Bressan, D., Battistoni, G. & Hannon, G. J. The dawn of spatial omics. Science 381, eabq4964 (2023).
Liu, L. et al. Spatiotemporal omics for biology and medicine. Cell 187, 4488–4519 (2024).
Jain, S. & Eadon, M. T. Spatial transcriptomics in health and disease. Nat. Rev. Nephrol. 20, 659–671 (2024).
Moses, L. & Pachter, L. Museum of spatial transcriptomics. Nat. Methods 19, 534–546 (2022).
Moffitt, J. R., Lundberg, E. & Heyn, H. The emerging landscape of spatial profiling technologies. Nat. Rev. Genet. 23, 741–759 (2022).
Alexandrov, T., Saez-Rodriguez, J. & Saka, S. K. Enablers and challenges of spatial omics, a melting pot of technologies. Mol. Syst. Biol. 19, e10571 (2023).
De Jonghe, J. et al. scTrends: a living review of commercial single-cell and spatial ’omic technologies. Cell Genom. 4, 100723 (2024).
Gulati, G. S., D’Silva, J. P., Liu, Y., Wang, L. & Newman, A. M. Profiling cell identity and tissue architecture with single-cell and spatial transcriptomics. Nat. Rev. Mol. Cell Biol. 26, 11–31 (2025).
Palla, G., Fischer, D. S., Regev, A. & Theis, F. J. Spatial components of molecular tissue biology. Nat. Biotechnol. 40, 308–318 (2022).
Xiao, Z. et al. 3D reconstruction of a gastrulating human embryo. Cell 187, 2855–2874.e19 (2024).
Xu, Y. et al. A single-cell transcriptome atlas profiles early organogenesis in human embryos. Nat. Cell Biol. 25, 604–615 (2023).
Cui, L. et al. Spatial transcriptomic characterization of a Carnegie stage 7 human embryo. Nat. Cell Biol. 27, 360–369 (2025).
Pavon, N. et al. Patterning ganglionic eminences in developing human brain organoids using a morphogen-gradient-inducing device. Cell Rep. Methods 4, 100689 (2024).
Sanchís-Calleja, F. et al. Decoding morphogen patterning of human neural organoids with a multiplexed single-cell transcriptomic screen. Preprint at bioRxiv https://doi.org/10.1101/2024.02.08.579413 (2024).
Bertacchi, M., Maharaux, G., Loubat, A., Jung, M. & Studer, M. FGF8-mediated gene regulation affects regional identity in human cerebral organoids. eLife 13, e98096 (2024).
Amin, N. D. et al. Generating human neural diversity with a multiplexed morphogen screen in organoids. Cell Stem Cell 31, 1831–1846.e9 (2024).
Chen, Z., Han, F., Du, Y., Shi, H. & Zhou, W. Hypoxic microenvironment in cancer: molecular mechanisms and therapeutic interventions. Signal Transduct. Target. Ther. 8, 70 (2023).
Martínez-Reyes, I. & Chandel, N. S. Cancer metabolism: looking forward. Nat. Rev. Cancer 21, 669–680 (2021).
Carmona-Fontaine, C. et al. Metabolic origins of spatial organization in the tumor microenvironment. Proc. Natl Acad. Sci. USA 114, 2934–2939 (2017).
Haley, M. J. et al. Hypoxia coordinates the spatial landscape of myeloid cells within glioblastoma to affect survival. Sci. Adv. 10, eadj3301 (2024).
Wang, W. et al. Identification of hypoxic macrophages in glioblastoma with therapeutic potential for vasculature normalization. Cancer Cell 42, 815–832.e12 (2024).
Rashidi, A. et al. Myeloid cell-derived creatine in the hypoxic niche promotes glioblastoma growth. Cell Metab. 36, 62–77.e8 (2024).
Sun, H. et al. The relevance between hypoxia-dependent spatial transcriptomics and the prognosis and efficacy of immunotherapy in claudin-low breast cancer. Front. Immunol. 13, 1042835 (2022).
Malagoli Tagliazucchi, G., Wiecek, A. J., Withnell, E. & Secrier, M. Genomic and microenvironmental heterogeneity shaping epithelial-to-mesenchymal trajectories in cancer. Nat. Commun. 14, 789 (2023).
Kureshi, C. T. & Dougan, S. K. Cytokines in cancer. Cancer Cell 43, 15–35 (2025).
Nirmal, A. J. et al. The spatial landscape of progression and immunoediting in primary melanoma at single-cell resolution. Cancer Discov. 12, 1518–1541 (2022).
Zhao, L. et al. Tertiary lymphoid structures in diseases: immune mechanisms and therapeutic advances. Signal Transduct. Target. Ther. 9, 225 (2024).
Wu, R. et al. Comprehensive analysis of spatial architecture in primary liver cancer. Sci. Adv. 7, eabg3750 (2021).
Jing, X. et al. Role of hypoxia in cancer therapy by regulating the tumor microenvironment. Mol. Cancer 18, 157 (2019).
Campillo, N. et al. Differential oxygenation in tumor microenvironment modulates macrophage and cancer cell crosstalk: Novel experimental setting and proof of concept. Front. Oncol. 9, 43 (2019).
Auxillos, J. et al. Spatially resolved analysis of microenvironmental gradient impact on cancer cell phenotypes. Sci. Adv. 10, eadn3448 (2024).
Keren, L. et al. A structured tumor-immune microenvironment in triple negative breast cancer revealed by multiplexed ion beam imaging. Cell 174, 1373–1387.e19 (2018).
Niethammer, P., Grabher, C., Look, A. T. & Mitchison, T. J. A tissue-scale gradient of hydrogen peroxide mediates rapid wound detection in zebrafish. Nature 459, 996–999 (2009).
Kueckelhaus, J. et al. Inferring histology-associated gene expression gradients in spatial transcriptomic studies. Nat. Commun. 15, 7280 (2024).
Li, C. et al. SpaceWalker enables interactive gradient exploration for spatial transcriptomics data. Cell Rep. Methods 3, 100645 (2023).
Chitra, U. et al. Mapping the topography of spatial gene expression with interpretable deep learning. Nat. Methods 22, 298–309 (2025).
Rood, J. E. et al. The Human Cell Atlas from a cell census to a unified foundation model. Nature 637, 1065–1071 (2025).
Dann, E. et al. Precise identification of cell states altered in disease using healthy single-cell references. Nat. Genet. 55, 1998–2008 (2023).
Tabula Sapiens Consortium. The tabula sapiens: a multiple-organ, single-cell transcriptomic atlas of humans. Science 376, eabl4896 (2022).
Barkley, D. et al. Cancer cell states recur across tumor types and form specific interactions with the tumor microenvironment. Nat. Genet. 54, 1192–1201 (2022).
Lomakin, A. et al. Spatial genomics maps the structure, nature and evolution of cancer clones. Nature 611, 594–602 (2022).
Mo, C.-K. et al. Tumour evolution and microenvironment interactions in 2D and 3D space. Nature 634, 1178–1186 (2024).
Erickson, A. et al. Spatially resolved clonal copy number alterations in benign and malignant tissue. Nature 608, 360–367 (2022).
Engblom, C. et al. Spatial transcriptomics of B cell and T cell receptors reveals lymphocyte clonal dynamics. Science 382, eadf8486 (2023).
Cords, L. et al. Cancer-associated fibroblast classification in single-cell and spatial proteomics data. Nat. Commun. 14, 4294 (2023).
Ma, C. et al. Pan-cancer spatially resolved single-cell analysis reveals the crosstalk between cancer-associated fibroblasts and tumor microenvironment. Mol. Cancer 22, 170 (2023).
Chu, X., Tian, Y. & Lv, C. Decoding the spatiotemporal heterogeneity of tumor-associated macrophages. Mol. Cancer 23, 150 (2024).
Matusiak, M. et al. Spatially segregated macrophage populations predict distinct outcomes in colon cancer. Cancer Discov. 14, 1418–1439 (2024).
Nasir, I. et al. Tumor macrophage functional heterogeneity can inform the development of novel cancer therapies. Trends Immunol. 44, 971–985 (2023).
Ianevski, A. et al. Single-cell transcriptomes identify patient-tailored therapies for selective co-inhibition of cancer clones. Nat. Commun. 15, 8579 (2024).
Tirosh, I. & Suva, M. L. Cancer cell states: lessons from ten years of single-cell RNA-sequencing of human tumors. Cancer Cell 42, 1497–1506 (2024).
Bai, Z. et al. Spatially exploring RNA biology in archival formalin-fixed paraffin-embedded tissues. Cell 187, 6760–6779.e24 (2024).
Russell, A. J. C. et al. Slide-tags enables single-nucleus barcoding for multimodal spatial genomics. Nature 625, 101–109 (2024).
Watson, B. R. et al. Spatial transcriptomics of healthy and fibrotic human liver at single-cell resolution. Nat. Commun. 16, 319 (2025).
Mayr, C. H. et al. Spatial transcriptomic characterization of pathologic niches in IPF. Sci. Adv. 10, eadl5473 (2024).
Franzén, L. et al. Mapping spatially resolved transcriptomes in human and mouse pulmonary fibrosis. Nat. Genet. 56, 1725–1736 (2024).
Kenigsbuch, M. et al. A shared disease-associated oligodendrocyte signature among multiple CNS pathologies. Nat. Neurosci. 25, 876–886 (2022).
Lázár, E. et al. Spatiotemporal gene expression and cellular dynamics of the developing human heart. Nat. Genet. https://doi.org/10.1038/s41588-025-02352-6 (2025).
He, P. et al. A human fetal lung cell atlas uncovers proximal-distal gradients of differentiation and key regulators of epithelial fates. Cell 185, 4841–4860.e25 (2022).
Suo, C. et al. Mapping the developing human immune system across organs. Science 376, eabo0510 (2022).
Greenbaum, S. et al. A spatially resolved timeline of the human maternal-fetal interface. Nature 619, 595–605 (2023).
Yayon, N. et al. A spatial human thymus cell atlas mapped to a continuous tissue axis. Nature 635, 708–718 (2024).
Asp, M. et al. A spatiotemporal organ-wide gene expression and cell atlas of the developing human heart. Cell 179, 1647–1660.e19 (2019).
Li, X. et al. Profiling spatiotemporal gene expression of the developing human spinal cord and implications for ependymoma origin. Nat. Neurosci. 26, 891–901 (2023).
Sountoulidis, A. et al. A topographic atlas defines developmental origins of cell heterogeneity in the human embryonic lung. Nat. Cell Biol. 25, 351–365 (2023).
Zhang, B. et al. A human embryonic limb cell atlas resolved in space and time. Nature 635, 668–678 (2024).
To, K. et al. A multi-omic atlas of human embryonic skeletal development. Nature 635, 657–667 (2024).
Quach, H. et al. Early human fetal lung atlas reveals the temporal dynamics of epithelial cell plasticity. Nat. Commun. 15, 5898 (2024).
Sariyar, S. et al. High-parametric protein maps reveal the spatial organization in early-developing human lung. Nat. Commun. 15, 9381 (2024).
Gopee, N. H. et al. A prenatal skin atlas reveals immune regulation of human skin morphogenesis. Nature 635, 679–689 (2024).
Cranley, J. et al. Multiomic analysis reveals developmental dynamics of the human heart in health and disease. Preprint at bioRxiv https://doi.org/10.1101/2024.04.29.591736 (2024).
Bayraktar, S. et al. High-resolution atlas of the developing human heart and the great vessels. Preprint at bioRxiv https://doi.org/10.1101/2024.04.27.591127 (2024).
Valm, A. M. et al. Systems-level analysis of microbial community organization through combinatorial labeling and spectral imaging. Proc. Natl Acad. Sci. USA 108, 4152–4157 (2011).
Shi, H. et al. Highly multiplexed spatial mapping of microbial communities. Nature 588, 676–681 (2020).
Lyu, L. et al. Simultaneous profiling of host expression and microbial abundance by spatial metatranscriptome sequencing. Genome Res. 33, 401–411 (2023).
Zhu, B. et al. A multi-omics spatial framework for host-microbiome dissection within the intestinal tissue microenvironment. Nat. Commun. 16, 1230 (2025).
Saarenpää, S. et al. Spatial metatranscriptomics resolves host-bacteria-fungi interactomes. Nat. Biotechnol. 42, 1384–1393 (2024).
Sarfatis, A., Wang, Y., Twumasi-Ankrah, N. & Moffitt, J. R. Highly multiplexed spatial transcriptomics in bacteria. Science 387, eadr0932 (2025).
Sounart, H. et al. Dual spatially resolved transcriptomics for human host-pathogen colocalization studies in FFPE tissue sections. Genome Biol. 24, 237 (2023).
Rendeiro, A. F. et al. The spatial landscape of lung pathology during COVID-19 progression. Nature 593, 564–569 (2021).
Lee, J. T. H. et al. Integrated histopathology, spatial and single cell transcriptomics resolve cellular drivers of early and late alveolar damage in COVID-19. Nat. Commun. 16, 1979 (2025).
Zhang, T. et al. Brain-wide alterations revealed by spatial transcriptomics and proteomics in COVID-19 infection. Nat. Aging 4, 1598–1618 (2024).
Pita-Juarez, Y. et al. A single-nucleus and spatial transcriptomic atlas of the COVID-19 liver reveals topological, functional, and regenerative organ disruption in patients. Genome Biol. 26, 56 (2025).
Chen, J., Larsson, L., Swarbrick, A. & Lundeberg, J. Spatial landscapes of cancers: insights and opportunities. Nat. Rev. Clin. Oncol. 21, 660–674 (2024).
Gong, D., Arbesfeld-Qiu, J. M., Perrault, E., Bae, J. W. & Hwang, W. L. Spatial oncology: translating contextual biology to the clinic. Cancer Cell 42, 1653–1675 (2024).
Xu, M., Zhang, T., Xia, R., Wei, Y. & Wei, X. Targeting the tumor stroma for cancer therapy. Mol. Cancer 21, 208 (2022).
Bilotta, M. T., Antignani, A. & Fitzgerald, D. J. Managing the TME to improve the efficacy of cancer therapy. Front. Immunol. 13, 954992 (2022).
Rui, R., Zhou, L. & He, S. Cancer immunotherapies: advances and bottlenecks. Front. Immunol. 14, 1212476 (2023).
Marusyk, A., Janiszewska, M. & Polyak, K. Intratumor heterogeneity: the Rosetta stone of therapy resistance. Cancer Cell 37, 471–484 (2020).
Galon, J. & Bruni, D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 18, 197–218 (2019).
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).
Helmink, B. A. et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 577, 549–555 (2020).
Vanhersecke, L. et al. Mature tertiary lymphoid structures predict immune checkpoint inhibitor efficacy in solid tumors independently of PD-L1 expression. Nat. Cancer 2, 794–802 (2021).
Dong, Y., Wang, T. & Wu, H. Tertiary lymphoid structures in autoimmune diseases. Front. Immunol. 14, 1322035 (2023).
Sato, Y., Silina, K., van den Broek, M., Hirahara, K. & Yanagita, M. The roles of tertiary lymphoid structures in chronic diseases. Nat. Rev. Nephrol. 19, 525–537 (2023).
Castillo, R. L. et al. Spatial transcriptomics stratifies psoriatic disease severity by emergent cellular ecosystems. Sci. Immunol. 8, eabq7991 (2023).
Nayar, S. et al. Molecular and spatial analysis of tertiary lymphoid structures in Sjogren’s syndrome. Nat. Commun. 16, 5 (2025).
Liu, S. et al. Spatial maps of T cell receptors and transcriptomes reveal distinct immune niches and interactions in the adaptive immune response. Immunity 55, 1940–1952.e5 (2022).
Andersson, A. et al. Spatial deconvolution of HER2-positive breast cancer delineates tumor-associated cell type interactions. Nat. Commun. 12, 6012 (2021).
Bandyopadhyay, S. et al. Mapping the cellular biogeography of human bone marrow niches using single-cell transcriptomics and proteomic imaging. Cell 187, 3120–3140.e29 (2024).
Dasdemir, E. et al. Spatial transcriptomics reveals inflammation and trans-differentiation states of acute myeloid leukemia in extramedullary and medullary tissues. Preprint at bioRxiv https://doi.org/10.1101/2024.11.11.622999 (2024).
Vadakekolathu, J. et al. Immune landscapes predict chemotherapy resistance and immunotherapy response in acute myeloid leukemia. Sci. Transl. Med. 12, eaaz0463 (2020).
Rutella, S. et al. Immune dysfunction signatures predict outcomes and define checkpoint blockade-unresponsive microenvironments in acute myeloid leukemia. J. Clin. Invest. 132, e159579 (2022).
Koedijk, J. B. et al. A multidimensional analysis reveals distinct immune phenotypes and the composition of immune aggregates in pediatric acute myeloid leukemia. Leukemia 38, 2332–2343 (2024).
Wu, Y. et al. Multimodal transcriptomics reveal neurogenic aging trajectories and age-related regional inflammation in the dentate gyrus. Nat. Neurosci. 28, 415–430 (2025).
Sun, E. D. et al. Spatial transcriptomic clocks reveal cell proximity effects in brain ageing. Nature 638, 160–171 (2025).
Chen, W.-T. et al. Spatial transcriptomics and in situ sequencing to study Alzheimer’s disease. Cell 182, 976–991.e19 (2020).
Avey, D. R. et al. Uncovering plaque-glia niches in human Alzheimer’s disease brains using spatial transcriptomics. Mol. Neurodegener. Adv. 1, 2 (2025).
Miyoshi, E. et al. Spatial and single-nucleus transcriptomic analysis of genetic and sporadic forms of Alzheimer’s disease. Nat. Genet. 56, 2704–2717 (2024).
Kamath, T. et al. Single-cell genomic profiling of human dopamine neurons identifies a population that selectively degenerates in Parkinson’s disease. Nat. Neurosci. 25, 588–595 (2022).
Ma, M. et al. The spatial landscape of glial pathology and T-cell response in Parkinson’s disease substantia nigra. Nat. Commun. 16, 7146 (2025).
Liu, Z. et al. Spatiotemporal single-cell roadmap of human skin wound healing. Cell Stem Cell 32, 479–498 (2025).
Theocharidis, G. et al. Single cell transcriptomic landscape of diabetic foot ulcers. Nat. Commun. 13, 181 (2022).
Chan, A. S. F. et al. Spatio-temporal dynamics of the fibrotic niche in cardiac repair. Preprint at bioRxiv https://doi.org/10.1101/2024.11.10.622609 (2024).
Yamada, S. et al. Spatiotemporal transcriptome analysis reveals critical roles for mechano-sensing genes at the border zone in remodeling after myocardial infarction. Nat. Cardiovasc. Res. 1, 1072–1083 (2022).
Wünnemann, F. et al. Spatial omics of acute myocardial infarction reveals a novel mode of immune cell infiltration. Preprint at bioRxiv https://doi.org/10.1101/2024.05.20.594955 (2024).
He, J. et al. Single-cell and spatial transcriptomic analyses reveals the dynamic transcript profiles of myocardial lymphangiogenesis post myocardial infarction. eLife 13, RP99192 (2024).
Kuppe, C. et al. Spatial multi-omic map of human myocardial infarction. Nature 608, 766–777 (2022).
Eyres, M. et al. Spatially resolved deconvolution of the fibrotic niche in lung fibrosis. Cell Rep. 40, 111230 (2022).
Vannan, A. et al. Spatial transcriptomics identifies molecular niche dysregulation associated with distal lung remodeling in pulmonary fibrosis. Nat. Genet. 57, 647–658 (2025).
Abedini, A. et al. Single-cell multi-omic and spatial profiling of human kidneys implicates the fibrotic microenvironment in kidney disease progression. Nat. Genet. 56, 1712–1724 (2024).
Lake, B. B. et al. An atlas of healthy and injured cell states and niches in the human kidney. Nature 619, 585–594 (2023).
Fischer, D. S., Villanueva, M. A., Winter, P. S. & Shalek, A. K. Adapting systems biology to address the complexity of human disease in the single-cell era. Nat. Rev. Genet. 26, 514–531 (2025).
Takahama, M. et al. A pairwise cytokine code explains the organism-wide response to sepsis. Nat. Immunol. 25, 226–239 (2024).
Pierre, A., Lancel, S. & Preau, S. Organ crosstalk and dysfunction in sepsis: harnessing emerging biotechnologies for future breakthroughs. Ann. Intensive Care 14, 161 (2024).
Lilja, S. et al. Multi-organ single-cell analysis reveals an on/off switch system with potential for personalized treatment of immunological diseases. Cell Rep. Med. 4, 100956 (2023).
Elmentaite, R., Domínguez Conde, C., Yang, L. & Teichmann, S. A. Single-cell atlases: shared and tissue-specific cell types across human organs. Nat. Rev. Genet. 23, 395–410 (2022).
Eraslan, G. et al. Single-nucleus cross-tissue molecular reference maps toward understanding disease gene function. Science 376, eabl4290 (2022).
Domínguez Conde, C. et al. Cross-tissue immune cell analysis reveals tissue-specific features in humans. Science 376, eabl5197 (2022).
Tsagiopoulou, M., Rashmi, S., Aguilar-Fernandez, S., Nieto, J. & Gut, I. G. Multi-organ single-cell transcriptomics of immune cells uncovered organ-specific gene expression and functions. Sci. Data 11, 316 (2024).
Mulder, K. et al. Cross-tissue single-cell landscape of human monocytes and macrophages in health and disease. Immunity 54, 1883–1900.e5 (2021).
Gao, Y. et al. Cross-tissue human fibroblast atlas reveals myofibroblast subtypes with distinct roles in immune modulation. Cancer Cell 42, 1764–1783.e10 (2024).
Korsunsky, I. et al. Cross-tissue, single-cell stromal atlas identifies shared pathological fibroblast phenotypes in four chronic inflammatory diseases. Med 3, 481–518.e14 (2022).
Wen, J. et al. The genetic architecture of biological age in nine human organ systems. Nat. Aging 4, 1290–1307 (2024).
Gao, Y. et al. Metastasis organotropism: redefining the congenial soil. Dev. Cell 49, 375–391 (2019).
Carrolo, M. et al. Metastatic organotropism: a brief overview. Front. Oncol. 14, 1358786 (2024).
Dunbar, K. J. et al. Regulation of metastatic organotropism. Trends Cancer 11, 216–231 (2025).
Kuett, L. et al. Distant metastases of breast cancer resemble primary tumors in cancer cell composition but differ in immune cell phenotypes. Cancer Res. 85, 15–31 (2025).
Klughammer, J. et al. A multi-modal single-cell and spatial expression map of metastatic breast cancer biopsies across clinicopathological features. Nat. Med. 30, 3236–3249 (2024).
Johnson, B. E. et al. An omic and multidimensional spatial atlas from serial biopsies of an evolving metastatic breast cancer. Cell Rep. Med. 3, 100525 (2022).
Brady, L. et al. Inter- and intra-tumor heterogeneity of metastatic prostate cancer determined by digital spatial gene expression profiling. Nat. Commun. 12, 1426 (2021).
Tagore, S. et al. Single-cell and spatial genomic landscape of non-small cell lung cancer brain metastases. Nat. Med. 31, 1351–1363 (2025).
Zhang, Q. et al. The spatial transcriptomic landscape of non-small cell lung cancer brain metastasis. Nat. Commun. 13, 5983 (2022).
Karimi, E. et al. Single-cell spatial immune landscapes of primary and metastatic brain tumours. Nature 614, 555–563 (2023).
Sundar, R. et al. Spatial profiling of gastric cancer patient-matched primary and locoregional metastases reveals principles of tumour dissemination. Gut 70, 1823–1832 (2021).
Zhao, J. J. et al. Spatially resolved niche and tumor microenvironmental alterations in gastric cancer peritoneal metastases. Gastroenterology 167, 1384–1398.e4 (2024).
Levy, J. J. et al. Identification of spatial proteomic signatures of colon tumor metastasis: a digital spatial profiling approach. Am. J. Pathol. 193, 778–795 (2023).
Sathe, A. et al. Colorectal cancer metastases in the liver establish immunosuppressive spatial networking between tumor-associated SPP1+ macrophages and fibroblasts. Clin. Cancer Res. 29, 244–260 (2023).
Wang, F. et al. Single-cell and spatial transcriptome analysis reveals the cellular heterogeneity of liver metastatic colorectal cancer. Sci. Adv. 9, eadf5464 (2023).
Wu, Y. et al. Spatiotemporal immune landscape of colorectal cancer liver metastasis at single-cell level. Cancer Discov. 12, 134–153 (2022).
Wei, C. et al. Delineating the early dissemination mechanisms of acral melanoma by integrating single-cell and spatial transcriptomic analyses. Nat. Commun. 14, 8119 (2023).
Biermann, J. et al. Dissecting the treatment-naive ecosystem of human melanoma brain metastasis. Cell 185, 2591–2608.e30 (2022).
Boe, R. H., Triandafillou, C. G., Lazcano, R., Wargo, J. A.& Raj, A. Spatial transcriptomics reveals influence of microenvironment on intrinsic fates in melanoma therapy resistance. Preprint at bioRxiv https://doi.org/10.1101/2024.06.30.601416 (2024).
Naulaerts, S. et al. Multiomics and spatial mapping characterizes human CD8+ T cell states in cancer. Sci. Transl. Med. 15, eadd1016 (2023).
Du, Y., Ding, X. & Ye, Y. The spatial multi-omics revolution in cancer therapy: precision redefined. Cell Rep. Med. 5, 101740 (2024).
Di Mauro, F. & Arbore, G. Spatial dissection of the immune landscape of solid tumors to advance precision medicine. Cancer Immunol. Res. 12, 800–813 (2024).
Bollhagen, A. & Bodenmiller, B. Highly multiplexed tissue imaging in precision oncology and translational cancer research. Cancer Discov. 14, 2071–2088 (2024).
Luecken, M. D. et al. Benchmarking atlas-level data integration in single-cell genomics. Nat. Methods 19, 41–50 (2022).
Clifton, K. et al. STalign: alignment of spatial transcriptomics data using diffeomorphic metric mapping. Nat. Commun. 14, 8123 (2023).
[No authors listed] STAligner enables the integration and alignment of multiple spatial transcriptomics datasets. Nat. Comput. Sci. 3, 831–832 (2023).
Mitchel, J., Gao, T., Cole, E., Petukhov, V. & Kharchenko, P. V. Impact of segmentation errors in analysis of spatial transcriptomics data. Preprint at bioRxiv https://doi.org/10.1101/2025.01.02.631135 (2025).
Park, J. et al. Cell segmentation-free inference of cell types from in situ transcriptomics data. Nat. Commun. 12, 3545 (2021).
Si, Y. et al. FICTURE: scalable segmentation-free analysis of submicron-resolution spatial transcriptomics. Nat. Methods 21, 1843–1854 (2024).
Alam, S. et al. Popari: modeling multisample variation in spatial transcriptomics. Preprint at bioRxiv https://doi.org/10.1101/2025.05.08.652741 (2025).
Holdener, C. & De Vlaminck, I. Smoothie: efficient inference of spatial co-expression networks from denoised spatial transcriptomics data. Preprint at bioRxiv https://doi.org/10.1101/2025.02.26.640406 (2025).
Rood, J. E. et al. Toward a common coordinate framework for the human body. Cell 179, 1455–1467 (2019).
Andersson, A. et al. A landmark-based common coordinate framework for spatial transcriptomics data. Preprint at bioRxiv https://doi.org/10.1101/2021.11.11.468178 (2021).
Börner, K. et al. Human biomolecular atlas program (HuBMAP): 3D human reference atlas construction and usage. Nat. Methods 22, 845–860 (2025).
Börner, K. et al. Anatomical structures, cell types and biomarkers of the human reference atlas. Nat. Cell Biol. 23, 1117–1128 (2021).
Mungall, C. J., Torniai, C., Gkoutos, G. V., Lewis, S. E. & Haendel, M. A. Uberon, an integrative multi-species anatomy ontology. Genome Biol. 13, R5 (2012).
Ekvall, M. et al. Spatial landmark detection and tissue registration with deep learning. Nat. Methods 21, 673–679 (2024).
Lu, M. Y. et al. A visual-language foundation model for computational pathology. Nat. Med. 30, 863–874 (2024).
Wang, X. et al. A pathology foundation model for cancer diagnosis and prognosis prediction. Nature 634, 970–978 (2024).
Chelebian, E., Avenel, C. & Wählby, C. Combining spatial transcriptomics with tissue morphology. Nat. Commun. 16, 4452 (2025).
Jaume, G. et al. HEST-1k: a dataset for spatial transcriptomics and histology image analysis. In 38th Conference on Neural Information Processing Systems (NeurIPS, 2024).
Chelebian, E. et al. Discovery of tumour indicating morphological changes in benign prostate biopsies through AI. Sci. Rep. 15, 30770 (2025).
He, B., Bergenstråhle, L. & Lundeberg, J. Integrating spatial gene expression and breast tumour morphology via deep learning. Nat. Biomed. Eng. 4, 827–834 (2020).
Bergenstråhle, L. et al. Super-resolved spatial transcriptomics by deep data fusion. Nat. Biotechnol. 40, 476–479 (2022).
Zhang, D. et al. Inferring super-resolution tissue architecture by integrating spatial transcriptomics with histology. Nat. Biotechnol. 42, 1372–1377 (2024).
Wang, C. et al. Benchmarking the translational potential of spatial gene expression prediction from histology. Nat. Commun. 16, 1544 (2025).
Hu, J. et al. SpaGCN: integrating gene expression, spatial location and histology to identify spatial domains and spatially variable genes by graph convolutional network. Nat. Methods 18, 1342–1351 (2021).
Monjo, T., Koido, M., Nagasawa, S., Suzuki, Y. & Kamatani, Y. Efficient prediction of a spatial transcriptomics profile better characterizes breast cancer tissue sections without costly experimentation. Sci. Rep. 12, 4133 (2022).
Cisternino, F. et al. Self-supervised learning for characterising histomorphological diversity and spatial RNA expression prediction across 23 human tissue types. Nat. Commun. 15, 5906 (2024).
He, S. et al. Starfysh integrates spatial transcriptomic and histologic data to reveal heterogeneous tumor-immune hubs. Nat. Biotechnol. 43, 223–235 (2025).
Chen, W. et al. A visual-omics foundation model to bridge histopathology with spatial transcriptomics. Nat. Methods 22, 1568–1582 (2025).
Hoang, D.-T. et al. A deep-learning framework to predict cancer treatment response from histopathology images through imputed transcriptomics. Nat. Cancer 5, 1305–1317 (2024).
Lu, M. Y. et al. A multimodal generative AI copilot for human pathology. Nature 634, 466–473 (2024).
Wang, H. et al. SpatialAgent: an autonomous AI agent for spatial biology. Preprint at bioRxiv https://doi.org/10.1101/2025.04.03.646459 (2025).
Vorontsov, E. et al. A foundation model for clinical-grade computational pathology and rare cancers detection. Nat. Med. 30, 2924–2935 (2024).
Biancalani, T. et al. Deep learning and alignment of spatially resolved single-cell transcriptomes with tangram. Nat. Methods 18, 1352–1362 (2021).
Wenckstern, J. et al. AI-powered virtual tissues from spatial proteomics for clinical diagnostics and biomedical discovery. Preprint at https://doi.org/10.48550/arXiv.2501.06039 (2025).
Bunne, C. et al. How to build the virtual cell with artificial intelligence: priorities and opportunities. Preprint at https://doi.org/10.48550/arXiv.2409.11654 (2024).
Zhang, J. et al. Tahoe-100M: a giga-scale single-cell perturbation atlas for context-dependent gene function and cellular modeling. Preprint at bioRxiv https://doi.org/10.1101/2025.02.20.639398 (2025).
Dhainaut, M. et al. Spatial CRISPR genomics identifies regulators of the tumor microenvironment. Cell 185, 1223–1239.e20 (2022).
Teo, A. Y. Y. et al. Identification of perturbation-responsive regions and genes in comparative spatial transcriptomics atlases. Preprint at bioRxiv https://doi.org/10.1101/2024.06.13.598641 (2024).
Megas, S. et al. Celcomen: spatial causal disentanglement for single-cell and tissue perturbation modeling. Preprint at https://doi.org/10.48550/arXiv.2409.05804 (2024).
Frey, N.C. et al. Lab-in-the-loop therapeutic antibody design with deep learning. Preprint at bioRxiv https://doi.org/10.1101/2025.02.19.639050 (2025).
Laubenbacher, R., Mehrad, B., Shmulevich, I. & Trayanova, N. Digital twins in medicine. Nat. Comput. Sci. 4, 184–191 (2024).
Bhatia, H. S. et al. Spatial proteomics in three-dimensional intact specimens. Cell 185, 5040–5058.e19 (2022).
Kanatani, S. et al. Whole-brain spatial transcriptional analysis at cellular resolution. Science 386, 907–915 (2024).
Gandin, V. et al. Deep-tissue transcriptomics and subcellular imaging at high spatial resolution. Science 388, eadq2084 (2025).
Fang, R. et al. Three-dimensional single-cell transcriptome imaging of thick tissues. eLife 12, RP90029 (2024).
Ertürk, A. Deep 3D histology powered by tissue clearing, omics and AI. Nat. Methods 21, 1153–1165 (2024).
Glaser, A. K. et al. Light-sheet microscopy for slide-free non-destructive pathology of large clinical specimens. Nat. Biomed. Eng. 1, 0084 (2017).
Wang, X. et al. Three-dimensional intact-tissue sequencing of single-cell transcriptional states. Science 361, eaat5691 (2018).
Zeira, R., Land, M., Strzalkowski, A. & Raphael, B. J. Alignment and integration of spatial transcriptomics data. Nat. Methods 19, 567–575 (2022).
Qiu, X. et al. Spatiotemporal modeling of molecular holograms. Cell 187, 7351–7373.e61 (2024).
Almagro-Pérez, C. et al. AI-driven 3D spatial transcriptomics. Preprint at https://doi.org/10.48550/arXiv.2502.17761 (2025).
Vandereyken, K., Sifrim, A., Thienpont, B. & Voet, T. Methods and applications for single-cell and spatial multi-omics. Nat. Rev. Genet. 24, 494–515 (2023).
Liu, X. et al. Spatial multi-omics: deciphering technological landscape of integration of multi-omics and its applications. J. Hematol. Oncol. 17, 72 (2024).
Kiessling, P. & Kuppe, C. Spatial multi-omics: novel tools to study the complexity of cardiovascular diseases. Genome Med. 16, 14 (2024).
Eisenstein, M. Seven technologies to watch in 2022. Nature 601, 658–661 (2022).
Bao, F. et al. Integrative spatial analysis of cell morphologies and transcriptional states with MUSE. Nat. Biotechnol. 40, 1200–1209 (2022).
Long, Y. et al. Deciphering spatial domains from spatial multi-omics with SpatialGlue. Nat. Methods 21, 1658–1667 (2024).
Coleman, K. et al. Resolving tissue complexity by multimodal spatial omics modeling with MISO. Nat. Methods 22, 530–538 (2025).
Wang, S., Lin, S. & Yang, C. The dawn of spatiotemporal transcriptomics. Biomed. Anal. 1, 154–161 (2024).
Velten, B. & Stegle, O. Principles and challenges of modeling temporal and spatial omics data. Nat. Methods 20, 1462–1474 (2023).
Ren, J. et al. Spatiotemporally resolved transcriptomics reveals the subcellular RNA kinetic landscape. Nat. Methods 20, 695–705 (2023).
Holler, K. et al. Spatio-temporal mRNA tracking in the early zebrafish embryo. Nat. Commun. 12, 3358 (2021).
Rietjens, R. G. J. & Heijs, B. In situ isotope tracing at single-cell resolution using mass spectrometry imaging. Methods Mol. Biol. 2855, 523–535 (2025).
Wang, L. et al. Spatially resolved isotope tracing reveals tissue metabolic activity. Nat. Methods 19, 223–230 (2022).
Buglakova, E. et al. Spatial single-cell isotope tracing reveals heterogeneity of de novo fatty acid synthesis in cancer. Nat. Metab. 6, 1695–1711 (2024).
Ratz, M. et al. Clonal relations in the mouse brain revealed by single-cell and spatial transcriptomics. Nat. Neurosci. 25, 285–294 (2022).
Chow, K.-H. K. et al. Imaging cell lineage with a synthetic digital recording system. Science 372, eabb3099 (2021).
He, Z. et al. Lineage recording in human cerebral organoids. Nat. Methods 19, 90–99 (2022).
Tong, L. et al. Patient-derived organoids in precision cancer medicine. Med 5, 1351–1377 (2024).
Wahle, P. et al. Multimodal spatiotemporal phenotyping of human retinal organoid development. Nat. Biotechnol. 41, 1765–1775 (2023).
Chiaradia, I. et al. Tissue morphology influences the temporal program of human brain organoid development. Cell Stem Cell 30, 1351–1367.e10 (2023).
Legnini, I. et al. Spatiotemporal, optogenetic control of gene expression in organoids. Nat. Methods 20, 1544–1552 (2023).
You, Y. et al. Systematic comparison of sequencing-based spatial transcriptomic methods. Nat. Methods 21, 1743–1754 (2024).
Ren, P. et al. Systematic benchmarking of high-throughput subcellular spatial transcriptomics platforms. Preprint at bioRxiv https://doi.org/10.1101/2024.12.23.630033 (2024).
Wang, H. et al. Systematic benchmarking of imaging spatial transcriptomics platforms in FFPE tissues. Preprint at bioRxiv https://doi.org/10.1101/2023.12.07.570603 (2023).
Kim, Y. et al. Seq-Scope protocol: repurposing illumina sequencing flow cells for high-resolution spatial transcriptomics. Preprint at bioRxiv https://doi.org/10.1101/2024.03.29.587285 (2024).
Poovathingal, S. et al. Nova-ST: nano-patterned ultra-dense platform for spatial transcriptomics. Cell Rep. Methods 4, 100831 (2024).
Schott, M. et al. Open-ST: high-resolution spatial transcriptomics in 3D. Cell 187, 3953–3972.e26 (2024).
Liao, R. et al. Sequencing-based spatial transcriptomics with scRNA-seq sensitivity. Preprint at bioRxiv https://doi.org/10.1101/2025.01.15.633111 (2025).
Liu, Y. et al. High-spatial-resolution multi-omics sequencing via deterministic barcoding in tissue. Cell 183, 1665–1681.e18 (2020).
Lebrigand, K. et al. The spatial landscape of gene expression isoforms in tissue sections. Nucleic Acids Res. 51, e47 (2023).
Zou, L. S. et al. Detection of allele-specific expression in spatial transcriptomics with spASE. Genome Biol. 25, 180 (2024).
Ma, C. et al. Inferring allele-specific copy number aberrations and tumor phylogeography from spatially resolved transcriptomics. Nat. Methods 21, 2239–2247 (2024).
Deng, Y. et al. Spatial-CUT&Tag: spatially resolved chromatin modification profiling at the cellular level. Science 375, 681–686 (2022).
Deng, Y. et al. Spatial profiling of chromatin accessibility in mouse and human tissues. Nature 609, 375–383 (2022).
Llorens-Bobadilla, E. et al. Solid-phase capture and profiling of open chromatin by spatial ATAC. Nat. Biotechnol. 41, 1085–1088 (2023).
Guo, P. et al. Multiplexed spatial mapping of chromatin features, transcriptome and proteins in tissues. Nat. Methods 22, 520–529 (2025).
Zhao, T. et al. Spatial genomics enables multi-modal study of clonal heterogeneity in tissues. Nature 601, 85–91 (2022).
Seferbekova, Z., Lomakin, A., Yates, L. R. & Gerstung, M. Spatial biology of cancer evolution. Nat. Rev. Genet. 24, 295–313 (2023).
[No authors listed] Method of the year 2024: spatial proteomics. Nat. Methods 21, 2195–2196 (2024).
Mund, A., Brunner, A.-D. & Mann, M. Unbiased spatial proteomics with single-cell resolution in tissues. Mol. Cell 82, 2335–2349 (2022).
de Souza, N., Zhao, S. & Bodenmiller, B. Multiplex protein imaging in tumour biology. Nat. Rev. Cancer 24, 171–191 (2024).
Jhaveri, N. et al. Mapping the spatial proteome of head and neck tumors: key immune mediators and metabolic determinants in the tumor microenvironment. GEN Biotechnol. 2, 418–434 (2023).
Yagnik, G., Liu, Z., Rothschild, K. J. & Lim, M. J. Highly multiplexed immunohistochemical MALDI-MS imaging of biomarkers in tissues. J. Am. Soc. Mass. Spectrom. 32, 977–988 (2021).
Claes, B. S. R. et al. MALDI-IHC-guided in-depth spatial proteomics: targeted and untargeted MSI combined. Anal. Chem. 95, 2329–2338 (2023).
Gut, G., Herrmann, M. D. & Pelkmans, L. Multiplexed protein maps link subcellular organization to cellular states. Science 361, eaar7042 (2018).
Quardokus, E. M. et al. Organ mapping antibody panels: a community resource for standardized multiplexed tissue imaging. Nat. Methods 20, 1174–1178 (2023).
Mund, A. et al. Deep visual proteomics defines single-cell identity and heterogeneity. Nat. Biotechnol. 40, 1231–1240 (2022).
Zhang, H. et al. Mass spectrometry imaging for spatially resolved multi-omics molecular mapping. npj Imaging 2, 20 (2024).
Alexandrov, T. Spatial metabolomics: from a niche field towards a driver of innovation. Nat. Metab. 5, 1443–1445 (2023).
Saharuka, V. et al. Large-scale evaluation of spatial metabolomics protocols and technologies. Preprint at bioRxiv https://doi.org/10.1101/2024.01.29.577354 (2024).
Singhal, V. et al. BANKSY unifies cell typing and tissue domain segmentation for scalable spatial omics data analysis. Nat. Genet. 56, 431–441 (2024).
Li, H. et al. A comprehensive benchmarking with practical guidelines for cellular deconvolution of spatial transcriptomics. Nat. Commun. 14, 1548 (2023).
Li, B. et al. Benchmarking spatial and single-cell transcriptomics integration methods for transcript distribution prediction and cell type deconvolution. Nat. Methods 19, 662–670 (2022).
Armingol, E., Baghdassarian, H. M. & Lewis, N. E. The diversification of methods for studying cell-cell interactions and communication. Nat. Rev. Genet. 25, 381–400 (2024).
Plummer, J. T., Vlachos, I. S. & Martelotto, L. G. Introducing the global alliance for spatial technologies (GESTALT). Nat. Genet. 57, 275–279 (2025).
Lim, J. et al. Transitioning single-cell genomics into the clinic. Nat. Rev. Genet. 24, 573–584 (2023).