Abstract
Although the first generation of cancer immunotherapeutics produced unprecedented improvements in clinical outcomes for individuals with cancer, novel strategies to increase treatment specificity, delivery efficiency and pharmacokinetics are still needed. In this Review, we describe the potential advantages and current limitations of nanomaterials for cancer immunotherapy and highlight rational uses of nanosystems to generate potent and durable antitumor immune responses. We close with a review of the current state of clinical development of nanomedicine for cancer immunotherapy.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 digital issues and online access to articles
$119.00 per year
only $9.92 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to the full article PDF.
USD 39.95
Prices may be subject to local taxes which are calculated during checkout



Similar content being viewed by others
References
Antonia, S. J. et al. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N. Engl. J. Med. 377, 1919–1929 (2017).
Pires da Silva, I. et al. Ipilimumab alone or ipilimumab plus anti-PD-1 therapy in patients with metastatic melanoma resistant to anti-PD-(L)1 monotherapy: a multicentre, retrospective, cohort study. Lancet Oncol. 22, 836–847 (2021).
Maude, S. L. et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 371, 1507–1517 (2014).
Gettinger, S. N. et al. Overall survival and long-term safety of nivolumab (anti-programmed death 1 antibody, BMS-936558, ONO-4538) in patients with previously treated advanced non-small-cell lung cancer. J. Clin. Oncol. 33, 2004–2012 (2015).
Borghaei, H. et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N. Engl. J. Med. 373, 1627–1639 (2015).
Carbone, D. P. et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N. Engl. J. Med. 376, 2415–2426 (2017).
Herbst, R. S. et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 387, 1540–1550 (2016).
Postow, M. A., Sidlow, R. & Hellmann, M. D. Immune-related adverse events associated with immune checkpoint blockade. N. Engl. J. Med. 378, 158–168 (2018).
Gangadhar, T. C. & Vonderheide, R. H. Mitigating the toxic effects of anticancer immunotherapy. Nat. Rev. Clin. Oncol. 11, 91–99 (2014).
Morgan, R. A. et al. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol. Ther. 18, 843–851 (2010).
Brentjens, R., Yeh, R., Bernal, Y., Riviere, I. & Sadelain, M. Treatment of chronic lymphocytic leukemia with genetically targeted autologous T cells: case report of an unforeseen adverse event in a phase I clinical trial. Mol. Ther. 18, 666–668 (2010).
Chauhan, V. P. & Jain, R. K. Strategies for advancing cancer nanomedicine. Nat. Mater. 12, 958–962 (2013).
Maeda, H., Nakamura, H. & Fang, J. The EPR effect for macromolecular drug delivery to solid tumors: improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor imaging in vivo. Adv. Drug Deliv. Rev. 65, 71–79 (2013).
Jiang, W., Kim, B. Y., Rutka, J. T. & Chan, W. C. Nanoparticle-mediated cellular response is size-dependent. Nat. Nanotechnol. 3, 145–150 (2008).
Li, Y., Kroger, M. & Liu, W. K. Shape effect in cellular uptake of PEGylated nanoparticles: comparison between sphere, rod, cube and disk. Nanoscale 7, 16631–16646 (2015).
Dong, S. et al. Adaptive design of mRNA-loaded extracellular vesicles for targeted immunotherapy of cancer. Nat. Commun. 14, 6610 (2023).
Sayour, E. J., Boczkowski, D., Mitchell, D. A. & Nair, S. K. Cancer mRNA vaccines: clinical advances and future opportunities. Nat. Rev. Clin. Oncol. 21, 489–500 (2024).
Wang, J. et al. Physical activation of innate immunity by spiky particles. Nat. Nanotechnol. 13, 1078–1086 (2018).
Zhu, M. et al. Cell-penetrating nanoparticles activate the inflammasome to enhance antibody production by targeting microtubule-associated protein 1-light chain 3 for degradation. ACS Nano 14, 3703–3717 (2020).
Sun, B. et al. Engineering an effective immune adjuvant by designed control of shape and crystallinity of aluminum oxyhydroxide nanoparticles. ACS Nano 7, 10834–10849 (2013).
Mendez-Gomez, H. R. et al. RNA aggregates harness the danger response for potent cancer immunotherapy. Cell 187, 2521–2535 (2024).
Grippin, A. J. et al. Dendritic cell-activating magnetic nanoparticles enable early prediction of antitumor response with magnetic resonance imaging. ACS Nano 13, 13884–13898 (2019).
Duan, X. et al. Photodynamic therapy mediated by nontoxic core-shell nanoparticles synergizes with immune checkpoint blockade to elicit antitumor immunity and antimetastatic effect on breast cancer. J. Am. Chem. Soc. 138, 16686–16695 (2016).
Wan, J., Zhang, X., Tang, D., Liu, T. & Xiao, H. Biodegradable NIR-II pseudo conjugate polymeric nanoparticles amplify photodynamic immunotherapy via alleviation of tumor hypoxia and tumor-associated macrophage reprogramming. Adv. Mater. 35, e2209799 (2023).
Hayashi, K. et al. Magnetically responsive smart nanoparticles for cancer treatment with a combination of magnetic hyperthermia and remote-control drug release. Theranostics 4, 834–844 (2014).
Marill, J., Mohamed Anesary, N. & Paris, S. DNA damage enhancement by radiotherapy-activated hafnium oxide nanoparticles improves cGAS–STING pathway activation in human colorectal cancer cells. Radiother. Oncol. 141, 262–266 (2019).
Bonvalot, S. et al. NBTXR3, a first-in-class radioenhancer hafnium oxide nanoparticle, plus radiotherapy versus radiotherapy alone in patients with locally advanced soft-tissue sarcoma (Act.In.Sarc): a multicentre, phase 2-3, randomised, controlled trial. Lancet Oncol. 20, 1148–1159 (2019).
Hewitt, S. L. et al. Durable anticancer immunity from intratumoral administration of IL-23, IL-36γ, and OX40L mRNAs. Sci. Transl. Med. 11, eaat9143 (2019).
Kang, M. et al. Nanocomplex-mediated in vivo programming to chimeric antigen receptor-M1 macrophages for cancer therapy. Adv. Mater. 33, e2103258 (2021).
Chen, C. et al. Intracavity generation of glioma stem cell-specific CAR macrophages primes locoregional immunity for postoperative glioblastoma therapy. Sci. Transl. Med. 14, eabn1128 (2022).
Stadler, C. R. et al. Elimination of large tumors in mice by mRNA-encoded bispecific antibodies. Nat. Med. 23, 815–817 (2017).
Shi, J., Kantoff, P. W., Wooster, R. & Farokhzad, O. C. Cancer nanomedicine: progress, challenges and opportunities. Nat. Rev. Cancer 17, 20–37 (2017).
Tenchov, R., Bird, R., Curtze, A. E. & Zhou, Q. Lipid nanoparticles from liposomes to mRNA vaccine delivery, a landscape of research diversity and advancement. ACS Nano 15, 16982–17015 (2021).
Wilhelm, S. et al. Analysis of nanoparticle delivery to tumours. Nat. Rev. Mater. 1, 16014 (2016).
Sindhwani, S. et al. The entry of nanoparticles into solid tumours. Nat. Mater. 19, 566–575 (2020).
Liang, Q. et al. The softness of tumour-cell-derived microparticles regulates their drug-delivery efficiency. Nat. Biomed. Eng. 3, 729–740 (2019).
Guo, P. et al. Nanoparticle elasticity directs tumor uptake. Nat. Commun. 9, 130 (2018).
Dane, E. L. et al. STING agonist delivery by tumour-penetrating PEG-lipid nanodiscs primes robust anticancer immunity. Nat. Mater. 21, 710–720 (2022).
Chen, Y. et al. Therapeutic remodeling of the tumor microenvironment enhances nanoparticle delivery. Adv. Sci. 6, 1802070 (2019).
Wang, Y. et al. Age-associated disparity in phagocytic clearance affects the efficacy of cancer nanotherapeutics. Nat. Nanotechnol. 19, 255–263 (2024).
Schudel, A., Francis, D. M. & Thomas, S. N. Material design for lymph node drug delivery. Nat. Rev. Mater. 4, 415–428 (2019).
Galstyan, A. et al. Blood–brain barrier permeable nano immunoconjugates induce local immune responses for glioma therapy. Nat. Commun. 10, 3850 (2019).
Zhang, F. et al. Uniform brain tumor distribution and tumor associated macrophage targeting of systemically administered dendrimers. Biomaterials 52, 507–516 (2015).
Sayour, E. J. et al. Systemic activation of antigen-presenting cells via RNA-loaded nanoparticles. Oncoimmunology 6, e1256527 (2017).
Kranz, L. M. et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 534, 396–401 (2016).
Sahin, U. et al. An RNA vaccine drives immunity in checkpoint-inhibitor-treated melanoma. Nature 585, 107–112 (2020).
Jneid, B. et al. Selective STING stimulation in dendritic cells primes antitumor T cell responses. Sci. Immunol. 8, eabn6612 (2023).
Yi, S. et al. Tailoring nanostructure morphology for enhanced targeting of dendritic cells in atherosclerosis. ACS Nano 10, 11290–11303 (2016).
Karabin, N. B. et al. Sustained micellar delivery via inducible transitions in nanostructure morphology. Nat. Commun. 9, 624 (2018).
Si, Y. et al. Adjuvant-free nanofiber vaccine induces in situ lung dendritic cell activation and TH17 responses. Sci. Adv. 6, eaba0995 (2020).
Guermonprez, P., Valladeau, J., Zitvogel, L., Thery, C. & Amigorena, S. Antigen presentation and T cell stimulation by dendritic cells. Annu. Rev. Immunol. 20, 621–667 (2002).
Rosenberg, S. A., Yang, J. C. & Restifo, N. P. Cancer immunotherapy: moving beyond current vaccines. Nat. Med. 10, 909–915 (2004).
Sampson, J. H. et al. Immunologic escape after prolonged progression-free survival with epidermal growth factor receptor variant III peptide vaccination in patients with newly diagnosed glioblastoma. J. Clin. Oncol. 28, 4722–4729 (2010).
Sayour, E. J. et al. Personalized tumor RNA loaded lipid-nanoparticles prime the systemic and intratumoral milieu for response to cancer immunotherapy. Nano Lett. 18, 6195–6206 (2018).
Rojas, L. A. et al. Personalized RNA neoantigen vaccines stimulate T cells in pancreatic cancer. Nature 618, 144–150 (2023).
Weber, J. S. et al. Individualised neoantigen therapy mRNA-4157 (V940) plus pembrolizumab versus pembrolizumab monotherapy in resected melanoma (KEYNOTE-942): a randomised, phase 2b study. Lancet 403, 632–644 (2024).
Gainor, J. F. et al. T cell responses to individualized neoantigen therapy mRNA-4157 (V940) alone or in combination with pembrolizumab in the phase 1 KEYNOTE-603 study. Cancer Discov. 14, 2029–2223 (2024).
Karikó, K. et al. Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with increased translational capacity and biological stability. Mol. Ther. 16, 1833–1840 (2008).
Carroll, E. C. et al. The vaccine adjuvant chitosan promotes cellular immunity via DNA sensor cGAS–STING-dependent induction of type I interferons. Immunity 44, 597–608 (2016).
Wegmann, F. et al. Polyethyleneimine is a potent mucosal adjuvant for viral glycoprotein antigens. Nat. Biotechnol. 30, 883–888 (2012).
Wei, X. et al. Cationic nanocarriers induce cell necrosis through impairment of Na+/K+-ATPase and cause subsequent inflammatory response. Cell Res. 25, 237–253 (2015).
Luo, M. et al. A STING-activating nanovaccine for cancer immunotherapy. Nat. Nanotechnol. 12, 648–654 (2017).
Miao, L. et al. Delivery of mRNA vaccines with heterocyclic lipids increases anti-tumor efficacy by STING-mediated immune cell activation. Nat. Biotechnol. 37, 1174–1185 (2019).
Wang, J. et al. STING licensing of type I dendritic cells potentiates antitumor immunity. Sci. Immunol. 9, eadj3945 (2024).
Huang, Z. et al. Anti-tumor immune responses of tumor-associated macrophages via Toll-like receptor 4 triggered by cationic polymers. Biomaterials 34, 746–755 (2013).
Marabelle, A., Kohrt, H., Caux, C. & Levy, R. Intratumoral immunization: a new paradigm for cancer therapy. Clin. Cancer Res. 20, 1747–1756 (2014).
Lu, Y. et al. Immunological conversion of solid tumours using a bispecific nanobioconjugate for cancer immunotherapy. Nat. Nanotechnol. 17, 1332–1341 (2022).
Min, Y. et al. Antigen-capturing nanoparticles improve the abscopal effect and cancer immunotherapy. Nat. Nanotechnol. 12, 877–882 (2017).
Shae, D. et al. Endosomolytic polymersomes increase the activity of cyclic dinucleotide STING agonists to enhance cancer immunotherapy. Nat. Nanotechnol. 14, 269–278 (2019).
Ruffell, B. & Coussens, L. M. Macrophages and therapeutic resistance in cancer. Cancer Cell 27, 462–472 (2015).
Bronte, V. & Murray, P. J. Understanding local macrophage phenotypes in disease: modulating macrophage function to treat cancer. Nat. Med. 21, 117–119 (2015).
Decout, A., Katz, J. D., Venkatraman, S. & Ablasser, A. The cGAS–STING pathway as a therapeutic target in inflammatory diseases. Nat. Rev. Immunol. 21, 548–569 (2021).
Samson, N. & Ablasser, A. The cGAS–STING pathway and cancer. Nat. Cancer 3, 1452–1463 (2022).
Li, X. et al. Cancer immunotherapy based on image-guided STING activation by nucleotide nanocomplex-decorated ultrasound microbubbles. Nat. Nanotechnol. 17, 891–899 (2022).
Liu, D. et al. Tumor microenvironment-responsive nanoparticles amplifying STING signaling pathway for cancer immunotherapy. Adv. Mater. 36, e2304845 (2024).
Wang, X. et al. A protein-based cGAS–STING nanoagonist enhances T cell-mediated anti-tumor immune responses. Nat. Commun. 13, 5685 (2022).
Rodell, C. B. et al. TLR7/8-agonist-loaded nanoparticles promote the polarization of tumour-associated macrophages to enhance cancer immunotherapy. Nat. Biomed. Eng. 2, 578–588 (2018).
Tahtinen, S. et al. IL-1 and IL-1ra are key regulators of the inflammatory response to RNA vaccines. Nat. Immunol. 23, 532–542 (2022).
Feng, M. et al. Phagocytosis checkpoints as new targets for cancer immunotherapy. Nat. Rev. Cancer 19, 568–586 (2019).
von Roemeling, C. A. et al. Therapeutic modulation of phagocytosis in glioblastoma can activate both innate and adaptive antitumour immunity. Nat. Commun. 11, 1508 (2020).
Koh, E. et al. Exosome-SIRPα, a CD47 blockade increases cancer cell phagocytosis. Biomaterials 121, 121–129 (2017).
Lee, N. K. et al. Caspase-cleavable peptide–doxorubicin conjugate in combination with CD47-antagonizing nanocage therapeutics for immune-mediated elimination of colorectal cancer. Biomaterials 277, 121105 (2021).
Kulkarni, A. et al. A designer self-assembled supramolecule amplifies macrophage immune responses against aggressive cancer. Nat. Biomed. Eng. 2, 589–599 (2018).
Barkal, A. A. et al. CD24 signalling through macrophage Siglec-10 is a target for cancer immunotherapy. Nature 572, 392–396 (2019).
Yuan, H. et al. Multivalent bi-specific nanobioconjugate engager for targeted cancer immunotherapy. Nat. Nanotechnol. 12, 763–769 (2017).
Lei, A. et al. A second-generation M1-polarized CAR macrophage with antitumor efficacy. Nat. Immunol. 25, 102–116 (2024).
Klichinsky, M. et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat. Biotechnol. 38, 947–953 (2020).
Sloas, C. et al. Macrophages expressing synthetic cytokine receptors reverse IL10-mediated immunosuppression within solid tumors and promote adaptive immunity. Cancer Res. 84, 5249 (2024).
Kim, K. S. et al. Cationic nanoparticle-mediated activation of natural killer cells for effective cancer immunotherapy. ACS Appl. Mater. Interfaces 12, 56731–56740 (2020).
Shin, H. et al. Enhancing CAR-NK cells against solid tumors through chemical and genetic fortification with DOTAP-functionalized lipid nanoparticles. Adv. Funct. Mater. 34, 2315721 (2024).
Myers, J. A. & Miller, J. S. Exploring the NK cell platform for cancer immunotherapy. Nat. Rev. Clin. Oncol. 18, 85–100 (2021).
Au, K. M., Park, S. I. & Wang, A. Z. Trispecific natural killer cell nanoengagers for targeted chemoimmunotherapy. Sci. Adv. 6, eaba8564 (2020).
Wang, D. Y. et al. Fatal toxic effects associated with immune checkpoint inhibitors: a systematic review and meta-analysis. JAMA Oncol. 4, 1721–1728 (2018).
Johnson, D. B., Nebhan, C. A., Moslehi, J. J. & Balko, J. M. Immune-checkpoint inhibitors: long-term implications of toxicity. Nat. Rev. Clin. Oncol. 19, 254–267 (2022).
Spain, L., Diem, S. & Larkin, J. Management of toxicities of immune checkpoint inhibitors. Cancer Treat. Rev. 44, 51–60 (2016).
Cappell, K. M. & Kochenderfer, J. N. Long-term outcomes following CAR T cell therapy: what we know so far. Nat. Rev. Clin. Oncol. 20, 359–371 (2023).
Kalos, M. et al. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci. Transl. Med. 3, 95ra73 (2011).
Porter, D. L., Levine, B. L., Kalos, M., Bagg, A. & June, C. H. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N. Engl. J. Med. 365, 725–733 (2011).
Rafiq, S., Hackett, C. S. & Brentjens, R. J. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat. Rev. Clin. Oncol. 17, 147–167 (2020).
June, C. H., Riddell, S. R. & Schumacher, T. N. Adoptive cellular therapy: a race to the finish line. Sci. Transl. Med. 7, 280ps287 (2015).
Fesnak, A. D., June, C. H. & Levine, B. L. Engineered T cells: the promise and challenges of cancer immunotherapy. Nat. Rev. Cancer 16, 566–581 (2016).
Larson, R. C. & Maus, M. V. Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nat. Rev. Cancer 21, 145–161 (2021).
Maggs, L., Cattaneo, G., Dal, A. E., Moghaddam, A. S. & Ferrone, S. CAR T cell-based immunotherapy for the treatment of glioblastoma. Front. Neurosci. 15, 662064 (2021).
Imai, C. et al. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia 18, 676–684 (2004).
Krause, A. et al. Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes. J. Exp. Med. 188, 619–626 (1998).
van der Stegen, S. J., Hamieh, M. & Sadelain, M. The pharmacology of second-generation chimeric antigen receptors. Nat. Rev. Drug Discov. 14, 499–509 (2015).
Stephan, M. T., Moon, J. J., Um, S. H., Bershteyn, A. & Irvine, D. J. Therapeutic cell engineering with surface-conjugated synthetic nanoparticles. Nat. Med. 16, 1035–1041 (2010).
Tang, L. et al. Enhancing T cell therapy through TCR-signaling-responsive nanoparticle drug delivery. Nat. Biotechnol. 36, 707–716 (2018).
Guo, Y. et al. Metabolic reprogramming of terminally exhausted CD8+ T cells by IL-10 enhances anti-tumor immunity. Nat. Immunol. 22, 746–756 (2021).
Smith, T. T. et al. In situ programming of leukaemia-specific T cells using synthetic DNA nanocarriers. Nat. Nanotechnol. 12, 813–820 (2017).
Zhu, C. et al. Injectable supramolecular hydrogels for in situ programming of CAR-T cells toward solid tumor immunotherapy. Adv. Mater. 36, e2310078 (2024).
Verdun, N. & Marks, P. Secondary cancers after chimeric antigen receptor T-cell therapy. N. Engl. J. Med. 390, 584–586 (2024).
Billingsley, M. M. et al. Ionizable lipid nanoparticle-mediated mRNA delivery for human CAR T cell engineering. Nano Lett. 20, 1578–1589 (2020).
Parayath, N. N., Stephan, S. B., Koehne, A. L., Nelson, P. S. & Stephan, M. T. In vitro-transcribed antigen receptor mRNA nanocarriers for transient expression in circulating T cells in vivo. Nat. Commun. 11, 6080 (2020).
Mabry, R. et al. 1222 In situ CAR therapy using oRNA lipid nanoparticles regresses tumors in mice. J. Immunother. Cancer 10, A1265 (2022).
Tombacz, I. et al. Highly efficient CD4+ T cell targeting and genetic recombination using engineered CD4+ cell-homing mRNA–LNPs. Mol. Ther. 29, 3293–3304 (2021).
Haist, M., Mailander, V. & Bros, M. Nanodrugs targeting T cells in tumor therapy. Front. Immunol. 13, 912594 (2022).
O’Donnell, J. S., Teng, M. W. L. & Smyth, M. J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 16, 151–167 (2019).
Park, J. et al. Combination delivery of TGF-β inhibitor and IL-2 by nanoscale liposomal polymeric gels enhances tumour immunotherapy. Nat. Mater. 11, 895–905 (2012).
Schmid, D. et al. T cell-targeting nanoparticles focus delivery of immunotherapy to improve antitumor immunity. Nat. Commun. 8, 1747 (2017).
Shi, C. et al. Targeting the activity of T cells by membrane surface redox regulation for cancer theranostics. Nat. Nanotechnol. 18, 86–97 (2023).
Jiang, C.-T. et al. Immunomodulating nano-adaptors potentiate antibody-based cancer immunotherapy. Nat. Commun. 12, 1359 (2021).
Alhallak, K. et al. Nanoparticle T-cell engagers as a modular platform for cancer immunotherapy. Leukemia 35, 2346–2357 (2021).
Zhang, D. et al. Enhancing CRISPR/Cas gene editing through modulating cellular mechanical properties for cancer therapy. Nat. Nanotechnol. 17, 777–787 (2022).
Hu, X. et al. An artificial metabzyme for tumour-cell-specific metabolic therapy. Nat. Nanotechnol. 19, 1712–1722 (2024).
Li, S. et al. Prolonged activation of innate immune pathways by a polyvalent STING agonist. Nat. Biomed. Eng. 5, 455–466 (2021).
Sun, X. et al. Amplifying STING activation by cyclic dinucleotide–manganese particles for local and systemic cancer metalloimmunotherapy. Nat. Nanotechnol. 16, 1260–1270 (2021).
Wang, Y. et al. Universal STING mimic boosts antitumour immunity via preferential activation of tumour control signalling pathways. Nat. Nanotechnol. 19, 856–866 (2024).
Powderly, J. D. et al. Phase 1/2 study of mRNA-4359 administered alone and in combination with immune checkpoint blockade in adult participants with advanced solid tumors. J. Clin. Oncol. 41, TPS2676 (2023).
Sethna, Z. et al. RNA neoantigen vaccines prime long-lived CD8+ T cells in pancreatic cancer. Nature 639, 1042–1051 (2025).
Lopez, J. et al. Autogene cevumeran with or without atezolizumab in advanced solid tumors: a phase 1 trial. Nat. Med. 31, 152–164 (2025).
Guan, C. et al. RNA-based immunostimulatory liposomal spherical nucleic acids as potent TLR7/8 modulators. Small 14, e1803284 (2018).
Daniel, W. L., Lorch, U., Mix, S. & Bexon, A. S. A first-in-human phase 1 study of cavrotolimod, a TLR9 agonist spherical nucleic acid, in healthy participants: evidence of immune activation. Front. Immunol. 13, 1073777 (2022).
Duurland, C. L. et al. INT-1B3, an LNP formulated miR-193a-3p mimic, promotes anti-tumor immunity by enhancing T cell mediated immune responses via modulation of the tumor microenvironment and induction of immunogenic cell death. Oncotarget 15, 470–485 (2024).
Jang, S. C. et al. ExoSTING, an extracellular vesicle loaded with STING agonists, promotes tumor immune surveillance. Commun. Biol. 4, 497 (2021).
Waldron, J. Codiak files for bankruptcy after exosome-focused biotech unable to satisfy ‘financial needs’. Fierce Biotech (27 March 2023).
Diab, A. et al. Bempegaldesleukin plus nivolumab in untreated advanced melanoma: the open-label, phase III PIVOT IO 001 trial results. J. Clin. Oncol. 41, 4756–4767 (2023).
Diab, A. et al. PIVOT IO 001: first disclosure of efficacy and safety of bempegaldesleukin (BEMPEG) plus nivolumab (NIVO) vs NIVO monotherapy in advanced melanoma (MEL). Ann. Oncol. 33, S901 (2022).
Diab, A. et al. Bempegaldesleukin (NKTR-214) plus nivolumab in patients with advanced solid tumors: phase I dose-escalation study of safety, efficacy, and immune activation (PIVOT-02). Cancer Discov. 10, 1158–1173 (2020).
Diab, A. et al. Immune monitoring after NKTR-214 plus nivolumab (PIVOT-02) in previously untreated patients with metastatic stage IV melanoma. J. Transl. Med. 17 (2019).
Diab, A. et al. Bempegaldesleukin plus nivolumab in first-line metastatic melanoma. J. Clin. Oncol. 39, 2914–2925 (2021).
Alvarez, R. D. et al. A phase II trial of intraperitoneal EGEN-001, an IL-12 plasmid formulated with PEG–PEI–cholesterol lipopolymer in the treatment of persistent or recurrent epithelial ovarian, fallopian tube or primary peritoneal cancer: a Gynecologic Oncology Group study. Gynecol. Oncol. 133, 433–438 (2014).
Stadler, C. R. et al. Preclinical efficacy and pharmacokinetics of an RNA-encoded T cell-engaging bispecific antibody targeting human claudin 6. Sci. Transl. Med. 16, eadl2720 (2024).
Le, N. T. A phase I study to evaluate the safety and tolerability of JCXH-211 (a self-replicating mRNA encoding IL-12) intratumoral injection in patients with malignant solid tumors: results from the phase Ia dose escalation. J. Clin. Oncol. 42, 2539 (2024).
Wang, Z. H. et al. Intravenous administration of IL-12 encoding self-replicating RNA–lipid nanoparticle complex leads to safe and effective antitumor responses. Sci. Rep. 14, 7366 (2024).
Patel, M. et al. 539 Phase 1 study of mRNA-2752, a lipid nanoparticle encapsulating mRNAs encoding human OX40L/IL-23/IL-36γ, for intratumoral (ITu) injection ± durvalumab in advanced solid tumors and lymphoma. J. ImmunoTher. Cancer 9, A569 (2021).
Moderna R&D Day highlights progress and strategic priorities; https://investors.modernatx.com/news/news-details/2024/Moderna-RD-Day-Highlights-Progress-and-Strategic-Priorities/default.aspx (2024).
Bentebibel, S. E. et al. A first-in-human study and biomarker analysis of NKTR-214, a novel IL-2Rβγ-biased cytokine, in patients with advanced or metastatic solid tumors. Cancer Discov. 9, 711–721 (2019).
Hamilton, E. et al. 801 PRIME IL-15 (RPTR-147): preliminary clinical results and biomarker analysis from a first-in-human phase 1 study of IL-15 loaded peripherally-derived autologous T cell therapy in solid tumor patients. J. ImmunoTher. Cancer 8, A479–A480 (2020).
Haanen, J. 611O. Updated results from BNT211-01 (NCT04503278), an ongoing, first-in-human, phase 1 study evaluating safety and efficacy of CLDN6 CAR T cells and a CLDN6-encoding mRNA vaccine in patients with relapsed/refractory CLDN6+ solid tumors. Ann. Oncol. 35, S489–S490 (2024).
MacDiarmid, J. A. et al. Sequential treatment of drug-resistant tumors with targeted minicells containing siRNA or a cytotoxic drug. Nat. Biotechnol. 27, 643–651 (2009).
van Zandwijk, N. et al. Safety and activity of microRNA-loaded minicells in patients with recurrent malignant pleural mesothelioma: a first-in-man, phase 1, open-label, dose-escalation study. Lancet Oncol. 18, 1386–1396 (2017).
Ganju, V. et al. Phase I/IIa trial in advanced pancreatic ductal adenocarcinoma treated with cytotoxic drug-packaged, EGFR-targeted nanocells and glycolipid-packaged nanocells. Clin. Cancer Res. 30, 304–314 (2024).
He, H., Liu, L., Morin, E. E., Liu, M. & Schwendeman, A. Survey of clinical translation of cancer nanomedicines—lessons learned from successes and failures. Acc. Chem. Res. 52, 2445–2461 (2019).
Albanese, A., Tang, P. S. & Chan, W. C. The effect of nanoparticle size, shape, and surface chemistry on biological systems. Annu. Rev. Biomed. Eng. 14, 1–16 (2012).
Faria, M. et al. Minimum information reporting in bio-nano experimental literature. Nat. Nanotechnol. 13, 777–785 (2018).
Leong, H. S. et al. On the issue of transparency and reproducibility in nanomedicine. Nat. Nanotechnol. 14, 629–635 (2019).
Grippin, A. J., Sayour, E. J. & Mitchell, D. A. Translational nanoparticle engineering for cancer vaccines. Oncoimmunology 6, e1290036 (2017).
Mulhopt, S. et al. Characterization of nanoparticle batch-to-batch variability. Nanomaterials 8, 311 (2018).
Kurzrock, R. et al. Moving beyond 3 + 3: the future of clinical trial design. Am. Soc. Clin. Oncol. Educ. Book 41, e133–e144 (2021).
McQuade, J. L., Daniel, C. R., Helmink, B. A. & Wargo, J. A. Modulating the microbiome to improve therapeutic response in cancer. Lancet Oncol. 20, e77–e91 (2019).
Rosshart, S. P. et al. Laboratory mice born to wild mice have natural microbiota and model human immune responses. Science 365, eaaq4361 (2019).
Nejman, D. et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science 368, 973–980 (2020).
Lei, K. et al. Cancer-cell stiffening via cholesterol depletion enhances adoptive T-cell immunotherapy. Nat. Biomed. Eng. 5, 1411–1425 (2021).
Fuhs, T. et al. Rigid tumours contain soft cancer cells. Nat. Phys. 18, 1510–1519 (2022).
Sceneay, J. et al. Interferon signaling is diminished with age and is associated with immune checkpoint blockade efficacy in triple-negative breast cancer. Cancer Discov. 9, 1208–1227 (2019).
Conforti, F. et al. Cancer immunotherapy efficacy and patients’ sex: a systematic review and meta-analysis. Lancet Oncol. 19, 737–746 (2018).
Serpooshan, V. et al. Effect of cell sex on uptake of nanoparticles: the overlooked factor at the nanobio interface. ACS Nano 12, 2253–2266 (2018).
Kim, J., Eygeris, Y., Gupta, M. & Sahay, G. Self-assembled mRNA vaccines. Adv. Drug Deliv. Rev. 170, 83–112 (2021).
Miao, L. et al. Synergistic lipid compositions for albumin receptor mediated delivery of mRNA to the liver. Nat. Commun. 11, 2424 (2020).
Wittrup, A. et al. Visualizing lipid-formulated siRNA release from endosomes and target gene knockdown. Nat. Biotechnol. 33, 870–876 (2015).
Semple, S. C. et al. Rational design of cationic lipids for siRNA delivery. Nat. Biotechnol. 28, 172–176 (2010).
Acknowledgements
We thank C. F. Wogan of the Division of Radiation Oncology at MD Anderson Cancer Center for editing this manuscript. This work is supported, in part, by National Institutes of Health grants R01CA291876, R01CA284108, R01NS117828 and T32-CA196561-08, American Cancer Society grant RSG-22-052-01-IBCD, Cancer Prevention and Research Institute of Texas grants RP240493 and RP250191, the American Brain Tumor Association, the Radiological Society of North America, the Red Gates Foundation and Colorectal Cancer Alliance. The content is solely the responsibility of the authors and does not necessarily represent the official views of the funding sources.
Author information
Authors and Affiliations
Contributions
A.J.G., W.J. and B.Y.S.K. conceived the project and were responsible for all phases of manuscript preparation. A.J.G., D.L., W.J. and B.Y.S.K. wrote the manuscript. A.G. and D.L. designed the figures. All authors searched for literature and edited the manuscript.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Cancer thanks Dmitri Simberg and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Grippin, A.J., Lee, D., Parkes, E.E. et al. Nanotechnology for immuno-oncology. Nat Cancer 6, 1311–1325 (2025). https://doi.org/10.1038/s43018-025-01025-x
Received:
Accepted:
Published:
Version of record:
Issue date:
DOI: https://doi.org/10.1038/s43018-025-01025-x
This article is cited by
-
AI-engineered multifunctional nanoplatforms: synergistically bridging precision diagnosis and intelligent therapy in next-generation oncology
Journal of Nanobiotechnology (2025)


