Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Perspective
  • Published:

Spatial mapping of protein composition and tissue organization: a primer for multiplexed antibody-based imaging

Abstract

Tissues and organs are composed of distinct cell types that must operate in concert to perform physiological functions. Efforts to create high-dimensional biomarker catalogs of these cells have been largely based on single-cell sequencing approaches, which lack the spatial context required to understand critical cellular communication and correlated structural organization. To probe in situ biology with sufficient depth, several multiplexed protein imaging methods have been recently developed. Though these technologies differ in strategy and mode of immunolabeling and detection tags, they commonly utilize antibodies directed against protein biomarkers to provide detailed spatial and functional maps of complex tissues. As these promising antibody-based multiplexing approaches become more widely adopted, new frameworks and considerations are critical for training future users, generating molecular tools, validating antibody panels, and harmonizing datasets. In this Perspective, we provide essential resources, key considerations for obtaining robust and reproducible imaging data, and specialized knowledge from domain experts and technology developers.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Obtaining high-content imaging data using a wide range of multiplexed antibody-based imaging platforms.
Fig. 2: Considerations for the choice and implementation of multiplexed antibody-based imaging technologies into existing workflows.
Fig. 3: Phases of panel development and validation for multiplexed antibody-based imaging assays.
Fig. 4: Process of conjugating antibodies with modifiers for multiplexing.

Similar content being viewed by others

References

  1. Stack, E. C., Wang, C., Roman, K. A. & Hoyt, C. C. Multiplexed immunohistochemistry, imaging, and quantitation: A review, with an assessment of Tyramide signal amplification, multispectral imaging and multiplex analysis. Methods 70, 46–58 (2014).

    Article  CAS  PubMed  Google Scholar 

  2. Thul, P. J. et al. A subcellular map of the human proteome. Science 356, eaal3321 (2017).

    Article  PubMed  Google Scholar 

  3. Gawad, C., Koh, W. & Quake, S. R. Single-cell genome sequencing: current state of the science. Nat. Rev. Genet. 17, 175–188 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Massonnet, P. & Heeren, R. M. A. A concise tutorial review of TOF-SIMS based molecular and cellular imaging. J. Anal. Spectrom. 34, 2217–2228 (2019).

    Article  CAS  Google Scholar 

  5. Neumann, E. K., Do, T. D., Comi, T. J. & Sweedler, J. V. Exploring the fundamental structures of life: non-targeted, chemical analysis of single cells and subcellular structures. Angew. Chem. Int. Ed. 58, 9348–9364 (2019).

    Article  CAS  Google Scholar 

  6. Zhang, L. & Vertes, A. Single-cell mass spectrometry approaches to explore cellular heterogeneity. Angew. Chem. Int. Ed. 57, 4466–4477 (2018).

    Article  CAS  Google Scholar 

  7. Porwit, A. & Béné, M. C. Multiparameter flow cytometry applications in the diagnosis of mixed phenotype acute leukemia. Cytom. Part B: Clin. Cytom. 96, 183–194 (2019).

    Article  Google Scholar 

  8. Bandura, D. R. et al. Mass cytometry: technique for real time single cell multitarget immunoassay based on inductively coupled plasma time-of-flight mass spectrometry. Anal. Chem. 81, 6813–6822 (2009).

    Article  CAS  PubMed  Google Scholar 

  9. Kolodziejczyk, A. A., Kim, J. K., Svensson, V., Marioni, J. C. & Teichmann, S. A. The technology and biology of single-cell RNA sequencing. Mol. cell 58, 610–620 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. Stoeckius, M. et al. Simultaneous epitope and transcriptome measurement in single cells. Nat. Methods 14, 865–868 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Taube, J. M. et al. The Society for Immunotherapy of Cancer statement on best practices for multiplex immunohistochemistry (IHC) and immunofluorescence (IF) staining and validation. J. Immunother. Cancer 8, e000155 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Goltsev, Y. et al. Deep profiling of mouse splenic architecture with CODEX multiplexed imaging. Cell 174, 968–981.e915 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. van den Brink, S. C. et al. Single-cell sequencing reveals dissociation-induced gene expression in tissue subpopulations. Nat. Methods 14, 935–936 (2017).

    Article  PubMed  Google Scholar 

  14. Gerner, M. Y., Kastenmuller, W., Ifrim, I., Kabat, J. & Germain, R. N. Histo-cytometry: a method for highly multiplex quantitative tissue imaging analysis applied to dendritic cell subset microanatomy in lymph nodes. Immunity 37, 364–376 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Asp, M., Bergenstråhle, J. & Lundeberg, J. Spatially resolved transcriptomes—next generation tools for tissue exploration. BioEssays 42, 1900221 (2020).

    Article  Google Scholar 

  16. Marx, V. Method of the year: spatially resolved transcriptomics. Nat. Methods 18, 9–14 (2021).

    Article  CAS  PubMed  Google Scholar 

  17. Uhlén, M. et al. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    Article  PubMed  Google Scholar 

  18. Uhlen, M. et al. A pathology atlas of the human cancer transcriptome. Science 357, eaan2507 (2017).

    Article  PubMed  Google Scholar 

  19. Tan, W. C. C. et al. Overview of multiplex immunohistochemistry/immunofluorescence techniques in the era of cancer immunotherapy. Cancer Commun. 40, 135–153 (2020).

    Article  Google Scholar 

  20. Bodenmiller, B. Multiplexed epitope-based tissue imaging for discovery and healthcare applications. Cell Syst. 2, 225–238 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Radtke, A. J. et al. IBEX: a versatile multiplex optical imaging approach for deep phenotyping and spatial analysis of cells in complex tissues. Proc. Natl Acad. Sci. 117, 33455–33465 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Gut, G., Herrmann, M. D. & Pelkmans, L. Multiplexed protein maps link subcellular organization to cellular states. Science 361, eaar7042 (2018).

    Article  PubMed  Google Scholar 

  23. Lin, J.-R., Fallahi-Sichani, M. & Sorger, P. K. Highly multiplexed imaging of single cells using a high-throughput cyclic immunofluorescence method. Nat. Commun. 6, 8390 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Lin, J.-R. et al. Highly multiplexed immunofluorescence imaging of human tissues and tumors using t-CyCIF and conventional optical microscopes. eLife 7, e31657 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Gerdes, M. J. et al. Highly multiplexed single-cell analysis of formalin-fixed, paraffin-embedded cancer tissue. Proc. Natl Acad. Sci. USA 110, 11982–11987 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Schubert, W. et al. Analyzing proteome topology and function by automated multidimensional fluorescence microscopy. Nat. Biotechnol. 24, 1270–1278 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Wählby, C., Erlandsson, F., Bengtsson, E. & Zetterberg, A. Sequential immunofluorescence staining and image analysis for detection of large numbers of antigens in individual cell nuclei. Cytometry 47, 32–41 (2002).

    Article  PubMed  Google Scholar 

  28. Zrazhevskiy, P. & Gao, X. Quantum dot imaging platform for single-cell molecular profiling. Nat. Commun. 4, 1–12 (2013).

    Article  Google Scholar 

  29. Du, Z. et al. Qualifying antibodies for image-based immune profiling and multiplexed tissue imaging. Nat. Protoc. 14, 2900–2930 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Wang, Y. et al. Rapid sequential in situ multiplexing with DNA exchange imaging in neuronal cells and tissues. Nano Lett. 17, 6131–6139 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Saka, S. K. et al. Immuno-SABER enables highly multiplexed and amplified protein imaging in tissues. Nat. Biotechnol. 37, 1080–1090 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Schürch, C. M. et al. Coordinated cellular neighborhoods orchestrate antitumoral immunity at the colorectal cancer invasive front. Cell 182, 1341–1359.e1319 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Neumann, E. K. et al. Highly multiplexed immunofluorescence of the human kidney using co-detection by indexing. Kidney Int. https://doi.org/10.1016/j.kint.2021.08.033 (2021).

  34. Lin, R. et al. A hybridization-chain-reaction-based method for amplifying immunosignals. Nat. Methods 15, 275–278 (2018).

    Article  PubMed  Google Scholar 

  35. Wang, Y. et al. Multiplexed in situ protein imaging using DNA-barcoded antibodies with extended hybridization chain reactions. Preprint at bioRxiv https://doi.org/10.1101/274456 (2020).

  36. Tsujikawa, T. et al. Quantitative multiplex immunohistochemistry reveals myeloid-inflamed tumor-immune complexity associated with poor prognosis. Cell Rep. 19, 203–217 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Angelo, M. et al. Multiplexed ion beam imaging of human breast tumors. Nat. Med. 20, 436–442 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Giesen, C. et al. Highly multiplexed imaging of tumor tissues with subcellular resolution by mass cytometry. Nat. Methods 11, 417–422 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Chang, Q. et al. Imaging mass cytometry. Cytom. Part A 91, 160–169 (2017).

    Article  Google Scholar 

  40. Wei, L. et al. Super-multiplex vibrational imaging. Nature 544, 465–470 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Wong, H. S. et al. A local regulatory T cell feedback circuit maintains immune homeostasis by pruning self-activated T cells. Cell, 3981–3997 (2021).

  42. Chung, K. et al. Structural and molecular interrogation of intact biological systems. Nature 497, 332–337 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Li, W., Germain, R. N. & Gerner, M. Y. Multiplex, quantitative cellular analysis in large tissue volumes with clearing-enhanced 3D microscopy (Ce3D). Proc. Natl Acad. Sci. USA 114, E7321–E7330 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Murray, E. et al. Simple, scalable proteomic imaging for high-dimensional profiling of intact systems. Cell 163, 1500–1514 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ku, T. et al. Multiplexed and scalable super-resolution imaging of three-dimensional protein localization in size-adjustable tissues. Nat. Biotechnol. 34, 973–981 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Park, Y.-G. et al. Protection of tissue physicochemical properties using polyfunctional crosslinkers. Nat. Biotechnol. 37, 73–83 (2019).

    Article  CAS  Google Scholar 

  47. Tillberg, P. W. et al. Protein-retention expansion microscopy of cells and tissues labeled using standard fluorescent proteins and antibodies. Nat. Biotechnol. 34, 987–992 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Zhang, J. et al. Evaluation of the tumor-targeting efficiency and intratumor heterogeneity of anticancer drugs using quantitative mass spectrometry imaging. Theranostics 10, 2621–2630 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Yun, D. H. et al. Ultrafast immunostaining of organ-scale tissues for scalable proteomic phenotyping. Preprint at bioRxiv https://doi.org/10.1101/660373 (2019).

  50. Kim, S.-Y. et al. Stochastic electrotransport selectively enhances the transport of highly electromobile molecules. Proc. Natl Acad. Sci. USA 112, E6274–E6283 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Davis, A. S. et al. Characterizing and diminishing autofluorescence in formalin-fixed paraffin-embedded human respiratory tissue. J. Histochemistry Cytochemistry 62, 405–423 (2014).

    Article  CAS  Google Scholar 

  52. Merritt, C. R. et al. Multiplex digital spatial profiling of proteins and RNA in fixed tissue. Nat. Biotechnol. 38, 586–599 (2020).

    Article  CAS  PubMed  Google Scholar 

  53. Shi, S.-R., Shi, Y. & Taylor, C. R. Antigen retrieval immunohistochemistry: review and future prospects in research and diagnosis over two decades. J. Histochem. Cytochem. 59, 13–32 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Shi, S.-R. et al. Evaluation of the value of frozen tissue section used as ‘gold standard’ for immunohistochemistry. Am. J. Clin. Pathol. 129, 358–366 (2008).

    Article  CAS  PubMed  Google Scholar 

  55. Neumann, E. K., Comi, T. J., Rubakhin, S. S. & Sweedler, J. V. Lipid heterogeneity between astrocytes and neurons revealed by single-cell MALDI-MS combined with immunocytochemical classification. Angew. Chem. Int. Ed. 58, 5910–5914 (2019).

    Article  CAS  Google Scholar 

  56. Radtke, A. J. et al. IBEX: an iterative immunolabeling and chemical bleaching method for high-content imaging of diverse tissues. Nat. Protoc. https://doi.org/10.1038/s41596-021-00644-9 (2021).

  57. Muzzey, D. & Oudenaarden, A. V. Quantitative time-lapse fluorescence microscopy in single cells. Annu. Rev. Cell Dev. Biol. 25, 301–327 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Santi, P. A. Light sheet fluorescence microscopy: a review. J. Histochem. Cytochem. 59, 129–138 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Leung, B. O. & Chou, K. C. Review of super-resolution fluorescence microscopy for biology. Appl. Spectrosc. 65, 967–980 (2011).

    Article  CAS  PubMed  Google Scholar 

  60. Shakya, R., Nguyen, T. H., Waterhouse, N. & Khanna, R. Immune contexture analysis in immuno-oncology: applications and challenges of multiplex fluorescent immunohistochemistry. Clin. Transl. Immunol. 9, e1183 (2020).

    Article  Google Scholar 

  61. Samusik, N., Good, Z., Spitzer, M. H., Davis, K. L. & Nolan, G. P. Automated mapping of phenotype space with single-cell data. Nat. Methods 13, 493–496 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Brummelman, J. et al. Development, application and computational analysis of high-dimensional fluorescent antibody panels for single-cell flow cytometry. Nat. Protoc. 14, 1946–1969 (2019).

    Article  CAS  PubMed  Google Scholar 

  63. Szabó, Á. et al. The effect of fluorophore conjugation on antibody affinity and the photophysical properties of dyes. Biophys. J. 114, 688–700 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Adusumalli, S. R. et al. Chemoselective and site‐selective lysine‐directed lysine modification enables single‐site labeling of native proteins. Angew. Chem. Int. Ed. 59, 10332–10336 (2020).

    Article  CAS  Google Scholar 

  65. Matos, M. J. et al. Chemo-and regioselective lysine modification on native proteins. JACS 140, 4004–4017 (2018).

    Article  CAS  Google Scholar 

  66. Cremers, G. A. O., Rosier, B. J. H. M., Riera Brillas, R., Albertazzi, L. & de Greef, T. F. A. Efficient small-scale conjugation of DNA to primary antibodies for multiplexed cellular targeting. Bioconjug Chem. 30, 2384–2392 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Sograte-Idrissi, S. et al. Circumvention of common labelling artefacts using secondary nanobodies. Nanoscale 12, 10226–10239 (2020).

    Article  CAS  PubMed  Google Scholar 

  68. Rajagopalan, A. et al. SeqStain is an efficient method for multiplexed, spatialomic profiling of human and murine tissues. Cell Rep. Methods 1, 100006 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Brun, M.-P. & Gauzy-Lazo, L. in Antibody–Drug Conjugates 173–187 (Springer, 2013).

  70. Datta-Mannan, A. et al. The properties of cysteine-conjugated antibody–drug conjugates are impacted by the IgG subclass. AAPS J. 20, 103 (2018).

    Article  PubMed  Google Scholar 

  71. Uhlen, M. et al. A proposal for validation of antibodies. Nat. Methods 13, 823–827 (2016).

    Article  CAS  PubMed  Google Scholar 

  72. Bordeaux, J. et al. Antibody validation. BioTechniques 48, 197–209 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Pagès, F. et al. International validation of the consensus Immunoscore for the classification of colon cancer: a prognostic and accuracy study. Lancet 391, 2128–2139 (2018).

    Article  PubMed  Google Scholar 

  74. Stadler, C. et al. Systematic validation of antibody binding and protein subcellular localization using siRNA and confocal microscopy. J. Proteom. 75, 2236–2251 (2012).

    Article  CAS  Google Scholar 

  75. Giuliano, C. J., Lin, A., Girish, V. & Sheltzer, J. M. Generating single cell-derived knockout clones in mammalian cells with CRISPR/Cas9. Curr. Protoc. Mol. Biol. 128, e100 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Hewitt, S. M., Baskin, D. G., Frevert, C. W., Stahl, W. L. & Rosa-Molinar, E. Controls for immunohistochemistry: The Histochemical Society’s standards of practice for validation of immunohistochemical assays. J. Histochem. Cytochem. 62, 693–697 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Gustavson, M. D., Rimm, D. L. & Dolled-Filhart, M. Tissue microarrays: leaping the gap between research and clinical adoption. Personalized Med. 10, 441–451 (2013).

    Article  CAS  Google Scholar 

  78. Camp, R. L., Chung, G. G. & Rimm, D. L. Automated subcellular localization and quantification of protein expression in tissue microarrays. Nat. Med. 8, 1323–1328 (2002).

    Article  CAS  PubMed  Google Scholar 

  79. Martinez-Morilla, S. et al. Biomarker discovery in patients with immunotherapy-treated melanoma with imaging mass cytometry. Clin. Cancer Res. 27, 1987–1996 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Baker, M. 1,500 scientists lift the lid on reproducibility. Nature 533, 452 (2016).

    Article  CAS  PubMed  Google Scholar 

  81. Jarvis, M. F. & Williams, M. Irreproducibility in preclinical biomedical research: perceptions, uncertainties, and knowledge gaps. Trends Pharmacol. Sci. 37, 290–302 (2016).

    Article  CAS  PubMed  Google Scholar 

  82. Snyder, M. P. et al. The human body at cellular resolution: the NIH Human Biomolecular Atlas Program. Nature 574, 187–192 (2019).

    Article  Google Scholar 

  83. Rozenblatt-Rosen, O. et al. The human tumor atlas network: charting tumor transitions across space and time at single-cell resolution. Cell 181, 236–249 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Clark, K. et al. The Cancer Imaging Archive (TCIA): maintaining and operating a public information repository. J. Digit. Imaging 26, 1045–1057 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  85. Williams, E. et al. Image Data Resource: a bioimage data integration and publication platform. Nat. Methods 14, 775–781 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Schneider, C. A., Rasband, W. S. & Eliceiri, K. W. NIH Image to ImageJ: 25 years of image analysis. Nat. Methods 9, 671–675 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Rashid, R. et al. Highly multiplexed immunofluorescence images and single-cell data of immune markers in tonsil and lung cancer. Sci. Data 6, 323 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  88. Czech, E., Aksoy, B. A., Aksoy, P. & Hammerbacher, J. Cytokit: a single-cell analysis toolkit for high dimensional fluorescent microscopy imaging. BMC Bioinf. 20, 448 (2019).

    Article  Google Scholar 

  89. McQuin, C. et al. CellProfiler 3.0: next-generation image processing for biology. PLoS Biol. 16, e2005970 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  90. Stringer, C., Wang, T., Michaelos, M. & Pachitariu, M. Cellpose: a generalist algorithm for cellular segmentation. Nat. Methods 18, 100–106 (2021).

    Article  CAS  PubMed  Google Scholar 

  91. Bankhead, P. et al. QuPath: open source software for digital pathology image analysis. Sci. Rep. 7, 16878 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  92. Bannon, D. et al. DeepCell Kiosk: scaling deep learning-enabled cellular image analysis with Kubernetes. Nat. Methods 18, 43–45 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Sommer, C., Straehle, C., Koethe, U. & Hamprecht, F. A. in 2011 IEEE International Symposium on Biomedical Imaging: From Nano to Macro. 230–233 (IEEE, 2011).

  94. Schapiro, D. et al. histoCAT: analysis of cell phenotypes and interactions in multiplex image cytometry data. Nat. Methods 14, 873–876 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Schapiro, D. et al. MCMICRO: A scalable, modular image-processing pipeline for multiplexed tissue imaging. Preprint at bioRxiv https://doi.org/10.1101/2021.03.15.435473 (2021).

  96. Palla, G. et al. Squidpy: a scalable framework for spatial single cell analysis. Preprint at bioRxiv https://doi.org/10.1101/2021.02.19.431994 (2021).

  97. Stoltzfus, C. R. et al. CytoMAP: a spatial analysis toolbox reveals features of myeloid cell organization in lymphoid tissues. Cell Rep. 31, 107523 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Paintdakhi, A. et al. Oufti: an integrated software package for high-accuracy, high-throughput quantitative microscopy analysis. Mol. Microbiol. 99, 767–777 (2016).

    Article  CAS  PubMed  Google Scholar 

  99. Sage, D. et al. DeconvolutionLab2: an open-source software for deconvolution microscopy. Methods 115, 28–41 (2017).

    Article  CAS  PubMed  Google Scholar 

  100. Kulikov, V. et al. DoGNet: a deep architecture for synapse detection in multiplexed fluorescence images. PLoS Comput. Biol. 15, e1007012 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Risom, T. et al. Transition to invasive breast cancer is associated with progressive changes in the structure and composition of tumor stroma. Preprint at bioRxiv https://doi.org/10.1101/2021.01.05.425362 (2021).

  102. Kramer, B. A. & Pelkmans, L. Cellular state determines the multimodal signaling response of single cells. Preprint at bioRxiv https://doi.org/10.1101/2019.12.18.880930 (2019).

  103. Yu, X., Yang, Y.-P., Dikici, E., Deo, S. K. & Daunert, S. Beyond antibodies as binding partners: the role of antibody mimetics in bioanalysis. Annu Rev. Anal. Chem. 10, 293–320 (2017).

    Article  CAS  Google Scholar 

  104. Berry, S. et al. Analysis of multispectral imaging with the AstroPath platform informs efficacy of PD-1 blockade. Science 372, eaba2609 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Wilkinson, M. D. et al. The FAIR Guiding Principles for scientific data management and stewardship. Sci. Data 3, 160018 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  106. Schapiro, D. et al. MITI Minimum Information guidelines for highly multiplexed tissue images. Preprint at https://arxiv.org/abs/2108.09499 (2021).

  107. Moore, J. et al. OME-NGFF: scalable format strategies for interoperable bioimaging data. Preprint at bioRxiv https://doi.org/10.1101/2021.03.31.437929 (2021).

  108. Nirmal, A. J. et al. The spatial landscape of progression and immunoediting in primary melanoma at single cell resolution. Preprint at bioRxiv https://doi.org/10.1101/2021.05.23.445310 (2021).

  109. Liu, Y. et al. High-spatial-resolution multi-omics sequencing via deterministic barcoding in tissue. Cell 183, 1665–1681(2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful for engaging and thoughtful discussions from the Affinity Reagent Imaging and Validation Working Group, HuBMAP Consortium. The authors would like to acknowledge funding from the following sources: NIH U54 DK120058, NIH U54 EY032442, NIH R01 AI145992, NIH R01 AI138581 (R. M. C. and J. M. S.), NIH T32ES007028 (E. K. N), NIH U54 HG010426-01 (M. P. S. and G. P. N.), NIH UG3 HL145600-01, NIH UH3 CA246633-01 (R. M. A), NIH UH3 CA246635-01 (N. L. K.), Swedish Research Council 2018-06461 (E. L.), Erling Persson Family Foundation (E. L.), Wallenberg Foundation (E. L.), NIH UH3 CA246594-01 (A. S. and E. M.), NIH T32CA196585 and ACS PF-20-032-01-CSM (J. W. H.), and NIH UH3 CA255133-03 (S.K.S.) and European Molecular Biology Laboratory (S. K. S.). This work was supported, in part, by the Intramural Research Program of the NIH, NIAID and NCI. We thank J. Hernandez and J. Davis (National Cancer Institute, NIH) for providing de-identified human tissues featured in this work, and K. Prummel (EMBL) for comments on the manuscript. Figures were made using the tools on Biorender.com.

Author information

Authors and Affiliations

Authors

Contributions

J. W. H., E. K. N., A. J. R, J. M. C., and S. K. S. conceived the plan, designed figures, and wrote the manuscript. R. T. B. designed display items and helped write the manuscript. A. A., K. C., E. M., J. H., A. E. W., J. F., J. C., A. S., R. M. C., R. M. A., G. P. N., K. C., S. M. H., R. N. G., J. M. S., E. L., M. P. S., and N. L. K. provided domain expertise and assisted with the conception and writing of the manuscript.

Corresponding authors

Correspondence to Andrea J. Radtke or Sinem K. Saka.

Ethics declarations

Competing interests

A. E. W. is an employee and shareholder of Abcam plc. J. F. is an employee of Cell Signaling Technologies. J. C. is an employee and shareholder of BioLegend. J. H. is an employee of Bio-Techne. E. S. is an employee of Thermo Scientific. E. M. and A. S. are current or past employees of GE Research. K. C. is an inventor for patent applications covering some technologies described in this paper and a cofounder of LifeCanvas Technologies. G. P. N. is inventor on a US patent, covering some technologies described in this paper, has equity in and/or is a member of the scientific advisory board of Akoya Biosciences. S. K. S. is an inventor for patent applications related to some of the methods described here.

Additional information

Peer review information Nature Methods thanks Trevor McKee, David Rimm and Takahiro Tsujikawa for their contribution to the peer review of this work. Rita Strack was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary Fig. 1, Tables 1–6 and References

Reporting Summary

Supplementary Dataset

List of community-validated antibody clones targeting common human and mouse protein markers across different highly multiplexed imaging platforms

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hickey, J.W., Neumann, E.K., Radtke, A.J. et al. Spatial mapping of protein composition and tissue organization: a primer for multiplexed antibody-based imaging. Nat Methods 19, 284–295 (2022). https://doi.org/10.1038/s41592-021-01316-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-021-01316-y

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing