lipid metabolism as an emerging therapeutic strategy to target cancer stem cells. Stem Cells 38, 6–14 (2020). https://doi.org/10.1002/stem.3101
- B.J. Hershey, R. Vazzana, D.L. Joppi, K.M. Havas, Lipid droplets define a sub-population of breast cancer stem cells. J. Clin. Med. 9, 87 (2019). https://doi.org/10.3390/jcm9010087
- S.J. Dixon, G.E. Winter, L.S. Musavi, E.D. Lee, B. Snijder et al., Human haploid cell genetics reveals roles for lipid metabolism genes in nonapoptotic cell death. ACS Chem. Biol. 10, 1604–1609 (2015). https://doi.org/10.1021/acschembio.5b00245
- L. Zhao, Y. Peng, S. He, R. Li, Z. Wang et al., Apatinib induced ferroptosis by lipid peroxidation in gastric cancer. Gastric Cancer 24, 642–654 (2021). https://doi.org/10.1007/s10120-021-01159-8
- J. Li, S. Huang, Q. Wang, D. Zhou, B. Zhao et al., Andrographolide promoted ferroptosis to repress the development of non-small cell lung cancer through activation of the mitochondrial dysfunction. Phytomedicine 109, 154601 (2023). https://doi.org/10.1016/j.phymed.2022.154601
- F. Yang, Y. Xiao, J.-H. Ding, X. Jin, D. Ma et al., Ferroptosis heterogeneity in triple-negative breast cancer reveals an innovative immunotherapy combination strategy. Cell Metab. 35, 84-100.e8 (2023). https://doi.org/10.1016/j.cmet.2022.09.021
- G.-Q. Chen, F.A. Benthani, J. Wu, D. Liang, Z.-X. Bian et al., Artemisinin compounds sensitize cancer cells to ferroptosis by regulating iron homeostasis. Cell Death Differ. 27, 242–254 (2020). https://doi.org/10.1038/s41418-019-0352-3
- S. Huang, H. Le, G. Hong, G. Chen, F. Zhang et al., An all-in-one biomimetic iron-small interfering RNA nanoplatform induces ferroptosis for cancer therapy. Acta Biomater. 148, 244–257 (2022). https://doi.org/10.1016/j.actbio.2022.06.017
- F. Zhang, F. Li, G.-H. Lu, W. Nie, L. Zhang et al., Engineering magnetosomes for ferroptosis/immunomodulation synergism in cancer. ACS Nano 13, 5662–5673 (2019). https://doi.org/10.1021/acsnano.9b00892
- Z. Shen, T. Liu, Y. Li, J. Lau, Z. Yang et al., Fenton-reaction-acceleratable magnetic nanops for ferroptosis therapy of orthotopic brain tumors. ACS Nano 12, 11355–11365 (2018). https://doi.org/10.1021/acsnano.8b06201
- Y. Zou, W.S. Henry, E.L. Ricq, E.T. Graham, V.V. Phadnis et al., Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature 585, 603–608 (2020). https://doi.org/10.1038/s41586-020-2732-8
- Y.Y. Tyurina, A.A. Kapralov, V.A. Tyurin, G. Shurin, A.A. Amoscato et al., Redox phospholipidomics discovers pro-ferroptotic death signals in A375 melanoma cells in vitro and in vivo. Redox Biol. 61, 102650 (2023). https://doi.org/10.1016/j.redox.2023.102650
- K.A. Jasim, A.J. Gesquiere, Ultrastable and biofunctionalizable conjugated polymer nanops with encapsulated iron for ferroptosis assisted chemodynamic therapy. Mol. Pharm. 16, 4852–4866 (2019). https://doi.org/10.1021/acs.molpharmaceut.9b00737
- S. Koo, O.K. Park, J. Kim, S.I. Han, T.Y. Yoo et al., Enhanced chemodynamic therapy by Cu-Fe peroxide nanops: tumor microenvironment-mediated synergistic Fenton reaction. ACS Nano 16, 2535–2545 (2022). https://doi.org/10.1021/acsnano.1c09171
- N. Wang, Q. Zeng, R. Zhang, D. Xing, T. Zhang, Eradication of solid tumors by chemodynamic theranostics with H2O2-catalyzed hydroxyl radical burst. Theranostics 11, 2334–2348 (2021). https://doi.org/10.7150/thno.49277
- M. Zahiri, M. Falsafi, K. Lamei, K. Abnous, S.M. Taghdisi et al., Targeted biomimetic hollow mesoporous organosilica nanops for delivery of doxorubicin to colon adenocarcinoma: in vitro and in vivo evaluation. Microporous Mesoporous Mater. 335, 111841 (2022). https://doi.org/10.1016/j.micromeso.2022.111841
- L. Huang, J. Zhu, W. Xiong, J. Feng, J. Yang et al., Tumor-generated reactive oxygen species storm for high-performance ferroptosis therapy. ACS Nano 17, 11492–11506 (2023). https://doi.org/10.1021/acsnano.3c01369
- S. Bai, Y. Lan, S. Fu, H. Cheng, Z. Lu et al., Connecting calcium-based nanomaterials and cancer: from diagnosis to therapy. Nano-Micro Lett. 14, 145 (2022). https://doi.org/10.1007/s40820-022-00894-6
- Z. Chen, J.-J. Yin, Y.-T. Zhou, Y. Zhang, L. Song et al., Dual enzyme-like activities of iron oxide nanops and their implication for diminishing cytotoxicity. ACS Nano 6, 4001–4012 (2012). https://doi.org/10.1021/nn300291r
- L. Gao, J. Zhuang, L. Nie, J. Zhang, Y. Zhang et al., Intrinsic peroxidase-like activity of ferromagnetic nanops. Nat. Nanotechnol. 2, 577–583 (2007). https://doi.org/10.1038/nnano.2007.260
- L.S. Li, E.A. Bey, Y. Dong, J. Meng, B. Patra et al., Modulating endogenous NQO1 levels identifies key regulatory mechanisms of action of β-lapachone for pancreatic cancer therapy. Clin. Cancer Res. 17, 275–285 (2011). https://doi.org/10.1158/1078-0432.CCR-10-1983
- H. Yeo, C.A. Lyssiotis, Y. Zhang, H. Ying, J.M. Asara et al., FoxO3 coordinates metabolic pathways to maintain redox balance in neural stem cells. EMBO J. 32, 2589–2602 (2013). https://doi.org/10.1038/emboj.2013.186
- Q. Chen, J. Zhou, Z. Chen, Q. Luo, J. Xu et al., Tumor-specific expansion of oxidative stress by glutathione depletion and use of a Fenton nanoagent for enhanced chemodynamic therapy. ACS Appl. Mater. Interfaces 11, 30551–30565 (2019). https://doi.org/10.1021/acsami.9b09323
- W. Li, S. Yin, Y. Shen, H. Li, L. Yuan et al., Molecular engineering of pH-responsive NIR oxazine assemblies for evoking tumor ferroptosis via triggering lysosomal dysfunction. J. Am. Chem. Soc. 145, 3736–3747 (2023). https://doi.org/10.1021/jacs.2c13222
- Y. Xie, S. Zhu, X. Song, X. Sun, Y. Fan et al., The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell Rep. 20, 1692–1704 (2017). https://doi.org/10.1016/j.celrep.2017.07.055
- H. Chen, F. Peng, J. Xu, G. Wang, Y. Zhao, Increased expression of GPX4 promotes the tumorigenesis of thyroid cancer by inhibiting ferroptosis and predicts poor clinical outcomes. Aging 15, 230–245 (2023). https://doi.org/10.18632/aging.204473
- X. Wei, X. Yi, X.-H. Zhu, D.-S. Jiang, Posttranslational modifications in ferroptosis. Oxid. Med. Cell. Longev. 2020, 8832043 (2020). https://doi.org/10.1155/2020/8832043
- C. Gai, C. Liu, X. Wu, M. Yu, J. Zheng et al., MT1DP loaded by folate-modified liposomes sensitizes erastin-induced ferroptosis via regulating miR-365a-3p/NRF2 axis in non-small cell lung cancer cells. Cell Death Dis. 11, 751 (2020). https://doi.org/10.1038/s41419-020-02939-3
- A. Brown, S. Kumar, P.B. Tchounwou, Cisplatin-based chemotherapy of human cancers. J. Cancer Sci. Ther. 11, 97 (2019). https://doi.org/10.4172/1948-5956.1000592
- G. Bronte, D. Andreis, S. Bravaccini, R. Maltoni, L. Cecconetto et al., Sorafenib for the treatment of breast cancer. Expert Opin. Pharmacother. 18, 621–630 (2017). https://doi.org/10.1080/14656566.2017.1309024
- C. Bae, H. Kim, Y.M. Kook, C. Lee, C. Kim et al., Induction of ferroptosis using functionalized iron-based nanops for anti-cancer therapy. Mater. Today Bio 17, 100457 (2022). https://doi.org/10.1016/j.mtbio.2022.100457
- M. Huo, L. Wang, Y. Chen, J. Shi, Tumor-selective catalytic nanomedicine by nanocatalyst delivery. Nat. Commun. 8, 357 (2017). https://doi.org/10.1038/s41467-017-00424-8
- L. Wang, M. Huo, Y. Chen, J. Shi, Iron-engineered mesoporous silica nanocatalyst with biodegradable and catalytic framework for tumor-specific therapy. Biomaterials 163, 1–13 (2018). https://doi.org/10.1016/j.biomaterials.2018.02.018
- X. Qian, J. Zhang, Z. Gu, Y. Chen, Nanocatalysts-augmented Fenton chemical reaction for nanocatalytic tumor therapy. Biomaterials 211, 1–13 (2019). https://doi.org/10.1016/j.biomaterials.2019.04.023
- Z. Zhou, J. Song, R. Tian, Z. Yang, G. Yu et al., Activatable singlet oxygen generation from lipid hydroperoxide nanops for cancer therapy. Angew. Chem. Int. Ed. 56, 6492–6496 (2017). https://doi.org/10.1002/anie.201701181
- A. Sheikh, S. Md, P. Kesharwani, Aptamer grafted nanop as targeted therapeutic tool for the treatment of breast cancer. Biomed. Pharmacother. 146, 112530 (2022). https://doi.org/10.1016/j.biopha.2021.112530
- M. Fatima, A. Sheikh, N. Hasan, A. Sahebkar, Y. Riadi et al., Folic acid conjugated poly(amidoamine) dendrimer as a smart nanocarriers for tracing, imaging, and treating cancers over-expressing folate receptors. Eur. Polym. J. 170, 111156 (2022). https://doi.org/10.1016/j.eurpolymj.2022.111156
- Y. Du, J. Zhou, F. He, P. Zang, H. Gong et al., A bright future: advanced nanotechnology-assisted microwave therapy. Nano Today 52, 101963 (2023). https://doi.org/10.1016/j.nantod.2023.101963
- A.J. Howarth, Y. Liu, P. Li, Z. Li, T.C. Wang et al., Chemical, thermal and mechanical stabilities of metal–organic frameworks. Nat. Rev. Mater. 1, 15018 (2016). https://doi.org/10.1038/natrevmats.2015.18
- P. Venkatesan, N. Thirumalaivasan, H.-P. Yu, P.-S. Lai, S.-P. Wu, Redox stimuli delivery vehicle based on transferrin-capped MSNPs for targeted drug delivery in cancer therapy. ACS Appl. Bio Mater. 2, 1623–1633 (2019). https://doi.org/10.1021/acsabm.9b00036
- K. Lu, T. Aung, N. Guo, R. Weichselbaum, W. Lin, Nanoscale metal-organic frameworks for therapeutic, imaging, and sensing applications. Adv. Mater. 30, e1707634 (2018). https://doi.org/10.1002/adma.201707634
- L. Jiao, H.-L. Jiang, Metal-organic-framework-based single-atom catalysts for energy applications. Chem 5, 786–804 (2019). https://doi.org/10.1016/j.chempr.2018.12.011
- M. Falsafi, M. Zahiri, A.S. Saljooghi, K. Abnous, S.M. Taghdisi et al., Aptamer targeted red blood cell membrane-coated porphyrinic copper-based MOF for guided photochemotherapy against metastatic breast cancer. Micropor Mesopor Mater. 325, 111337 (2021). https://doi.org/10.1016/j.micromeso.2021.111337
- L.F. Ye, K.R. Chaudhary, F. Zandkarimi, A.D. Harken, C.J. Kinslow et al., Radiation-induced lipid peroxidation triggers ferroptosis and synergizes with ferroptosis inducers. ACS Chem. Biol. 15, 469–484 (2020). https://doi.org/10.1021/acschembio.9b00939
- G. Lei, C. Mao, Y. Yan, L. Zhuang, B. Gan, Ferroptosis, radiotherapy, and combination therapeutic strategies. Protein Cell 12, 836–857 (2021). https://doi.org/10.1007/s13238-021-00841-y
- G. Lei, Y. Zhang, T. Hong, X. Zhang, X. Liu et al., Ferroptosis as a mechanism to mediate p53 function in tumor radiosensitivity. Oncogene 40, 3533–3547 (2021). https://doi.org/10.1038/s41388-021-01790-w
- G. Lei, Y. Zhang, P. Koppula, X. Liu, J. Zhang et al., The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 30, 146–162 (2020). https://doi.org/10.1038/s41422-019-0263-3
- Y. Wu, C. Yu, M. Luo, C. Cen, J. Qiu et al., Ferroptosis in cancer treatment: another way to Rome. Front. Oncol. 10, 571127 (2020). https://doi.org/10.3389/fonc.2020.571127
- Y. Liang, C. Peng, N. Su, Q. Li, S. Chen et al., Tumor microenvironments self-activated cascade catalytic nanoscale metal organic frameworks as ferroptosis inducer for radiosensitization. Chem. Eng. J. 437, 135309 (2022). https://doi.org/10.1016/j.cej.2022.135309
- B. Hassannia, P. Vandenabeele, T. Vanden Berghe, Targeting ferroptosis to iron out cancer. Cancer Cell 35, 830–849 (2019). https://doi.org/10.1016/j.ccell.2019.04.002
- T. Liu, W. Liu, M. Zhang, W. Yu, F. Gao et al., Ferrous-supply-regeneration nanoengineering for cancer-cell-specific ferroptosis in combination with imaging-guided photodynamic therapy. ACS Nano 12, 12181–12192 (2018). https://doi.org/10.1021/acsnano.8b05860
- M. Mu, Y. Wang, S. Zhao, X. Li, R. Fan et al., Engineering a pH/glutathione-responsive tea polyphenol nanodevice as an apoptosis/ferroptosis-inducing agent. ACS Appl. Bio Mater. 3, 4128–4138 (2020). https://doi.org/10.1021/acsabm.0c00225
- W. Xu, T. Wang, J. Qian, J. Wang, G. Hou et al., Fe(II)-hydrazide coordinated all-active metal organic framework for photothermally enhanced tumor penetration and ferroptosis-apoptosis synergistic therapy. Chem. Eng. J. 437, 135311 (2022). https://doi.org/10.1016/j.cej.2022.135311
- C. Liang, X. Zhang, M. Yang, X. Dong, Recent progress in ferroptosis inducers for cancer therapy. Adv. Mater. 31, e1904197 (2019). https://doi.org/10.1002/adma.201904197
- H. He, L. Du, H. Guo, Y. An, L. Lu et al., Redox responsive metal organic framework nanops induces ferroptosis for cancer therapy. Small 16, e2001251 (2020). https://doi.org/10.1002/smll.202001251
- Q. Guan, R. Guo, S. Huang, F. Zhang, J. Liu et al., Mesoporous polydopamine carrying sorafenib and SPIO nanops for MRI-guided ferroptosis cancer therapy. J. Control. Release 320, 392–403 (2020). https://doi.org/10.1016/j.jconrel.2020.01.048
- X. Hu, R. Li, W. Wu, K. Fang, Z. Zhu et al., A Fe(III)-porphyrin-oxaliplatin(IV) nanoplatform for enhanced ferroptosis and combined therapy. J. Control. Release 348, 660–671 (2022). https://doi.org/10.1016/j.jconrel.2022.06.019
- H. Peng, X. Zhang, P. Yang, J. Zhao, W. Zhang et al., Defect self-assembly of metal-organic framework triggers ferroptosis to overcome resistance. Bioact. Mater. 19, 1–11 (2021). https://doi.org/10.1016/j.bioactmat.2021.12.018
- A. Jain, P. Kesharwani, N.K. Garg, A. Jain, S.A. Jain et al., Galactose engineered solid lipid nanops for targeted delivery of doxorubicin. Colloids Surf. B Biointerf 134, 47–58 (2015). https://doi.org/10.1016/j.colsurfb.2015.06.027
- N. Soni, N. Soni, H. Pandey, R. Maheshwari, P. Kesharwani et al., Augmented delivery of gemcitabine in lung cancer cells exploring mannose anchored solid lipid nanops. J. Colloid Interface Sci. 481, 107–116 (2016). https://doi.org/10.1016/j.jcis.2016.07.020
- A. Jain, N.K. Garg, A. Jain, P. Kesharwani, A.K. Jain et al., A synergistic approach of adapalene-loaded nanostructured lipid carriers, and vitamin C co-administration for treating acne. Drug Dev. Ind. Pharm. 42, 897–905 (2016). https://doi.org/10.3109/03639045.2015.1104343
- A. Jain, G. Sharma, V. Kushwah, N.K. Garg, P. Kesharwani et al., Methotrexate and beta-carotene loaded-lipid polymer hybrid nanops: a preclinical study for breast cancer. Nanomedicine 12, 1851–1872 (2017). https://doi.org/10.2217/nnm-2017-0011
- S. Md, S. Haque, T. Madheswaran, F. Zeeshan, V.S. Meka et al., Lipid based nanocarriers system for topical delivery of photosensitizers. Drug Discov. Today 22, 1274–1283 (2017). https://doi.org/10.1016/j.drudis.2017.04.010
- N. Hasan, M. Imran, P. Kesharwani, K. Khanna, R. Karwasra et al., Intranasal delivery of Naloxone-loaded solid lipid nanops as a promising simple and non-invasive approach for the management of opioid overdose. Int. J. Pharm. 599, 120428 (2021). https://doi.org/10.1016/j.ijpharm.2021.120428
- G.S. Bhagwat, R.B. Athawale, R.P. Gude, S. Md, N.A. Alhakamy et al., Formulation and development of transferrin targeted solid lipid nanops for breast cancer therapy. Front. Pharmacol. 11, 614290 (2020). https://doi.org/10.3389/fphar.2020.614290
- A. Aziz, U. Rehman, A. Sheikh, M.A.S. Abourehab, P. Kesharwani, Lipid-based nanocarrier mediated CRISPR/Cas9 delivery for cancer therapy. J. Biomater. Sci. Polym. Ed. 34, 398–418 (2023). https://doi.org/10.1080/09205063.2022.2121592
- F. Mohammadpour, H. Kamali, L. Gholami, A.P. McCloskey, P. Kesharwani et al., Solid lipid nanops: a promising tool for insulin delivery. Expert Opin. Drug Deliv. 19, 1577–1595 (2022). https://doi.org/10.1080/17425247.2022.2138328
- D.C. Drummond, C.O. Noble, Z. Guo, K. Hong, J.W. Park et al., Development of a highly active nanoliposomal irinotecan using a novel intraliposomal stabilization strategy. Cancer Res. 66, 3271–3277 (2006). https://doi.org/10.1158/0008-5472.CAN-05-4007
- C. Zylberberg, K. Gaskill, S. Pasley, S. Matosevic, Engineering liposomal nanops for targeted gene therapy. Gene Ther. 24, 441–452 (2017). https://doi.org/10.1038/gt.2017.41
- X. Chen, Y. Zhang, C. Tang, C. Tian, Q. Sun et al., Co-delivery of paclitaxel and anti-survivin siRNA via redox-sensitive oligopeptide liposomes for the synergistic treatment of breast cancer and metastasis. Int. J. Pharm. 529, 102–115 (2017). https://doi.org/10.1016/j.ijpharm.2017.06.071
- F. Persano, G. Gigli, S. Leporatti, Lipid-polymer hybrid nanops in cancer therapy: current overview and future directions. Nano Ex. 2, 012006 (2021). https://doi.org/10.1088/2632-959x/abeb4b
- Z. Li, C. Wang, C. Dai, R. Hu, L. Ding et al., Engineering dual catalytic nanomedicine for autophagy-augmented and ferroptosis-involved cancer nanotherapy. Biomaterials 287, 121668 (2022). https://doi.org/10.1016/j.biomaterials.2022.121668
- P. Ghasemiyeh, S. Mohammadi-Samani, Solid lipid nanops and nanostructured lipid carriers as novel drug delivery systems: applications, advantages and disadvantages. Res. Pharm. Sci. 13, 288–303 (2018). https://doi.org/10.4103/1735-5362.235156
- H. Yuan, L.-L. Wang, Y.-Z. Du, J. You, F.-Q. Hu et al., Preparation and characteristics of nanostructured lipid carriers for control-releasing progesterone by melt-emulsification. Coll. Surf. B Biointerf. 60, 174–179 (2007). https://doi.org/10.1016/j.colsurfb.2007.06.011
- E. Esposito, M. Drechsler, R. Cortesi, C. Nastruzzi, Encapsulation of cannabinoid drugs in nanostructured lipid carriers. Eur. J. Pharm. Biopharm. 102, 87–91 (2016). https://doi.org/10.1016/j.ejpb.2016.03.005
- J. Zhang, J. Yang, T. Zuo, S. Ma, N. Xokrat et al., Heparanase-driven sequential released nanops for ferroptosis and tumor microenvironment modulations synergism in breast cancer therapy. Biomaterials 266, 120429 (2021). https://doi.org/10.1016/j.biomaterials.2020.120429
- L. Zeng, B.H.J. Gowda, M.G. Ahmed, M.A.S. Abourehab, Z.S. Chen et al., Advancements in nanop-based treatment approaches for skin cancer therapy. Mol. Cancer 22, 10 (2023). https://doi.org/10.1186/s12943-022-01708-4
- Z. Liu, N. Parveen, U. Rehman, A. Aziz, A. Sheikh et al., Unravelling the enigma of siRNA and aptamer mediated therapies against pancreatic cancer. Mol. Cancer 22, 8 (2023). https://doi.org/10.1186/s12943-022-01696-5
- U. Rehman, M.A.S. Abourehab, A. Alexander, P. Kesharwani, Polymeric micelles assisted combinatorial therapy: is it new hope for pancreatic cancer? Eur. Polym. J. 184, 111784 (2023). https://doi.org/10.1016/j.eurpolymj.2022.111784
- N. Parveen, M.A.S. Abourehab, R. Shukla, P.V. Thanikachalam, G.K. Jain et al., Immunoliposomes as an emerging nanocarrier for breast cancer therapy. Eur. Polym. J. 184, 111781 (2023). https://doi.org/10.1016/j.eurpolymj.2022.111781
- A.K. Jain, S. Jain, M.A.S. Abourehab, P. Mehta, P. Kesharwani, An insight on topically applied formulations for management of various skin disorders. J. Biomater. Sci. Polym. Ed. 33, 2406–2432 (2022). https://doi.org/10.1080/09205063.2022.2103625
- Y. Wang, T. Liu, X. Li, H. Sheng, X. Ma et al., Ferroptosis-inducing nanomedicine for cancer therapy. Front. Pharmacol. 12, 735965 (2021). https://doi.org/10.3389/fphar.2021.735965
- J. Gao, T. Luo, J. Wang, Gene interfered-ferroptosis therapy for cancers. Nat. Commun. 12, 5311 (2021). https://doi.org/10.1038/s41467-021-25632-1
- M. Cheng, B. Zhang, W. Cui, M.L. Gross, Laser-initiated radical trifluoromethylation of peptides and proteins: application to mass-spectrometry-based protein footprinting. Angew. Chem. Int. Ed. 56, 14007–14010 (2017). https://doi.org/10.1002/anie.201706697
- H. Chen, J. Wen, Iron oxide nanops loaded with paclitaxel inhibits glioblastoma by enhancing autophagy-dependent ferroptosis pathway. Eur. J. Pharmacol. 921, 174860 (2022). https://doi.org/10.1016/j.ejphar.2022.174860
- A. Ruiz-de-Angulo, M. Bilbao-Asensio, J. Cronin, S.J. Evans, M.J.D. Clift et al., Chemically programmed vaccines: iron catalysis in nanops enhances combination immunotherapy and immunotherapy-promoted tumor ferroptosis. iScience 23, 101499 (2020). https://doi.org/10.1016/j.isci.2020.101499
- X. Zhu, Q. Chen, L. Xie, W. Chen, Y. Jiang et al., Iron ion and sulfasalazine-loaded polydopamine nanops for Fenton reaction and glutathione peroxidase 4 inactivation for enhanced cancer ferrotherapy. Acta Biomater. 145, 210–221 (2022). https://doi.org/10.1016/j.actbio.2022.04.024
- J. Li, S. Wang, X. Lin, Y. Cao, Z. Cai et al., Red blood cell-mimic nanocatalyst triggering radical storm to augment cancer immunotherapy. Nano-Micro Lett. 14, 57 (2022). https://doi.org/10.1007/s40820-022-00801-z
- J. Li, W. Lu, Y. Yang, R. Xiang, Y. Ling et al., Hybrid nanomaterials for cancer immunotherapy. Adv. Sci. 10, e2204932 (2023). https://doi.org/10.1002/advs.202204932
- R. Thangam, K.D. Patel, H. Kang, R. Paulmurugan, Advances in engineered polymer nanop tracking platforms towards cancer immunotherapy-current status and future perspectives. Vaccines 9, 935 (2021). https://doi.org/10.3390/vaccines9080935
- X. Cai, L. Ruan, D. Wang, J. Zhang, J. Tang et al., Boosting chemotherapy of bladder cancer cells by ferroptosis using intelligent magnetic targeting nanops. Colloids Surf. B Biointerf 234, 113664 (2024). https://doi.org/10.1016/j.colsurfb.2023.113664
- R. Thangam, M.S. Kim, G. Bae, Y. Kim, N. Kang et al., Remote switching of elastic movement of decorated ligand nanostructures controls the adhesion-regulated polarization of host macrophages. Adv. Funct. Mater. 31, 2008698 (2021). https://doi.org/10.1002/adfm.202008698
- S. Min, Y.S. Jeon, H. Choi, C. Khatua, N. Li et al., Large and externally positioned ligand-coated nanopatches facilitate the adhesion-dependent regenerative polarization of host macrophages. Nano Lett. 20, 7272–7280 (2020). https://doi.org/10.1021/acs.nanolett.0c02655
- W. Yin, J. Chang, J. Sun, Y. Zhao, S. Chen et al., Arginine nanops mediated closed-loop ferroptosis enhancement for T cell activity boosting in cancer immunotherapy. Appl. Mater. Today 36, 102047 (2024). https://doi.org/10.1016/j.apmt.2023.102047
- J. Wu, X. Wang, Q. Wang, Z. Lou, S. Li et al., Nanomaterials with enzyme-like characteristics (nanozymes): next-generation artificial enzymes (II). Chem. Soc. Rev. 48, 1004–1076 (2019). https://doi.org/10.1039/c8cs00457a
- Y. Lin, J. Ren, X. Qu, Nano-gold as artificial enzymes: hidden talents. Adv. Mater. 26, 4200–4217 (2014). https://doi.org/10.1002/adma.201400238
- Q. Ma, Y. Liu, H. Zhu, L. Zhang, X. Liao, Nanozymes in tumor theranostics. Front Oncol. 11, 666017 (2021). https://doi.org/10.3389/fonc.2021.666017
- S. Dong, Y. Dong, B. Liu, J. Liu, S. Liu et al., Guiding transition metal-doped hollow cerium tandem nanozymes with elaborately regulated multi-enzymatic activities for intensive chemodynamic therapy. Adv. Mater. 34, e2107054 (2022). https://doi.org/10.1002/adma.202107054
- R.K. Sindhu, A. Najda, P. Kaur, M. Shah, H. Singh et al., Potentiality of nanoenzymes for cancer treatment and other diseases: current status and future challenges. Materials 14, 5965 (2021). https://doi.org/10.3390/ma14205965
- X. Zhang, X. Chen, Y. Zhao, Nanozymes: versatile platforms for cancer diagnosis and therapy. Nanomicro Lett. 14, 95 (2022). https://doi.org/10.1007/s40820-022-00828-2
- P. Wang, T. Wang, J. Hong, X. Yan, M. Liang, Nanozymes: a new disease imaging strategy. Front. Bioeng. Biotechnol. 8, 15 (2020). https://doi.org/10.3389/fbioe.2020.00015
- M. Liang, X. Yan, Nanozymes: from new concepts, mechanisms, and standards to applications. Acc. Chem. Res. 52, 2190–2200 (2019). https://doi.org/10.1021/acs.accounts.9b00140
- X. Ren, D. Chen, Y. Wang, H. Li, Y. Zhang et al., Nanozymes-recent development and biomedical applications. J. Nanobiotechnol. 20, 92 (2022). https://doi.org/10.1186/s12951-022-01295-y
- Y. Zhang, Y. Jin, H. Cui, X. Yan, K. Fan, Nanozyme-based catalytic theranostics. RSC Adv. 10, 10–20 (2020). https://doi.org/10.1039/C9RA09021E
- H. Ge, J. Du, J. Zheng, N. Xu, Q. Yao et al., Effective treatment of cisplatin-resistant ovarian tumors with a MoS2-based sonosensitizer and nanoenzyme capable of reversing the resistant-microenvironment and enhancing ferroptosis and apoptosis. Chem. Eng. J. 446, 137040 (2022). https://doi.org/10.1016/j.cej.2022.137040
- Y. Liu, W. Zhen, Y. Wang, J. Liu, L. Jin et al., One-dimensional Fe2 P acts as a Fenton agent in response to NIR II light and ultrasound for deep tumor synergetic theranostics. Angew. Chem. Int. Ed. 58, 2407–2412 (2019). https://doi.org/10.1002/anie.201813702
- W.-P. Li, C.-H. Su, Y.-C. Chang, Y.-J. Lin, C.-S. Yeh, Ultrasound-induced reactive oxygen species mediated therapy and imaging using a Fenton reaction activable polymersome. ACS Nano 10, 2017–2027 (2016). https://doi.org/10.1021/acsnano.5b06175
- L. Zhang, S.-S. Wan, C.-X. Li, L. Xu, H. Cheng et al., An adenosine triphosphate-responsive autocatalytic Fenton nanop for tumor ablation with self-supplied H2O2 and acceleration of Fe(III)/Fe(II) conversion. Nano Lett. 18, 7609–7618 (2018). https://doi.org/10.1021/acs.nanolett.8b03178
- L. Xing, X.-Y. Liu, T.-J. Zhou, X. Wan, Y. Wang et al., Photothermal nanozyme-ignited Fenton reaction-independent ferroptosis for breast cancer therapy. J. Control. Release 339, 14–26 (2021). https://doi.org/10.1016/j.jconrel.2021.09.019
- G. Zhang, N. Li, Y. Qi, Q. Zhao, J. Zhan et al., Synergistic ferroptosis-gemcitabine chemotherapy of the gemcitabine loaded carbonaceous nanozymes to enhance the treatment and magnetic resonance imaging monitoring of pancreatic cancer. Acta Biomater. 142, 284–297 (2022). https://doi.org/10.1016/j.actbio.2022.02.006
- M.J. Ko, S. Min, H. Hong, W. Yoo, J. Joo et al., Magnetic nanops for ferroptosis cancer therapy with diagnostic imaging. Bioact. Mater. 32, 66–97 (2023). https://doi.org/10.1016/j.bioactmat.2023.09.015
- H. Gavilán, S.K. Avugadda, T. Fernández-Cabada, N. Soni, M. Cassani et al., Magnetic nanops and clusters for magnetic hyperthermia: optimizing their heat performance and developing combinatorial therapies to tackle cancer. Chem. Soc. Rev. 50, 11614–11667 (2021). https://doi.org/10.1039/d1cs00427a
- S. Dutta, S. Noh, R.S. Gual, X. Chen, S. Pané et al., Recent developments in metallic degradable micromotors for biomedical and environmental remediation applications. Nano-Micro Lett. 16, 41 (2023). https://doi.org/10.1007/s40820-023-01259-3
- L. Yang, K.D. Patel, C. Rathnam, R. Thangam, Y. Hou et al., Harnessing the therapeutic potential of extracellular vesicles for biomedical applications using multifunctional magnetic nanomaterials. Small 18, e2104783 (2022). https://doi.org/10.1002/smll.202104783
- X. Zhao, K. Guo, K. Zhang, S. Duan, M. Chen et al., Orchestrated yolk-shell nanohybrids regulate macrophage polarization and dendritic cell maturation for oncotherapy with augmented antitumor immunity. Adv. Mater. 34, e2108263 (2022). https://doi.org/10.1002/adma.202108263
- J. Zhang, K. Zhou, J. Lin, X. Yao, D. Ju et al., Ferroptosis-enhanced chemotherapy for triple-negative breast cancer with magnetic composite nanops. Biomaterials 303, 122395 (2023). https://doi.org/10.1016/j.biomaterials.2023.122395
- Z.-J. Zhang, Z.-T. Liu, Y.-P. Huang, W. Nguyen, Y.-X. Wang et al., Magnetic resonance and fluorescence imaging superparamagnetic nanops induce apoptosis and ferroptosis through photodynamic therapy to treat colorectal cancer. Mater. Today Phys. 36, 101150 (2023). https://doi.org/10.1016/j.mtphys.2023.101150
- Z.-H. Li, Y. Chen, X. Zeng, X.-Z. Zhang, Ultra-small FePt/siRNA loaded mesoporous silica nanoplatform to deplete cysteine for enhanced ferroptosis in breast tumor therapy. Nano Today 38, 101150 (2021). https://doi.org/10.1016/j.nantod.2021.101150
- J. Yang, L. Ding, L. Yu, Y. Wang, M. Ge et al., Nanomedicine enables autophagy-enhanced cancer-cell ferroptosis. Sci. Bull. 66, 464–477 (2021). https://doi.org/10.1016/j.scib.2020.10.021
- S. Liang, X. Deng, P.-A. Ma, Z. Cheng, J. Lin, Recent advances in nanomaterial-assisted combinational sonodynamic cancer therapy. Adv. Mater. 32, e2003214 (2020). https://doi.org/10.1002/adma.202003214
- S. Liu, K. Dou, B. Liu, M. Pang, P.-A. Ma et al., Construction of multiform hollow-structured covalent organic frameworks via a facile and universal strategy for enhanced sonodynamic cancer therapy. Angew. Chem. Int. Ed. 62, e202301831 (2023). https://doi.org/10.1002/anie.202301831
- Y. Dong, S. Dong, B. Liu, C. Yu, J. Liu et al., 2D piezoelectric Bi2MoO6 nanoribbons for GSH-enhanced sonodynamic therapy. Adv. Mater. 33, 2106838 (2021). https://doi.org/10.1002/adma.202106838
- A.P. McHale, J.F. Callan, N. Nomikou, C. Fowley, B. Callan, Sonodynamic therapy: concept, mechanism and application to cancer treatment. Adv Exp Med Biol 880, 429–450 (2016). https://doi.org/10.1007/978-3-319-22536-4_22
- D. Costley, C.M. Ewan, C. Fowley, A.P. McHale, J. Atchison et al., Treating cancer with sonodynamic therapy: a review. Int. J. Hyperthermia 31, 107–117 (2015). https://doi.org/10.3109/02656736.2014.992484
- S. Dong, Y. Dong, Z. Zhao, J. Liu, S. Liu et al., “electron transport chain interference” strategy of amplified mild-photothermal therapy and defect-engineered multi-enzymatic activities for synergistic tumor-personalized suppression. J. Am. Chem. Soc. 145, 9488–9507 (2023). https://doi.org/10.1021/jacs.2c09608
- J. Yu, F. Zhu, Y. Yang, P. Zhang, Y. Zheng et al., Ultrasmall iron-doped zinc oxide nanops for ferroptosis assisted sono-chemodynamic cancer therapy. Colloids Surf. B Biointerfaces 232, 113606 (2023). https://doi.org/10.1016/j.colsurfb.2023.113606
- L. Zhou, C. Dong, L. Ding, W. Feng, L. Yu et al., Targeting ferroptosis synergistically sensitizes apoptotic sonodynamic anti-tumor nanotherapy. Nano Today 39, 101212 (2021). https://doi.org/10.1016/j.nantod.2021.101212
- Y. Cao, H.-Y. Huang, L.-Q. Chen, H.-H. Du, J.-H. Cui et al., Enhanced lysosomal escape of pH-responsive polyethylenimine-betaine functionalized carbon nanotube for the codelivery of survivin small interfering RNA and doxorubicin. ACS Appl. Mater. Interfaces 11, 9763–9776 (2019). https://doi.org/10.1021/acsami.8b20810
- P. Norouzi, H. Motasadizadeh, F. Atyabi, R. Dinarvand, M. Gholami et al., Combination therapy of breast cancer by codelivery of doxorubicin and survivin siRNA using polyethylenimine modified silk fibroin nanops. ACS Biomater. Sci. Eng. 7, 1074–1087 (2021). https://doi.org/10.1021/acsbiomaterials.0c01511
- J. Chen, Y. Wang, L. Han, R. Wang, C. Gong et al., A ferroptosis-inducing biomimetic nanocomposite for the treatment of drug-resistant prostate cancer. Mater. Today Bio 17, 100484 (2022). https://doi.org/10.1016/j.mtbio.2022.100484
- Z. Li, J. Bu, X. Zhu, H. Zhou, K. Ren et al., Anti-tumor immunity and ferroptosis of hepatocellular carcinoma are enhanced by combined therapy of sorafenib and delivering modified GO-based PD-L1 siRNAs. Biomater. Adv. 136, 212761 (2022). https://doi.org/10.1016/j.bioadv.2022.212761
- F. Wei, S. Kuang, T.W. Rees, X. Liao, J. Liu et al., Ruthenium(II) complexes coordinated to graphitic carbon nitride: oxygen self-sufficient photosensitizers which produce multiple ROS for photodynamic therapy in hypoxia. Biomaterials 276, 121064 (2021). https://doi.org/10.1016/j.biomaterials.2021.121064
- A.E. O’Connor, W.M. Gallagher, A.T. Byrne, Porphyrin and nonporphyrin photosensitizers in oncology: preclinical and clinical advances in photodynamic therapy. Photochem. Photobiol. 85, 1053–1074 (2009). https://doi.org/10.1111/j.1751-1097.2009.00585.x
- A. Sneider, R. Jadia, B. Piel, D. VanDyke, C. Tsiros et al., Engineering remotely triggered liposomes to target triple negative breast cancer. Oncomedicine 2, 1–13 (2017). https://doi.org/10.7150/oncm.17406
- M. Li, J. Sun, W. Zhang, Y. Zhao, S. Zhang et al., Drug delivery systems based on CD44-targeted glycosaminoglycans for cancer therapy. Carbohydr. Polym. 251, 117103 (2021). https://doi.org/10.1016/j.carbpol.2020.117103
- W. Tang, Z. Zhen, M. Wang, H. Wang, Y.-J. Chuang et al., Red blood cell-facilitated photodynamic therapy for cancer treatment. Adv. Funct. Mater. 26, 1757–1768 (2016). https://doi.org/10.1002/adfm.201504803
- F. Wei, J. Karges, J. Shen, L. Xie, K. Xiong et al., A mitochondria-localized oxygen self-sufficient two-photon nano-photosensitizer for ferroptosis-boosted photodynamic therapy under hypoxia. Nano Today 44, 101509 (2022). https://doi.org/10.1016/j.nantod.2022.101509
- Z. Dong, L. Feng, Y. Hao, Q. Li, M. Chen et al., Synthesis of CaCO3-based nanomedicine for enhanced sonodynamic therapy via amplification of tumor oxidative stress. Chem 6, 1391–1407 (2020). https://doi.org/10.1016/j.chempr.2020.02.020
- X. Jing, Y. Xu, D. Liu, Y. Wu, N. Zhou et al., Intelligent nanoflowers: a full tumor microenvironment-responsive multimodal cancer theranostic nanoplatform. Nanoscale 11, 15508–15518 (2019). https://doi.org/10.1039/c9nr04768a
- Z. Dong, L. Feng, Y. Chao, Y. Hao, M. Chen et al., Amplification of tumor oxidative stresses with liposomal Fenton catalyst and glutathione inhibitor for enhanced cancer chemotherapy and radiotherapy. Nano Lett. 19, 805–815 (2019). https://doi.org/10.1021/acs.nanolett.8b03905
- S. Luo, D. Ma, R. Wei, W. Yao, X. Pang et al., A tumor microenvironment responsive nanoplatform with oxidative stress amplification for effective MRI-based visual tumor ferroptosis. Acta Biomater. 138, 518–527 (2022). https://doi.org/10.1016/j.actbio.2021.11.007
- L. Zhou, F. Wei, J. Xiang, H. Li, C. Li et al., Enhancing the ROS generation ability of a rhodamine-decorated iridium(iii) complex by ligand regulation for endoplasmic reticulum-targeted photodynamic therapy. Chem. Sci. 11, 12212–12220 (2020). https://doi.org/10.1039/D0SC04751A
- Y. Yu, H. Jia, Y. Liu, L. Zhang, G. Feng et al., Recent progress in type I aggregation-induced emission photosensitizers for photodynamic therapy. Molecules 28, 332 (2022). https://doi.org/10.3390/molecules28010332
- X. Li, J.F. Lovell, J. Yoon, X. Chen, Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat. Rev. Clin. Oncol. 17, 657–674 (2020). https://doi.org/10.1038/s41571-020-0410-2
- S. Xu, Y. Yuan, X. Cai, C.-J. Zhang, F. Hu et al., Tuning the singlet-triplet energy gap: a unique approach to efficient photosensitizers with aggregation-induced emission (AIE) characteristics. Chem. Sci. 6, 5824–5830 (2015). https://doi.org/10.1039/c5sc01733e
- J.F. Lovell, T.W.B. Liu, J. Chen, G. Zheng, Activatable photosensitizers for imaging and therapy. Chem. Rev. 110, 2839–2857 (2010). https://doi.org/10.1021/cr900236h
- Z. Zhao, H. Zhang, J.W.Y. Lam, B.Z. Tang, Aggregation-induced emission: new vistas at the aggregate level. Angew. Chem. Int. Ed. 59, 9888–9907 (2020). https://doi.org/10.1002/anie.201916729
- R. Jiang, J. Dai, X. Dong, Q. Wang, Z. Meng et al., Improving image-guided surgical and immunological tumor treatment efficacy by photothermal and photodynamic therapies based on a multifunctional NIR AIEgen. Adv. Mater. 33, e2101158 (2021). https://doi.org/10.1002/adma.202101158
- B. Wang, L. Wang, H. Wu, X. Liu, J. Zhu et al., The commercial antibiotics with inherent AIE feature: in situ visualization of antibiotic metabolism and specifically differentiation of bacterial species and broad-spectrum therapy. Bioact. Mater. 23, 223–233 (2022). https://doi.org/10.1016/j.bioactmat.2022.11.002
- S. Liu, Y. Li, R.T.K. Kwok, J.W.Y. Lam, B.Z. Tang, Structural and process controls of AIEgens for NIR-II theranostics. Chem. Sci. 12, 3427–3436 (2020). https://doi.org/10.1039/d0sc02911d
- L. Liu, H. He, Z. Luo, H. Zhou, R. Liang et al., In situ photocatalyzed oxygen generation with photosynthetic bacteria to enable robust immunogenic photodynamic therapy in triple-negative breast cancer. Adv. Funct. Mater. 30, 1910176 (2020). https://doi.org/10.1002/adfm.201910176
- X. Yu, Y.-C. Zhang, X. Yang, Z. Huang, T. Zhang et al., Bonsai-inspired AIE nanohybrid photosensitizer based on vermiculite nanosheets for ferroptosis-assisted oxygen self-sufficient photodynamic cancer therapy. Nano Today 44, 101477 (2022). https://doi.org/10.1016/j.nantod.2022.101477
- W. Wang, J. Cai, J. Wen, X. Li, Y. Yu et al., Boosting ferroptosis via abplatin(iv) for treatment of platinum-resistant recurrent ovarian cancer. Nano Today 44, 101459 (2022). https://doi.org/10.1016/j.nantod.2022.101459
- Y. Han, Z. Dong, C. Wang, Q. Li, Y. Hao et al., Ferrous ions doped calcium carbonate nanops potentiate chemotherapy by inducing ferroptosis. J. Control. Release 348, 346–356 (2022). https://doi.org/10.1016/j.jconrel.2022.06.002
- Y. Liu, J. Wu, Y. Jin, W. Zhen, Y. Wang et al., Copper(I) phosphide nanocrystals for in situ self-generation magnetic resonance imaging-guided photothermal-enhanced chemodynamic synergetic therapy resisting deep-seated tumor. Adv. Funct. Mater. 29, 1904678 (2019). https://doi.org/10.1002/adfm.201904678
- A. Bagheri, H. Arandiyan, C. Boyer, M. Lim, Lanthanide-doped upconversion nanops: emerging intelligent light-activated drug delivery systems. Adv. Sci. 3, 1500437 (2016). https://doi.org/10.1002/advs.201500437
- W. Zeng, X. Wu, T. Chen, S. Sun, Z. Shi et al., Renal-clearable ultrasmall polypyrrole nanops with size-regulated property for second near-infrared light-mediated photothermal therapy. Adv. Funct. Mater. 31, 2008362 (2021). https://doi.org/10.1002/adfm.202008362
- M. Yu, J. Yu, Y. Yi, T. Chen, L. Yu et al., Oxidative stress-amplified nanomedicine for intensified ferroptosis-apoptosis combined tumor therapy. J. Control. Release 347, 104–114 (2022). https://doi.org/10.1016/j.jconrel.2022.04.047
- X. Zhang, S. Yang, Q. Wang, W. Ye, S. Liu et al., Tailored theranostic nanops cause efficient ferroptosis in head and neck squamous cell carcinoma through a reactive oxygen species “butterfly effect.” Chem. Eng. J. 423, 130083 (2021). https://doi.org/10.1016/j.cej.2021.130083
- C. Chen, W. Du, W. Jing, P. Sun, C. Shi et al., Leveraging tumor cell ferroptosis for colorectal cancer treatment via nanoelicitor-activated tumoricidal immunity. Chem. Eng. J. 430, 132983 (2022). https://doi.org/10.1016/j.cej.2021.132983
- Y. Dai, Z. Yang, S. Cheng, Z. Wang, R. Zhang et al., Toxic reactive oxygen species enhanced synergistic combination therapy by self-assembled metal-phenolic network nanops. Adv. Mater. 30, 1704877 (2018). https://doi.org/10.1002/adma.201704877
- P. Li, M. Gao, Z. Hu, T. Xu, J. Chen et al., Synergistic ferroptosis and macrophage re-polarization using engineering exosome-mimic M1 nanovesicles for cancer metastasis suppression. Chem. Eng. J. 409, 128217 (2021). https://doi.org/10.1016/j.cej.2020.128217
- J. Yang, S. Ma, R. Xu, Y. Wei, J. Zhang et al., Smart biomimetic metal organic frameworks based on ROS-ferroptosis-glycolysis regulation for enhanced tumor chemo-immunotherapy. J. Control. Release 334, 21–33 (2021). https://doi.org/10.1016/j.jconrel.2021.04.013
- A. Ghoochani, E.-C. Hsu, M. Aslan, M.A. Rice, H.M. Nguyen et al., Ferroptosis inducers are a novel therapeutic approach for advanced prostate cancer. Cancer Res. 81, 1583–1594 (2021). https://doi.org/10.1158/0008-5472.CAN-20-3477
- D. Tang, X. Chen, R. Kang, G. Kroemer, Ferroptosis: molecular mechanisms and health implications. Cell Res. 31, 107–125 (2021). https://doi.org/10.1038/s41422-020-00441-1
- E.H. Kim, D. Shin, J. Lee, A.R. Jung, J.L. Roh, CISD2 inhibition overcomes resistance to sulfasalazine-induced ferroptotic cell death in head and neck cancer. Cancer Lett. 432, 180–190 (2018). https://doi.org/10.1016/j.canlet.2018.06.018
- D. Li, Y. Li, The interaction between ferroptosis and lipid metabolism in cancer. Signal Transduct. Target. Ther. 5, 108 (2020). https://doi.org/10.1038/s41392-020-00216-5
- X. Zhang, K. Yu, L. Ma, Z. Qian, X. Tian et al., Endogenous glutamate determines ferroptosis sensitivity via ADCY10-dependent YAP suppression in lung adenocarcinoma. Theranostics 11, 5650–5674 (2021). https://doi.org/10.7150/thno.55482
- S. Park, J. Oh, M. Kim, E.-J. Jin, Bromelain effectively suppresses Kras-mutant colorectal cancer by stimulating ferroptosis. Anim. Cells Syst. 22, 334–340 (2018). https://doi.org/10.1080/19768354.2018.1512521
- Y. Sun, Y. He, J. Tong, D. Liu, H. Zhang et al., All-trans retinoic acid inhibits the malignant behaviors of hepatocarcinoma cells by regulating ferroptosis. Genes Dis. 9, 1742–1756 (2022). https://doi.org/10.1016/j.gendis.2022.04.011
- L. Sun, H. Wang, D. Xu, S. Yu, L. Zhang et al., Lapatinib induces mitochondrial dysfunction to enhance oxidative stress and ferroptosis in doxorubicin-induced cardiomyocytes via inhibition of PI3K/AKT signaling pathway. Bioengineered 13, 48–60 (2022). https://doi.org/10.1080/21655979.2021.2004980
- N. Eling, L. Reuter, J. Hazin, A. Hamacher-Brady, N.R. Brady, Identification of artesunate as a specific activator of ferroptosis in pancreatic cancer cells. Oncoscience 2, 517–532 (2015). https://doi.org/10.18632/oncoscience.160
- K.S. Kim, B. Choi, H. Choi, M.J. Ko, D.H. Kim et al., Enhanced natural killer cell anti-tumor activity with nanops mediated ferroptosis and potential therapeutic application in prostate cancer. J. Nanobiotechnol. 20, 428 (2022). https://doi.org/10.1186/s12951-022-01635-y
- T. Kasukabe, Y. Honma, J. Okabe-Kado, Y. Higuchi, N. Kato et al., Combined treatment with cotylenin A and phenethyl isothiocyanate induces strong antitumor activity mainly through the induction of ferroptotic cell death in human pancreatic cancer cells. Oncol. Rep. 36, 968–976 (2016). https://doi.org/10.3892/or.2016.4867
- D. Wang, W. Fang, C. Huang, Z. Chen, T. Nie et al., MR imaging guided iron-based nanoenzyme for synergistic Ferroptosis-Starvation therapy in triple negative breast cancer. Smart Mater. Med. 3, 159–167 (2022). https://doi.org/10.1016/j.smaim.2021.12.008
References
D.-W. Zheng, Q. Lei, J.-Y. Zhu, J.-X. Fan, C.-X. Li et al., Switching apoptosis to ferroptosis: metal-organic network for high-efficiency anticancer therapy. Nano Lett. 17, 284–291 (2017). https://doi.org/10.1021/acs.nanolett.6b04060
M.J. Davis, B.H. Ha, E.C. Holman, R. Halaban, J. Schlessinger et al., RAC1P29S is a spontaneously activating cancer-associated GTPase. Proc. Natl. Acad. Sci. U.S.A. 110, 912–917 (2013). https://doi.org/10.1073/pnas.1220895110
S.J. Dixon, K.M. Lemberg, M.R. Lamprecht, R. Skouta, E.M. Zaitsev et al., Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012). https://doi.org/10.1016/j.cell.2012.03.042
C. Cerella, C. Grandjenette, M. Dicato, M. Diederich, Roles of apoptosis and cellular senescence in cancer and aging. Curr. Drug Targets 17, 405–415 (2016). https://doi.org/10.2174/1389450116666150202155915
A. Sheikh, N.A. Alhakamy, S. Md, P. Kesharwani, Recent progress of RGD modified liposomes as multistage rocket against cancer. Front. Pharmacol. 12, 803304 (2022). https://doi.org/10.3389/fphar.2021.803304
M. Ishibashi, H. Tamura, M. Sunakawa, A. Kondo-Onodera, N. Okuyama et al., Myeloma drug resistance induced by binding of myeloma B7–H1 (PD-L1) to PD-1. Cancer Immunol. Res. 4, 779–788 (2016). https://doi.org/10.1158/2326-6066.CIR-15-0296
Z. Su, Z. Yang, L. Xie, J.P. DeWitt, Y. Chen, Cancer therapy in the necroptosis era. Cell Death Differ. 23, 748–756 (2016). https://doi.org/10.1038/cdd.2016.8
W.S. Yang, B.R. Stockwell, Ferroptosis: death by lipid peroxidation. Trends Cell Biol. 26, 165–176 (2016). https://doi.org/10.1016/j.tcb.2015.10.014
C. Nathan, A. Cunningham-Bussel, Beyond oxidative stress: an immunologist’s guide to reactive oxygen species. Nat. Rev. Immunol. 13, 349–361 (2013). https://doi.org/10.1038/nri3423
W.S. Yang, R. SriRamaratnam, M.E. Welsch, K. Shimada, R. Skouta et al., Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014). https://doi.org/10.1016/j.cell.2013.12.010
L. Jiang, N. Kon, T. Li, S.-J. Wang, T. Su et al., Ferroptosis as a p53-mediated activity during tumour suppression. Nature 520, 57–62 (2015). https://doi.org/10.1038/nature14344
A. Manohar, J. Park, D.D. Geleta, C. Krishnamoorthi, R. Thangam et al., Synthesis and characterization of ZnO nanops for photocatalysis, antibacterial and cytotoxicity in kidney cancer (A498) cell lines. J. Alloys Compd. 874, 159868 (2021). https://doi.org/10.1016/j.jallcom.2021.159868
H. Deng, J. Zhang, Y. Yang, J. Yang, Y. Wei et al., Chemodynamic and photothermal combination therapy based on dual-modified metal-organic framework for inducing tumor ferroptosis/pyroptosis. ACS Appl. Mater. Interfaces 14, 24089–24101 (2022). https://doi.org/10.1021/acsami.2c00574
L. Yu, Y. Xu, Z. Pu, H. Kang, M. Li et al., Photocatalytic superoxide radical generator that induces pyroptosis in cancer cells. J. Am. Chem. Soc. 144, 11326–11337 (2022). https://doi.org/10.1021/jacs.2c03256
Y. Du, J. Yang, F. He, X. Zhao, J. Zhou et al., Revealing the mutually enhanced mechanism of necroptosis and immunotherapy induced by defect engineering and piezoelectric effect. Adv. Mater. 36, e2304322 (2024). https://doi.org/10.1002/adma.202304322
P. Zheng, B. Ding, G. Zhu, C. Li, J. Lin, Biodegradable Ca2+ nanomodulators activate pyroptosis through mitochondrial Ca2+ overload for cancer immunotherapy. Angew. Chem. Int. Ed. 61, e202204904 (2022). https://doi.org/10.1002/anie.202204904
S.-J. Wang, D. Li, Y. Ou, L. Jiang, Y. Chen et al., Acetylation is crucial for p53-mediated ferroptosis and tumor suppression. Cell Rep. 17, 366–373 (2016). https://doi.org/10.1016/j.celrep.2016.09.022
Y. Xie, W. Hou, X. Song, Y. Yu, J. Huang et al., Ferroptosis: process and function. Cell Death Differ. 23, 369–379 (2016). https://doi.org/10.1038/cdd.2015.158
S. Mukherjee, S. Mukherjee, M.A.S. Abourehab, A. Sahebkar, P. Kesharwani, Exploring dendrimer-based drug delivery systems and their potential applications in cancer immunotherapy. Eur. Polym. J. 177, 111471 (2022). https://doi.org/10.1016/j.eurpolymj.2022.111471
Y. Du, R. Zhang, J. Yang, S. Liu, J. Zhou et al., A “closed-loop” therapeutic strategy based on mutually reinforced ferroptosis and immunotherapy. Adv. Funct. Mater. 32, 2111784 (2022). https://doi.org/10.1002/adfm.202111784
K. Zhang, Z. Ma, S. Li, Y. Wu, J. Zhang et al., Disruption of dual homeostasis by a metal-organic framework nanoreactor for ferroptosis-based immunotherapy of tumor. Biomaterials 284, 121502 (2022). https://doi.org/10.1016/j.biomaterials.2022.121502
H. Tang, D. Chen, C. Li, C. Zheng, X. Wu et al., Dual GSH-exhausting sorafenib loaded manganese-silica nanodrugs for inducing the ferroptosis of hepatocellular carcinoma cells. Int. J. Pharm. 572, 118782 (2019). https://doi.org/10.1016/j.ijpharm.2019.118782
P. Kesharwani, R.K. Tekade, V. Gajbhiye, K. Jain, N.K. Jain, Cancer targeting potential of some ligand-anchored poly(propylene imine) dendrimers: a comparison. Nanomed. Nanotechnol. Biol. Med. 7, 295–304 (2011). https://doi.org/10.1016/j.nano.2010.10.010
A. Sheikh, S. Md, N.A. Alhakamy, P. Kesharwani, Recent development of aptamer conjugated chitosan nanops as cancer therapeutics. Int. J. Pharm. 620, 121751 (2022). https://doi.org/10.1016/j.ijpharm.2022.121751
A. Sheikh, P. Kesharwani, An insight into aptamer engineered dendrimer for cancer therapy. Eur. Polym. J. 159, 110746 (2021). https://doi.org/10.1016/j.eurpolymj.2021.110746
P. Kesharwani, A. Sheikh, M.A.S. Abourehab, R. Salve, V. Gajbhiye, A combinatorial delivery of survivin targeted siRNA using cancer selective nanops for triple negative breast cancer therapy. J. Drug Deliv. Sci. Technol. 80, 104164 (2023). https://doi.org/10.1016/j.jddst.2023.104164
P. Kesharwani, K. Jain, N.K. Jain, Dendrimer as nanocarrier for drug delivery. Prog. Polym. Sci. 39, 268–307 (2014). https://doi.org/10.1016/j.progpolymsci.2013.07.005
P. Kesharwani, V. Gajbhiye, N.K. Jain, A review of nanocarriers for the delivery of small interfering RNA. Biomaterials 33, 7138–7150 (2012). https://doi.org/10.1016/j.biomaterials.2012.06.068
P. Kesharwani, A.K. Iyer, Recent advances in dendrimer-based nanovectors for tumor-targeted drug and gene delivery. Drug Discov. Today 20, 536–547 (2015). https://doi.org/10.1016/j.drudis.2014.12.012
R.A. Bapat, T.V. Chaubal, C.P. Joshi, P.R. Bapat, H. Choudhury et al., An overview of application of silver nanops for biomaterials in dentistry. Mater. Sci. Eng. C Mater. Biol. Appl. 91, 881–898 (2018). https://doi.org/10.1016/j.msec.2018.05.069
P. Kesharwani, S. Banerjee, U. Gupta, M.C.I. Mohd Amin, S. Padhye et al., PAMAM dendrimers as promising nanocarriers for RNAi therapeutics. Mater. Today 18, 565–572 (2015). https://doi.org/10.1016/j.mattod.2015.06.003
A.K. Sharma, A. Gothwal, P. Kesharwani, H. Alsaab, A.K. Iyer et al., Dendrimer nanoarchitectures for cancer diagnosis and anticancer drug delivery. Drug Discov. Today 22, 314–326 (2017). https://doi.org/10.1016/j.drudis.2016.09.013
P. Kesharwani, B. Gorain, S.Y. Low, S.A. Tan, E.C.S. Ling et al., Nanotechnology based approaches for anti-diabetic drugs delivery. Diabetes Res. Clin. Pract. 136, 52–77 (2018). https://doi.org/10.1016/j.diabres.2017.11.018
P. Kesharwani, A. Gothwal, A.K. Iyer, K. Jain, M.K. Chourasia et al., Dendrimer nanohybrid carrier systems: an expanding horizon for targeted drug and gene delivery. Drug Discov. Today 23, 300–314 (2018). https://doi.org/10.1016/j.drudis.2017.06.009
P. Kesharwani, H. Choudhury, J.G. Meher, M. Pandey, B. Gorain, Dendrimer-entrapped gold nanops as promising nanocarriers for anticancer therapeutics and imaging. Prog. Mater. Sci. 103, 484–508 (2019). https://doi.org/10.1016/j.pmatsci.2019.03.003
P. Kesharwani, R. Ghanghoria, N.K. Jain, Carbon nanotube exploration in cancer cell lines. Drug Discov. Today 17, 1023–1030 (2012). https://doi.org/10.1016/j.drudis.2012.05.003
P. Kesharwani, V. Mishra, N.K. Jain, Validating the anticancer potential of carbon nanotube-based therapeutics through cell line testing. Drug Discov. Today 20, 1049–1060 (2015). https://doi.org/10.1016/j.drudis.2015.05.004
M. Yuan, M. Kermanian, T. Agarwal, Z. Yang, S. Yousefiasl et al., Defect engineering in biomedical sciences. Adv. Mater. 35, e2304176 (2023). https://doi.org/10.1002/adma.202304176
R. Jiang, X. Li, D. Hu, M. Zhu, D. Zhou et al., Rapamycin-reinforced ferroptosis assisted by a lysosome-controlled disintegratable micelle in autophagy-dependent/independent manners. Appl. Mater. Today 23, 101066 (2021). https://doi.org/10.1016/j.apmt.2021.101066
G. Zhu, H. Chi, M. Liu, Y. Yin, H. Diao et al., Multifunctional “ball-rod” Janus nanops boosting Fenton reaction for ferroptosis therapy of non-small cell lung cancer. J. Colloid Interface Sci. 621, 12–23 (2022). https://doi.org/10.1016/j.jcis.2022.04.021
H. Zheng, J. Jiang, S. Xu, W. Liu, Q. Xie et al., Nanop-induced ferroptosis: detection methods, mechanisms and applications. Nanoscale 13, 2266–2285 (2021). https://doi.org/10.1039/d0nr08478f
H. Yu, J. Yan, Z. Li, L. Yang, F. Ju et al., Recent trends in emerging strategies for ferroptosis-based cancer therapy. Nanoscale Adv. 5, 1271–1290 (2023). https://doi.org/10.1039/d2na00719c
C. Zhang, X. Liu, S. Jin, Y. Chen, R. Guo, Ferroptosis in cancer therapy: a novel approach to reversing drug resistance. Mol. Cancer 21, 47 (2022). https://doi.org/10.1186/s12943-022-01530-y
C. Qiao, H. Wang, Q. Guan, M. Wei, Z. Li, Ferroptosis-based nano delivery systems targeted therapy for colorectal cancer: insights and future perspectives. Asian J. Pharm. Sci. 17, 613–629 (2022). https://doi.org/10.1016/j.ajps.2022.09.002
K. Liu, L. Huang, S. Qi, S. Liu, W. Xie et al., Ferroptosis: the entanglement between traditional drugs and nanodrugs in tumor therapy. Adv. Healthc. Mater. 12, 2203085 (2023). https://doi.org/10.1002/adhm.202203085
G. Lei, L. Zhuang, B. Gan, Targeting ferroptosis as a vulnerability in cancer. Nat. Rev. Cancer 22, 381–396 (2022). https://doi.org/10.1038/s41568-022-00459-0
N. Kang, S. Son, S. Min, H. Hong, C. Kim et al., Stimuli-responsive ferroptosis for cancer therapy. Chem. Soc. Rev. 52, 3955–3972 (2023). https://doi.org/10.1039/d3cs00001j
X. Jiang, B.R. Stockwell, M. Conrad, Ferroptosis: mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 22, 266–282 (2021). https://doi.org/10.1038/s41580-020-00324-8
M.J. Ko, W. Yoo, S. Min, Y. Zhang, J. Joo et al., Photonic control of image-guided ferroptosis cancer nanomedicine. Coord. Chem. Rev. 500, 215532 (2024). https://doi.org/10.1016/J.CCR.2023.215532
M. Jung, C. Mertens, E. Tomat, B. Brüne, Iron as a central player and promising target in cancer progression. Int. J. Mol. Sci. 20, 273 (2019). https://doi.org/10.3390/ijms20020273
S.V. Torti, F.M. Torti, Iron and cancer: more ore to be mined. Nat. Rev. Cancer 13, 342–355 (2013). https://doi.org/10.1038/nrc3495
T.A. Rouault, N. Maio, Biogenesis and functions of mammalian iron-sulfur proteins in the regulation of iron homeostasis and pivotal metabolic pathways. J. Biol. Chem. 292, 12744–12753 (2017). https://doi.org/10.1074/jbc.R117.789537
A. Leftin, N. Ben-Chetrit, F. Klemm, J.A. Joyce, J.A. Koutcher, Iron imaging reveals tumor and metastasis macrophage hemosiderin deposits in breast cancer. PLoS ONE 12, e0184765 (2017). https://doi.org/10.1371/journal.pone.0184765
S. Dev, J.L. Babitt, Overview of iron metabolism in health and disease. Hemodial. Int. 21, S6–S20 (2017). https://doi.org/10.1111/hdi.12542
A.R. Bogdan, M. Miyazawa, K. Hashimoto, Y. Tsuji, Regulators of iron homeostasis: new players in metabolism, cell death, and disease. Trends Biochem. Sci. 41, 274–286 (2016). https://doi.org/10.1016/j.tibs.2015.11.012
N. Wilkinson, K. Pantopoulos, The IRP/IRE system in vivo: insights from mouse models. Front. Pharmacol. 5, 176 (2014). https://doi.org/10.3389/fphar.2014.00176
C. Raggi, E. Gammella, M. Correnti, P. Buratti, E. Forti et al., Dysregulation of iron metabolism in cholangiocarcinoma stem-like cells. Sci. Rep. 7, 17667 (2017). https://doi.org/10.1038/s41598-017-17804-1
Z. Rychtarcikova, S. Lettlova, V. Tomkova, V. Korenkova, L. Langerova et al., Tumor-initiating cells of breast and prostate origin show alterations in the expression of genes related to iron metabolism. Oncotarget 8, 6376–6398 (2017). https://doi.org/10.18632/oncotarget.14093
T.B. Toh, J.J. Lim, E.K. Chow, Epigenetics in cancer stem cells. Mol. Cancer 16, 29 (2017). https://doi.org/10.1186/s12943-017-0596-9
E.N. Wainwright, P. Scaffidi, Epigenetics and cancer stem cells: unleashing, hijacking, and restricting cellular plasticity. Trends Cancer 3, 372–386 (2017). https://doi.org/10.1016/j.trecan.2017.04.004
J.P.F. Angeli, M. Schneider, B. Proneth, Y.Y. Tyurina, V.A. Tyurin et al., Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat. Cell Biol. 16, 1180–1191 (2014). https://doi.org/10.1038/ncb3064
Y. Mou, J. Wang, J. Wu, D. He, C. Zhang et al., Ferroptosis, a new form of cell death: opportunities and challenges in cancer. J. Hematol. Oncol. 12, 34 (2019). https://doi.org/10.1186/s13045-019-0720-y
B. Lu, X.B. Chen, M.D. Ying, Q.J. He, J. Cao et al., The role of ferroptosis in cancer development and treatment response. Front. Pharmacol. 8, 992 (2018). https://doi.org/10.3389/fphar.2017.00992
Y. Chen, Z. Yao, P. Liu, Q. Hu, Y. Huang et al., A self-assembly nano-prodrug for triple-negative breast cancer combined treatment by ferroptosis therapy and chemotherapy. Acta Biomater. 159, 275–288 (2023). https://doi.org/10.1016/j.actbio.2023.01.050
K. Kazan, S. Kalaipandian, Ferroptosis: yet another way to die. Trends Plant Sci. 24, 479–481 (2019). https://doi.org/10.1016/j.tplants.2019.03.005
J.P. Friedmann Angeli, D.V. Krysko, M. Conrad, Ferroptosis at the crossroads of cancer-acquired drug resistance and immune evasion. Nat. Rev. Cancer 19, 405–414 (2019). https://doi.org/10.1038/s41568-019-0149-1
M. Conlon, S.J. Dixon, Ferroptosis-like death in plant cells. Mol. Cell. Oncol. 4, e1302906 (2017). https://doi.org/10.1080/23723556.2017.1302906
X. Lang, M.D. Green, W. Wang, J. Yu, J.E. Choi et al., Radiotherapy and immunotherapy promote tumoral lipid oxidation and ferroptosis via synergistic repression of SLC7A11. Cancer Discov. 9, 1673–1685 (2019). https://doi.org/10.1158/2159-8290.CD-19-0338
F. Kuang, J. Liu, D. Tang, R. Kang, Oxidative damage and antioxidant defense in ferroptosis. Front. Cell Dev. Biol. 8, 586578 (2020). https://doi.org/10.3389/fcell.2020.586578
F. Ursini, M. Maiorino, C. Gregolin, The selenoenzyme phospholipid hydroperoxide glutathione peroxidase. Biochim. Biophys. Acta BBA Gen. Subj. 839, 62–70 (1985). https://doi.org/10.1016/0304-4165(85)90182-5
W.S. Yang, K.J. Kim, M.M. Gaschler, M. Patel, M.S. Shchepinov et al., Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc. Natl. Acad. Sci. U.S.A. 113, E4966–E4975 (2016). https://doi.org/10.1073/pnas.1603244113
X. Sui, R. Zhang, S. Liu, T. Duan, L. Zhai et al., RSL3 drives ferroptosis through GPX4 inactivation and ROS production in colorectal cancer. Front. Pharmacol. 9, 1371 (2018). https://doi.org/10.3389/fphar.2018.01371
J. Yang, J. Mo, J. Dai, C. Ye, W. Cen et al., Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis. 12, 1079 (2021). https://doi.org/10.1038/s41419-021-04367-3
B. Han, Y. Liu, Q. Zhang, L. Liang, Propofol decreases cisplatin resistance of non-small cell lung cancer by inducing GPX4-mediated ferroptosis through the miR-744-5p/miR-615-3p axis. J. Proteomics 274, 104777 (2023). https://doi.org/10.1016/j.jprot.2022.104777
S. Cai, Z. Ding, X. Liu, J. Zeng, Trabectedin induces ferroptosis via regulation of HIF-1α/IRP1/TFR1 and Keap1/Nrf2/GPX4 axis in non-small cell lung cancer cells. Chem. Biol. Interact. 369, 110262 (2023). https://doi.org/10.1016/j.cbi.2022.110262
W. Zhang, B. Jiang, Y. Liu, L. Xu, M. Wan, Bufotalin induces ferroptosis in non-small cell lung cancer cells by facilitating the ubiquitination and degradation of GPX4. Free Radic. Biol. Med. 180, 75–84 (2022). https://doi.org/10.1016/j.freeradbiomed.2022.01.009
R. Yu, Y. Zhou, S. Shi, X. Wang, S. Huang et al., Icariside II induces ferroptosis in renal cell carcinoma cells by regulating the miR-324-3p/GPX4 axis. Phytomedicine 102, 154182 (2022). https://doi.org/10.1016/j.phymed.2022.154182
M. Hashemzaei, A.K. Barani, M. Iranshahi, R. Rezaee, K. Tsarouhas et al., Effects of resveratrol on carbon monoxide-induced cardiotoxicity in rats. Environ. Toxicol. Pharmacol. 46, 110–115 (2016). https://doi.org/10.1016/j.etap.2016.07.010
T.M. Bahr, R.D. Christensen, D.M. Ward, F. Meng, L.K. Jackson et al., Ferritin in serum and urine: a pilot study. Blood Cells Mol. Dis. 76, 59–62 (2019). https://doi.org/10.1016/j.bcmd.2019.02.001
Y. Qin, Y. Qiao, D. Wang, C. Tang, G. Yan, Ferritinophagy and ferroptosis in cardiovascular disease: mechanisms and potential applications. Biomed. Pharmacother. 141, 111872 (2021). https://doi.org/10.1016/j.biopha.2021.111872
K. Shimada, R. Skouta, A. Kaplan, W.S. Yang, M. Hayano et al., Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat. Chem. Biol. 12, 497–503 (2016). https://doi.org/10.1038/nchembio.2079
Z. Wu, Y. Geng, X. Lu, Y. Shi, G. Wu et al., Chaperone-mediated autophagy is involved in the execution of ferroptosis. Proc. Natl. Acad. Sci. U.S.A. 116, 2996–3005 (2019). https://doi.org/10.1073/pnas.1819728116
M. Conrad, D.A. Pratt, The chemical basis of ferroptosis. Nat. Chem. Biol. 15, 1137–1147 (2019). https://doi.org/10.1038/s41589-019-0408-1
B.R. Stockwell, Ferroptosis turns 10: emerging mechanisms, physiological functions, and therapeutic applications. Cell 185, 2401–2421 (2022). https://doi.org/10.1016/j.cell.2022.06.003
Y. Zou, H. Li, E.T. Graham, A.A. Deik, J.K. Eaton et al., Cytochrome P450 oxidoreductase contributes tophospholipid peroxidation in ferroptosis. Nat. Chem. Biol. 16, 302–309 (2020). https://doi.org/10.1038/s41589-020-0472-6
V.E. Kagan, G. Mao, F. Qu, J.P.F. Angeli, S. Doll et al., Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat. Chem. Biol. 13, 81–90 (2017). https://doi.org/10.1038/nchembio.2238
R. Shintoku, Y. Takigawa, K. Yamada, C. Kubota, Y. Yoshimoto et al., Lipoxygenase-mediated generation of lipid peroxides enhances ferroptosis induced by erastin and RSL3. Cancer Sci. 108, 2187–2194 (2017). https://doi.org/10.1111/cas.13380
L. Tirinato, C. Liberale, S. Di Franco, P. Candeloro, A. Benfante et al., Lipid droplets: a new player in colorectal cancer stem cells unveiled by spectroscopic imaging. Stem Cells 33, 35–44 (2015). https://doi.org/10.1002/stem.1837
M. Visweswaran, F. Arfuso, S. Warrier, A. Dharmarajan, Aberrant lipid metabolism as an emerging therapeutic strategy to target cancer stem cells. Stem Cells 38, 6–14 (2020). https://doi.org/10.1002/stem.3101
B.J. Hershey, R. Vazzana, D.L. Joppi, K.M. Havas, Lipid droplets define a sub-population of breast cancer stem cells. J. Clin. Med. 9, 87 (2019). https://doi.org/10.3390/jcm9010087
S.J. Dixon, G.E. Winter, L.S. Musavi, E.D. Lee, B. Snijder et al., Human haploid cell genetics reveals roles for lipid metabolism genes in nonapoptotic cell death. ACS Chem. Biol. 10, 1604–1609 (2015). https://doi.org/10.1021/acschembio.5b00245
L. Zhao, Y. Peng, S. He, R. Li, Z. Wang et al., Apatinib induced ferroptosis by lipid peroxidation in gastric cancer. Gastric Cancer 24, 642–654 (2021). https://doi.org/10.1007/s10120-021-01159-8
J. Li, S. Huang, Q. Wang, D. Zhou, B. Zhao et al., Andrographolide promoted ferroptosis to repress the development of non-small cell lung cancer through activation of the mitochondrial dysfunction. Phytomedicine 109, 154601 (2023). https://doi.org/10.1016/j.phymed.2022.154601
F. Yang, Y. Xiao, J.-H. Ding, X. Jin, D. Ma et al., Ferroptosis heterogeneity in triple-negative breast cancer reveals an innovative immunotherapy combination strategy. Cell Metab. 35, 84-100.e8 (2023). https://doi.org/10.1016/j.cmet.2022.09.021
G.-Q. Chen, F.A. Benthani, J. Wu, D. Liang, Z.-X. Bian et al., Artemisinin compounds sensitize cancer cells to ferroptosis by regulating iron homeostasis. Cell Death Differ. 27, 242–254 (2020). https://doi.org/10.1038/s41418-019-0352-3
S. Huang, H. Le, G. Hong, G. Chen, F. Zhang et al., An all-in-one biomimetic iron-small interfering RNA nanoplatform induces ferroptosis for cancer therapy. Acta Biomater. 148, 244–257 (2022). https://doi.org/10.1016/j.actbio.2022.06.017
F. Zhang, F. Li, G.-H. Lu, W. Nie, L. Zhang et al., Engineering magnetosomes for ferroptosis/immunomodulation synergism in cancer. ACS Nano 13, 5662–5673 (2019). https://doi.org/10.1021/acsnano.9b00892
Z. Shen, T. Liu, Y. Li, J. Lau, Z. Yang et al., Fenton-reaction-acceleratable magnetic nanops for ferroptosis therapy of orthotopic brain tumors. ACS Nano 12, 11355–11365 (2018). https://doi.org/10.1021/acsnano.8b06201
Y. Zou, W.S. Henry, E.L. Ricq, E.T. Graham, V.V. Phadnis et al., Plasticity of ether lipids promotes ferroptosis susceptibility and evasion. Nature 585, 603–608 (2020). https://doi.org/10.1038/s41586-020-2732-8
Y.Y. Tyurina, A.A. Kapralov, V.A. Tyurin, G. Shurin, A.A. Amoscato et al., Redox phospholipidomics discovers pro-ferroptotic death signals in A375 melanoma cells in vitro and in vivo. Redox Biol. 61, 102650 (2023). https://doi.org/10.1016/j.redox.2023.102650
K.A. Jasim, A.J. Gesquiere, Ultrastable and biofunctionalizable conjugated polymer nanops with encapsulated iron for ferroptosis assisted chemodynamic therapy. Mol. Pharm. 16, 4852–4866 (2019). https://doi.org/10.1021/acs.molpharmaceut.9b00737
S. Koo, O.K. Park, J. Kim, S.I. Han, T.Y. Yoo et al., Enhanced chemodynamic therapy by Cu-Fe peroxide nanops: tumor microenvironment-mediated synergistic Fenton reaction. ACS Nano 16, 2535–2545 (2022). https://doi.org/10.1021/acsnano.1c09171
N. Wang, Q. Zeng, R. Zhang, D. Xing, T. Zhang, Eradication of solid tumors by chemodynamic theranostics with H2O2-catalyzed hydroxyl radical burst. Theranostics 11, 2334–2348 (2021). https://doi.org/10.7150/thno.49277
M. Zahiri, M. Falsafi, K. Lamei, K. Abnous, S.M. Taghdisi et al., Targeted biomimetic hollow mesoporous organosilica nanops for delivery of doxorubicin to colon adenocarcinoma: in vitro and in vivo evaluation. Microporous Mesoporous Mater. 335, 111841 (2022). https://doi.org/10.1016/j.micromeso.2022.111841
L. Huang, J. Zhu, W. Xiong, J. Feng, J. Yang et al., Tumor-generated reactive oxygen species storm for high-performance ferroptosis therapy. ACS Nano 17, 11492–11506 (2023). https://doi.org/10.1021/acsnano.3c01369
S. Bai, Y. Lan, S. Fu, H. Cheng, Z. Lu et al., Connecting calcium-based nanomaterials and cancer: from diagnosis to therapy. Nano-Micro Lett. 14, 145 (2022). https://doi.org/10.1007/s40820-022-00894-6
Z. Chen, J.-J. Yin, Y.-T. Zhou, Y. Zhang, L. Song et al., Dual enzyme-like activities of iron oxide nanops and their implication for diminishing cytotoxicity. ACS Nano 6, 4001–4012 (2012). https://doi.org/10.1021/nn300291r
L. Gao, J. Zhuang, L. Nie, J. Zhang, Y. Zhang et al., Intrinsic peroxidase-like activity of ferromagnetic nanops. Nat. Nanotechnol. 2, 577–583 (2007). https://doi.org/10.1038/nnano.2007.260
L.S. Li, E.A. Bey, Y. Dong, J. Meng, B. Patra et al., Modulating endogenous NQO1 levels identifies key regulatory mechanisms of action of β-lapachone for pancreatic cancer therapy. Clin. Cancer Res. 17, 275–285 (2011). https://doi.org/10.1158/1078-0432.CCR-10-1983
H. Yeo, C.A. Lyssiotis, Y. Zhang, H. Ying, J.M. Asara et al., FoxO3 coordinates metabolic pathways to maintain redox balance in neural stem cells. EMBO J. 32, 2589–2602 (2013). https://doi.org/10.1038/emboj.2013.186
Q. Chen, J. Zhou, Z. Chen, Q. Luo, J. Xu et al., Tumor-specific expansion of oxidative stress by glutathione depletion and use of a Fenton nanoagent for enhanced chemodynamic therapy. ACS Appl. Mater. Interfaces 11, 30551–30565 (2019). https://doi.org/10.1021/acsami.9b09323
W. Li, S. Yin, Y. Shen, H. Li, L. Yuan et al., Molecular engineering of pH-responsive NIR oxazine assemblies for evoking tumor ferroptosis via triggering lysosomal dysfunction. J. Am. Chem. Soc. 145, 3736–3747 (2023). https://doi.org/10.1021/jacs.2c13222
Y. Xie, S. Zhu, X. Song, X. Sun, Y. Fan et al., The tumor suppressor p53 limits ferroptosis by blocking DPP4 activity. Cell Rep. 20, 1692–1704 (2017). https://doi.org/10.1016/j.celrep.2017.07.055
H. Chen, F. Peng, J. Xu, G. Wang, Y. Zhao, Increased expression of GPX4 promotes the tumorigenesis of thyroid cancer by inhibiting ferroptosis and predicts poor clinical outcomes. Aging 15, 230–245 (2023). https://doi.org/10.18632/aging.204473
X. Wei, X. Yi, X.-H. Zhu, D.-S. Jiang, Posttranslational modifications in ferroptosis. Oxid. Med. Cell. Longev. 2020, 8832043 (2020). https://doi.org/10.1155/2020/8832043
C. Gai, C. Liu, X. Wu, M. Yu, J. Zheng et al., MT1DP loaded by folate-modified liposomes sensitizes erastin-induced ferroptosis via regulating miR-365a-3p/NRF2 axis in non-small cell lung cancer cells. Cell Death Dis. 11, 751 (2020). https://doi.org/10.1038/s41419-020-02939-3
A. Brown, S. Kumar, P.B. Tchounwou, Cisplatin-based chemotherapy of human cancers. J. Cancer Sci. Ther. 11, 97 (2019). https://doi.org/10.4172/1948-5956.1000592
G. Bronte, D. Andreis, S. Bravaccini, R. Maltoni, L. Cecconetto et al., Sorafenib for the treatment of breast cancer. Expert Opin. Pharmacother. 18, 621–630 (2017). https://doi.org/10.1080/14656566.2017.1309024
C. Bae, H. Kim, Y.M. Kook, C. Lee, C. Kim et al., Induction of ferroptosis using functionalized iron-based nanops for anti-cancer therapy. Mater. Today Bio 17, 100457 (2022). https://doi.org/10.1016/j.mtbio.2022.100457
M. Huo, L. Wang, Y. Chen, J. Shi, Tumor-selective catalytic nanomedicine by nanocatalyst delivery. Nat. Commun. 8, 357 (2017). https://doi.org/10.1038/s41467-017-00424-8
L. Wang, M. Huo, Y. Chen, J. Shi, Iron-engineered mesoporous silica nanocatalyst with biodegradable and catalytic framework for tumor-specific therapy. Biomaterials 163, 1–13 (2018). https://doi.org/10.1016/j.biomaterials.2018.02.018
X. Qian, J. Zhang, Z. Gu, Y. Chen, Nanocatalysts-augmented Fenton chemical reaction for nanocatalytic tumor therapy. Biomaterials 211, 1–13 (2019). https://doi.org/10.1016/j.biomaterials.2019.04.023
Z. Zhou, J. Song, R. Tian, Z. Yang, G. Yu et al., Activatable singlet oxygen generation from lipid hydroperoxide nanops for cancer therapy. Angew. Chem. Int. Ed. 56, 6492–6496 (2017). https://doi.org/10.1002/anie.201701181
A. Sheikh, S. Md, P. Kesharwani, Aptamer grafted nanop as targeted therapeutic tool for the treatment of breast cancer. Biomed. Pharmacother. 146, 112530 (2022). https://doi.org/10.1016/j.biopha.2021.112530
M. Fatima, A. Sheikh, N. Hasan, A. Sahebkar, Y. Riadi et al., Folic acid conjugated poly(amidoamine) dendrimer as a smart nanocarriers for tracing, imaging, and treating cancers over-expressing folate receptors. Eur. Polym. J. 170, 111156 (2022). https://doi.org/10.1016/j.eurpolymj.2022.111156
Y. Du, J. Zhou, F. He, P. Zang, H. Gong et al., A bright future: advanced nanotechnology-assisted microwave therapy. Nano Today 52, 101963 (2023). https://doi.org/10.1016/j.nantod.2023.101963
A.J. Howarth, Y. Liu, P. Li, Z. Li, T.C. Wang et al., Chemical, thermal and mechanical stabilities of metal–organic frameworks. Nat. Rev. Mater. 1, 15018 (2016). https://doi.org/10.1038/natrevmats.2015.18
P. Venkatesan, N. Thirumalaivasan, H.-P. Yu, P.-S. Lai, S.-P. Wu, Redox stimuli delivery vehicle based on transferrin-capped MSNPs for targeted drug delivery in cancer therapy. ACS Appl. Bio Mater. 2, 1623–1633 (2019). https://doi.org/10.1021/acsabm.9b00036
K. Lu, T. Aung, N. Guo, R. Weichselbaum, W. Lin, Nanoscale metal-organic frameworks for therapeutic, imaging, and sensing applications. Adv. Mater. 30, e1707634 (2018). https://doi.org/10.1002/adma.201707634
L. Jiao, H.-L. Jiang, Metal-organic-framework-based single-atom catalysts for energy applications. Chem 5, 786–804 (2019). https://doi.org/10.1016/j.chempr.2018.12.011
M. Falsafi, M. Zahiri, A.S. Saljooghi, K. Abnous, S.M. Taghdisi et al., Aptamer targeted red blood cell membrane-coated porphyrinic copper-based MOF for guided photochemotherapy against metastatic breast cancer. Micropor Mesopor Mater. 325, 111337 (2021). https://doi.org/10.1016/j.micromeso.2021.111337
L.F. Ye, K.R. Chaudhary, F. Zandkarimi, A.D. Harken, C.J. Kinslow et al., Radiation-induced lipid peroxidation triggers ferroptosis and synergizes with ferroptosis inducers. ACS Chem. Biol. 15, 469–484 (2020). https://doi.org/10.1021/acschembio.9b00939
G. Lei, C. Mao, Y. Yan, L. Zhuang, B. Gan, Ferroptosis, radiotherapy, and combination therapeutic strategies. Protein Cell 12, 836–857 (2021). https://doi.org/10.1007/s13238-021-00841-y
G. Lei, Y. Zhang, T. Hong, X. Zhang, X. Liu et al., Ferroptosis as a mechanism to mediate p53 function in tumor radiosensitivity. Oncogene 40, 3533–3547 (2021). https://doi.org/10.1038/s41388-021-01790-w
G. Lei, Y. Zhang, P. Koppula, X. Liu, J. Zhang et al., The role of ferroptosis in ionizing radiation-induced cell death and tumor suppression. Cell Res. 30, 146–162 (2020). https://doi.org/10.1038/s41422-019-0263-3
Y. Wu, C. Yu, M. Luo, C. Cen, J. Qiu et al., Ferroptosis in cancer treatment: another way to Rome. Front. Oncol. 10, 571127 (2020). https://doi.org/10.3389/fonc.2020.571127
Y. Liang, C. Peng, N. Su, Q. Li, S. Chen et al., Tumor microenvironments self-activated cascade catalytic nanoscale metal organic frameworks as ferroptosis inducer for radiosensitization. Chem. Eng. J. 437, 135309 (2022). https://doi.org/10.1016/j.cej.2022.135309
B. Hassannia, P. Vandenabeele, T. Vanden Berghe, Targeting ferroptosis to iron out cancer. Cancer Cell 35, 830–849 (2019). https://doi.org/10.1016/j.ccell.2019.04.002
T. Liu, W. Liu, M. Zhang, W. Yu, F. Gao et al., Ferrous-supply-regeneration nanoengineering for cancer-cell-specific ferroptosis in combination with imaging-guided photodynamic therapy. ACS Nano 12, 12181–12192 (2018). https://doi.org/10.1021/acsnano.8b05860
M. Mu, Y. Wang, S. Zhao, X. Li, R. Fan et al., Engineering a pH/glutathione-responsive tea polyphenol nanodevice as an apoptosis/ferroptosis-inducing agent. ACS Appl. Bio Mater. 3, 4128–4138 (2020). https://doi.org/10.1021/acsabm.0c00225
W. Xu, T. Wang, J. Qian, J. Wang, G. Hou et al., Fe(II)-hydrazide coordinated all-active metal organic framework for photothermally enhanced tumor penetration and ferroptosis-apoptosis synergistic therapy. Chem. Eng. J. 437, 135311 (2022). https://doi.org/10.1016/j.cej.2022.135311
C. Liang, X. Zhang, M. Yang, X. Dong, Recent progress in ferroptosis inducers for cancer therapy. Adv. Mater. 31, e1904197 (2019). https://doi.org/10.1002/adma.201904197
H. He, L. Du, H. Guo, Y. An, L. Lu et al., Redox responsive metal organic framework nanops induces ferroptosis for cancer therapy. Small 16, e2001251 (2020). https://doi.org/10.1002/smll.202001251
Q. Guan, R. Guo, S. Huang, F. Zhang, J. Liu et al., Mesoporous polydopamine carrying sorafenib and SPIO nanops for MRI-guided ferroptosis cancer therapy. J. Control. Release 320, 392–403 (2020). https://doi.org/10.1016/j.jconrel.2020.01.048
X. Hu, R. Li, W. Wu, K. Fang, Z. Zhu et al., A Fe(III)-porphyrin-oxaliplatin(IV) nanoplatform for enhanced ferroptosis and combined therapy. J. Control. Release 348, 660–671 (2022). https://doi.org/10.1016/j.jconrel.2022.06.019
H. Peng, X. Zhang, P. Yang, J. Zhao, W. Zhang et al., Defect self-assembly of metal-organic framework triggers ferroptosis to overcome resistance. Bioact. Mater. 19, 1–11 (2021). https://doi.org/10.1016/j.bioactmat.2021.12.018
A. Jain, P. Kesharwani, N.K. Garg, A. Jain, S.A. Jain et al., Galactose engineered solid lipid nanops for targeted delivery of doxorubicin. Colloids Surf. B Biointerf 134, 47–58 (2015). https://doi.org/10.1016/j.colsurfb.2015.06.027
N. Soni, N. Soni, H. Pandey, R. Maheshwari, P. Kesharwani et al., Augmented delivery of gemcitabine in lung cancer cells exploring mannose anchored solid lipid nanops. J. Colloid Interface Sci. 481, 107–116 (2016). https://doi.org/10.1016/j.jcis.2016.07.020
A. Jain, N.K. Garg, A. Jain, P. Kesharwani, A.K. Jain et al., A synergistic approach of adapalene-loaded nanostructured lipid carriers, and vitamin C co-administration for treating acne. Drug Dev. Ind. Pharm. 42, 897–905 (2016). https://doi.org/10.3109/03639045.2015.1104343
A. Jain, G. Sharma, V. Kushwah, N.K. Garg, P. Kesharwani et al., Methotrexate and beta-carotene loaded-lipid polymer hybrid nanops: a preclinical study for breast cancer. Nanomedicine 12, 1851–1872 (2017). https://doi.org/10.2217/nnm-2017-0011
S. Md, S. Haque, T. Madheswaran, F. Zeeshan, V.S. Meka et al., Lipid based nanocarriers system for topical delivery of photosensitizers. Drug Discov. Today 22, 1274–1283 (2017). https://doi.org/10.1016/j.drudis.2017.04.010
N. Hasan, M. Imran, P. Kesharwani, K. Khanna, R. Karwasra et al., Intranasal delivery of Naloxone-loaded solid lipid nanops as a promising simple and non-invasive approach for the management of opioid overdose. Int. J. Pharm. 599, 120428 (2021). https://doi.org/10.1016/j.ijpharm.2021.120428
G.S. Bhagwat, R.B. Athawale, R.P. Gude, S. Md, N.A. Alhakamy et al., Formulation and development of transferrin targeted solid lipid nanops for breast cancer therapy. Front. Pharmacol. 11, 614290 (2020). https://doi.org/10.3389/fphar.2020.614290
A. Aziz, U. Rehman, A. Sheikh, M.A.S. Abourehab, P. Kesharwani, Lipid-based nanocarrier mediated CRISPR/Cas9 delivery for cancer therapy. J. Biomater. Sci. Polym. Ed. 34, 398–418 (2023). https://doi.org/10.1080/09205063.2022.2121592
F. Mohammadpour, H. Kamali, L. Gholami, A.P. McCloskey, P. Kesharwani et al., Solid lipid nanops: a promising tool for insulin delivery. Expert Opin. Drug Deliv. 19, 1577–1595 (2022). https://doi.org/10.1080/17425247.2022.2138328
D.C. Drummond, C.O. Noble, Z. Guo, K. Hong, J.W. Park et al., Development of a highly active nanoliposomal irinotecan using a novel intraliposomal stabilization strategy. Cancer Res. 66, 3271–3277 (2006). https://doi.org/10.1158/0008-5472.CAN-05-4007
C. Zylberberg, K. Gaskill, S. Pasley, S. Matosevic, Engineering liposomal nanops for targeted gene therapy. Gene Ther. 24, 441–452 (2017). https://doi.org/10.1038/gt.2017.41
X. Chen, Y. Zhang, C. Tang, C. Tian, Q. Sun et al., Co-delivery of paclitaxel and anti-survivin siRNA via redox-sensitive oligopeptide liposomes for the synergistic treatment of breast cancer and metastasis. Int. J. Pharm. 529, 102–115 (2017). https://doi.org/10.1016/j.ijpharm.2017.06.071
F. Persano, G. Gigli, S. Leporatti, Lipid-polymer hybrid nanops in cancer therapy: current overview and future directions. Nano Ex. 2, 012006 (2021). https://doi.org/10.1088/2632-959x/abeb4b
Z. Li, C. Wang, C. Dai, R. Hu, L. Ding et al., Engineering dual catalytic nanomedicine for autophagy-augmented and ferroptosis-involved cancer nanotherapy. Biomaterials 287, 121668 (2022). https://doi.org/10.1016/j.biomaterials.2022.121668
P. Ghasemiyeh, S. Mohammadi-Samani, Solid lipid nanops and nanostructured lipid carriers as novel drug delivery systems: applications, advantages and disadvantages. Res. Pharm. Sci. 13, 288–303 (2018). https://doi.org/10.4103/1735-5362.235156
H. Yuan, L.-L. Wang, Y.-Z. Du, J. You, F.-Q. Hu et al., Preparation and characteristics of nanostructured lipid carriers for control-releasing progesterone by melt-emulsification. Coll. Surf. B Biointerf. 60, 174–179 (2007). https://doi.org/10.1016/j.colsurfb.2007.06.011
E. Esposito, M. Drechsler, R. Cortesi, C. Nastruzzi, Encapsulation of cannabinoid drugs in nanostructured lipid carriers. Eur. J. Pharm. Biopharm. 102, 87–91 (2016). https://doi.org/10.1016/j.ejpb.2016.03.005
J. Zhang, J. Yang, T. Zuo, S. Ma, N. Xokrat et al., Heparanase-driven sequential released nanops for ferroptosis and tumor microenvironment modulations synergism in breast cancer therapy. Biomaterials 266, 120429 (2021). https://doi.org/10.1016/j.biomaterials.2020.120429
L. Zeng, B.H.J. Gowda, M.G. Ahmed, M.A.S. Abourehab, Z.S. Chen et al., Advancements in nanop-based treatment approaches for skin cancer therapy. Mol. Cancer 22, 10 (2023). https://doi.org/10.1186/s12943-022-01708-4
Z. Liu, N. Parveen, U. Rehman, A. Aziz, A. Sheikh et al., Unravelling the enigma of siRNA and aptamer mediated therapies against pancreatic cancer. Mol. Cancer 22, 8 (2023). https://doi.org/10.1186/s12943-022-01696-5
U. Rehman, M.A.S. Abourehab, A. Alexander, P. Kesharwani, Polymeric micelles assisted combinatorial therapy: is it new hope for pancreatic cancer? Eur. Polym. J. 184, 111784 (2023). https://doi.org/10.1016/j.eurpolymj.2022.111784
N. Parveen, M.A.S. Abourehab, R. Shukla, P.V. Thanikachalam, G.K. Jain et al., Immunoliposomes as an emerging nanocarrier for breast cancer therapy. Eur. Polym. J. 184, 111781 (2023). https://doi.org/10.1016/j.eurpolymj.2022.111781
A.K. Jain, S. Jain, M.A.S. Abourehab, P. Mehta, P. Kesharwani, An insight on topically applied formulations for management of various skin disorders. J. Biomater. Sci. Polym. Ed. 33, 2406–2432 (2022). https://doi.org/10.1080/09205063.2022.2103625
Y. Wang, T. Liu, X. Li, H. Sheng, X. Ma et al., Ferroptosis-inducing nanomedicine for cancer therapy. Front. Pharmacol. 12, 735965 (2021). https://doi.org/10.3389/fphar.2021.735965
J. Gao, T. Luo, J. Wang, Gene interfered-ferroptosis therapy for cancers. Nat. Commun. 12, 5311 (2021). https://doi.org/10.1038/s41467-021-25632-1
M. Cheng, B. Zhang, W. Cui, M.L. Gross, Laser-initiated radical trifluoromethylation of peptides and proteins: application to mass-spectrometry-based protein footprinting. Angew. Chem. Int. Ed. 56, 14007–14010 (2017). https://doi.org/10.1002/anie.201706697
H. Chen, J. Wen, Iron oxide nanops loaded with paclitaxel inhibits glioblastoma by enhancing autophagy-dependent ferroptosis pathway. Eur. J. Pharmacol. 921, 174860 (2022). https://doi.org/10.1016/j.ejphar.2022.174860
A. Ruiz-de-Angulo, M. Bilbao-Asensio, J. Cronin, S.J. Evans, M.J.D. Clift et al., Chemically programmed vaccines: iron catalysis in nanops enhances combination immunotherapy and immunotherapy-promoted tumor ferroptosis. iScience 23, 101499 (2020). https://doi.org/10.1016/j.isci.2020.101499
X. Zhu, Q. Chen, L. Xie, W. Chen, Y. Jiang et al., Iron ion and sulfasalazine-loaded polydopamine nanops for Fenton reaction and glutathione peroxidase 4 inactivation for enhanced cancer ferrotherapy. Acta Biomater. 145, 210–221 (2022). https://doi.org/10.1016/j.actbio.2022.04.024
J. Li, S. Wang, X. Lin, Y. Cao, Z. Cai et al., Red blood cell-mimic nanocatalyst triggering radical storm to augment cancer immunotherapy. Nano-Micro Lett. 14, 57 (2022). https://doi.org/10.1007/s40820-022-00801-z
J. Li, W. Lu, Y. Yang, R. Xiang, Y. Ling et al., Hybrid nanomaterials for cancer immunotherapy. Adv. Sci. 10, e2204932 (2023). https://doi.org/10.1002/advs.202204932
R. Thangam, K.D. Patel, H. Kang, R. Paulmurugan, Advances in engineered polymer nanop tracking platforms towards cancer immunotherapy-current status and future perspectives. Vaccines 9, 935 (2021). https://doi.org/10.3390/vaccines9080935
X. Cai, L. Ruan, D. Wang, J. Zhang, J. Tang et al., Boosting chemotherapy of bladder cancer cells by ferroptosis using intelligent magnetic targeting nanops. Colloids Surf. B Biointerf 234, 113664 (2024). https://doi.org/10.1016/j.colsurfb.2023.113664
R. Thangam, M.S. Kim, G. Bae, Y. Kim, N. Kang et al., Remote switching of elastic movement of decorated ligand nanostructures controls the adhesion-regulated polarization of host macrophages. Adv. Funct. Mater. 31, 2008698 (2021). https://doi.org/10.1002/adfm.202008698
S. Min, Y.S. Jeon, H. Choi, C. Khatua, N. Li et al., Large and externally positioned ligand-coated nanopatches facilitate the adhesion-dependent regenerative polarization of host macrophages. Nano Lett. 20, 7272–7280 (2020). https://doi.org/10.1021/acs.nanolett.0c02655
W. Yin, J. Chang, J. Sun, Y. Zhao, S. Chen et al., Arginine nanops mediated closed-loop ferroptosis enhancement for T cell activity boosting in cancer immunotherapy. Appl. Mater. Today 36, 102047 (2024). https://doi.org/10.1016/j.apmt.2023.102047
J. Wu, X. Wang, Q. Wang, Z. Lou, S. Li et al., Nanomaterials with enzyme-like characteristics (nanozymes): next-generation artificial enzymes (II). Chem. Soc. Rev. 48, 1004–1076 (2019). https://doi.org/10.1039/c8cs00457a
Y. Lin, J. Ren, X. Qu, Nano-gold as artificial enzymes: hidden talents. Adv. Mater. 26, 4200–4217 (2014). https://doi.org/10.1002/adma.201400238
Q. Ma, Y. Liu, H. Zhu, L. Zhang, X. Liao, Nanozymes in tumor theranostics. Front Oncol. 11, 666017 (2021). https://doi.org/10.3389/fonc.2021.666017
S. Dong, Y. Dong, B. Liu, J. Liu, S. Liu et al., Guiding transition metal-doped hollow cerium tandem nanozymes with elaborately regulated multi-enzymatic activities for intensive chemodynamic therapy. Adv. Mater. 34, e2107054 (2022). https://doi.org/10.1002/adma.202107054
R.K. Sindhu, A. Najda, P. Kaur, M. Shah, H. Singh et al., Potentiality of nanoenzymes for cancer treatment and other diseases: current status and future challenges. Materials 14, 5965 (2021). https://doi.org/10.3390/ma14205965
X. Zhang, X. Chen, Y. Zhao, Nanozymes: versatile platforms for cancer diagnosis and therapy. Nanomicro Lett. 14, 95 (2022). https://doi.org/10.1007/s40820-022-00828-2
P. Wang, T. Wang, J. Hong, X. Yan, M. Liang, Nanozymes: a new disease imaging strategy. Front. Bioeng. Biotechnol. 8, 15 (2020). https://doi.org/10.3389/fbioe.2020.00015
M. Liang, X. Yan, Nanozymes: from new concepts, mechanisms, and standards to applications. Acc. Chem. Res. 52, 2190–2200 (2019). https://doi.org/10.1021/acs.accounts.9b00140
X. Ren, D. Chen, Y. Wang, H. Li, Y. Zhang et al., Nanozymes-recent development and biomedical applications. J. Nanobiotechnol. 20, 92 (2022). https://doi.org/10.1186/s12951-022-01295-y
Y. Zhang, Y. Jin, H. Cui, X. Yan, K. Fan, Nanozyme-based catalytic theranostics. RSC Adv. 10, 10–20 (2020). https://doi.org/10.1039/C9RA09021E
H. Ge, J. Du, J. Zheng, N. Xu, Q. Yao et al., Effective treatment of cisplatin-resistant ovarian tumors with a MoS2-based sonosensitizer and nanoenzyme capable of reversing the resistant-microenvironment and enhancing ferroptosis and apoptosis. Chem. Eng. J. 446, 137040 (2022). https://doi.org/10.1016/j.cej.2022.137040
Y. Liu, W. Zhen, Y. Wang, J. Liu, L. Jin et al., One-dimensional Fe2 P acts as a Fenton agent in response to NIR II light and ultrasound for deep tumor synergetic theranostics. Angew. Chem. Int. Ed. 58, 2407–2412 (2019). https://doi.org/10.1002/anie.201813702
W.-P. Li, C.-H. Su, Y.-C. Chang, Y.-J. Lin, C.-S. Yeh, Ultrasound-induced reactive oxygen species mediated therapy and imaging using a Fenton reaction activable polymersome. ACS Nano 10, 2017–2027 (2016). https://doi.org/10.1021/acsnano.5b06175
L. Zhang, S.-S. Wan, C.-X. Li, L. Xu, H. Cheng et al., An adenosine triphosphate-responsive autocatalytic Fenton nanop for tumor ablation with self-supplied H2O2 and acceleration of Fe(III)/Fe(II) conversion. Nano Lett. 18, 7609–7618 (2018). https://doi.org/10.1021/acs.nanolett.8b03178
L. Xing, X.-Y. Liu, T.-J. Zhou, X. Wan, Y. Wang et al., Photothermal nanozyme-ignited Fenton reaction-independent ferroptosis for breast cancer therapy. J. Control. Release 339, 14–26 (2021). https://doi.org/10.1016/j.jconrel.2021.09.019
G. Zhang, N. Li, Y. Qi, Q. Zhao, J. Zhan et al., Synergistic ferroptosis-gemcitabine chemotherapy of the gemcitabine loaded carbonaceous nanozymes to enhance the treatment and magnetic resonance imaging monitoring of pancreatic cancer. Acta Biomater. 142, 284–297 (2022). https://doi.org/10.1016/j.actbio.2022.02.006
M.J. Ko, S. Min, H. Hong, W. Yoo, J. Joo et al., Magnetic nanops for ferroptosis cancer therapy with diagnostic imaging. Bioact. Mater. 32, 66–97 (2023). https://doi.org/10.1016/j.bioactmat.2023.09.015
H. Gavilán, S.K. Avugadda, T. Fernández-Cabada, N. Soni, M. Cassani et al., Magnetic nanops and clusters for magnetic hyperthermia: optimizing their heat performance and developing combinatorial therapies to tackle cancer. Chem. Soc. Rev. 50, 11614–11667 (2021). https://doi.org/10.1039/d1cs00427a
S. Dutta, S. Noh, R.S. Gual, X. Chen, S. Pané et al., Recent developments in metallic degradable micromotors for biomedical and environmental remediation applications. Nano-Micro Lett. 16, 41 (2023). https://doi.org/10.1007/s40820-023-01259-3
L. Yang, K.D. Patel, C. Rathnam, R. Thangam, Y. Hou et al., Harnessing the therapeutic potential of extracellular vesicles for biomedical applications using multifunctional magnetic nanomaterials. Small 18, e2104783 (2022). https://doi.org/10.1002/smll.202104783
X. Zhao, K. Guo, K. Zhang, S. Duan, M. Chen et al., Orchestrated yolk-shell nanohybrids regulate macrophage polarization and dendritic cell maturation for oncotherapy with augmented antitumor immunity. Adv. Mater. 34, e2108263 (2022). https://doi.org/10.1002/adma.202108263
J. Zhang, K. Zhou, J. Lin, X. Yao, D. Ju et al., Ferroptosis-enhanced chemotherapy for triple-negative breast cancer with magnetic composite nanops. Biomaterials 303, 122395 (2023). https://doi.org/10.1016/j.biomaterials.2023.122395
Z.-J. Zhang, Z.-T. Liu, Y.-P. Huang, W. Nguyen, Y.-X. Wang et al., Magnetic resonance and fluorescence imaging superparamagnetic nanops induce apoptosis and ferroptosis through photodynamic therapy to treat colorectal cancer. Mater. Today Phys. 36, 101150 (2023). https://doi.org/10.1016/j.mtphys.2023.101150
Z.-H. Li, Y. Chen, X. Zeng, X.-Z. Zhang, Ultra-small FePt/siRNA loaded mesoporous silica nanoplatform to deplete cysteine for enhanced ferroptosis in breast tumor therapy. Nano Today 38, 101150 (2021). https://doi.org/10.1016/j.nantod.2021.101150
J. Yang, L. Ding, L. Yu, Y. Wang, M. Ge et al., Nanomedicine enables autophagy-enhanced cancer-cell ferroptosis. Sci. Bull. 66, 464–477 (2021). https://doi.org/10.1016/j.scib.2020.10.021
S. Liang, X. Deng, P.-A. Ma, Z. Cheng, J. Lin, Recent advances in nanomaterial-assisted combinational sonodynamic cancer therapy. Adv. Mater. 32, e2003214 (2020). https://doi.org/10.1002/adma.202003214
S. Liu, K. Dou, B. Liu, M. Pang, P.-A. Ma et al., Construction of multiform hollow-structured covalent organic frameworks via a facile and universal strategy for enhanced sonodynamic cancer therapy. Angew. Chem. Int. Ed. 62, e202301831 (2023). https://doi.org/10.1002/anie.202301831
Y. Dong, S. Dong, B. Liu, C. Yu, J. Liu et al., 2D piezoelectric Bi2MoO6 nanoribbons for GSH-enhanced sonodynamic therapy. Adv. Mater. 33, 2106838 (2021). https://doi.org/10.1002/adma.202106838
A.P. McHale, J.F. Callan, N. Nomikou, C. Fowley, B. Callan, Sonodynamic therapy: concept, mechanism and application to cancer treatment. Adv Exp Med Biol 880, 429–450 (2016). https://doi.org/10.1007/978-3-319-22536-4_22
D. Costley, C.M. Ewan, C. Fowley, A.P. McHale, J. Atchison et al., Treating cancer with sonodynamic therapy: a review. Int. J. Hyperthermia 31, 107–117 (2015). https://doi.org/10.3109/02656736.2014.992484
S. Dong, Y. Dong, Z. Zhao, J. Liu, S. Liu et al., “electron transport chain interference” strategy of amplified mild-photothermal therapy and defect-engineered multi-enzymatic activities for synergistic tumor-personalized suppression. J. Am. Chem. Soc. 145, 9488–9507 (2023). https://doi.org/10.1021/jacs.2c09608
J. Yu, F. Zhu, Y. Yang, P. Zhang, Y. Zheng et al., Ultrasmall iron-doped zinc oxide nanops for ferroptosis assisted sono-chemodynamic cancer therapy. Colloids Surf. B Biointerfaces 232, 113606 (2023). https://doi.org/10.1016/j.colsurfb.2023.113606
L. Zhou, C. Dong, L. Ding, W. Feng, L. Yu et al., Targeting ferroptosis synergistically sensitizes apoptotic sonodynamic anti-tumor nanotherapy. Nano Today 39, 101212 (2021). https://doi.org/10.1016/j.nantod.2021.101212
Y. Cao, H.-Y. Huang, L.-Q. Chen, H.-H. Du, J.-H. Cui et al., Enhanced lysosomal escape of pH-responsive polyethylenimine-betaine functionalized carbon nanotube for the codelivery of survivin small interfering RNA and doxorubicin. ACS Appl. Mater. Interfaces 11, 9763–9776 (2019). https://doi.org/10.1021/acsami.8b20810
P. Norouzi, H. Motasadizadeh, F. Atyabi, R. Dinarvand, M. Gholami et al., Combination therapy of breast cancer by codelivery of doxorubicin and survivin siRNA using polyethylenimine modified silk fibroin nanops. ACS Biomater. Sci. Eng. 7, 1074–1087 (2021). https://doi.org/10.1021/acsbiomaterials.0c01511
J. Chen, Y. Wang, L. Han, R. Wang, C. Gong et al., A ferroptosis-inducing biomimetic nanocomposite for the treatment of drug-resistant prostate cancer. Mater. Today Bio 17, 100484 (2022). https://doi.org/10.1016/j.mtbio.2022.100484
Z. Li, J. Bu, X. Zhu, H. Zhou, K. Ren et al., Anti-tumor immunity and ferroptosis of hepatocellular carcinoma are enhanced by combined therapy of sorafenib and delivering modified GO-based PD-L1 siRNAs. Biomater. Adv. 136, 212761 (2022). https://doi.org/10.1016/j.bioadv.2022.212761
F. Wei, S. Kuang, T.W. Rees, X. Liao, J. Liu et al., Ruthenium(II) complexes coordinated to graphitic carbon nitride: oxygen self-sufficient photosensitizers which produce multiple ROS for photodynamic therapy in hypoxia. Biomaterials 276, 121064 (2021). https://doi.org/10.1016/j.biomaterials.2021.121064
A.E. O’Connor, W.M. Gallagher, A.T. Byrne, Porphyrin and nonporphyrin photosensitizers in oncology: preclinical and clinical advances in photodynamic therapy. Photochem. Photobiol. 85, 1053–1074 (2009). https://doi.org/10.1111/j.1751-1097.2009.00585.x
A. Sneider, R. Jadia, B. Piel, D. VanDyke, C. Tsiros et al., Engineering remotely triggered liposomes to target triple negative breast cancer. Oncomedicine 2, 1–13 (2017). https://doi.org/10.7150/oncm.17406
M. Li, J. Sun, W. Zhang, Y. Zhao, S. Zhang et al., Drug delivery systems based on CD44-targeted glycosaminoglycans for cancer therapy. Carbohydr. Polym. 251, 117103 (2021). https://doi.org/10.1016/j.carbpol.2020.117103
W. Tang, Z. Zhen, M. Wang, H. Wang, Y.-J. Chuang et al., Red blood cell-facilitated photodynamic therapy for cancer treatment. Adv. Funct. Mater. 26, 1757–1768 (2016). https://doi.org/10.1002/adfm.201504803
F. Wei, J. Karges, J. Shen, L. Xie, K. Xiong et al., A mitochondria-localized oxygen self-sufficient two-photon nano-photosensitizer for ferroptosis-boosted photodynamic therapy under hypoxia. Nano Today 44, 101509 (2022). https://doi.org/10.1016/j.nantod.2022.101509
Z. Dong, L. Feng, Y. Hao, Q. Li, M. Chen et al., Synthesis of CaCO3-based nanomedicine for enhanced sonodynamic therapy via amplification of tumor oxidative stress. Chem 6, 1391–1407 (2020). https://doi.org/10.1016/j.chempr.2020.02.020
X. Jing, Y. Xu, D. Liu, Y. Wu, N. Zhou et al., Intelligent nanoflowers: a full tumor microenvironment-responsive multimodal cancer theranostic nanoplatform. Nanoscale 11, 15508–15518 (2019). https://doi.org/10.1039/c9nr04768a
Z. Dong, L. Feng, Y. Chao, Y. Hao, M. Chen et al., Amplification of tumor oxidative stresses with liposomal Fenton catalyst and glutathione inhibitor for enhanced cancer chemotherapy and radiotherapy. Nano Lett. 19, 805–815 (2019). https://doi.org/10.1021/acs.nanolett.8b03905
S. Luo, D. Ma, R. Wei, W. Yao, X. Pang et al., A tumor microenvironment responsive nanoplatform with oxidative stress amplification for effective MRI-based visual tumor ferroptosis. Acta Biomater. 138, 518–527 (2022). https://doi.org/10.1016/j.actbio.2021.11.007
L. Zhou, F. Wei, J. Xiang, H. Li, C. Li et al., Enhancing the ROS generation ability of a rhodamine-decorated iridium(iii) complex by ligand regulation for endoplasmic reticulum-targeted photodynamic therapy. Chem. Sci. 11, 12212–12220 (2020). https://doi.org/10.1039/D0SC04751A
Y. Yu, H. Jia, Y. Liu, L. Zhang, G. Feng et al., Recent progress in type I aggregation-induced emission photosensitizers for photodynamic therapy. Molecules 28, 332 (2022). https://doi.org/10.3390/molecules28010332
X. Li, J.F. Lovell, J. Yoon, X. Chen, Clinical development and potential of photothermal and photodynamic therapies for cancer. Nat. Rev. Clin. Oncol. 17, 657–674 (2020). https://doi.org/10.1038/s41571-020-0410-2
S. Xu, Y. Yuan, X. Cai, C.-J. Zhang, F. Hu et al., Tuning the singlet-triplet energy gap: a unique approach to efficient photosensitizers with aggregation-induced emission (AIE) characteristics. Chem. Sci. 6, 5824–5830 (2015). https://doi.org/10.1039/c5sc01733e
J.F. Lovell, T.W.B. Liu, J. Chen, G. Zheng, Activatable photosensitizers for imaging and therapy. Chem. Rev. 110, 2839–2857 (2010). https://doi.org/10.1021/cr900236h
Z. Zhao, H. Zhang, J.W.Y. Lam, B.Z. Tang, Aggregation-induced emission: new vistas at the aggregate level. Angew. Chem. Int. Ed. 59, 9888–9907 (2020). https://doi.org/10.1002/anie.201916729
R. Jiang, J. Dai, X. Dong, Q. Wang, Z. Meng et al., Improving image-guided surgical and immunological tumor treatment efficacy by photothermal and photodynamic therapies based on a multifunctional NIR AIEgen. Adv. Mater. 33, e2101158 (2021). https://doi.org/10.1002/adma.202101158
B. Wang, L. Wang, H. Wu, X. Liu, J. Zhu et al., The commercial antibiotics with inherent AIE feature: in situ visualization of antibiotic metabolism and specifically differentiation of bacterial species and broad-spectrum therapy. Bioact. Mater. 23, 223–233 (2022). https://doi.org/10.1016/j.bioactmat.2022.11.002
S. Liu, Y. Li, R.T.K. Kwok, J.W.Y. Lam, B.Z. Tang, Structural and process controls of AIEgens for NIR-II theranostics. Chem. Sci. 12, 3427–3436 (2020). https://doi.org/10.1039/d0sc02911d
L. Liu, H. He, Z. Luo, H. Zhou, R. Liang et al., In situ photocatalyzed oxygen generation with photosynthetic bacteria to enable robust immunogenic photodynamic therapy in triple-negative breast cancer. Adv. Funct. Mater. 30, 1910176 (2020). https://doi.org/10.1002/adfm.201910176
X. Yu, Y.-C. Zhang, X. Yang, Z. Huang, T. Zhang et al., Bonsai-inspired AIE nanohybrid photosensitizer based on vermiculite nanosheets for ferroptosis-assisted oxygen self-sufficient photodynamic cancer therapy. Nano Today 44, 101477 (2022). https://doi.org/10.1016/j.nantod.2022.101477
W. Wang, J. Cai, J. Wen, X. Li, Y. Yu et al., Boosting ferroptosis via abplatin(iv) for treatment of platinum-resistant recurrent ovarian cancer. Nano Today 44, 101459 (2022). https://doi.org/10.1016/j.nantod.2022.101459
Y. Han, Z. Dong, C. Wang, Q. Li, Y. Hao et al., Ferrous ions doped calcium carbonate nanops potentiate chemotherapy by inducing ferroptosis. J. Control. Release 348, 346–356 (2022). https://doi.org/10.1016/j.jconrel.2022.06.002
Y. Liu, J. Wu, Y. Jin, W. Zhen, Y. Wang et al., Copper(I) phosphide nanocrystals for in situ self-generation magnetic resonance imaging-guided photothermal-enhanced chemodynamic synergetic therapy resisting deep-seated tumor. Adv. Funct. Mater. 29, 1904678 (2019). https://doi.org/10.1002/adfm.201904678
A. Bagheri, H. Arandiyan, C. Boyer, M. Lim, Lanthanide-doped upconversion nanops: emerging intelligent light-activated drug delivery systems. Adv. Sci. 3, 1500437 (2016). https://doi.org/10.1002/advs.201500437
W. Zeng, X. Wu, T. Chen, S. Sun, Z. Shi et al., Renal-clearable ultrasmall polypyrrole nanops with size-regulated property for second near-infrared light-mediated photothermal therapy. Adv. Funct. Mater. 31, 2008362 (2021). https://doi.org/10.1002/adfm.202008362
M. Yu, J. Yu, Y. Yi, T. Chen, L. Yu et al., Oxidative stress-amplified nanomedicine for intensified ferroptosis-apoptosis combined tumor therapy. J. Control. Release 347, 104–114 (2022). https://doi.org/10.1016/j.jconrel.2022.04.047
X. Zhang, S. Yang, Q. Wang, W. Ye, S. Liu et al., Tailored theranostic nanops cause efficient ferroptosis in head and neck squamous cell carcinoma through a reactive oxygen species “butterfly effect.” Chem. Eng. J. 423, 130083 (2021). https://doi.org/10.1016/j.cej.2021.130083
C. Chen, W. Du, W. Jing, P. Sun, C. Shi et al., Leveraging tumor cell ferroptosis for colorectal cancer treatment via nanoelicitor-activated tumoricidal immunity. Chem. Eng. J. 430, 132983 (2022). https://doi.org/10.1016/j.cej.2021.132983
Y. Dai, Z. Yang, S. Cheng, Z. Wang, R. Zhang et al., Toxic reactive oxygen species enhanced synergistic combination therapy by self-assembled metal-phenolic network nanops. Adv. Mater. 30, 1704877 (2018). https://doi.org/10.1002/adma.201704877
P. Li, M. Gao, Z. Hu, T. Xu, J. Chen et al., Synergistic ferroptosis and macrophage re-polarization using engineering exosome-mimic M1 nanovesicles for cancer metastasis suppression. Chem. Eng. J. 409, 128217 (2021). https://doi.org/10.1016/j.cej.2020.128217
J. Yang, S. Ma, R. Xu, Y. Wei, J. Zhang et al., Smart biomimetic metal organic frameworks based on ROS-ferroptosis-glycolysis regulation for enhanced tumor chemo-immunotherapy. J. Control. Release 334, 21–33 (2021). https://doi.org/10.1016/j.jconrel.2021.04.013
A. Ghoochani, E.-C. Hsu, M. Aslan, M.A. Rice, H.M. Nguyen et al., Ferroptosis inducers are a novel therapeutic approach for advanced prostate cancer. Cancer Res. 81, 1583–1594 (2021). https://doi.org/10.1158/0008-5472.CAN-20-3477
D. Tang, X. Chen, R. Kang, G. Kroemer, Ferroptosis: molecular mechanisms and health implications. Cell Res. 31, 107–125 (2021). https://doi.org/10.1038/s41422-020-00441-1
E.H. Kim, D. Shin, J. Lee, A.R. Jung, J.L. Roh, CISD2 inhibition overcomes resistance to sulfasalazine-induced ferroptotic cell death in head and neck cancer. Cancer Lett. 432, 180–190 (2018). https://doi.org/10.1016/j.canlet.2018.06.018
D. Li, Y. Li, The interaction between ferroptosis and lipid metabolism in cancer. Signal Transduct. Target. Ther. 5, 108 (2020). https://doi.org/10.1038/s41392-020-00216-5
X. Zhang, K. Yu, L. Ma, Z. Qian, X. Tian et al., Endogenous glutamate determines ferroptosis sensitivity via ADCY10-dependent YAP suppression in lung adenocarcinoma. Theranostics 11, 5650–5674 (2021). https://doi.org/10.7150/thno.55482
S. Park, J. Oh, M. Kim, E.-J. Jin, Bromelain effectively suppresses Kras-mutant colorectal cancer by stimulating ferroptosis. Anim. Cells Syst. 22, 334–340 (2018). https://doi.org/10.1080/19768354.2018.1512521
Y. Sun, Y. He, J. Tong, D. Liu, H. Zhang et al., All-trans retinoic acid inhibits the malignant behaviors of hepatocarcinoma cells by regulating ferroptosis. Genes Dis. 9, 1742–1756 (2022). https://doi.org/10.1016/j.gendis.2022.04.011
L. Sun, H. Wang, D. Xu, S. Yu, L. Zhang et al., Lapatinib induces mitochondrial dysfunction to enhance oxidative stress and ferroptosis in doxorubicin-induced cardiomyocytes via inhibition of PI3K/AKT signaling pathway. Bioengineered 13, 48–60 (2022). https://doi.org/10.1080/21655979.2021.2004980
N. Eling, L. Reuter, J. Hazin, A. Hamacher-Brady, N.R. Brady, Identification of artesunate as a specific activator of ferroptosis in pancreatic cancer cells. Oncoscience 2, 517–532 (2015). https://doi.org/10.18632/oncoscience.160
K.S. Kim, B. Choi, H. Choi, M.J. Ko, D.H. Kim et al., Enhanced natural killer cell anti-tumor activity with nanops mediated ferroptosis and potential therapeutic application in prostate cancer. J. Nanobiotechnol. 20, 428 (2022). https://doi.org/10.1186/s12951-022-01635-y
T. Kasukabe, Y. Honma, J. Okabe-Kado, Y. Higuchi, N. Kato et al., Combined treatment with cotylenin A and phenethyl isothiocyanate induces strong antitumor activity mainly through the induction of ferroptotic cell death in human pancreatic cancer cells. Oncol. Rep. 36, 968–976 (2016). https://doi.org/10.3892/or.2016.4867
D. Wang, W. Fang, C. Huang, Z. Chen, T. Nie et al., MR imaging guided iron-based nanoenzyme for synergistic Ferroptosis-Starvation therapy in triple negative breast cancer. Smart Mater. Med. 3, 159–167 (2022). https://doi.org/10.1016/j.smaim.2021.12.008