Le, D. T. et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 372, 2509–2520 (2015).
Pelka, Okay. et al. Spatially organized multicellular immune hubs in human colorectal most cancers. Cell 184, 4734–4752 (2021).
Mihm, M. C. Jr. & Mulé, J. J. Reflections on the histopathology of tumor-infiltrating lymphocytes in melanoma and the host immune response. Most cancers Immunol. Res. 3, 827–835 (2015).
Angell, H. Okay., Bruni, D., Barrett, J. C., Herbst, R. & Galon, J. The immunoscore: colon most cancers and past. Clin. Most cancers Res. 26, 332–339 (2020).
Thommen, D. S. et al. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung most cancers handled with PD-1 blockade. Nat. Med. 24, 994–1004 (2018).
Moldoveanu, D. et al. Spatially mapping the immune panorama of melanoma utilizing imaging mass cytometry. Sci. Immunol. 7, eabi5072 (2022).
Litchfield, Okay. et al. Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition. Cell 184, 596–614 (2021).
Cristescu, R. et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 362, eaar3593 (2018).
Ayers, M. et al. IFN-γ-related mRNA profile predicts medical response to PD-1 blockade. J. Clin. Make investments. 127, 2930–2940 (2017).
Reschke, R. et al. Immune cell and tumor cell-derived CXCL10 is indicative of immunotherapy response in metastatic melanoma. J. Immunother. Most cancers 9, e003521 (2021).
Sade-Feldman, M. et al. Defining T cell states related to response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013 (2018).
Krishna, S. et al. Stem-like CD8 T cells mediate response of adoptive cell immunotherapy in opposition to human most cancers. Science 370, 1328–1334 (2020).
Siddiqui, I. et al. Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like properties promote tumor management in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195–211 (2019).
Kurtulus, S. et al. Checkpoint blockade immunotherapy induces dynamic adjustments in PD-1-CD8+ tumor-infiltrating T cells. Immunity 50, 181–194 (2019).
Miller, B. C. et al. Subsets of exhausted CD8+ T cells differentially mediate tumor management and reply to checkpoint blockade. Nat. Immunol. 20, 326–336 (2019).
Reck, M. et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung most cancers. N. Engl. J. Med. 375, 1823–1833 (2016).
Liu, B., Zhang, Y., Wang, D., Hu, X. & Zhang, Z. Single-cell meta-analyses reveal responses of tumor-reactive CXCL13+ T cells to immune-checkpoint blockade. Nat. Most cancers 3, 1123–1136 (2022).
Caushi, J. X. et al. Transcriptional applications of neoantigen-specific TIL in anti-PD-1-treated lung cancers. Nature 596, 126–132 (2021).
Oliveira, G. et al. Phenotype, specificity and avidity of antitumour CD8+ T cells in melanoma. Nature 596, 119–125 (2021).
Lowery, F. J. et al. Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers. Science 375, 877–884 (2022).
Hanada, Okay.-I. et al. A phenotypic signature that identifies neoantigen-reactive T cells in contemporary human lung cancers. Most cancers Cell 40, 479–493 (2022).
Eisenhauer, E. A. et al. New response analysis standards in strong tumours: revised RECIST guideline (model 1.1). Eur. J. Most cancers 45, 228–247 (2009).
Groom, J. R. & Luster, A. D. CXCR3 in T cell perform. Exp. Cell. Res. 317, 620–631 (2011).
Chen, Okay. H., Boettiger, A. N., Moffitt, J. R., Wang, S. & Zhuang, X. RNA imaging. Spatially resolved, extremely multiplexed RNA profiling in single cells. Science 348, aaa6090 (2015).
Wu, T. et al. The TCF1–Bcl6 axis counteracts kind I interferon to repress exhaustion and preserve T cell stemness. Sci. Immunol. 1, eaai8593 (2016).
Im, S. J. et al. Defining CD8+ T cells that present the proliferative burst after PD-1 remedy. Nature 537, 417–421 (2016).
Eberhardt, C. S. et al. Useful HPV-specific PD-1+ stem-like CD8 T cells in head and neck most cancers. Nature 597, 279–284 (2021).
Lee, J. et al. IL-15 promotes self-renewal of progenitor exhausted CD8 T cells throughout persistent antigenic stimulation. Entrance. Immunol. 14, 1117092 (2023).
Xue, D. et al. A tumor-specific pro-IL-12 prompts preexisting cytotoxic T cells to manage established tumors. Sci. Immunol. 7, eabi6899 (2022).
Maier, B. et al. A conserved dendritic-cell regulatory program limits antitumour immunity. Nature 580, 257–262 (2020).
Qi, J. et al. Single-cell and spatial evaluation reveal interplay of FAP+ fibroblasts and SPP1+ macrophages in colorectal most cancers. Nat. Commun. 13, 1742 (2022).
Kim, N. et al. Single-cell RNA sequencing demonstrates the molecular and mobile reprogramming of metastatic lung adenocarcinoma. Nat. Commun. 11, 2285 (2020).
Tune, A., Nikolcheva, T. & Krensky, A. M. Transcriptional regulation of RANTES expression in T lymphocytes. Immunol. Rev. 177, 236–245 (2000).
Li, H. et al. Dysfunctional CD8 T cells kind a proliferative, dynamically regulated compartment inside human melanoma. Cell 176, 775–789 (2019).
Dubois, S. P., Waldmann, T. A. & Müller, J. R. Survival adjustment of mature dendritic cells by IL-15. Proc. Natl Acad. Sci. USA 102, 8662–8667 (2005).
Di Pilato, M. et al. CXCR6 positions cytotoxic T cells to obtain vital survival alerts within the tumor microenvironment. Cell 184, 4512–4530 (2021).
Cohen, M. et al. The interplay of CD4+ helper T cells with dendritic cells shapes the tumor microenvironment and immune checkpoint blockade response. Nat. Most cancers 3, 303–317 (2022).
Rapp, M. et al. CCL22 controls immunity by selling regulatory T cell communication with dendritic cells in lymph nodes. J. Exp. Med. 216, 1170–1181 (2019).
Oh, D. Y. et al. Intratumoral CD4+ T cells mediate anti-tumor cytotoxicity in human bladder most cancers. Cell 181, 1612–1625 (2020).
Lei, X. et al. CD4+ helper T cells endow cDC1 with cancer-impeding capabilities within the human tumor micro-environment. Nat. Commun. 14, 217 (2023).
Home, I. G. et al. Macrophage-derived CXCL9 and CXCL10 are required for antitumor immune responses following immune checkpoint blockade. Clin. Most cancers Res. 26, 487–504 (2020).
Chow, M. T. et al. Intratumoral exercise of the CXCR3 chemokine system is required for the efficacy of anti-PD-1 remedy. Immunity 50, 1498–1512 (2019).
Nixon, B. G. et al. Tumor-associated macrophages expressing the transcription issue IRF8 promote T cell exhaustion in most cancers. Immunity 55, 2044–2058 (2022).
Kersten, Okay. et al. Spatiotemporal co-dependency between macrophages and exhausted CD8+ T cells in most cancers. Most cancers Cell 40, 624–638 (2022).
Cang, Z. et al. Screening cell-cell communication in spatial transcriptomics through collective optimum transport. Nat. Strategies 20, 218–228 (2023).
Cheng, H.-W. et al. CCL19-producing fibroblastic stromal cells restrain lung carcinoma development by selling native antitumor T-cell responses. J. Allergy Clin. Immunol. 142, 1257–1271 (2018).
Peng, Y. et al. Single-cell profiling of tumor-infiltrating TCF1/TCF7+ T cells reveals a T lymphocyte subset related to tertiary lymphoid constructions/organs and a superior prognosis in oral most cancers. Oral. Oncol. 119, 105348 (2021).
Im, S. J. et al. Traits and anatomic location of PD-1+TCF1+ stem-like CD8 T cells in persistent viral an infection and most cancers. Proc. Natl Acad. Sci. USA 120, e2221985120 (2023).
Rangel-Moreno, J., Moyron-Quiroz, J. E., Hartson, L., Kusser, Okay. & Randall, T. D. Pulmonary expression of CXC chemokine ligand 13, CC chemokine ligand 19, and CC chemokine ligand 21 is crucial for native immunity to influenza. Proc. Natl Acad. Sci. USA 104, 10577–10582 (2007).
Sato, Y. et al. Heterogeneous fibroblasts underlie age-dependent tertiary lymphoid tissues within the kidney. JCI Perception 1, e87680 (2016).
Sautès-Fridman, C., Petitprez, F., Calderaro, J. & Fridman, W. H. Tertiary lymphoid constructions within the period of most cancers immunotherapy. Nat. Rev. Most cancers 19, 307–325 (2019).
Schumacher, T. N. & Thommen, D. S. Tertiary lymphoid constructions in most cancers. Science 375, eabf9419 (2022).
Merritt, C. R. et al. Multiplex digital spatial profiling of proteins and RNA in fastened tissue. Nat. Biotechnol. 38, 586–599 (2020).
Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set assortment. Cell Syst. 1, 417–425 (2015).
Wherry, E. J. et al. Molecular signature of CD8+ T cell exhaustion throughout persistent viral an infection. Immunity 27, 670–684 (2007).
Hoch, T. et al. Multiplexed imaging mass cytometry of the chemokine milieus in melanoma characterizes options of the response to immunotherapy. Sci. Immunol. 7, eabk1692 (2022).
Schulz, D. et al. Simultaneous multiplexed imaging of mRNA and proteins with subcellular decision in breast most cancers tissue samples by mass cytometry. Cell Syst. 6, 25–36 (2018).
Ardighieri, L. et al. Infiltration by CXCL10 secreting macrophages is related to antitumor immunity and response to remedy in ovarian most cancers subtypes. Entrance. Immunol. 12, 690201 (2021).
Alspach, E. et al. MHC-II neoantigens form tumour immunity and response to immunotherapy. Nature 574, 696–701 (2019).
Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014).
Ryan, N. et al. STAT1 inhibits T-cell exhaustion and myeloid derived suppressor cell accumulation to advertise antitumor immune responses in head and neck squamous cell carcinoma. Int. J. Most cancers 146, 1717–1729 (2020).
Meissl, Okay., Macho-Maschler, S., Müller, M. & Strobl, B. The great and the unhealthy faces of STAT1 in strong tumours. Cytokine 89, 12–20 (2017).
Prokhnevska, N. et al. CD8+ T cell activation in most cancers contains an preliminary activation part in lymph nodes adopted by effector differentiation inside the tumor. Immunity 56, 107–124 (2023).
Huang, Q. et al. The primordial differentiation of tumor-specific reminiscence CD8+ T cells as bona fide responders to PD-1/PD-L1 blockade in draining lymph nodes. Cell 185, 4049–4066 (2022).
Jansen, C. S. et al. An intra-tumoral area of interest maintains and differentiates stem-like CD8 T cells. Nature 576, 465–470 (2019).
Grant, S. M., Lou, M., Yao, L., Germain, R. N. & Radtke, A. J. The lymph node at a look–how spatial group optimizes the immune response. J. Cell Sci. 133, jcs241828 (2020).
Duckworth, B. C. & Groom, J. R. Conversations that depend: mobile interactions that drive T cell destiny. Immunol. Rev. 300, 203–219 (2021).
Gommerman, J. L. & Browning, J. L. Lymphotoxin/mild, lymphoid microenvironments and autoimmune illness. Nat. Rev. Immunol. 3, 642–655 (2003).
Piao, W. et al. Regulatory T cells situation lymphatic endothelia for enhanced transendothelial migration. Cell Rep. 30, 1052–1062 (2020).
Dejardin, E. et al. The lymphotoxin-beta receptor induces totally different patterns of gene expression through two NF-κB pathways. Immunity 17, 525–535 (2002).
Schaeuble, Okay. et al. Perivascular fibroblasts of the growing spleen act as LTα1β2-dependent precursors of each T and B zone organizer cells. Cell Rep. 21, 2500–2514 (2017).
Bar-Ephraïm, Y. E. & Mebius, R. E. Innate lymphoid cells in secondary lymphoid organs. Immunol. Rev. 271, 185–199 (2016).
Magen, A. et al. Intratumoral dendritic cell-CD4+ T helper cell niches allow CD8+ T cell differentiation following PD-1 blockade in hepatocellular carcinoma. Nat. Med. 29, 1389–1399 (2023).
Garon, E. B. et al. Pembrolizumab for the therapy of non-small-cell lung most cancers. N. Engl. J. Med. 372, 2018–2028 (2015).
Filbin, M. R. et al. Longitudinal proteomic evaluation of extreme COVID-19 reveals survival-associated signatures, tissue-specific cell loss of life, and cell–cell interactions. Cell Rep. Med. 2, 100287 (2021).
Traag, V. A., Waltman, L. & van Eck, N. J. From Louvain to Leiden: guaranteeing well-connected communities. Sci. Rep. 9, 5233 (2019).
Subramanian, A. et al. Gene set enrichment evaluation: a knowledge-based strategy for decoding genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).
Stringer, C., Wang, T., Michaelos, M. & Pachitariu, M. Cellpose: a generalist algorithm for mobile segmentation. Nat. Strategies 18, 100–106 (2021).
Petukhov, V. et al. Cell segmentation in imaging-based spatial transcriptomics. Nat. Biotechnol. 40, 345–354 (2022).
Korsunsky, I. et al. Cross-tissue, single-cell stromal atlas identifies shared pathological fibroblast phenotypes in 4 persistent inflammatory illnesses. Med. 3, 481–518 (2022).
Melville, J. uwot: the uniform manifold approximation and projection (UMAP) methodology for dimensionality discount. https://github.com/jlmelville/uwot (2020).
Csardi, G. et al. The igraph software program package deal for complicated community analysis. InterJournal Complicated Syst. 1695, 1–9 (2006).
Bates, D., Mächler, M., Bolker, B. & Walker, S. Becoming linear mixed-effects fashions utilizing lme4. J. Stat. Softw. https://doi.org/10.18637/jss.v067.i01 (2015).
Gelman, A. & Su, Y. -S. arm: knowledge evaluation utilizing regression and multilevel/hierarchical fashions. https://CRAN.R-project.org/package deal=arm (2020).
Korsunsky, I. et al. Quick, delicate and correct integration of single-cell knowledge with Concord. Nat. Strategies 16, 1289–1296 (2019).
Lee, D. T. & Schachter, B. J. Two algorithms for setting up a Delaunay triangulation. Int. J. Comput. Inf. Sci. 9, 219–242 (1980).
Keren, L. et al. A structured tumor-immune microenvironment in triple unfavorable breast most cancers revealed by multiplexed ion beam imaging. Cell 174, 1373–1387 (2018).

