Abstract
Tumors evolve to avoid immune destruction and establish an immunosuppressive microenvironment. Syngeneic mouse tumor models are critical for understanding tumor immune evasion and testing cancer immunotherapy. Derived from established mouse tumor cell lines that can already evade the immune system, these models cannot simulate early phases of immunoediting during initial tumorigenesis. We developed a syngeneic mouse teratoma model derived from noncancerous mouse embryonic stem cells and conducted a genome-wide CRISPR screen to identify genes that impact early phases of cancer immunoediting. We found that loss of pro-apoptotic tumor suppressor genes, including Trp53, increased necrosis in teratomas, releasing APOE lipid particles into the extracellular milieu. Infiltrating T cells drawn to tumor necrotic regions accumulated lipids and became dysfunctional. Blocking lipid uptake in T cells or reducing necrosis in teratomas by inactivating the mitochondrial permeability transition pore (mPTP) restored immunosurveillance. Because mouse teratomas were highly enriched for brain tissues, we next examined the tumor-immune interaction in human glioblastoma (GBM). Indeed, infiltrating T cells in TP53-mutated human GBM accumulated APOE and were dysfunctional. Anti-APOE and anti-PDCD1 antibodies synergistically boosted anti-GBM immunity and prolonged survival in mice. Our results link mPTP-mediated tumor necrosis to immune evasion and suggest that targeting the uptake of lipids released by necrotic tumor cells by infiltrating immune cells can enhance cancer immunotherapy.
This is a preview of subscription content, access via your institution
Access options
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 full article PDF
Prices may be subject to local taxes which are calculated during checkout







Similar content being viewed by others
Data availability
The omics data and analyses have been deposited to FigShare with the dataset collection identifier 6929467 or through the following link https://figshare.com/account/home#/collections/6929467, including datasets of CRISPR screening in mouse teratomas (https://doi.org/10.6084/m9.figshare.24559687); scRNA-seq datasets of mouse teratomas (https://doi.org/10.6084/m9.figshare.27314364), mouse GBM (https://doi.org/10.6084/m9.figshare.27314304), and lipidomics results of teratoma interstitial fluid (https://doi.org/10.6084/m9.figshare.28703174). The raw data of scRNA-seq for Chinese GBM patients have been uploaded to Genome Sequence Archive for Human (https://ngdc.cncb.ac.cn/gsa-human/browse/HRA012282).
References
Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).
Hanahan, D. Hallmarks of cancer: new dimensions. Cancer Discov. 12, 31–46 (2022).
Dunn, G. P., Bruce, A. T., Ikeda, H., Old, L. J. & Schreiber, R. D. Cancer immunoediting: from immunosurveillance to tumor escape. Nat. Immunol. 3, 991–998 (2002).
Dunn, G. P., Old, L. J. & Schreiber, R. D. The three Es of cancer immunoediting. Annu. Rev. Immunol. 22, 329–360 (2004).
Vesely, M. D., Kershaw, M. H., Schreiber, R. D. & Smyth, M. J. Natural innate and adaptive immunity to cancer. Annu. Rev. Immunol. 29, 235–271 (2011).
Schreiber, R. D., Old, L. J. & Smyth, M. J. Cancer immunoediting: integrating immunity’s roles in cancer suppression and promotion. Science 331, 1565–1570 (2011).
Gubin, M. M. & Vesely, M. D. Cancer immunoediting in the era of immuno-oncology. Clin. Cancer Res. 28, 3917–3928 (2022).
Jhunjhunwala, S., Hammer, C. & Delamarre, L. Antigen presentation in cancer: insights into tumour immunogenicity and immune evasion. Nat. Rev. Cancer 21, 298–312 (2021).
McGranahan, N. et al. Allele-specific HLA loss and immune escape in lung cancer evolution. Cell 171, 1259–1271.e11 (2017).
He, X. & Xu, C. Immune checkpoint signaling and cancer immunotherapy. Cell Res. 30, 660–669 (2020).
Sharma, P. et al. Immune checkpoint therapy-current perspectives and future directions. Cell 186, 1652–1669 (2023).
Taylor, A., Verhagen, J., Blaser, K., Akdis, M. & Akdis, C. A. Mechanisms of immune suppression by interleukin-10 and transforming growth factor-beta: the role of T regulatory cells. Immunology 117, 433–442 (2006).
Seruga, B., Zhang, H., Bernstein, L. J. & Tannock, I. F. Cytokines and their relationship to the symptoms and outcome of cancer. Nat. Rev. Cancer 8, 887–899 (2008).
Ma, X. et al. Cholesterol induces CD8(+) T cell exhaustion in the tumor microenvironment. Cell Metab. 30, 143–156.e5 (2019).
Hicks, K. C., Tyurina, Y. Y., Kagan, V. E. & Gabrilovich, D. I. Myeloid cell-derived oxidized lipids and regulation of the tumor microenvironment. Cancer Res. 82, 187–194 (2022).
Allinen, M. et al. Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell 6, 17–32 (2004).
Hanahan, D. & Coussens, L. M. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell 21, 309–322 (2012).
McAllister, S. S. & Weinberg, R. A. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat. Cell Biol. 16, 717–727 (2014).
Xia, A., Zhang, Y., Xu, J., Yin, T. & Lu, X. J. T Cell dysfunction in cancer immunity and immunotherapy. Front. Immunol. 10, 1719 (2019).
Zeng, Z. et al. TISMO: syngeneic mouse tumor database to model tumor immunity and immunotherapy response. Nucleic Acids Res. 50, D1391–D1397 (2022).
Chen, S. et al. Genome-wide CRISPR screen in a mouse model of tumor growth and metastasis. Cell 160, 1246–1260 (2015).
Sun, Y. et al. Targeting TBK1 to overcome resistance to cancer immunotherapy. Nature 615, 158–167 (2023).
Chow, R. D. et al. AAV-mediated direct in vivo CRISPR screen identifies functional suppressors in glioblastoma. Nat. Neurosci. 20, 1329–1341 (2017).
Martin, T. D. et al. The adaptive immune system is a major driver of selection for tumor suppressor gene inactivation. Science 373, 1327–1335 (2021).
Evans, M. J. & Kaufman, M. H. Establishment in culture of pluripotential cells from mouse embryos. Nature 292, 154–156 (1981).
McDonald, D. et al. Defining the teratoma as a model for multi-lineage human development. Cell 183, 1402–1419.e18 (2020).
Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat. Biotechnol. 34, 184–191 (2016).
Muller, F. J., Goldmann, J., Loser, P. & Loring, J. F. A call to standardize teratoma assays used to define human pluripotent cell lines. Cell Stem Cell 6, 412–414 (2010).
Wang, B. et al. Integrative analysis of pooled CRISPR genetic screens using MAGeCKFlute. Nat. Protoc. 14, 756–780 (2019).
Zhang, D., Zaugg, K., Mak, T. W. & Elledge, S. J. A role for the deubiquitinating enzyme USP28 in control of the DNA-damage response. Cell 126, 529–542 (2006).
Cuella-Martin, R. et al. 53BP1 integrates DNA repair and p53-dependent cell fate decisions via distinct mechanisms. Mol. Cell 64, 51–64 (2016).
Kastenhuber, E. R. & Lowe, S. W. Putting p53 in Context. Cell 170, 1062–1078 (2017).
el-Deiry, W. S. et al. WAF1, a potential mediator of p53 tumor suppression. Cell 75, 817–825 (1993).
Nakano, K. & Vousden, K. H. PUMA, a novel proapoptotic gene, is induced by p53. Mol. Cell 7, 683–694 (2001).
Aubrey, B. J., Kelly, G. L., Janic, A., Herold, M. J. & Strasser, A. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression?. Cell Death Differ. 25, 104–113 (2018).
McArthur, K. et al. BAK/BAX macropores facilitate mitochondrial herniation and mtDNA efflux during apoptosis. Science 359, eaao6047 (2018).
Wei, M. C. et al. Proapoptotic BAX and BAK: a requisite gateway to mitochondrial dysfunction and death. Science 292, 727–730 (2001).
Vandenabeele, P., Bultynck, G. & Savvides, S. N. Pore-forming proteins as drivers of membrane permeabilization in cell death pathways. Nat. Rev. Mol. Cell Biol. 24, 312–333 (2023).
Stambolic, V. et al. Negative regulation of PKB/Akt-dependent cell survival by the tumor suppressor PTEN. Cell 95, 29–39 (1998).
Lee, Y. R., Chen, M. & Pandolfi, P. P. The functions and regulation of the PTEN tumour suppressor: new modes and prospects. Nat. Rev. Mol. Cell Biol. 19, 547–562 (2018).
Mancias, J. D., Wang, X., Gygi, S. P., Harper, J. W. & Kimmelman, A. C. Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature 509, 105–109 (2014).
Blagih, J., Buck, M. D. & Vousden, K. H. p53, cancer and the immune response. J. Cell Sci. 133, jcs237453 (2020).
Kather, J. N. et al. Topography of cancer-associated immune cells in human solid tumors. eLife 7, e36967 (2018).
Smith-Garvin, J. E., Koretzky, G. A. & Jordan, M. S. T cell activation. Annu. Rev. Immunol. 27, 591–619 (2009).
Philip, M. & Schietinger, A. CD8(+) T cell differentiation and dysfunction in cancer. Nat. Rev. Immunol. 22, 209–223 (2022).
Wang, M. & Kaufman, R. J. Protein misfolding in the endoplasmic reticulum as a conduit to human disease. Nature 529, 326–335 (2016).
Han, J. et al. ER-stress-induced transcriptional regulation increases protein synthesis leading to cell death. Nat. Cell Biol. 15, 481–490 (2013).
Han, J. & Kaufman, R. J. The role of ER stress in lipid metabolism and lipotoxicity. J. Lipid Res. 57, 1329–1338 (2016).
Xu, S. et al. Uptake of oxidized lipids by the scavenger receptor CD36 promotes lipid peroxidation and dysfunction in CD8(+) T cells in tumors. Immunity 54, 1561–1577.e7 (2021).
Manzo, T. et al. Accumulation of long-chain fatty acids in the tumor microenvironment drives dysfunction in intrapancreatic CD8+ T cells. J. Exp. Med. 217, e20191920 (2017).
Yang, W. et al. Potentiating the antitumour response of CD8(+) T cells by modulating cholesterol metabolism. Nature 531, 651–655 (2016).
Noy, R. & Pollard, J. W. Tumor-associated macrophages: from mechanisms to therapy. Immunity 41, 49–61 (2014).
Seimon, T. A. et al. Atherogenic lipids and lipoproteins trigger CD36-TLR2-dependent apoptosis in macrophages undergoing endoplasmic reticulum stress. Cell Metab. 12, 467–482 (2010).
Di Conza, G. et al. Tumor-induced reshuffling of lipid composition on the endoplasmic reticulum membrane sustains macrophage survival and pro-tumorigenic activity. Nat. Immunol. 22, 1403–1415 (2021).
Goldstein, I. et al. p53, a novel regulator of lipid metabolism pathways. J. Hepatol. 56, 656–662 (2012).
Freed-Pastor, W. A. et al. Mutant p53 disrupts mammary tissue architecture via the mevalonate pathway. Cell 148, 244–258 (2012).
DeBose-Boyd, R. A. & Ye, J. SREBPs in lipid metabolism, insulin signaling, and beyond. Trends Biochem. Sci. 43, 358–368 (2018).
Infante, R. E. et al. NPC2 facilitates bidirectional transfer of cholesterol between NPC1 and lipid bilayers, a step in cholesterol egress from lysosomes. Proc. Natl. Acad. Sci. USA 105, 15287–15292 (2008).
Yamazaki, Y., Zhao, N., Caulfield, T. R., Liu, C. C. & Bu, G. Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat. Rev. Neurol. 15, 501–518 (2019).
Yang, Y., Lee, M. & Fairn, G. D. Phospholipid subcellular localization and dynamics. J. Biol. Chem. 293, 6230–6240 (2018).
Ioannou, M. S. et al. Neuron-astrocyte metabolic coupling protects against activity-induced fatty acid toxicity. Cell 177, 1522–1535.e14 (2019).
Estes, R. E., Lin, B., Khera, A. & Davis, M. Y. Lipid metabolism influence on neurodegenerative disease progression: is the vehicle as important as the cargo?. Front. Mol. Neurosci. 14, 788695 (2021).
Wahrle, S. E. et al. ABCA1 is required for normal central nervous system ApoE levels and for lipidation of astrocyte-secreted apoE. J. Biol. Chem. 279, 40987–40993 (2004).
Moore, K. J., Sheedy, F. J. & Fisher, E. A. Macrophages in atherosclerosis: a dynamic balance. Nat. Rev. Immunol. 13, 709–721 (2013).
Hotamisligil, G. S. Endoplasmic reticulum stress and atherosclerosis. Nat. Med. 16, 396–399 (2010).
Golstein, P. & Kroemer, G. Cell death by necrosis: towards a molecular definition. Trends Biochem. Sci. 32, 37–43 (2007).
Zong, W. X. & Thompson, C. B. Necrotic death as a cell fate. Genes Dev. 20, 1–15 (2006).
Zhang, Y., Chen, X., Gueydan, C. & Han, J. Plasma membrane changes during programmed cell deaths. Cell Res. 28, 9–21 (2018).
Vanden Berghe, T. et al. Necroptosis, necrosis and secondary necrosis converge on similar cellular disintegration features. Cell Death Differ. 17, 922–930 (2010).
Dondelinger, Y. et al. NINJ1 is activated by cell swelling to regulate plasma membrane permeabilization during regulated necrosis. Cell Death Dis. 14, 755 (2023).
David, L. et al. NINJ1 mediates plasma membrane rupture by cutting and releasing membrane disks. Cell 187, 2224–2235.e16 (2024).
Sahoo, B., Mou, Z., Liu, W., Dubyak, G. & Dai, X. How NINJ1 mediates plasma membrane rupture and why NINJ2 cannot. Cell 188, 292–302.e11 (2025).
Linkermann, A. et al. Two independent pathways of regulated necrosis mediate ischemia-reperfusion injury. Proc. Natl. Acad. Sci. USA 110, 12024–12029 (2013).
Zhang, T. et al. Prolonged hypoxia alleviates prolyl hydroxylation-mediated suppression of RIPK1 to promote necroptosis and inflammation. Nat. Cell Biol. 25, 950–962 (2023).
Bonora, M., Giorgi, C. & Pinton, P. Molecular mechanisms and consequences of mitochondrial permeability transition. Nat. Rev. Mol. Cell Biol. 23, 266–285 (2022).
Nakagawa, T. et al. Cyclophilin D-dependent mitochondrial permeability transition regulates some necrotic but not apoptotic cell death. Nature 434, 652–658 (2005).
Zorov, D. B., Juhaszova, M. & Sollott, S. J. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol. Rev. 94, 909–950 (2014).
Karch, J. et al. Inhibition of mitochondrial permeability transition by deletion of the ANT family and CypD. Sci. Adv. 5, eaaw4597 (2019).
Baines, C. P. et al. Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature 434, 658–662 (2005).
Ma, X. et al. CD36-mediated ferroptosis dampens intratumoral CD8(+) T cell effector function and impairs their antitumor ability. Cell Metab. 33, 1001–1012.e5 (2021).
Liao, F. et al. Targeting of nonlipidated, aggregated apoE with antibodies inhibits amyloid accumulation. J. Clin. Invest. 128, 2144–2155 (2018).
Xiong, M. et al. APOE immunotherapy reduces cerebral amyloid angiopathy and amyloid plaques while improving cerebrovascular function. Sci. Transl. Med. 13, eabd7522 (2021).
Pardoll, D. M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 12, 252–264 (2012).
Cillo, A. R. et al. Blockade of LAG-3 and PD-1 leads to co-expression of cytotoxic and exhaustion gene modules in CD8(+) T cells to promote antitumor immunity. Cell 187, 4373–4388.e15 (2024).
Singh, R., Letai, A. & Sarosiek, K. Regulation of apoptosis in health and disease: the balancing act of BCL-2 family proteins. Nat. Rev. Mol. Cell Biol. 20, 175–193 (2019).
Carneiro, B. A. & El-Deiry, W. S. Targeting apoptosis in cancer therapy. Nat. Rev. Clin. Oncol. 17, 395–417 (2020).
Vousden, K. H. & Prives, C. Blinded by the light: the growing complexity of p53. Cell 137, 413–431 (2009).
Zhang, X. D., Wu, J. J., Gillespie, S., Borrow, J. & Hersey, P. Human melanoma cells selected for resistance to apoptosis by prolonged exposure to tumor necrosis factor-related apoptosis-inducing ligand are more vulnerable to necrotic cell death induced by cisplatin. Clin. Cancer Res. 12, 1355–1364 (2006).
White, E. Autophagic cell death unraveled: pharmacological inhibition of apoptosis and autophagy enables necrosis. Autophagy 4, 399–401 (2008).
Nikoletopoulou, V., Markaki, M., Palikaras, K. & Tavernarakis, N. Crosstalk between apoptosis, necrosis and autophagy. Biochim. Biophys. Acta 1833, 3448–3459 (2013).
Scaffidi, P., Misteli, T. & Bianchi, M. E. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 418, 191–195 (2002).
Iyer, S. S. et al. Necrotic cells trigger a sterile inflammatory response through the Nlrp3 inflammasome. Proc. Natl. Acad. Sci. USA 106, 20388–20393 (2009).
Yang, L. G., March, Z. M., Stephenson, R. A. & Narayan, P. S. Apolipoprotein E in lipid metabolism and neurodegenerative disease. Trends Endocrinol. Metab. 34, 430–445 (2023).
Cantuti-Castelvetri, L. et al. Defective cholesterol clearance limits remyelination in the aged central nervous system. Science 359, 684–688 (2018).
Liu, L., MacKenzie, K. R., Putluri, N., Maletic-Savatic, M. & Bellen, H. J. The glia-neuron lactate shuttle and elevated ROS promote lipid synthesis in neurons and lipid droplet accumulation in glia via APOE/D. Cell Metab. 26, 719–737.e6 (2017).
Chen, Y. et al. Cholesterol inhibits TCR signaling by directly restricting TCR-CD3 core tunnel motility. Mol. Cell 82, 1278–1287.e5 (2022).
Molnar, E. et al. Cholesterol and sphingomyelin drive ligand-independent T-cell antigen receptor nanoclustering. J. Biol. Chem. 287, 42664–42674 (2012).
Brennan, C. W. et al. The somatic genomic landscape of glioblastoma. Cell 155, 462–477 (2013).
Lim, M. et al. Phase III trial of chemoradiotherapy with temozolomide plus nivolumab or placebo for newly diagnosed glioblastoma with methylated MGMT promoter. Neuro. Oncol. 24, 1935–1949 (2022).
Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).
Suva, M. L. et al. Reconstructing and reprogramming the tumor-propagating potential of glioblastoma stem-like cells. Cell 157, 580–594 (2014).
Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).
Jiang, S. et al. Cell Taxonomy: a curated repository of cell types with multifaceted characterization. Nucleic Acids Res. 51, D853–D860 (2023).
Zeisel, A. et al. Molecular architecture of the mouse nervous system. Cell 174, 999–1014.e22 (2018).
Cahoy, J. D. et al. A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J. Neurosci. 28, 264–278 (2008).
Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).
Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291.e9 (2019).
Lopez, R., Regier, J., Cole, M. B., Jordan, M. I. & Yosef, N. Deep generative modeling for single-cell transcriptomics. Nat. Methods 15, 1053–1058 (2018).
Traag, V. A., Waltman, L. & van Eck, N. J. From Louvain to Leiden: guaranteeing well-connected communities. Sci. Rep. 9, 5233 (2019).
Wu, T. et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation 2, 100141 (2021).
Wen, B., Mei, Z., Zeng, C. & Liu, S. metaX: a flexible and comprehensive software for processing metabolomics data. BMC Bioinform. 18, 183 (2017).
Acknowledgements
We thank Yang Yang and Bing Li for their help with the amplification of the Brie mouse CRISPR knockout library and Eunhee Choi, Sushama Sivakumar, and the staff at the Animal Resource Center at UT Southwestern Medical Center for technical assistance with the teratoma assay. We are grateful to the Histo Pathology Core at UT Southwestern Medical Center for assistance with teratoma sectioning and to the McDermott Sequencing Core for next-generation sequencing. We thank Yang-Xin Fu for reading the manuscript critically.
Funding
This study was supported by the National Natural Science Foundation of China (Project 32130053 to H.Y., 82303354 to Yapeng J., and 82273493 to Z.Z.). H.Y. is supported by the New Cornerstone Science Foundation through the New Cornerstone Investigator Program.
Author information
Authors and Affiliations
Contributions
Yapeng J. designed and performed all experiments and analyzed the data. A.A.S. and C.X. analyzed the CRISPR screen. A.A.S., C.X., J.J., L.H., and W.P. analyzed the whole-exome and scRNA-seq data. B.M.E. performed histopathological analysis of mouse teratomas. P.L., M.Z., S.H., M.W., Yuchen J., X. Liu, D.Y., Y.G., Q.X., and Z.Z. performed histopathological analysis of human and mouse GBM. H.Y., X. Luo, and Z.Z. supervised this research. Yapeng J. and H.Y. wrote the manuscript with input from all authors.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
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
Ji, Y., Jiang, J., Hu, L. et al. Targeting necrotic lipid release in tumors enhances immunosurveillance and cancer immunotherapy of glioblastoma. Cell Res (2025). https://doi.org/10.1038/s41422-025-01155-y
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41422-025-01155-y