Palucka, Okay. & Banchereau, J. Most cancers immunotherapy by way of dendritic cells. Nat. Rev. Most cancers 12, 265–277 (2012).
Ye, B., Smerin, D., Gao, Q., Kang, C. & Xiong, X. Excessive-throughput sequencing of the immune repertoire in oncology: purposes for scientific prognosis, monitoring, and immunotherapies. Most cancers Lett. 416, 42–56 (2018).
Srivatsan, S. et al. Allogeneic tumor cell vaccines: the promise and limitations in scientific trials. Hum. Vaccines Immunother. 10, 52–63 (2014).
Frey, A. B. & Monu, N. Signaling defects in anti‐tumor T cells. Immunol. Rev. 222, 192–205 (2008).
Jensen-Jarolim, E. & Singer, J. Most cancers vaccines inducing antibody manufacturing: extra execs than cons. Skilled Rev. Vaccines 10, 1281–1289 (2011).
Herrmann, I. Okay., Wooden, M. J. A. & Fuhrmann, G. Extracellular vesicles as a next-generation drug supply platform. Nat. Nanotechnol. 16, 748–759 (2021).
Zhang, X., Cui, H., Zhang, W., Li, Z. & Gao, J. Engineered tumor cell-derived vaccines in opposition to most cancers: the artwork of combating poison with poison. Bioact. Mater. 22, 491–517 (2023).
van Niel, G. et al. Challenges and instructions in finding out cell–cell communication by extracellular vesicles. Nat. Rev. Mol. Cell Biol. 23, 369–382 (2022).
Kumari, P. et al. Host extracellular vesicles confer cytosolic entry to systemic LPS licensing non-canonical inflammasome sensing and pyroptosis. Nat. Cell Biol. 25, 1860–1872 (2023).
Bhatta, R. et al. Metabolic tagging of extracellular vesicles and growth of enhanced extracellular vesicle based mostly most cancers vaccines. Nat. Commun. 14, 8047 (2023).
Wang, S. et al. Macrophage-tumor chimeric exosomes accumulate in lymph node and tumor to activate the immune response and the tumor microenvironment. Sci. Transl. Med. 13, eabb6981 (2021).
Nam, G. H. et al. Rising prospects of exosomes for most cancers therapy: from standard remedy to immunotherapy. Adv. Mater. 32, 2002440 (2020).
Li, S., Xu, J., Qian, J. & Gao, X. Engineering extracellular vesicles for most cancers remedy: current advances and challenges in scientific translation. Biomater. Sci. 8, 6978–6991 (2020).
Santos, P. & Almeida, F. Exosome-based vaccines: historical past, present state, and scientific trials. Entrance. Immunol. 12, 711565 (2021).
Ahmadi, M., Abbasi, R. & Rezaie, J. Tumor immune escape: extracellular vesicles roles and therapeutics software. Cell Commun. Sign. 22, 9 (2024).
Chen, G. et al. Exosomal PD-L1 contributes to immunosuppression and is related to anti-PD-1 response. Nature 560, 382–386 (2018).
Yu, P. et al. Pyroptosis: mechanisms and illnesses. Sign Transduct. Goal. Ther. 6, 128 (2021).
Frank, D. & Vince, J. E. Pyroptosis versus necroptosis: similarities, variations, and crosstalk. Cell Loss of life Differ. 26, 99–114 (2019).
Faria, S. S. et al. NLRP3 inflammasome-mediated cytokine manufacturing and pyroptosis cell demise in breast most cancers. J. Biomed. Sci. 28, 26 (2021).
Li, Z. T. et al. Enhancing gasdermin-induced tumor pyroptosis by means of stopping ESCRT-dependent cell membrane restore augments antitumor immune response. Nat. Commun. 13, 6321 (2022).
Zhang, Z. et al. Gasdermin E suppresses tumour development by activating anti-tumour immunity. Nature 579, 415–420 (2020).
Horrevorts, S. Okay. et al. Glycan-modified apoptotic melanoma-derived extracellular vesicles as antigen supply for anti-tumor vaccination. Cancers 11, 1266 (2019).
Schnurr, M. et al. Apoptotic pancreatic tumor cells are superior to cell lysates in selling cross-priming of cytotoxic T cells and activate NK and γδ T cells. Most cancers Res. 62, 2347–2352 (2002).
Shi, J. et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell demise. Nature 526, 660–665 (2015).
Wang, G. et al. Tumour extracellular vesicles and particles induce liver metabolic dysfunction. Nature 618, 374–382 (2023).
Wang, Y. L. et al. Delivering antisense oligonucleotides throughout the blood-brain barrier by tumor cell-derived small apoptotic our bodies. Adv. Sci. 8, 2004929 (2021).
Krysko, D. V. et al. Immunogenic cell demise and DAMPs in most cancers remedy. Nat. Rev. Most cancers 12, 860–875 (2012).
Chen, Y. L. et al. Immuno-modulators improve antigen-specific immunity and anti-tumor results of mesothelin-specific chimeric DNA vaccine by means of selling DC maturation. Most cancers Lett. 425, 152–163 (2018).
Mossoba, M. E. et al. Tumor safety following vaccination with low doses of lentivirally transduced DCs expressing the self-antigen erbB2. Mol. Ther. 16, 607–617 (2008).
Toldo, S. & Abbate, A. The position of the NLRP3 inflammasome and pyroptosis in cardiovascular illnesses. Nat. Rev. Cardiol. 21, 219–237 (2024).
Zamani, P., Oskuee, R. Okay., Atkin, S. L., Navashenaq, J. G. & Sahebkar, A. MicroRNAs as vital regulators of the NLRP3 inflammasome. Prog. Biophys. Mol. Bio. 150, 50–61 (2020).
Ding, S., Liu, D., Wang, L., Wang, G. & Zhu, Y. Inhibiting microRNA-29a protects myocardial ischemia-reperfusion damage by concentrating on SIRT1 and suppressing oxidative stress and NLRP3-mediated pyroptosis pathway. J. Pharmacol. Exp. Ther. 372, 128–135 (2020).
Wang, Z., Solar, L., Jia, Okay., Wang, H. & Wang, X. miR-9-5p modulates the development of Parkinson’s illness by concentrating on SIRT1. Neurosci. Lett. 701, 226–233 (2019).
Bai, D. et al. ALDOA maintains NLRP3 inflammasome activation by controlling AMPK activation. Autophagy 18, 1673–1693 (2022).
Karki, R. et al. NLRC3 is an inhibitory sensor of PI3K-mTOR pathways in most cancers. Nature 540, 583–587 (2016).
Sharma, B. R. & Kanneganti, T. D. NLRP3 inflammasome in most cancers and metabolic illnesses. Nat. Immunol. 22, 550–559 (2021).
Zhang, X. et al. Cell microparticles loaded with tumor antigen and resiquimod reprogram tumor-associated macrophages and promote stem-like CD8+ T cells to spice up anti-PD-1 remedy. Nat. Commun. 14, 5653 (2023).
Chang, B. A., Cross, J. L., Najar, H. M. & Dutz, J. P. Topical resiquimod promotes priming of CTL to parenteral antigens. Vaccine 27, 5791–5799 (2009).
Rodell, C. B. et al. TLR7/8-agonist-loaded nanoparticles promote the polarization of tumour-associated macrophages to boost most cancers immunotherapy. Nat. Biomed. Eng. 2, 578–588 (2018).
Kuai, R., Ochyl, L. J., Bahjat, Okay. S., Schwendeman, A. & Moon, J. J. Designer vaccine nanodiscs for customized most cancers immunotherapy. Nat. Mater. 16, 489–496 (2017).
Hu, Z., Ott, P. A. & Wu, C. J. In the direction of customized, tumour-specific, therapeutic vaccines for most cancers. Nat. Rev. Immunol. 18, 168–182 (2018).
Saxena, M., van der Burg, S. H., Melief, C. J. M. & Bhardwaj, N. Therapeutic most cancers vaccines. Nat. Rev. Most cancers 21, 360–378 (2021).
Wang, T. et al. A most cancers vaccine-mediated postoperative immunotherapy for recurrent and metastatic tumors. Nat. Commun. 9, 1532 (2018).
Wright, S. S. et al. Transplantation of gasdermin pores by extracellular vesicles propagates pyroptosis to bystander cells. Cell 188, P280-291.E17 (2024).
Bergsbaken, T., Fink, S. L. & Cookson, B. T. Pyroptosis: host cell demise and irritation. Nat. Rev. Microbiol. 7, 99–109 (2009).
Ruhl, S. et al. ESCRT-dependent membrane restore negatively regulates pyroptosis downstream of GSDMD activation. Science 362, 956–960 (2018).
Vietri, M., Radulovic, M. & Stenmark, H. The various capabilities of ESCRTs. Nat. Rev. Mol. Cell Biol. 21, 25–42 (2020).
Baxter, A. A. et al. Evaluation of extracellular vesicles generated from monocytes below situations of lytic cell demise. Sci. Rep. 9, 7538 (2019).
Zhang, Y. et al. Inflammasome-derived exosomes activate NF-κB signaling in macrophages. J. Proteome Res. 16, 170–178 (2017).