Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that are present at low abundance in the circulation and tissues; they serve as crucial immune sentinels by continually sampling their environment, migrating to secondary lymphoid organs and shaping adaptive immune responses through antigen presentation. Owing to their ability to orchestrate tolerogenic or immunogenic responses to a specific antigen, DCs have a pivotal role in antitumour immunity and the response to immune checkpoint blockade and other immunotherapeutic approaches. The multifaceted functions of DCs are acquired through a complex, multistage process called maturation. Although the role of inflammatory triggers in driving DC maturation was established decades ago, less is known about DC maturation in non-inflammatory contexts, such as during homeostasis and in cancer. The advent of single-cell technologies has enabled an unbiased, high-dimensional characterization of various DC states, including mature DCs. This approach has clarified the molecular programmes associated with DC maturation and also revealed how cancers exploit these pathways to subvert immune surveillance. In this Review, we discuss the mechanisms by which cancer disrupts DC maturation and highlight emerging therapeutic opportunities to modulate DC states. These insights could inform the development of DC-centric immunotherapies, expanding the arsenal of strategies to enhance antitumour immunity.
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 print issues and online access
$259.00 per year
only $21.58 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
References
Steinman, R. M. & Cohn, Z. A. Identification of a novel cell type in peripheral lymphoid organs of mice. J. Exp. Med. 137, 1142–1162 (1973).
Cabeza-Cabrerizo, M., Cardoso, A., Minutti, C. M., da Costa, M. P. & Sousa, C. R. E. Dendritic cells revisited. Annu. Rev. Immunol. 39, 131–166 (2021).
Merad, M., Sathe, P., Helft, J., Miller, J. & Mortha, A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu. Rev. Immunol. 31, 563–604 (2013).
Rakoff-Nahoum, S. & Medzhitov, R. Toll-like receptors and cancer. Nat. Rev. Cancer 9, 57–63 (2009).
Prete, A. D. et al. Dendritic cell subsets in cancer immunity and tumor antigen sensing. Cell. Mol. Immunol. 20, 432–447 (2023).
Dalod, M., Chelbi, R., Malissen, B. & Lawrence, T. Dendritic cell maturation: functional specialization through signaling specificity and transcriptional programming. EMBO J. 33, 1104–1116 (2014).
Cao, L. L. & Kagan, J. C. Targeting innate immune pathways for cancer immunotherapy. Immunity 56, 2206–2217 (2023).
Fitzgerald, K. A. & Kagan, J. C. Toll-like receptors and the control of immunity. Cell 180, 1044–1066 (2020).
Wculek, S. K. et al. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 20, 7–24 (2020).
Salmon, H. et al. Expansion and activation of CD103+ dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity 44, 924–938 (2016).
Roberts, E. W. et al. Critical role for CD103+/CD141+ dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma. Cancer Cell 30, 324–336 (2016).
Sánchez-Paulete, A. R. et al. Cancer immunotherapy with immunomodulatory anti-CD137 and anti-PD-1 monoclonal antibodies requires BATF3-dependent dendritic cells. Cancer Discov. 6, 71–79 (2016).
Mellman, I. & Steinman, R. M. Dendritic cells specialized and regulated antigen processing machines. Cell 106, 255–258 (2001).
Sousa, C. R. e. Dendritic cells in a mature age. Nat. Rev. Immunol. 6, 476–483 (2006).
Pittet, M. J., Pilato, M. D., Garris, C. & Mempel, T. R. Dendritic cells as shepherds of T cell immunity in cancer. Immunity 56, 2218–2230 (2023).
Mellman, I., Chen, D. S., Powles, T. & Turley, S. J. The cancer-immunity cycle: indication, genotype, and immunotype. Immunity 56, 2188–2205 (2023).
Broz, M. L. et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 26, 638–652 (2014).
Engelhardt, J. J. et al. Marginating dendritic cells of the tumor microenvironment cross-present tumor antigens and stably engage tumor-specific T cells. Cancer Cell 21, 402–417 (2012).
Spranger, S., Dai, D., Horton, B. & Gajewski, T. F. Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 31, 711–723.e4 (2017).
Böttcher, J. P. et al. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 172, 1022–1037.e14 (2018).
Brown, C. C. et al. Transcriptional basis of mouse and human dendritic cell heterogeneity. Cell 179, 846–863.e24 (2019).
Rodrigues, P. F. et al. Progenitors of distinct lineages shape the diversity of mature type 2 conventional dendritic cells. Immunity 57, 1567–1585.e5 (2024).
Lewis, K. L. et al. Notch2 receptor signaling controls functional differentiation of dendritic cells in the spleen and intestine. Immunity 35, 780–791 (2011).
Tussiwand, R. et al. Klf4 expression in conventional dendritic cells is required for T helper 2 cell responses. Immunity 42, 916–928 (2015).
Minutti, C. M. et al. Distinct ontogenetic lineages dictate cDC2 heterogeneity. Nat. Immunol. 25, 448–461 (2024).
Binnewies, M. et al. Unleashing type-2 dendritic cells to drive protective antitumor CD4+ T cell immunity. Cell 177, 556–571.e16 (2019).
Kvedaraite, E. & Ginhoux, F. Human dendritic cells in cancer. Sci. Immunol. 7, eabm9409 (2022).
Schlitzer, A., McGovern, N. & Ginhoux, F. Dendritic cells and monocyte-derived cells: two complementary and integrated functional systems. Semin. Cell Dev. Biol. 41, 9–22 (2015).
León, B., López-Bravo, M. & Ardavín, C. Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against leishmania. Immunity 26, 519–531 (2007).
Cheong, C. et al. Microbial stimulation fully differentiates monocytes to DC-SIGN/CD209+ dendritic cells for immune T cell areas. Cell 143, 416–429 (2010).
Segura, E. et al. Human inflammatory dendritic cells induce Th17 cell differentiation. Immunity 38, 336–348 (2013).
Nizzoli, G. et al. Human CD1c+ dendritic cells secrete high levels of IL-12 and potently prime cytotoxic T-cell responses. Blood 122, 932–942 (2013).
Bourdely, P. et al. Transcriptional and functional analysis of CD1c+ human dendritic cells identifies a CD163+ subset priming CD8+CD103+ T cells. Immunity 53, 335–352.e8 (2020).
Bosteels, C. et al. Inflammatory type 2 cDCs acquire features of cDC1s and macrophages to orchestrate immunity to respiratory virus infection. Immunity 52, 1039–1056.e9 (2020).
Liu, Z. et al. Dendritic cell type 3 arises from Ly6C+ monocyte-dendritic cell progenitors. Immunity 56, 1761–1777.e6 (2023).
Yun, T. J. et al. Human plasmacytoid dendritic cells mount a distinct antiviral response to virus-infected cells. Sci. Immunol. 6, eabc7302 (2021).
Ito, T., Kanzler, H., Duramad, O., Cao, W. & Liu, Y.-J. Specialization, kinetics, and repertoire of type 1 interferon responses by human plasmacytoid predendritic cells. Blood 107, 2423–2431 (2006).
Ziegler-Heitbrock, L., Ohteki, T., Ginhoux, F., Shortman, K. & Spits, H. Reclassifying plasmacytoid dendritic cells as innate lymphocytes. Nat. Rev. Immunol. 23, 1–2 (2023).
Ziegler-Heitbrock, L., Ohteki, T., Ginhoux, F., Shortman, K. & Spits, H. Reply to ‘Reclassification of plasmacytoid dendritic cells as innate lymphocytes is premature.’. Nat. Rev. Immunol. 23, 338–339 (2023).
Reizis, B. et al. Reclassification of plasmacytoid dendritic cells as innate lymphocytes is premature. Nat. Rev. Immunol. 23, 336–337 (2023).
Park, M. D. et al. On the biology and therapeutic modulation of macrophages and dendritic cells in cancer. Annu. Rev. Cancer Biol. 7, 291–311 (2023).
Hegde, S. et al. Dendritic cell paucity leads to dysfunctional immune surveillance in pancreatic cancer. Cancer Cell 37, 289–307.e9 (2020).
Cueto, F. J. & Sancho, D. The Flt3L/Flt3 axis in dendritic cell biology and cancer immunotherapy. Cancers 13, 1525 (2021).
Meyer, M. A. et al. Breast and pancreatic cancer interrupt IRF8-dependent dendritic cell development to overcome immune surveillance. Nat. Commun. 9, 1250 (2018).
Kim, S. et al. IL-6 selectively suppresses cDC1 specification via C/EBPβ. J. Exp. Med. 220, e20221757 (2023).
Liu, T.-T. et al. Ablation of cDC2 development by triple mutations within the Zeb2 enhancer. Nature 607, 142–148 (2022).
Murphy, T. L. & Murphy, K. M. Dendritic cells in cancer immunology. Cell. Mol. Immunol. 19, 3–13 (2022).
Durai, V. et al. Cryptic activation of an Irf8 enhancer governs cDC1 fate specification. Nat. Immunol. 20, 1161–1173 (2019).
Zhang, S., Audiger, C., Chopin, M. & Nutt, S. L. Transcriptional regulation of dendritic cell development and function. Front. Immunol. 14, 1182553 (2023).
Li, J. et al. Mature dendritic cells enriched in immunoregulatory molecules (mregDCs): a novel population in the tumour microenvironment and immunotherapy target. Clin. Transl. Med. 13, e1199 (2023).
Cheng, S. et al. A pan-cancer single-cell transcriptional atlas of tumor infiltrating myeloid cells. Cell 184, 792–809.e23 (2021).
Leader, A. M. et al. Single-cell analysis of human non-small cell lung cancer lesions refines tumor classification and patient stratification. Cancer Cell 39, 1594–1609.e12 (2021).
Zilionis, R. et al. Single-cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity 50, 1317–1334.e10 (2019).
Gerhard, G. M., Bill, R., Messemaker, M., Klein, A. M. & Pittet, M. J. Tumor-infiltrating dendritic cell states are conserved across solid human cancers. J. Exp. Med. 218, e20200264 (2020).
Ginhoux, F. & Guilliams, M. Tissue-resident macrophage ontogeny and homeostasis. Immunity 44, 439–449 (2016).
Bigley, V. et al. Langerin‐expressing dendritic cells in human tissues are related to CD1c+ dendritic cells and distinct from Langerhans cells and CD141high XCR1+ dendritic cells. J. Leukoc. Biol. 97, 627–634 (2015).
Yu, C. I. et al. Human CD1c+ dendritic cells drive the differentiation of CD103+ CD8+ mucosal effector T cells via the cytokine TGF-β. Immunity 38, 818–830 (2013).
Sato-Hashimoto, M. et al. Microglial SIRPα regulates the emergence of CD11c+ microglia and demyelination damage in white matter. eLife 8, e42025 (2019).
Xuan, S. et al. Langerin-expressing dendritic cells in pulmonary immune-related diseases. Front. Med. 9, 909057 (2022).
Hubert, M. et al. IFN-III is selectively produced by cDC1 and predicts good clinical outcome in breast cancer. Sci. Immunol. 5, eaav3942 (2020).
Apetoh, L. et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 13, 1050–1059 (2007).
Fucikova, J. et al. Human tumor cells killed by anthracyclines induce a tumor-specific immune response. Cancer Res. 71, 4821–4833 (2011).
Elliott, M. R. et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature 461, 282–286 (2009).
Ma, M., Jiang, W. & Zhou, R. DAMPs and DAMP-sensing receptors in inflammation and diseases. Immunity 57, 752–771 (2024).
Dvorkin, S., Cambier, S., Volkman, H. E. & Stetson, D. B. New frontiers in the cGAS–STING intracellular DNA-sensing pathway. Immunity 57, 718–730 (2024).
Sousa, C. R. E., Yamasaki, S. & Brown, G. D. Myeloid C-type lectin receptors in innate immune recognition. Immunity 57, 700–717 (2024).
Sundaram, B., Tweedell, R. E., Kumar, S. P. & Kanneganti, T.-D. The NLR family of innate immune and cell death sensors. Immunity 57, 674–699 (2024).
Yoneyama, M., Kato, H. & Fujita, T. Physiological functions of RIG-I-like receptors. Immunity 57, 731–751 (2024).
Hu, W. et al. Differential outcome of TRIF-mediated signaling in TLR4 and TLR3 induced DC maturation. Proc. Natl Acad. Sci. USA 112, 13994–13999 (2015).
Carozza, J. A. et al. Extracellular cGAMP is a cancer-cell-produced immunotransmitter involved in radiation-induced anticancer immunity. Nat. Cancer 1, 184–196 (2020).
Corrales, L. et al. Antagonism of the STING pathway via activation of the AIM2 inflammasome by intracellular DNA. J. Immunol. 196, 3191–3198 (2016).
Sharma, N. et al. Differential expression profile of NLRs and AIM2 in glioma and implications for NLRP12 in glioblastoma. Sci. Rep. 9, 8480 (2019).
Khan, M. et al. Pyroptosis relates to tumor microenvironment remodeling and prognosis: a pan-cancer perspective. Front. Immunol. 13, 1062225 (2022).
Wu, L. et al. Noncanonical MAVS signaling restrains dendritic cell-driven antitumor immunity by inhibiting IL-12. Sci. Immunol. 8, eadf4919 (2023).
Tang, M. et al. Toll-like receptor 2 activation promotes tumor dendritic cell dysfunction by regulating IL-6 and IL-10 receptor signaling. Cell Rep. 13, 2851–2864 (2015).
Ahn, J., Konno, H. & Barber, G. N. Diverse roles of STING-dependent signaling on the development of cancer. Oncogene 34, 5302–5308 (2015).
Ahn, J. et al. Inflammation-driven carcinogenesis is mediated through STING. Nat. Commun. 5, 5166 (2014).
Huang, B., Zhao, J., Unkeless, J. C., Feng, Z. H. & Xiong, H. TLR signaling by tumor and immune cells: a double-edged sword. Oncogene 27, 218–224 (2008).
Mehrotra, P. & Ravichandran, K. S. Drugging the efferocytosis process: concepts and opportunities. Nat. Rev. Drug. Discov. 21, 601–620 (2022).
Doran, A. C., Yurdagul, A. & Tabas, I. Efferocytosis in health and disease. Nat. Rev. Immunol. 20, 254–267 (2020).
Chen, J., Zhao, Y. & Liu, Y. The role of nucleotides and purinergic signaling in apoptotic cell clearance — implications for chronic inflammatory diseases. Front. Immunol. 5, 656 (2014).
Truman, L. A. et al. CX3CL1/fractalkine is released from apoptotic lymphocytes to stimulate macrophage chemotaxis. Blood 112, 5026–5036 (2008).
Gude, D. R. et al. Apoptosis induces expression of sphingosine kinase 1 to release sphingosine‐1‐phosphate as a “come‐and‐get‐me” signal. FASEB J. 22, 2629–2638 (2008).
Weigert, A. et al. Cleavage of sphingosine kinase 2 by caspase-1 provokes its release from apoptotic cells. Blood 115, 3531–3540 (2010).
Lauber, K. et al. Apoptotic cells induce migration of phagocytes via caspase-3-mediated release of a lipid attraction signal. Cell 113, 717–730 (2003).
Peter, C. et al. Migration to spoptotic “find-me” signals is mediated via the phagocyte receptor G2A. J. Biol. Chem. 283, 5296–5305 (2008).
Ghiringhelli, F. et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1β-dependent adaptive immunity against tumors. Nat. Med. 15, 1170–1178 (2009). This paper elucidates the pivotal role of the NLRP3 inflammasome in bridging the innate and adaptive immune responses to dying tumour cells.
Silva-Vilches, C., Ring, S. & Mahnke, K. ATP and its metabolite adenosine as regulators of dendritic cell activity. Front. Immunol. 9, 2581 (2018).
Cekic, C. & Linden, J. Adenosine A2A receptors intrinsically regulate CD8+ T cells in the tumor microenvironment. Cancer Res. 74, 7239–7249 (2014).
Borodovsky, A. et al. Small molecule AZD4635 inhibitor of A2AR signaling rescues immune cell function including CD103+ dendritic cells enhancing anti-tumor immunity. J. Immunother. Cancer 8, e000417 (2020).
Segawa, K. & Nagata, S. An spoptotic ‘eat me’ signal: phosphatidylserine exposure. Trends Cell Biol. 25, 639–650 (2015).
Kobayashi, N. et al. TIM-1 and TIM-4 glycoproteins bind phosphatidylserine and mediate uptake of apoptotic cells. Immunity 27, 927–940 (2007).
Nakayama, M. et al. Tim-3 mediates phagocytosis of apoptotic cells and cross-presentation. Blood 113, 3821–3830 (2009).
Park, S.-Y. et al. Rapid cell corpse clearance by stabilin-2, a membrane phosphatidylserine receptor. Cell Death Differ. 15, 192–201 (2008).
Lemke, G. & Rothlin, C. V. Immunobiology of the TAM receptors. Nat. Rev. Immunol. 8, 327–336 (2008).
Fadok, V. A., Bratton, D. L., Frasch, S. C., Warner, M. L. & Henson, P. M. The role of phosphatidylserine in recognition of apoptotic cells by phagocytes. Cell Death Differ. 5, 551–562 (1998).
Sen, P. et al. Apoptotic cells induce Mer tyrosine kinase-dependent blockade of NF-κB activation in dendritic cells. Blood 109, 653–660 (2006).
Wallet, M. A. et al. MerTK is required for apoptotic cell-induced T cell tolerance. J. Exp. Med. 205, 219–232 (2008).
Rothlin, C. V., Ghosh, S., Zuniga, E. I., Oldstone, M. B. A. & Lemke, G. TAM receptors are pleiotropic inhibitors of the innate immune response. Cell 131, 1124–1136 (2007).
Stach, C. M. et al. Treatment with annexin V increases immunogenicity of apoptotic human T-cells in Balb/c mice. Cell Death Differ. 7, 911–915 (2000).
Bondanza, A. et al. Inhibition of phosphatidylserine recognition heightens the immunogenicity of irradiated lymphoma cells in vivo. J. Exp. Med. 200, 1157–1165 (2004).
Blander, J. M. MerTK blockade fuels anti-tumor immunity. Immunity 52, 212–214 (2020).
Gardner, A. et al. TIM-3 blockade enhances IL-12-dependent antitumor immunity by promoting CD8+ T cell and XCR1+ dendritic cell spatial co-localization. J. Immunother. Cancer 10, e003571 (2022).
Pulido, Á. et al. TIM-3 regulates CD103+ dendritic cell function and response to chemotherapy in breast cancer. Cancer Cell 33, 60–74.e6 (2018).
Pulido, Á. et al. The inhibitory receptor TIM-3 limits activation of the cGAS–STING pathway in intra-tumoral dendritic cells by suppressing extracellular DNA uptake. Immunity 54, 1154–1167.e7 (2021). This study unveils a novel mechanism in which blockade of the inhibitory receptor TIM3 enhances cancer immunotherapy efficacy via activation of the cGAS–STING pathway in cDC1s.
Dixon, K. O. et al. TIM-3 restrains anti-tumour immunity by regulating inflammasome activation. Nature 595, 101–106 (2021).
Li, H. et al. AXL targeting restores PD-1 blockade sensitivity of STK11/LKB1 mutant NSCLC through expansion of TCF1+ CD8 T cells. Cell Rep. Med. 3, 100554 (2022). This study elucidates the critical role of AXL inhibition in overcoming immune suppression in non-small-cell lung cancer with STK11–LKB1 mutations, through increased type I interferon secretion from DCs and expansion of tumour-associated TCF1+PD1+CD8 T cells.
Curigliano, G. et al. Phase I/Ib clinical trial of sabatolimab, an anti-TIM-3 antibody, alone and in combination with spartalizumab, an anti-PD-1 antibody, in advanced solid tumors. Clin. Cancer Res. 27, 3620–3629 (2021).
Morais, A. L. G., de, Cerdá, S. & de.Miguel, M. New checkpoint inhibitors on the road: targeting TIM-3 in solid tumors. Curr. Oncol. Rep. 24, 651–658 (2022).
Sancho, D. et al. Identification of a dendritic cell receptor that couples sensing of necrosis to immunity. Nature 458, 899–903 (2009). This study highlights the crucial role of CLEC9A in mediating the recognition of necrotic cells by CD8α+ DCs, demonstrating its involvement in cross-presentation of dead cell-associated antigens.
Tullett, K. M., Lahoud, M. H. & Radford, K. J. Harnessing human cross-presenting CLEC9A+XCR1+ dendritic cells for immunotherapy. Front. Immunol. 5, 239 (2014).
Cueto, F. J., Fresno, Cdel & Sancho, D. DNGR-1, a dendritic cell-specific sensor of tissue damage that dually modulates immunity and inflammation. Front. Immunol. 10, 3146 (2020).
Caronni, N. et al. TIM4 expression by dendritic cells mediates uptake of tumor-associated antigens and anti-tumor responses. Nat. Commun. 12, 2237 (2021).
Canton, J. et al. The receptor DNGR-1 signals for phagosomal rupture to promote cross-presentation of dead-cell-associated antigens. Nat. Immunol. 22, 140–153 (2021).
Giampazolias, E. et al. Secreted gelsolin inhibits DNGR-1-dependent cross-presentation and cancer immunity. Cell 184, 4016–4031.e22 (2021). This study uncovers a novel mechanism by which secreted gelsolin suppresses the cross-presentation of dead cell-associated antigens by cDC1, leading to enhanced anticancer CD8+ T cell responses.
Xu, F. et al. Mevalonate blockade in cancer cells triggers CLEC9A+ dendritic cell-mediated antitumor immunity. Cancer Res. 81, 4514–4528 (2021).
Li, D. & Wu, M. Pattern recognition receptors in health and diseases. Signal Transduct. Target. Ther. 6, 291 (2021).
Lind, N. A., Rael, V. E., Pestal, K., Liu, B. & Barton, G. M. Regulation of the nucleic acid-sensing Toll-like receptors. Nat. Rev. Immunol. 22, 224–235 (2022).
Savini, M., Zhao, Q. & Wang, M. C. Lysosomes: signaling hubs for metabolic sensing and longevity. Trends Cell Biol. 29, 876–887 (2019).
Bosteels, V. et al. LXR signaling controls homeostatic dendritic cell maturation. Sci. Immunol. 8, eadd3955 (2023). This study elucidates a novel mechanism by which the engulfment of apoptotic cells or cholesterol-rich LNPs triggers a homeostatic maturation process in cDC1s via activation of the LXR pathway.
Han, C. Z. & Ravichandran, K. S. Metabolic connections during apoptotic cell engulfment. Cell 147, 1442–1445 (2011).
Fond, A. M., Lee, C. S., Schulman, I. G., Kiss, R. S. & Ravichandran, K. S. Apoptotic cells trigger a membrane-initiated pathway to increase ABCA1. J. Clin. Invest. 125, 2748–2758 (2015).
Morioka, S. et al. Efferocytosis induces a novel SLC program to promote glucose uptake and lactate release. Nature 563, 714–718 (2018). This study showcases how each step of efferocytosis induces a highly choreographed transcriptional programme that dictates the metabolic rewiring of phagocytes.
Viaud, M. et al. Lysosomal cholesterol hydrolysis couples efferocytosis to anti-inflammatory oxysterol production. Circ. Res. 122, 1369–1384 (2018).
Mukundan, L. et al. PPAR-δ senses and orchestrates clearance of apoptotic cells to promote tolerance. Nat. Med. 15, 1266–1272 (2009).
Deng, L. et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity 41, 843–852 (2014).
Woo, S.-R. et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity 41, 830–842 (2014). This study unveils a critical role for the STING pathway in mediating spontaneous CD8+ T cell priming against tumours, demonstrating that tumour cell-derived DNA activates this pathway via cGAS and promotes type I interferon production.
Xu, M. M. et al. Dendritic cells but not macrophages sense tumor mitochondrial DNA for cross-priming through signal regulatory protein α signaling. Immunity 47, 363–373.e5 (2017). This study demonstrates how a phagocytic modulator acting within the engulfment phase of efferocytosis fine-tunes the subsequent digestion phase to modulate detection of immunogenic DAMPs within the internalized cargo.
Cummings, R. J. et al. Different tissue phagocytes sample apoptotic cells to direct distinct homeostasis programs. Nature 539, 565–569 (2016).
Maier, B. et al. A conserved dendritic-cell regulatory program limits antitumour immunity. Nature 580, 257–262 (2020). This study describes the mregDC programme in both steady state and tumoural contexts.
Steinman, R. M., Turley, S., Mellman, I. & Inaba, K. The induction of tolerance by dendritic cells that have captured apoptotic cells. J. Exp. Med. 191, 411–416 (2000).
Ardouin, L. et al. Broad and largely concordant molecular changes characterize tolerogenic and immunogenic dendritic cell maturation in thymus and periphery. Immunity 45, 305–318 (2016).
Miller, J. et al. Deciphering the transcriptional network of the dendritic cell lineage. Nat. Immunol. 13, 888–899 (2012).
Belabed, M. et al. Cholesterol mobilization regulates dendritic cell maturation and the immunogenic response to cancer. Nat. Immunol. https://doi.org/10.1038/s41590-024-02065-8 (2025).
Hammer, G. E. & Ma, A. Molecular control of steady-state dendritic cell maturation and immune homeostasis. Annu. Rev. Immunol. 31, 743–791 (2013).
Cohen, M. et al. The interaction of CD4+ helper T cells with dendritic cells shapes the tumor microenvironment and immune checkpoint blockade response. Nat. Cancer 3, 303–317 (2022).
Hu, J. et al. Tumor microenvironment remodeling after neoadjuvant immunotherapy in non-small cell lung cancer revealed by single-cell RNA sequencing. Genome Med. 15, 14 (2023).
Chen, J. H. et al. Human lung cancer harbors spatially organized stem-immunity hubs associated with response to immunotherapy. Nat. Immunol. 25, 644–658 (2024).
Bassez, A. et al. A single-cell map of intratumoral changes during anti-PD1 treatment of patients with breast cancer. Nat. Med. 27, 820–832 (2021).
Rawat, K. et al. CCL5-producing migratory dendritic cells guide CCR5+ monocytes into the draining lymph nodes. J. Exp. Med. 220, e20222129 (2023).
Fassett, M. S. et al. IL-31-dependent neurogenic inflammation restrains cutaneous type 2 immune response in allergic dermatitis. Sci. Immunol. 8, eabi6887 (2023).
Mulder, K. et al. Cross-tissue single-cell landscape of human monocytes and macrophages in health and disease. Immunity 54, 1883–1900.e5 (2021).
Baharom, F. et al. Systemic vaccination induces CD8+ T cells and remodels the tumor microenvironment. Cell 185, 4317–4332.e15 (2022).
Oba, T. et al. Overcoming primary and acquired resistance to anti-PD-L1 therapy by induction and activation of tumor-residing cDC1s. Nat. Commun. 11, 5415 (2020).
Lee, C. Y. C. et al. Tumour-retained activated CCR7+ dendritic cells are heterogeneous and regulate local anti-tumour cytolytic activity. Nat. Commun. 15, 682 (2024).
Zavitsanou, A.-M. et al. KEAP1 mutation in lung adenocarcinoma promotes immune evasion and immunotherapy resistance. Cell Rep. 42, 113295 (2023).
Magen, A. et al. Intratumoral dendritic cell–CD4+ T helper cell niches enable CD8+ T cell differentiation following PD-1 blockade in hepatocellular carcinoma. Nat. Med. 29, 1389–1399 (2023).
Boslem, E. et al. Therapeutic blockade of ER stress and inflammation prevents NASH and progression to HCC. Sci. Adv. 9, eadh0831 (2023).
Plebanek, M. P. et al. A lactate–SREBP2 signaling axis drives tolerogenic dendritic cell maturation and promotes cancer progression. Sci. Immunol. 9, eadi4191 (2024).
Chudnovskiy, A. et al. Proximity-dependent labeling identifies dendritic cells that drive the tumor-specific CD4+ T cell response. Sci. Immunol. 9, eadq8843 (2024).
Mair, F. et al. Extricating human tumour immune alterations from tissue inflammation. Nature 605, 728–735 (2022).
Luoma, A. M. et al. Tissue-resident memory and circulating T cells are early responders to pre-surgical cancer immunotherapy. Cell 185, 2918–2935.e29 (2022).
Minohara, K. et al. Mature dendritic cells enriched in regulatory molecules may control regulatory T cells and the prognosis of head and neck cancer. Cancer Sci. 114, 1256–1269 (2023).
You, S. et al. Lymphatic-localized Treg-mregDC crosstalk limits antigen trafficking and restrains anti-tumor immunity. Cancer Cell 42, 1415–1433.e12 (2024).
Baldominos, P. et al. Quiescent cancer cells resist T cell attack by forming an immunosuppressive niche. Cell 185, 1694–1708.e19 (2022).
Birocchi, F. et al. Targeted inducible delivery of immunoactivating cytokines reprograms glioblastoma microenvironment and inhibits growth in mouse models. Sci. Transl. Med. 14, eabl4106 (2022).
Zheng, X. et al. Single-cell analyses implicate ascites in remodeling the ecosystems of primary and metastatic tumors in ovarian cancer. Nat. Cancer 4, 1138–1156 (2023).
Tang, F. et al. A pan-cancer single-cell panorama of human natural killer cells. Cell 186, 4235–4251.e20 (2023).
Tran, M. A. et al. Urine scRNAseq reveals new insights into the bladder tumor immune microenvironment. J. Exp. Med. 221, e20240045 (2024).
Chen, S. T. et al. A shift in lung macrophage composition is associated with COVID-19 severity and recovery. Sci. Transl. Med. 14, eabn5168 (2022).
Nakamizo, S. et al. Single-cell analysis of human skin identifies CD14+ type 3 dendritic cells co-producing IL1B and IL23A in psoriasis. J. Exp. Med. 218, e20202345 (2021).
Zernecke, A. et al. Integrated single-cell analysis-based classification of vascular mononuclear phagocytes in mouse and human atherosclerosis. Cardiovasc. Res. 119, 1676–1689 (2022).
Duong, E. et al. Type I interferon activates MHC class I-dressed CD11b+ conventional dendritic cells to promote protective anti-tumor CD8+ T cell immunity. Immunity 55, 308–323.e9 (2022).
Meiser, P. et al. A distinct stimulatory cDC1 subpopulation amplifies CD8+ T cell responses in tumors for protective anti-cancer immunity. Cancer Cell 41, 1498–1515.e10 (2023).
To, T. K. J. et al. Single-cell transcriptomics reveal distinct subsets of activated dendritic cells in the tumor microenvironment. Preprint at bioRxiv https://doi.org/10.1101/2022.09.13.507746 (2022).
Montoya, M. et al. Type I interferons produced by dendritic cells promote their phenotypic and functional activation. Blood 99, 3263–3271 (2002).
Luft, T. et al. Type I IFNs enhance the terminal differentiation of dendritic cells. J. Immunol. 161, 1947–1953 (1998).
Simmons, D. P. et al. Type I IFN drives a distinctive dendritic cell maturation phenotype that allows continued class II MHC synthesis and antigen processing. J. Immunol. 188, 3116–3126 (2012).
Spadaro, F. et al. IFN-α enhances cross-presentation in human dendritic cells by modulating antigen survival, endocytic routing, and processing. Blood 119, 1407–1417 (2012).
Longhi, M. P. et al. Dendritic cells require a systemic type I interferon response to mature and induce CD4+ Th1 immunity with poly IC as adjuvant. J. Exp. Med. 206, 1589–1602 (2009).
Webb, L. M. et al. Type I interferon is required for T helper (Th) 2 induction by dendritic cells. EMBO J. 36, 2404–2418 (2017).
Cucak, H., Yrlid, U., Reizis, B., Kalinke, U. & Johansson-Lindbom, B. Type I interferon signaling in dendritic cells stimulates the development of lymph-node-resident T follicular helper cells. Immunity 31, 491–501 (2009).
Gautier, G. et al. A type I interferon autocrine–paracrine loop is involved in Toll-like receptor-induced interleukin-12p70 secretion by dendritic cells. J. Exp. Med. 201, 1435–1446 (2005).
Schaupp, L. et al. Microbiota-induced type I interferons instruct a poised basal state of dendritic cells. Cell 181, 1080–1096.e19 (2020). This seminal study shows the source of type I interferon and how it is required to poise DCs into a transcriptional, epigenetic and metabolic state capable of responding to subsequent PRR signalling.
Diamond, M. S. et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J. Exp. Med. 208, 1989–2003 (2011). Together with Fuertes et al. (2011), this study demonstrates how type I interferon is critical in dictating DC–T cell interactions that form the basis of antitumour immunity.
Fuertes, M. B. et al. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8α+ dendritic cells. J. Exp. Med. 208, 2005–2016 (2011).
Mattiuz, R. et al. Type 1 conventional dendritic cells and interferons are required for spontaneous CD4+ and CD8+ T‐cell protective responses to breast cancer. Clin. Transl. Immunol. 10, e1305 (2021).
Ghislat, G. et al. NF-κB-dependent IRF1 activation programs cDC1 dendritic cells to drive antitumor immunity. Sci. Immunol. 6, eabg3570 (2021). This study demonstrates how an intact NF-κB signalling pathway is required for DCs to respond to interferon signalling for immunogenic maturation against tumour antigens.
Boukhaled, G. M., Harding, S. & Brooks, D. G. Opposing roles of type I interferons in cancer immunity. Annu. Rev. Pathol. Mech. Dis. 16, 167–198 (2020).
Barrat, F. J., Crow, M. K. & Ivashkiv, L. B. Interferon target-gene expression and epigenomic signatures in health and disease. Nat. Immunol. 20, 1574–1583 (2019).
Früh, K. & Yang, Y. Antigen presentation by MHC class I and its regulation by interferon γ. Curr. Opin. Immunol. 11, 76–81 (1999).
Meissner, T. B. et al. NLR family member NLRC5 is a transcriptional regulator of MHC class I genes. Proc. Natl Acad. Sci. USA 107, 13794–13799 (2010).
Steimle, V., Siegrist, C.-A., Mottet, A., Lisowska-Grospierre, B. & Mach, B. Regulation of MHC class II expression by interferon-γ mediated by the transactivator gene CIITA. Science 265, 106–109 (1994).
Alspach, E., Lussier, D. M. & Schreiber, R. D. Interferon γ and its important roles in promoting and inhibiting spontaneous and therapeutic cancer immunity. Cold Spring Harb. Perspect. Biol. 11, a028480 (2019).
Kalaora, S. et al. Immunoproteasome expression is associated with better prognosis and response to checkpoint therapies in melanoma. Nat. Commun. 11, 896 (2020).
Nirschl, C. J. et al. IFNγ-dependent tissue-immune homeostasis is co-opted in the tumor microenvironment. Cell 170, 127–141.e15 (2017).
Ge, M. Q. et al. NK cells regulate CD8+ T cell priming and dendritic cell migration during influenza a infection by IFN-γ and perforin-dependent mechanisms. J. Immunol. 189, 2099–2109 (2012).
Lee, S. H. et al. Identifying the initiating events of anti-listeria responses using mice with conditional loss of IFN-γ receptor subunit 1 (IFNGR1). J. Immunol. 191, 4223–4234 (2013).
Wojno, E. D. T., Hunter, C. A. & Stumhofer, J. S. The immunobiology of the interleukin-12 family: room for discovery. Immunity 50, 851–870 (2019).
Gocher, A. M., Workman, C. J. & Vignali, D. A. A. Interferon-γ: teammate or opponent in the tumour microenvironment? Nat. Rev. Immunol. 22, 158–172 (2022).
Chow, M. T. et al. Intratumoral activity of the CXCR3 chemokine system is required for the efficacy of anti-PD-1 therapy. Immunity 50, 1498–1512.e5 (2019).
Garris, C. S. et al. Successful anti-PD-1 cancer immunotherapy requires T cell-dendritic cell crosstalk involving the cytokines IFN-γ and IL-12. Immunity 49, 1148–1161.e7 (2018). This study describes a feedforward loop of IFNγ and IL-12 signalling between T cells and DCs that is important for development of antitumour immunity and mediates responsiveness to immunotherapy.
Ayers, M. et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 127, 2930–2940 (2017).
Grasso, C. S. et al. Conserved interferon-γ signaling drives clinical response to immune checkpoint blockade therapy in melanoma. Cancer Cell 38, 500–515.e3 (2020).
Liu, Y. et al. Blockade of IDO–kynurenine–AhR metabolic circuitry abrogates IFN-γ-induced immunologic dormancy of tumor-repopulating cells. Nat. Commun. 8, 15207 (2017).
Allen, E. M. V. et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 350, 207–211 (2015).
Benci, J. L. et al. Tumor interferon signaling regulates a multigenic resistance program to immune checkpoint blockade. Cell 167, 1540–1554.e12 (2016).
Paschen, A., Melero, I. & Ribas, A. Central role of the antigen-presentation and interferon-γ pathways in resistance to immune checkpoint blockade. Annu. Rev. Cancer Biol. 6, 85–102 (2021).
Castro, F., Cardoso, A. P., Gonçalves, R. M., Serre, K. & Oliveira, M. J. Interferon-γ at the crossroads of tumor immune surveillance or evasion. Front. Immunol. 9, 847 (2018).
Peng, Q. et al. PD-L1 on dendritic cells attenuates T cell activation and regulates response to immune checkpoint blockade. Nat. Commun. 11, 4835 (2020).
Harden, J. L. et al. Dichotomous effects of IFN-γ on dendritic cell function determine the extent of IL-12-driven antitumor T cell immunity. J. Immunol. 187, 126–132 (2011).
Li, Q., Harden, J. L., Anderson, C. D. & Egilmez, N. K. Tolerogenic phenotype of IFN-γ-induced IDO+ dendritic cells is maintained via an autocrine IDO–kynurenine/AhR–IDO loop. J. Immunol. 197, 962–970 (2016).
Platten, M., Nollen, E. A. A., Röhrig, U. F., Fallarino, F. & Opitz, C. A. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat. Rev. Drug. Discov. 18, 379–401 (2019).
Benci, J. L. et al. Opposing functions of interferon coordinate adaptive and innate immune responses to cancer immune checkpoint blockade. Cell 178, 933–948.e14 (2019). This study demonstrates how IFNγ signalling, depending on the cell type within the TME, can have opposing effects on dictating the immunogenicity of the tumour.
Dhainaut, M. et al. Spatial CRISPR genomics identifies regulators of the tumor microenvironment. Cell 185, 1223–1239.e20 (2022).
Hayden, M. S. & Ghosh, S. NF-κB in immunobiology. Cell Res. 21, 223 244 (2011).
Ouaaz, F., Arron, J., Zheng, Y., Choi, Y. & Beg, A. A. Dendritic cell development and survival require distinct NF-κB subunits. Immunity 16, 257–270 (2002).
Kriehuber, E. et al. Balance between NF-κB and JNK/AP-1 activity controls dendritic cell life and death. Blood 106, 175–183 (2005).
Dissanayake, D. et al. Nuclear factor-κB1 controls the functional maturation of dendritic cells and prevents the activation of autoreactive T cells. Nat. Med. 17, 1663–1667 (2011).
Larghi, P. et al. The p50 subunit of NF-κB orchestrates dendritic cell lifespan and activation of adaptive immunity. PLoS ONE 7, e45279 (2012).
Zhong, H., May, M. J., Jimi, E. & Ghosh, S. The phosphorylation status of nuclear NF-κB determines its association with CBP/p300 or HDAC-1. Mol. Cell 9, 625–636 (2002).
Cheng, C. S. et al. The specificity of innate immune responses is enforced by repression of interferon response elements by NF-κB p50. Sci. Signal. 4, ra11 (2011).
Kool, M. et al. The ubiquitin-editing protein A20 prevents dendritic cell activation, recognition of apoptotic cells, and systemic autoimmunity. Immunity 35, 82–96 (2011).
Hammer, G. E. et al. Expression of A20 by dendritic cells preserves immune homeostasis and prevents colitis and spondyloarthritis. Nat. Immunol. 12, 1184–1193 (2011).
Ma, A. & Malynn, B. A. A20: linking a complex regulator of ubiquitylation to immunity and human disease. Nat. Rev. Immunol. 12, 774–785 (2012).
Baratin, M. et al. Homeostatic NF-κB signaling in steady-state migratory dendritic cells regulates immune homeostasis and tolerance. Immunity 42, 627–639 (2015). This paper is an excellent demonstration of how interrogating a transcriptional programme can uncover a potentially pivotal signalling network (that is, the cNF-κB pathway) that regulates DC maturation and function.
Klein, C., Brinkmann, U., Reichert, J. M. & Kontermann, R. E. The present and future of bispecific antibodies for cancer therapy. Nat. Rev. Drug Discov. 23, 301–319 (2024).
Hou, X., Zaks, T., Langer, R. & Dong, Y. Lipid nanoparticles for mRNA delivery. Nat. Rev. Mater. 6, 1078–1094 (2021).
Shinkura, R. et al. Alymphoplasia is caused by a point mutation in the mouse gene encoding NF-κB-inducing kinase. Nat. Genet. 22, 74–77 (1999).
Sun, S.-C. The non-canonical NF-κB pathway in immunity and inflammation. Nat. Rev. Immunol. 17, 545–558 (2017). This is a definitive review on the role of ncNF-κB pathway in immunity.
Sallusto, F. & Lanzavecchia, A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor α. J. Exp. Med. 179, 1109–1118 (1994).
Caux, C. et al. Activation of human dendritic cells through CD40 cross-linking. J. Exp. Med. 180, 1263–1272 (1994).
Schulz, O. et al. CD40 triggering of heterodimeric IL-12 p70 production by dendritic cells in vivo requires a microbial priming signal. Immunity 13, 453–462 (2000).
Cella, M. et al. Ligation of CD40 on dendritic cells triggers production of high levels of interleukin-12 and enhances T cell stimulatory capacity: T-T help via APC activation. J. Exp. Med. 184, 747–752 (1996).
Ferris, S. T. et al. cDC1 prime and are licensed by CD4+ T cells to induce anti-tumour immunity. Nature 584, 624–629 (2020). This study is an elegant demonstration of how cDC1s prime CD4 and CD8 T cells necessary for tumour rejection.
Wu, R. et al. Mechanisms of CD40-dependent cDC1 licensing beyond costimulation. Nat. Immunol. 23, 1536–1550 (2022). This study shows how CD40 signalling, beyond its well-established role in co-stimulation, is critical for DC survival and metabolic rewiring.
Jie, Z. et al. NIK signaling axis regulates dendritic cell function in intestinal immunity and homeostasis. Nat. Immunol. 19, 1224–1235 (2018).
Hofmann, J., Mair, F., Greter, M., Schmidt-Supprian, M. & Becher, B. NIK signaling in dendritic cells but not in T cells is required for the development of effector T cells and cell-mediated immune responses. J. Exp. Med. 208, 1917–1929 (2011).
Lind, E. F. et al. Dendritic cells require the NF-κB2 pathway for cross-presentation of soluble antigens. J. Immunol. 181, 354–363 (2008).
Katakam, A. K. et al. Dendritic cells require NIK for CD40-dependent cross-priming of CD8+ T cells. Proc. Natl Acad. Sci. USA 112, 14664–14669 (2015).
Du, X. et al. Hippo/Mst signalling couples metabolic state and immune function of CD8α+ dendritic cells. Nature 558, 141–145 (2018). This study is an elegant demonstration of how activation of ncNF-κB is linked with rewiring of the metabolic state of DCs upon its maturation.
Jin, J. et al. Noncanonical NF-κB pathway controls the production of type I interferons in antiviral innate immunity. Immunity 40, 342–354 (2014).
Navarro, H. I. et al. RelB-deficient autoinflammatory pathology presents as interferonopathy, but in mice is interferon-independent. J. Allergy Clin. Immunol. 152, 1261–1272 (2023).
Bullock, T. N. J. CD40 stimulation as a molecular adjuvant for cancer vaccines and other immunotherapies. Cell. Mol. Immunol. 19, 14–22 (2022).
Vonderheide, R. H. CD40 agonist antibodies in cancer immunotherapy. Annu. Rev. Med. 71, 47–58 (2019).
Zhang, Y. et al. Close the cancer–immunity cycle by integrating lipid nanoparticle–mRNA formulations and dendritic cell therapy. Nat. Nanotechnol. 18, 1364–1374 (2023).
Salomon, R. et al. Bispecific antibodies increase the therapeutic window of CD40 agonists through selective dendritic cell targeting. Nat. Cancer 3, 287–302 (2022).
Morrish, E., Brumatti, G. & Silke, J. Future therapeutic directions for smac-mimetics. Cells 9, 406 (2020).
Hou, Y. et al. Non-canonical NF-κB antagonizes STING sensor-mediated DNA sensing in radiotherapy. Immunity 49, 490–503.e4 (2018).
Reinfeld, B. I. et al. Cell-programmed nutrient partitioning in the tumour microenvironment. Nature 593, 282–288 (2021). This formative study demonstrates how different cell types within the TME compete for different metabolites.
Wculek, S. K., Khouili, S. C., Priego, E., Heras-Murillo, I. & Sancho, D. Metabolic control of dendritic cell functions: digesting information. Front. Immunol. 10, 775 (2019).
Thwe, P. M. et al. Cell-intrinsic glycogen metabolism supports early glycolytic reprogramming required for dendritic cell immune responses. Cell Metab. 26, 558–567.e5 (2017).
Maschalidi, S. et al. Targeting SLC7A11 improves efferocytosis by dendritic cells and wound healing in diabetes. Nature 606, 776–784 (2022).
Everts, B. et al. TLR-driven early glycolytic reprogramming via the kinases TBK1-IKKɛ supports the anabolic demands of dendritic cell activation. Nat. Immunol. 15, 323–332 (2014). This groundbreaking study characterizes the metabolic rewiring that occurs upon activation in DCs and demonstrates their need to generate TCA intermediates for downstream function.
Maroof, A., English, N. R., Bedford, P. A., Gabrilovich, D. I. & Knight, S. C. Developing dendritic cells become ‘lacy’ cells packed with fat and glycogen. Immunology 115, 473–483 (2005).
Caronni, N. et al. Downregulation of membrane trafficking proteins and lactate conditioning determine loss of dendritic cell function in lung cancer. Cancer Res. 78, 1685–1699 (2018).
Zhang, Z. et al. Antigen presentation by dendritic cells in tumors is disrupted by altered metabolism that involves pyruvate kinase M2 and its interaction with SOCS3. Cancer Res. 70, 89–98 (2010).
Altman, B. J., Stine, Z. E. & Dang, C. V. From Krebs to clinic: glutamine metabolism to cancer therapy. Nat. Rev. Cancer 16, 619–634 (2016).
Guo, C. et al. SLC38A2 and glutamine signalling in cDC1s dictate anti-tumour immunity. Nature 620, 200–208 (2023).
Lawless, S. J. et al. Glucose represses dendritic cell-induced T cell responses. Nat. Commun. 8, 15620 (2017).
Nobs, S. P. et al. Lung dendritic-cell metabolism underlies susceptibility to viral infection in diabetes. Nature 624, 645–652 (2023).
Pearce, E. J. & Everts, B. Dendritic cell metabolism. Nat. Rev. Immunol. 15, 18–29 (2015).
Ibrahim, J. et al. Dendritic cell populations with different concentrations of lipid regulate tolerance and immunity in mouse and human liver. Gastroenterology 143, 1061–1072 (2012).
Bonacina, F. et al. Myeloid apolipoprotein E controls dendritic cell antigen presentation and T cell activation. Nat. Commun. 9, 3083 (2018).
Westerterp, M. et al. Cholesterol accumulation in dendritic cells links the inflammasome to acquired immunity. Cell Metab. 25, 1294–1304.e6 (2017).
Bougnères, L. et al. A role for lipid bodies in the cross-presentation of phagocytosed antigens by MHC class I in dendritic cells. Immunity 31, 232–244 (2009). This study demonstrates the critical role of lipid bodies in antigen presentation.
Peng, X., He, Y., Huang, J., Tao, Y. & Liu, S. Metabolism of dendritic cells in tumor microenvironment: for immunotherapy. Front. Immunol. 12, 613492 (2021).
Herber, D. L. et al. Lipid accumulation and dendritic cell dysfunction in cancer. Nat. Med. 16, 880–886 (2010).
Cubillos-Ruiz, J. R. et al. ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell 161, 1527–1538 (2015).
Cao, W. et al. Oxidized lipids block antigen cross-presentation by dendritic cells in cancer. J. Immunol. 192, 2920–2931 (2014).
Veglia, F. et al. Lipid bodies containing oxidatively truncated lipids block antigen cross-presentation by dendritic cells in cancer. Nat. Commun. 8, 2122 (2017).
Mogilenko, D. A. et al. Metabolic and innate immune cues merge into a specific inflammatory response via the UPR. Cell 177, 1201–1216.e19 (2019).
Villablanca, E. J. et al. Tumor-mediated liver X receptor-α activation inhibits CC chemokine receptor-7 expression on dendritic cells and dampens antitumor responses. Nat. Med. 16, 98–105 (2010). This study demonstrates how cholesterol metabolism regulates DC maturation and function and how such processes are exploited by tumours.
Prendergast, G. C., Malachowski, W. P., DuHadaway, J. B. & Muller, A. J. Discovery of IDO1 inhibitors: from bench to bedside. Cancer Res. 77, 6795–6811 (2017).
Mondanelli, G., Iacono, A., Allegrucci, M., Puccetti, P. & Grohmann, U. Immunoregulatory interplay between arginine and tryptophan metabolism in health and disease. Front. Immunol. 10, 1565 (2019).
Gargaro, M. et al. Indoleamine 2,3-dioxygenase 1 activation in mature cDC1 promotes tolerogenic education of inflammatory cDC2 via metabolic communication. Immunity 55, 1032–1050.e14 (2022).
Long, G. V. et al. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. Lancet Oncol. 20, 1083–1097 (2019).
Eynde, B. J. V., den, Baren, Nvan & Baurain, J.-F. Is there a clinical future for IDO1 inhibitors after the failure of epacadostat in melanoma? Annu. Rev. Cancer Biol. 4, 241–256 (2019).
Sadik, A. et al. IL4I1 is a metabolic immune checkpoint that activates the AHR and promotes tumor progression. Cell 182, 1252–1270.e34 (2020).
Odunsi, K. et al. Metabolic adaptation of ovarian tumors in patients treated with an IDO1 inhibitor constrains antitumor immune responses. Sci. Transl. Med. 14, eabg8402 (2022).
Förster, R. et al. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell 99, 23–33 (1999).
Ohl, L. et al. CCR7 governs skin dendritic cell migration under inflammatory and steady-state conditions. Immunity 21, 279–288 (2004).
Yin, X., Chen, S. & Eisenbarth, S. C. Dendritic cell regulation of T helper cells. Annu. Rev. Immunol. 39, 759–790 (2021).
Eisenbarth, S. C. Dendritic cell subsets in T cell programming: location dictates function. Nat. Rev. Immunol. 19, 89–103 (2019).
Hannedouche, S. et al. Oxysterols direct immune cell migration via EBI2. Nature 475, 524–527 (2011). This study demonstrates how oxysterol can help to localize DCs at an optimal position within lymph nodes to be able to interact with both T and B cells.
Waldmann, T. A., Dubois, S., Miljkovic, M. D. & Conlon, K. C. IL-15 in the combination immunotherapy of cancer. Front. Immunol. 11, 868 (2020).
Pilato, M. D. et al. CXCR6 positions cytotoxic T cells to receive critical survival signals in the tumor microenvironment. Cell 184, 4512–4530.e22 (2021).
Philip, M. & Schietinger, A. CD8+ T cell differentiation and dysfunction in cancer. Nat. Rev. Immunol. 22, 209–223 (2022).
Propper, D. J. & Balkwill, F. R. Harnessing cytokines and chemokines for cancer therapy. Nat. Rev. Clin. Oncol. 19, 237–253 (2022).
Dähling, S. et al. Type 1 conventional dendritic cells maintain and guide the differentiation of precursors of exhausted T cells in distinct cellular niches. Immunity 55, 656–670.e8 (2022).
Schenkel, J. M. et al. Conventional type I dendritic cells maintain a reservoir of proliferative tumor-antigen specific TCF-1+ CD8+ T cells in tumor-draining lymph nodes. Immunity 54, 2338–2353.e6 (2021).
Spranger, S., Bao, R. & Gajewski, T. F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 523, 231–235 (2015).
Prokhnevska, N. et al. CD8+ T cell activation in cancer comprises an initial activation phase in lymph nodes followed by effector differentiation within the tumor. Immunity 56, 107–124.e5 (2023).
Giladi, A. et al. Dissecting cellular crosstalk by sequencing physically interacting cells. Nat. Biotechnol. 38, 629–637 (2020).
Nakandakari-Higa, S. et al. Universal recording of immune cell interactions in vivo. Nature 627, 399–406 (2024).
Bodenmiller, B. Multiplexed epitope-based tissue imaging for discovery and healthcare applications. Cell Syst. 2, 225–238 (2016).
Tian, L., Chen, F. & Macosko, E. Z. The expanding vistas of spatial transcriptomics. Nat. Biotechnol. 41, 773–782 (2023).
Jansen, C. S. et al. An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature 576, 465–470 (2019).
Joshi, N. S. et al. Regulatory T cells in tumor-associated tertiary lymphoid structures suppress anti-tumor T cell responses. Immunity 43, 579–590 (2015).
Schumacher, T. N. & Thommen, D. S. Tertiary lymphoid structures in cancer. Science 375, eabf9419 (2022).
Teillaud, J.-L., Houel, A., Panouillot, M., Riffard, C. & Dieu-Nosjean, M.-C. Tertiary lymphoid structures in anticancer immunity. Nat. Rev. Cancer 24, 629–646 (2024).
Ayala, M. A. M. et al. CXCR3 expression in regulatory T cells drives interactions with type I dendritic cells in tumors to restrict CD8+ T cell antitumor immunity. Immunity 56, 1613–1630.e5 (2023).
Goc, J. et al. Dendritic cells in tumor-associated tertiary lymphoid structures signal a Th1 cytotoxic immune contexture and license the positive prognostic value of infiltrating CD8+ T cells. Cancer Res. 74, 705–715 (2014).
Goc, J., Fridman, W.-H., Sautès-Fridman, C. & Dieu-Nosjean, M.-C. Characteristics of tertiary lymphoid structures in primary cancers. OncoImmunology 2, e26836 (2013).
Alexandre, Y. O. et al. XCR1+ dendritic cells promote memory CD8+ T cell recall upon secondary infections with Listeria monocytogenes or certain viruses. J. Exp. Med. 213, 75–92 (2016).
Cook, S. J. et al. Differential chemokine receptor expression and usage by pre‐cDC1 and pre‐cDC2. Immunol. Cell Biol. 96, 1131–1139 (2018).
Moussion, C. & Girard, J.-P. Dendritic cells control lymphocyte entry to lymph nodes through high endothelial venules. Nature 479, 542–546 (2011). This study shows that DC-derived lymphotoxin is critical for high endothelial venule maintenance.
Lavin, Y. et al. Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses. Cell 169, 750–765.e17 (2017).
Martinet, L. et al. High endothelial venule blood vessels for tumor-infiltrating lymphocytes are associated with lymphotoxin β-producing dendritic cells in human breast cancer. J. Immunol. 191, 2001–2008 (2013).
Martinet, L. et al. High endothelial venules (HEVs) in human melanoma lesions. OncoImmunology 1, 829–839 (2012).
Qian, J. et al. A pan-cancer blueprint of the heterogeneous tumor microenvironment revealed by single-cell profiling. Cell Res. 30, 745–762 (2020).
Wu, S.-Y. et al. CCL19+ dendritic cells potentiate clinical benefit of anti-PD-(L)1 immunotherapy in triple-negative breast cancer. Med 4, 373–393.e8 (2023).
Franken, A. et al. CD4+ T cell activation distinguishes response to anti-PD-L1+anti-CTLA4 therapy from anti-PD-L1 monotherapy. Immunity 57, 541–558.e7 (2024).
Kim, N. et al. Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma. Nat. Commun. 11, 2285 (2020).
Cillo, A. R. et al. Immune landscape of viral- and carcinogen-driven head and neck cancer. Immunity 52, 183–199.e9 (2020).
Wang, W. et al. Lymphatic endothelial transcription factor Tbx1 promotes an immunosuppressive microenvironment to facilitate post-myocardial infarction repair. Immunity 56, 2342–2357.e10 (2023).
Cottam, M. A., Caslin, H. L., Winn, N. C. & Hasty, A. H. Multiomics reveals persistence of obesity-associated immune cell phenotypes in adipose tissue during weight loss and weight regain in mice. Nat. Commun. 13, 2950 (2022).
Wang, J. et al. Single-cell multiomics defines tolerogenic extrathymic Aire-expressing populations with unique homology to thymic epithelium. Sci. Immunol. 6, eabl5053 (2021).
Reynolds, G. et al. Developmental cell programs are co-opted in inflammatory skin disease. Science 371, eaba6500 (2021).
Gu, W., Eke, C., Santiago, E. G., Olaloye, O. & Konnikova, L. Single-cell atlas of the small intestine throughout the human lifespan demonstrates unique features of fetal immune cells. Mucosal Immunol. 17, 599–617 (2024).
Alam, J. et al. Single-cell transcriptional profiling of murine conjunctival immune cells reveals distinct populations expressing homeostatic and regulatory genes. Mucosal Immunol. 15, 620–628 (2022).
Clappaert, E. J. et al. Flt3L therapy increases the abundance of Treg-promoting CCR7+ cDCs in preclinical cancer models. Front. Immunol. 14, 1166180 (2023).
Tietscher, S. et al. A comprehensive single-cell map of T cell exhaustion-associated immune environments in human breast cancer. Nat. Commun. 14, 98 (2023).
Liu, L. et al. Single cell sequencing reveals that CD39 inhibition mediates changes to the tumor microenvironment. Nat. Commun. 13, 6740 (2022).
Antunes, A. R. P. et al. Single-cell profiling of myeloid cells in glioblastoma across species and disease stage reveals macrophage competition and specialization. Nat. Neurosci. 24, 595–610 (2021).
Lee, A. H. et al. Neoadjuvant PD-1 blockade induces T cell and cDC1 activation but fails to overcome the immunosuppressive tumor associated macrophages in recurrent glioblastoma. Nat. Commun. 12, 6938 (2021).
Ji, A. L. et al. Multimodal analysis of composition and spatial architecture in human squamous cell carcinoma. Cell 182, 497–514.e22 (2020).
Silva-Sanchez, A. et al. Activation of regulatory dendritic cells by Mertk coincides with a temporal wave of apoptosis in neonatal lungs. Sci. Immunol. 8, eadc9081 (2023).
Thumkeo, D. et al. PGE2–EP2/EP4 signaling elicits immunosuppression by driving the mregDC–Treg axis in inflammatory tumor microenvironment. Cell Rep. 39, 110914 (2022).
Smalley, I. et al. Single-cell characterization of the immune microenvironment of melanoma brain and leptomeningeal metastases. Clin. Cancer Res. 27, 4109–4125 (2021).
Chen, Y.-P. et al. Single-cell transcriptomics reveals regulators underlying immune cell diversity and immune subtypes associated with prognosis in nasopharyngeal carcinoma. Cell Res. 30, 1024–1042 (2020).
Herrera, F. G. et al. Low dose radiotherapy reverses tumor immune desertification and resistance to immunotherapy. Cancer Discov. 12, 108–133 (2021).
Combes, A. J. et al. Discovering dominant tumor immune archetypes in a pan-cancer census. Cell 185, 184–203.e19 (2022).
Sinjab, A. et al. Resolving the spatial and cellular architecture of lung adenocarcinoma by multiregion single-cell sequencing. Cancer Discov. 11, 2506–2523 (2021).
Zhang, Y. et al. Single-cell analyses reveal key immune cell subsets associated with response to PD-L1 blockade in triple-negative breast cancer. Cancer Cell 39, 1578–1593.e8 (2021).
Zhang, Q. et al. Landscape and dynamics of single immune cells in hepatocellular carcinoma. Cell 179, 829–845.e20 (2019).
Liu, Y. et al. Immune phenotypic linkage between colorectal cancer and liver metastasis. Cancer Cell 40, 424–437.e5 (2022).
Chen, Z. et al. Single-cell RNA sequencing highlights the role of inflammatory cancer-associated fibroblasts in bladder urothelial carcinoma. Nat. Commun. 11, 5077 (2020).
Zheng, Y. et al. Immune suppressive landscape in the human esophageal squamous cell carcinoma microenvironment. Nat. Commun. 11, 6268 (2020).
Bill, R. et al. CXCL9:SPP1 macrophage polarity identifies a network of cellular programs that control human cancers. Science 381, 515–524 (2023).
Liu, Y. et al. Tumour heterogeneity and intercellular networks of nasopharyngeal carcinoma at single cell resolution. Nat. Commun. 12, 741 (2021).
Sáez, P. J. et al. ATP promotes the fast migration of dendritic cells through the activity of pannexin 1 channels and P2X7 receptors. Sci. Signal. 10, eaah7107 (2017).
Brausi, M. et al. Side effects of Bacillus Calmette-Guérin (BCG) in the treatment of intermediate- and high-risk Ta, T1 papillary carcinoma of the bladder: results of the EORTC genito-urinary cancers group randomised phase 3 study comparing one-third dose with full dose and 1 year with 3 years of maintenance BCG. Eur. Urol. 65, 69–76 (2014).
Geisse, J. et al. Imiquimod 5% cream for the treatment of superficial basal cell carcinoma: results from two phase III, randomized, vehicle-controlled studies. J. Am. Acad. Dermatol. 50, 722–733 (2004).
Brody, J. D. et al. In situ vaccination with a TLR9 agonist induces systemic lymphoma regression: a phase I/II study. J. Clin. Oncol. 28, 4324–4332 (2010).
Bakhribah, H. et al. A phase I study of the Toll-like receptor 5 (TLR5) agonist, entolimod in patients (pts) with advanced cancers. J. Clin. Oncol. 33, 3063 (2015).
Grimm, C. et al. Treatment of cervical intraepithelial neoplasia with topical imiquimod: a randomized controlled trial. Obstet. Gynecol. 120, 152–159 (2012).
Pollack, I. F. et al. Immune responses and outcome after vaccination with glioma-associated antigen peptides and poly-ICLC in a pilot study for pediatric recurrent low-grade gliomas. Neurooncol. 18, 1157–1168 (2016).
Mehrotra, S. et al. Vaccination with poly(IC:LC) and peptide-pulsed autologous dendritic cells in patients with pancreatic cancer. J. Hematol. Oncol. 10, 82 (2017).
Mett, V. et al. Mobilan: a recombinant adenovirus carrying Toll-like receptor 5 self-activating cassette for cancer immunotherapy. Oncogene 37, 439–449 (2018).
Nooka, A. K. et al. Assessment of safety and immunogenicity of PVX-410 vaccine with or without lenalidomide in patients with smoldering multiple myeloma: a nonrandomized clinical trial. JAMA Oncol. 4, e183267 (2018).
Bhatia, S. et al. Intratumoral G100, a TLR4 agonist, induces antitumor immune responses and tumor regression in patients with merkel cell carcinoma. Clin. Cancer Res. 25, 1185–1195 (2019).
Kim, Y. H. et al. In situ vaccination against mycosis fungoides by intratumoral injection of a TLR9 agonist combined with radiation: a phase 1/2 study. Blood 119, 355–363 (2012).
Manegold, C. et al. Randomized phase II trial of a Toll-like receptor 9 agonist oligodeoxynucleotide, PF-3512676, in combination with first-line taxane plus platinum chemotherapy for advanced-stage non-small-cell lung cancer. J. Clin. Oncol. 26, 3979–3986 (2008).
Weber, J. S. et al. Randomized phase 2/3 trial of CpG oligodeoxynucleotide PF‐3512676 alone or with dacarbazine for patients with unresectable stage III and IV melanoma. Cancer 115, 3944–3954 (2009).
Hammerich, L. et al. Systemic clinical tumor regressions and potentiation of PD1 blockade with in situ vaccination. Nat. Med. 25, 814–824 (2019).
Pavlick, A. et al. Combined vaccination with NY-ESO-1 protein, poly-ICLC, and montanide improves humoral and cellular immune responses in patients with high-risk melanoma. Cancer Immunol. Res. 8, 70–80 (2020).
Ribas, A. et al. SD-101 in combination with pembrolizumab in advanced melanoma: results of a phase Ib, multicenter study. Cancer Discov. 8, 1250–1257 (2018).
Sagiv-Barfi, I. et al. Eradication of spontaneous malignancy by local immunotherapy. Sci. Transl. Med. 10, eaan4488 (2018).
Rolfo, C., Giovannetti, E., Martinez, P., McCue, S. & Naing, A. Applications and clinical trial landscape using Toll-like receptor agonists to reduce the toll of cancer. NPJ Precis. Oncol. 7, 26 (2023).
Man, S. M. & Jenkins, B. J. Context-dependent functions of pattern recognition receptors in cancer. Nat. Rev. Cancer 22, 397–413 (2022).
Jneid, B. et al. Selective STING stimulation in dendritic cells primes antitumor T cell responses. Sci. Immunol. 8, eabn6612 (2023).
Lanng, K. R. B., Lauridsen, E. L. & Jakobsen, M. R. The balance of STING signaling orchestrates immunity in cancer. Nat. Immunol. 25, 1144–1157 (2024).
Samson, N. & Ablasser, A. The cGAS–STING pathway and cancer. Nat. Cancer 3, 1452–1463 (2022).
Wang, J. et al. STING licensing of type I dendritic cells potentiates antitumor immunity. Sci. Immunol. 9, eadj3945 (2024).
Sokolowska, O. & Nowis, D. STING signaling in cancer cells: important or not? Arch. Immunol. Ther. Exp. 66, 125–132 (2018).
Sivick, K. E. et al. Magnitude of therapeutic STING activation determines CD8+ T cell-mediated anti-tumor immunity. Cell Rep. 25, 3074–3085.e5 (2018).
Gaidt, M. M. et al. The DNA inflammasome in human myeloid cells is initiated by a STING-cell death program upstream of NLRP3. Cell 171, 1110–1124.e18 (2017).
Kabelitz, D. et al. Signal strength of STING activation determines cytokine plasticity and cell death in human monocytes. Sci. Rep. 12, 17827 (2022).
Wu, J. et al. STING-mediated disruption of calcium homeostasis chronically activates ER stress and primes T cell death. J. Exp. Med. 216, 867–883 (2019).
Li, J. et al. Non-cell-autonomous cancer progression from chromosomal instability. Nature 620, 1080–1088 (2023).
Harrington, K. J. et al. Preliminary results of the first-in-human (FIH) study of MK-1454, an agonist of stimulator of interferon genes (STING), as monotherapy or in combination with pembrolizumab (pembro) in patients with advanced solid tumors or lymphomas. Ann. Oncol. 29, viii712 (2018).
Meric-Bernstam, F. et al. Phase Ib study of MIW815 (ADU-S100) in combination with spartalizumab (PDR001) in patients (pts) with advanced/metastatic solid tumors or lymphomas. J. Clin. Oncol. 37, 2507 (2019).
Chau, C. H., Steeg, P. S. & Figg, W. D. Antibody–drug conjugates for cancer. Lancet 394, 793–804 (2019).
Acknowledgements
The authors acknowledge F. Ginhoux for insightful feedback on the manuscript. The authors also acknowledge the following funding sources. C.Y.M. is supported by National Institutes of Health (NIH) grant F30CA287638. R.M. is supported by the 2021 AACR-AstraZeneca Immuno-oncology Research Fellowship (21-40-12-MATT) and by the Portuguese Foundation for Science and Technology (2023.15874.PEX). D.P. is supported by Damon Runyon Rachleff Innovator Award, an NIH DP2 New Innovator Award (1DP2AI177905-0) and the Blavatnik Foundation. M.M. is partially supported by NIH grants CA257195, CA254104 and CA154947.
Author information
Authors and Affiliations
Contributions
C.Y.M., M.B., R.M. and M.D.P. researched data for the article. All authors contributed substantially to discussion of the content. All authors wrote the article. All authors reviewed and/or edited the manuscript before submission.
Corresponding author
Ethics declarations
Competing interests
M.M. serves on the scientific advisory board and/or holds stock from Compugen, Myeloid Therapeutics, Asher Bio, Dren Bio, Oncoresponse, Owkin, OSE, DemBio, Larkspur, Innate Pharma and Genenta; and receives funding for contracted research from Regeneron and Boerhinger Ingelheim. The above interests are not directly relevant to this article. The other authors declare no competing interests.
Peer review
Peer review information
Nature Reviews Cancer thanks Caetano Reis e Sousa, who co-reviewed with Victor Bosteels and Cecile Piot; 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.
Supplementary information
Glossary
- Bispecific antibodies
-
(BsAbs). Engineered antibodies that contain two binding sites directed at two different epitopes.
- CCR7-mediated migration
-
A main mode of migration for dendritic cells to the secondary lymphoid organs upon maturation in response to CCL19 and CCL22, ligands for CCR7.
- cGAS–STING pathway
-
A signalling pathway involving cyclic GMP–AMP synthase (cGAS) and stimulator of interferon genes (STING) that detects cytosolic DNA and activates the production of type I interferons in response to infection or cellular stress.
- Damage-associated molecular patterns
-
(DAMPs). Molecules released by dying cells that trigger immune responses.
- DC maturation
-
A process by which dendritic cells (DCs) gain the ability to interact with other haematopoietic cells (predominantly T cells) and shape the downstream immune response towards either a tolerogenic or immunogenic outcome.
- Exhausted T cell progenitors
-
A distinct subpopulation of T cells found in chronic infection and cancer that can self-renew, proliferate and differentiate into either terminally exhausted T cells or cytolytic effector T cell fates.
- Immunogenic responses
-
In dendritic cell-mediated immunity, immunogenic responses arise when a dendritic cell-presented antigen elicits T cell activation, proliferation and release of immunostimulatory cytokines.
- Indoleamine 2,3-dioxygenase 1
-
(IDO1). An enzyme that degrades tryptophan to kynurenines, which can promote activation and differentiation of regulatory T cells.
- Lipid nanoparticles
-
(LNPs). Nanoparticles composed of various mixtures of lipids optimized for nanoparticle stability, cell entry and endosomal escape; these entities are as a technical platform to deliver drugs and/or genetic material (for example, mRNA).
- Metabolic rewiring
-
Changes in the metabolic state of a cell, often brought on to adapt to environmental perturbations or to facilitate various functions of a cell upon differentiation or transition to another cell state.
- Ontogeny
-
The developmental lineage that gives rise to a specific cell type.
- Pathogen-associated molecular patterns
-
(PAMPs). Highly conserved, molecular motifs that derive from microorganisms and are recognized by pattern recognition receptors.
- Pattern recognition receptors
-
(PRRs). A diverse group of receptors expressed both on the cell surface and inside the cell that recognize pathogen-associated molecular patterns and damage-associated molecular patterns and initiate immune responses.
- Phagosomal acidification
-
The phagosome is a cellular compartment containing engulfed material and its acidification facilitates the degradation of internalized cargo by acidic hydrolases.
- SMAC mimetics
-
A class of small-molecule inhibitors that structurally resemble the amino-terminal inhibitor of apoptosis (IAP)-binding motif and can inhibit various IAP proteins.
- SOCS1 and SOCS3
-
Proteins that regulate cytokine signalling by inhibiting the JAK–STAT pathway.
- Tertiary lymphoid structures
-
(TLSs). Ectopic lymphoid structures found in inflammatory tissue sites that feature several organizational aspects akin to a lymph node including T cells, B cells, dendritic cells and high endothelial venules.
- Tolerogenic responses
-
In dendritic cell-mediated immunity, tolerogenic responses arise when a dendritic cell-presented antigen induces a T cell regulatory response.
- Transcriptional programme
-
A set of genes, of which changes in expression are highly correlated with one another; is differentially expressed by a distinct cluster of cells; and is linked with a putative function (or functions) of the cell cluster in question.
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
Moon, C.Y., Belabed, M., Park, M.D. et al. Dendritic cell maturation in cancer. Nat Rev Cancer 25, 225–248 (2025). https://doi.org/10.1038/s41568-024-00787-3
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41568-024-00787-3
This article is cited by
-
The distinct landscape of tumor immune microenvironment in homologous recombination deficient cancers
Biomarker Research (2025)
-
Single-cell transcriptional dissection illuminates an evolution of immunosuppressive microenvironment during pancreatic ductal adenocarcinoma metastasis
Signal Transduction and Targeted Therapy (2025)
-
Grundlagen der Immunresistenz onkologischer Erkrankungen
Die Onkologie (2025)