Abstract
Chimeric antigen receptor (CAR) T cell immunotherapy relies on CAR targeting of tumor-associated antigens; however, heterogenous antigen expression, interpatient variation and off-tumor expression by healthy cells remain barriers. Here we develop synthetic antigens to sensitize solid tumors for recognition and elimination by CAR T cells. Unlike tumor-associated antigens, we design synthetic antigens that are orthogonal to endogenous proteins to eliminate off-tumor targeting and that have a small genetic footprint to facilitate efficient tumor delivery to tumors by lipid nanoparticles. Using a camelid single-___domain antibody (VHH) as a synthetic antigen, we show that adoptive transfer of anti-VHH CAR T cells to female mice bearing VHH-expressing tumors reduced tumor burden in multiple syngeneic and xenograft models of cancer, improved survival, induced epitope spread, protected against tumor rechallenge and mitigated antigen escape in heterogenous tumors. Our work supports the in situ delivery of synthetic antigens to treat antigen-low or antigen-negative tumors with CAR T cells.
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
27,99 € / 30 days
cancel any time
Subscribe to this journal
Receive 12 digital issues and online access to articles
118,99 € per year
only 9,92 € per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout







Similar content being viewed by others
Data availability
All raw IVIS images for Extended Data Fig. 8 are available from the Georgia Institute of Technology Research Repository ([email protected]) upon publication of this paper. The remaining data are available within the article and Supplementary Information or upon reasonable request to the corresponding author. Source data are provided with this paper.
References
Lim, W. A. & June, C. H. The principles of engineering immune cells to treat cancer. Cell 168, 724–740 (2017).
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).
Hou, A. J., Chen, L. C. & Chen, Y. Y. Navigating CAR-T cells through the solid-tumour microenvironment. Nat. Rev. Drug Discov. 20, 531–550 (2021).
Martinez, M. & Moon, E. K. CAR T cells for solid tumors: new strategies for finding, infiltrating, and surviving in the tumor microenvironment. Front. Immunol. 10, 128 (2019).
Majzner, R. G. & Mackall, C. L. Tumor antigen escape from CAR T-cell therapy. Cancer Discov. 8, 1219–1226 (2018).
Majzner, R. G. et al. Tuning the antigen density requirement for CAR T-cell activity. Cancer Discov. 10, 702–723 (2020).
Hamieh, M. et al. CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape. Nature 568, 112–116 (2019).
Wudhikarn, K. et al. Infection during the first year in patients treated with CD19 CAR T cells for diffuse large B cell lymphoma. Blood Cancer J. 10, 79 (2020).
Nahas, G. et al. Persistent cytopenias after chimeric antigen receptor t-cell immunotherapy for CD19+ aggressive lymphoma: a single institution experience. Biol. Blood Marrow Transplant. 25, S180 (2019).
Fried, S. et al. Early and late hematologic toxicity following CD19 CAR-T cells. Bone Marrow Transplant. 54, 1643–1650 (2019).
Parker, K. R. et al. Single-cell analyses identify brain mural cells expressing CD19 as potential off-tumor targets for CAR-T immunotherapies. Cell 183, 126–142.e17 (2020).
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).
Ramakrishna, S., Barsan, V. & Mackall, C. Prospects and challenges for use of CAR T cell therapies in solid tumors. Expert Opin. Biol. Ther. 20, 503–516 (2020).
Wang, G. et al. Multiplexed activation of endogenous genes by CRISPRa elicits potent antitumor immunity. Nat. Immunol. 20, 1494–1505 (2019).
Cho, J. H., Collins, J. J. & Wong, W. W. Universal chimeric antigen receptors for multiplexed and logical control of T cell responses. Cell 173, 1426–1438.e11 (2018).
Roybal, K. T. et al. Precision tumor recognition by T cells with combinatorial antigen-sensing circuits. Cell 164, 770–779 (2016).
Lohmueller, J. J., Ham, J. D., Kvorjak, M. & Finn, O. J. mSA2 affinity-enhanced biotin-binding CAR T cells for universal tumor targeting. Oncoimmunology 7, e1368604 (2017).
Lee, Y. G. et al. Use of a single CAR T cell and several bispecific adapters facilitates eradication of multiple antigenically different solid tumors. Cancer Res. 79, 387–396 (2019).
Landgraf, K. E. et al. convertibleCARs: a chimeric antigen receptor system for flexible control of activity and antigen targeting. Commun. Biol. 3, 296 (2020).
Miller, I. C. et al. Enhanced intratumoural activity of CAR T cells engineered to produce immunomodulators under photothermal control. Nat. Biomed. Eng. 5, 1348–1359 (2021).
Zhang, A. Q. et al. Universal redirection of CAR T cells against solid tumours via membrane-inserted ligands for the CAR. Nat. Biomed. Eng. 7, 1113–1128 (2023).
Park, A. K. et al. Effective combination immunotherapy using oncolytic viruses to deliver CAR targets to solid tumors. Sci. Transl. Med. 12, eaaz1863 (2020).
Aalipour, A. et al. Viral delivery of CAR targets to solid tumors enables effective cell therapy. Mol. Ther. Oncolytics 17, 232–240 (2020).
Vincent, R. L. et al. Probiotic-guided CAR-T cells for solid tumor targeting. Science 382, 211–218 (2023).
Kunz, P. et al. The structural basis of nanobody unfolding reversibility and thermoresistance. Sci. Rep. 8, 7934 (2018).
Ingram, J. R., Schmidt, F. I. & Ploegh, H. L. Exploiting nanobodies’ singular traits. Annu. Rev. Immunol. 36, 695–715 (2018).
Peyvandi, F. et al. Caplacizumab for acquired thrombotic thrombocytopenic purpura. N. Engl. J. Med. 374, 511–522 (2016).
Tiwari, P. M. et al. Engineered mRNA-expressed antibodies prevent respiratory syncytial virus infection. Nat. Commun. 9, 3999 (2018).
Movahedi, K. et al. Nanobody-based targeting of the macrophage mannose receptor for effective in vivo imaging of tumor-associated macrophages. Cancer Res. 72, 4165–4177 (2012).
Keyaerts, M. et al. Phase I study of 68Ga-HER2-nanobody for PET/CT assessment of HER2 expression in breast carcinoma. J. Nucl. Med. 57, 27–33 (2016).
Nchinda, G. et al. The efficacy of DNA vaccination is enhanced in mice by targeting the encoded protein to dendritic cells. J. Clin. Invest. 118, 1427–1436 (2008).
Idoyaga, J. et al. Comparable T helper 1 (Th1) and CD8 T-cell immunity by targeting HIV gag p24 to CD8 dendritic cells within antibodies to Langerin, DEC205, and Clec9A. Proc. Natl Acad. Sci. USA 108, 2384–2389 (2011).
Xie, Y. J. et al. Nanobody-based CAR T cells that target the tumor microenvironment inhibit the growth of solid tumors in immunocompetent mice. Proc. Natl Acad. Sci. USA 116, 7624–7631 (2019).
Tanenbaum, M. E., Gilbert, L. A., Qi, L. S., Weissman, J. S. & Vale, R. D. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell 159, 635–646 (2014).
Hussack, G., Hirama, T., Ding, W., Mackenzie, R. & Tanha, J. Engineered single-___domain antibodies with high protease resistance and thermal stability. PLoS ONE 6, e28218 (2011).
Pinaud, F. & Dahan, M. Targeting and imaging single biomolecules in living cells by complementation-activated light microscopy with split-fluorescent proteins. Proc. Natl Acad. Sci. USA 108, E201–E210 (2011).
Du, Y., Pattnaik, A. K., Song, C., Yoo, D. & Li, G. Glycosyl-phosphatidylinositol (GPI)-anchored membrane association of the porcine reproductive and respiratory syndrome virus GP4 glycoprotein and its co-localization with CD163 in lipid rafts. Virology 424, 18–32 (2012).
Nickells, M. W., Alvarez, J. I., Lublin, D. M. & Atkinson, J. P. Characterization of DAF-2, a high molecular weight form of decay-accelerating factor (DAF; CD55), as a covalently cross-linked dimer of DAF-1. J. Immunol. 152, 676–685 (1994).
Rossey, I. et al. Potent single-___domain antibodies that arrest respiratory syncytial virus fusion protein in its prefusion state. Nat. Commun. 8, 14158 (2017).
Zehn, D., Cohen, C. J., Reiter, Y. & Walden, P. Extended presentation of specific MHC–peptide complexes by mature dendritic cells compared to other types of antigen-presenting cells. Eur. J. Immunol. 34, 1551–1560 (2004).
Yadav, M. et al. Predicting immunogenic tumour mutations by combining mass spectrometry and exome sequencing. Nature 515, 572–576 (2014).
Yin, L., Duan, J.-J., Bian, X.-W. & Yu, S.-C. Triple-negative breast cancer molecular subtyping and treatment progress. Breast Cancer Res. 22, 61 (2020).
Dees, S., Ganesan, R., Singh, S. & Grewal, I. S. Emerging CAR-T cell therapy for the treatment of triple-negative breast cancer. Mol. Cancer Ther. 19, 2409–2421 (2020).
Johnstone, C. N. et al. Functional and molecular characterisation of EO771.LMB tumours, a new C57BL/6-mouse-derived model of spontaneously metastatic mammary cancer. Dis. Model. Mech. 8, 237–251 (2015).
Wang, K., Wei, G. & Liu, D. CD19: a biomarker for B cell development, lymphoma diagnosis and therapy. Exp. Hematol. Oncol. 1, 36 (2012).
Dogan, A. et al. B-cell maturation antigen expression across hematologic cancers: a systematic literature review. Blood Cancer J. 10, 73 (2020).
Viardot, A., Locatelli, F., Stieglmaier, J., Zaman, F. & Jabbour, E. Concepts in immuno-oncology: tackling B cell malignancies with CD19-directed bispecific T cell engager therapies. Ann. Hematol. 99, 2215–2229 (2020).
Gerdes, M. J. et al. Emerging understanding of multiscale tumor heterogeneity. Front. Oncol. 4, 366 (2014).
Restle, D. et al. Organ-specific heterogeneity in tumor-infiltrating immune cells and cancer antigen expression in primary and autologous metastatic lung adenocarcinoma. J. Immunother. Cancer 11, e006609 (2023).
Chen, N., Li, X., Chintala, N. K., Tano, Z. E. & Adusumilli, P. S. Driving CARs on the uneven road of antigen heterogeneity in solid tumors. Curr. Opin. Immunol. 51, 103–110 (2018).
Sterner, R. C. & Sterner, R. M. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J. 11, 69 (2021).
El-Sayes, N., Vito, A. & Mossman, K. Tumor heterogeneity: a great barrier in the age of cancer immunotherapy. Cancers 13, 806 (2021).
Genoud, V. et al. Responsiveness to anti-PD-1 and anti-CTLA-4 immune checkpoint blockade in SB28 and GL261 mouse glioma models. Oncoimmunology 7, e1501137 (2018).
O’Rourke, D. M. et al. A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci. Transl. Med. 9, eaaa0984 (2017).
Seaman, B. J. et al. Audiovestibular dysfunction associated with adoptive cell immunotherapy for melanoma. Otolaryngol. Head Neck Surg. 147, 744–749 (2012).
Lamers, C. H. et al. Treatment of metastatic renal cell carcinoma with CAIX CAR-engineered T cells: clinical evaluation and management of on-target toxicity. Mol. Ther. 21, 904–912 (2013).
Nomura, N. et al. Prostate specific membrane antigen (PSMA) expression in primary gliomas and breast cancer brain metastases. Cancer Cell Int. 14, 26 (2014).
Gust, J., Taraseviciute, A. & Turtle, C. J. Neurotoxicity associated with CD19-targeted CAR-T cell therapies. CNS Drugs 32, 1091–1101 (2018).
Ruella, M. & June, C. H. Predicting dangerous rides in CAR T cells: bridging the gap between mice and humans. Mol. Ther. 26, 1401–1403 (2018).
Richman, S. A. et al. High-affinity GD2-specific CAR T cells induce fatal encephalitis in a preclinical neuroblastoma model. Cancer Immunol. Res. 6, 36–46 (2018).
Labanieh, L. et al. Enhanced safety and efficacy of protease-regulated CAR-T cell receptors. Cell 185, 1745–1763.e22 (2022).
Liu, X. et al. Affinity-tuned ErbB2 or EGFR chimeric antigen receptor T cells exhibit an increased therapeutic index against tumors in mice. Cancer Res. 75, 3596–3607 (2015).
Hernandez-Lopez, R. A. et al. T cell circuits that sense antigen density with an ultrasensitive threshold. Science 371, 1166–1171 (2021).
Singh, N. et al. Single chain variable fragment linker length regulates CAR biology and T cell efficacy. Blood 134, 247 (2019).
Millar, D. G. et al. Antibody-mediated delivery of viral epitopes to tumors harnesses CMV-specific T cells for cancer therapy. Nat. Biotechnol. 38, 420–425 (2020).
Marabelle, A., Tselikas, L., de Baere, T. & Houot, R. Intratumoral immunotherapy: using the tumor as the remedy. Ann. Oncol. 28, xii33–xii43 (2017).
Melero, I., Castanon, E., Alvarez, M., Champiat, S. & Marabelle, A. Intratumoural administration and tumour tissue targeting of cancer immunotherapies. Nat. Rev. Clin. Oncol. 18, 558–576 (2021).
Hong, W. X. et al. Intratumoral immunotherapy for early-stage solid tumors. Clin. Cancer Res. 26, 3091–3099 (2020).
Hammerich, L., Binder, A. & Brody, J. D. In situ vaccination: cancer immunotherapy both personalized and off-the-shelf. Mol. Oncol. 9, 1966–1981 (2015).
Johnson, D. B., Puzanov, I. & Kelley, M. C. Talimogene laherparepvec (T-VEC) for the treatment of advanced melanoma. Immunotherapy 7, 611–619 (2015).
Yang, T. Y. et al. Immunogenicity assessment of AAV-based gene therapies: an IQ consortium industry white paper. Mol. Ther. Methods Clin. Dev. 26, 471–494 (2022).
Anselmo, A. C. & Mitragotri, S. Nanoparticles in the clinic: an update. Bioeng. Transl. Med. 4, e10143 (2019).
Shirley, J. L., de Jong, Y. P., Terhorst, C. & Herzog, R. W. Immune responses to viral gene therapy vectors. Mol. Ther. 28, 709–722 (2020).
Chen, R. et al. Engineering circular RNA for enhanced protein production. Nat. Biotechnol. 41, 262–272 (2023).
Bloom, K., van den Berg, F. & Arbuthnot, P. Self-amplifying RNA vaccines for infectious diseases. Gene Ther. 28, 117–129 (2021).
Hou, X., Zaks, T., Langer, R. & Dong, Y. Lipid nanoparticles for mRNA delivery. Nat. Rev. Mater. 6, 1078–1094 (2021).
Huayamares, S. G. et al. High-throughput screens identify a lipid nanoparticle that preferentially delivers mRNA to human tumors in vivo. J. Control. Release 357, 394–403 (2023).
Johnson, L. A. et al. Rational development and characterization of humanized anti-EGFR variant III chimeric antigen receptor T cells for glioblastoma. Sci. Transl. Med. 7, 275ra222 (2015).
Schwartz, L. H. et al. RECIST 1.1—update and clarification: from the RECIST committee. Eur. J. Cancer 62, 132–137 (2016).
Acknowledgements
We thank D. R. Meyers (Emory) and C. D. Sago for helpful insights. This work was funded by the National Institutes of Health (NIH) Director’s New Innovator Award (DP2-HD091793), NIH Director’s Pioneer Award (DP1-CA280832), National Cancer Institute (R01CA273878), National Institute of Biomedical Imaging and Bioengineering (R01EB032822), National Center for Advancing Translational Sciences (UL1TR000454), Shurl and Kay Curci Foundation and NIH Shared Instrumentation Grant (1S10OD016264-01A1) and was partially performed at the Georgia Tech Institute for Nanotechnology, a member of the National Nanotechnology Coordinated Infrastructure, which is supported by the National Science Foundation (NSF; grant ECCS-1542174). L.G. was supported by the Alfred P. Sloan Foundation and the NIH GT BioMAT Training Grant under award number 5T32EB006343. L.G., A.S. and S.N.D. were supported by the NSF Graduate Research Fellowship under grant number DGE-1451512. A.H.Z. was supported by the NIH Kirschstein National Research Service Award program under award number F31-CA271803. C.A.T. was supported by the NIH Cell and Tissue Engineering training program under grant number 5T32GM145735. F.S. was supported by a postdoctoral fellowship from the Wallace H. Coulter Department of Biomedical Engineering and the College of Engineering at Peking University. This content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.
Author information
Authors and Affiliations
Contributions
L.G., A.H.Z. and G.A.K. designed the study and wrote the paper. L.G., A.H.Z., C.A.T., H.J.L. and Z.Z. analyzed the data and generated the figures. L.G., A.H.Z., C.A.T., H.J.L., E.K., Z.Z., N.S.C., C.S.C., S.F., S.A.O., A.S., S.N.D., H.P. and A.M.H. performed the experiments. L.G., A.H.Z., P.J.S. and G.A.K. supervised the studies. D.V., H.E.P. and F.S. contributed to the formulation and production of LNPs and provided helpful discussions. All authors reviewed the paper before submission.
Corresponding author
Ethics declarations
Competing interests
G.A.K. reports equity or consulting roles for Sunbird Bio, Port Therapeutics, Send Biotherapeutics and Ridge Biotechnologies. The terms of this arrangement were reviewed and approved by Georgia Tech in accordance with its conflict-of-interest policies. L.G., A.H.Z., D.V., P.J.S., C.S.C., H.J.L., C.A.T. and G.A.K. are listed as inventors on a patent application related to the results of this paper, titled ‘Synthetic Antigens as Chimeric Antigen Receptor (CAR) Ligands and Uses Thereof’ (US20230390335A1). The patent applicant is the Georgia Tech Research Corporation.
Peer review
Peer review information
Nature Cancer thanks Jan Melenhorst 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.
Extended data
Extended Data Fig. 1 Kinetics of expression of Kb-SIINFEKL pMHC complex.
Surface expression kinetics of the Kb-SIINFEKL pMHC complex on the surface of MC38 tumor cells following peptide pulsing with the SIINFEKL peptide for one hour at room temperature. Mean of n = 3 technical replicates from a single experiment.
Extended Data Fig. 2 Kinetics of expression of LNP mediated delivery of VHH synthetic antigen to tumor cells.
VHH expression on MDA-MB-468 (left) and E0771 (right) following VHH-LNP treatment at the indicated doses. VHH mean fluorescent intensity (MFI) quantified by flow cytometry at indicated days after transfection. Mean of n = 4 technical replicates from a single experiment.
Extended Data Fig. 3 Characterization of murine CAR T cells sourced from either pmel-1 or C57BL6/J mice.
(a) Staining of indicated murine T cell population with activation markers CD25 and CD69 following co-incubation with ST- or VHH-expressing E0771 tumor cells. Data is representative of 3 technical replicates from a single experiment. (b) IFN-γ secretion by Pmel-1 or C57BL/6J derived αVHH CAR T cells quantified following a 24 hr coculture with MC38-VHH tumor cells at a 1:1 E:T ratio. Mean of n = 3 technical replicates from a single experiment. (c) Cytotoxicity of Pmel-1 and C57BL/6 derived αVHH CAR T cells were assessed following a 24 hr coculture with MC38-VHH at a 1:1 E:T ratio. Mean of n = 3 technical replicates from a single experiment.
Extended Data Fig. 4 Murine αVHH CAR T cells are well tolerated by immunocompetent mice.
(a) Blood serum analysis 7 d post i.v. administration of αVHH CAR T cells, untransduced T cells, or saline into naïve C57BL6/J mice. One-way ANOVA, mean ± s.d., n = 4 mice.; n.s. = not significant. (b) Body weight measurements following i.v. administration of αVHH CAR T cells, wild-type T cells, or saline into naïve C57BL6/J mouse. Two-way ANOVA, mean ± s.d., n = 5 mice per cohort; n.s. = not significant. (c) PBMC analysis before and 3 days after ACT of saline, WT, αVHH CAR, or αCD19 CAR T cells into C57BL6/J mice. One-way ANOVA; mean ± s.e.m., n = 4 mice for αCD19 CAR treated mice, n = 5 mice in all other cohorts; n.s. = not significant ***p = 0.0002. (d) Blood serum analysis 3 d post i.v. administration of saline, WT, αVHH CAR, or αhHER2 CAR T cells into naïve B6-HER2 mice. One-way ANOVA; mean ± s.e.m., n = 3 mice for hHER2 and VHH treatments, n = 4 mice for Saline and WT treatments.; *p = 0.01, **p = 0.0065.
Extended Data Fig. 5 VHH expression alone does not alter tumor growth.
(a) VHH expression on wildtype or transduced MC38 and E0771 tumor cells. Data is representative of 2 independent experiments. (b) Tumor growth curves of wildtype MC38 (MC38-WT) or MC38 cells transduced to stably express VHH (MC38-VHH) without adoptive cell transfer of αVHH CAR T cells. n = 4 mice inoculated with MC38-WT and n = 3 mice inoculated with MC38-VHH. Two-way ANOVA, mean ± s.e.m. is depicted; n.s. = not significant (c) Tumor growth curves of wildtype E0771 (E0771-WT) or E0771 cells transduced to stably express VHH (E0771-VHH) without adoptive cell transfer of αVHH CAR T cells n = 4 mice inoculated with E0771-WT and n = 6 mice inoculated with E0771-VHH; Two-way ANOVA, mean ± s.e.m. is depicted; n.s. = not significant.
Extended Data Fig. 6 Adoptive transfer of αVHH CAR T cells to mice bearing wildtype tumors does not alter tumor growth.
Tumor growth curves of mice bearing wiltdype E0771 tumors treated intravenously with either saline, 5 × 106 untransduced (UTD) or αVHH CAR T cells. Two-way ANOVA, mean ± s.e.m., n = 5 mice; n.s. = not significant.
Extended Data Fig. 7 The antitumor activity αVHH CAR T cells is comparable to αHER2 CAR T cells against VHH + HER2+ tumors.
(a) Representative flow plots of HER2 and VHH expression on either wildtype or 468-HV tumor cells. Data is representative from a single experiment, repeated 2 times with similar results. (b) 468-HV tumor cells were cocultured with either human untransduced (UTD), αHER2 CAR, or αVHH CAR T cells at a 2:1 E:T ratio and assessed for cytotixicity after 24 hrs (Mean of n = 3 technical replicates) and (c) growth curves of 468-HV tumors following treatment with the UTD, αHER2 CAR, or αVHH CAR T cells. (Two-way ANOVA, mean ± s.e.m., n = 6 mice per cohort, αVHH (**p = 0.0080 on day 6, **p = 0.0015 on day 13, ***p = 0.0003 on day 18, ***p = 0.0005 on day 25, ***p = 0.0003 on day 33) and αHER2 (††p = 0.0054 on day 6, ††p = 0.0016 on day 13, †††p = 0.0003 on day 18, †††p = 0.0005 on day 25, †††p = 0.0003 on day 33) CAR T cell treatments are compared to UTD.
Extended Data Fig. 8 In vivo biodistribution of functional mRNA delivery mediated by CKK-LNPs following intratumoral injection.
(a) In vivo biodistribution following saline or a 2 µg intratumoral injection of nLuc mRNA-loaded LNPs (nLuc-LNPs) as measured by IVIS 24 hrs following injection. Luminesence in organs quantified (left) and representative (of n = 3 organs, each from individual mice) images are displayed on the right. mean ± s.d., multiple unpaired t test, n = 3 organs, each from individual mice; *p = 0.0397, ***p = 0.00047, ****p < 0.0001. (b) In vivo kinetics of nLuc expression following one (light blue) or two (dark blue) intratumoral injections of 5 µg nLuc-LNPs. Transfection kinetics quantified by IVIS. Two-way ANOVA, mean ± s.e.m., n = 4 mice; *p = 0.0196, **p = 0.0080.
Extended Data Fig. 9 αVHH CAR T cells reduce tumor burden of VHH LNP injected tumors.
(a) Tumor growth curves of wildtype E0771 (E0771-wt) tumors transfected with VHH-LNPs (CKK-E12) by intratumoral injection of 5 µg mRNA-loaded LNPs without adoptive cell transfer of αVHH CAR T cells. Two-way ANOVA, mean ± s.e.m. is depicted; n = 6 saline injected mice and n = 5 for VHH-LNP injected mice.; n.s. = not significant. (b) Tumor growth curves of wildtype E0771 tumor-bearing mice treated with intratumorally with either saline, Fluc-LNP or VHH-LNP and αVHH CAR T cells on days indicated by a dashed line (D0, D7, D14, D21, and D28). Two-way ANOVA, mean ± s.e.m., n = 5 mice. Saline vs. VHH-LNP: *p = 0.0176, ***p = 0.0006, ****p < 0.0001.
Extended Data Fig. 10 LNP-mediated synthetic antigen treatment augments antitumor immunity in a preclinical model of heterogenous glioblastoma tumors.
(a) EGFRvIII- (vIII-) or EGFRvIII+ (vIII+) SB28 tumor cells were transfected with 100 ng of VHH-LNP and evaluated for VHH expression by flow cytometry after 18 hrs, mean of n = 5 technical replicates from a single experiment (b) Mice bearing a heterogenous tumor mixture comprised of 90% vIII- and 10% vIII+ SB28 tumor cells were treated with saline or VHH-LNP followed by adoptive transfer of indicated CAR T cells (c) 24 hrs following intratumoral injection VHH-LNP (5 µg), SB28 tumors were dissociated and analyzed by flow cytometry for EGFRvIII (left) and VHH (right) expression, one-way unpaired T test, mean ± s.d., n = 3 dissociated tumors, each from individual mice; ***p = 0.0008, n.s.=nonsignficiant. (d) Representative flow plots of VHH expression on SB28 tumors treated with either saline or VHH-LNP. Data is representative of 3 tumors each isolated from one mouse (e) Tumor growth curves of heterogenous SB28 tumor-bearing mice treated with intratumorally with saline or VHH-LNP and systemically with either WT, αVHH, or αEGFRvIII CAR T cells days indicated days by dashed lines (D0, D7, & D14) n = 5 untreated and VHH-LNP + αVHH CAR treated mice, n = 6 mice with αEGFRvIII CAR T cell treatment, n = 7 mice with VHH-LNP + saline treatment. (f) Survival curves from (e) of heterogenous SB28 tumor-bearing mice following synthetic antigen treatment, log-rank (Mantel–Cox) test; **p = 0.0019 comparing EGFRvIII CAR treatment with LNP + αVHH CAR.
Supplementary information
Supplementary Information
Supplementary Figs. 1–6 and Tables 1–3.
Source data
Source Data Fig. 1
Statistical source data.
Source Data Fig. 2
Statistical source data.
Source Data Fig. 3
Statistical source data.
Source Data Fig. 4
Statistical source data.
Source Data Fig. 5
Statistical source data.
Source Data Fig. 6
Statistical source data.
Source Data Fig. 7
Statistical source data.
Source Data Extended Data Fig. 1
Statistical source data.
Source Data Extended Data Fig. 2
Statistical source data.
Source Data Extended Data Fig. 3
Statistical source data.
Source Data Extended Data Fig. 4
Statistical source data.
Source Data Extended Data Fig. 5
Statistical source data.
Source Data Extended Data Fig. 6
Statistical source data.
Source Data Extended Data Fig. 7
Statistical source data.
Source Data Extended Data Fig. 8
Statistical source data.
Source Data Extended Data Fig. 9
Statistical source data.
Source Data Extended Data Fig. 10
Statistical source data.
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
Gamboa, L., Zamat, A.H., Thiveaud, C.A. et al. Sensitizing solid tumors to CAR-mediated cytotoxicity by lipid nanoparticle delivery of synthetic antigens. Nat Cancer 6, 1073–1087 (2025). https://doi.org/10.1038/s43018-025-00968-5
Received:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s43018-025-00968-5