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.

  • Protocol
  • Published:

Qualifying antibodies for image-based immune profiling and multiplexed tissue imaging

Abstract

Multiplexed tissue imaging enables precise, spatially resolved enumeration and characterization of cell types and states in human resection specimens. A growing number of methods applicable to formalin-fixed, paraffin-embedded (FFPE) tissue sections have been described, the majority of which rely on antibodies for antigen detection and mapping. This protocol provides step-by-step procedures for confirming the selectivity and specificity of antibodies used in fluorescence-based tissue imaging and for the construction and validation of antibody panels. Although the protocol is implemented using tissue-based cyclic immunofluorescence (t-CyCIF) as an imaging platform, these antibody-testing methods are broadly applicable. We demonstrate assembly of a 16-antibody panel for enumerating and localizing T cells and B cells, macrophages, and cells expressing immune checkpoint regulators. The protocol is accessible to individuals with experience in microscopy and immunofluorescence; some experience in computation is required for data analysis. A typical 30-antibody dataset for 20 FFPE slides can be generated within 2 weeks.

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: Schematic of antibody validation approaches for multiplexed tissue imaging.
Fig. 2: Pixel-by-pixel validation of antibodies against PD-1 using t-CyCIF in human tonsil.
Fig. 3: Validation of antibodies for immune profiling by comparison to clinical-grade antibodies.
Fig. 4: Validation of antibodies for immune profiling by co-segregation of known markers and expected spatial localization.
Fig. 5: Spatially resolved cataloging of immune cells in lung cancer FFPE tissue and component analysis of the immune subpopulations by t-CyCIF.
Fig. 6: Illustration of low-frequency CD45+/CD3+/FOXP3+/CD4/CD8a+ T cells detected and confirmed by t-CyCIF.
Fig. 7: Illustration of low-frequency CD45+/CD3+/PD-1+/LAG3+ T cells detected and confirmed by t-CyCIF.
Fig. 8: Systematic automated identification of rare subpopulations using t-CyCIF.

Similar content being viewed by others

Data availability

The datasets generated and/or analyzed during the current study are available to view on cycif.org (https://www.cycif.org/featured-paper/du-and-lin-2019/figures/). The raw data are available from http://www.synapse.org/ (Synapse IDs: syn18684611 and syn17865732).

Code availability

Custom code used to analyze the data in this study is available on GitHub (https://github.com/sorgerlab/cycif).

References

  1. Cyriac, G. & Gandhi, L. Emerging biomarkers for immune checkpoint inhibition in lung cancer. Semin. Cancer Biol. 52, 269–277 (2018).

    Article  CAS  PubMed  Google Scholar 

  2. Epstein, J. I., Egevad, L., Humphrey, P. A., Montironi, R. & Members of the ISUP Immunohistochemistry in Diagnostic Urologic Pathology Group. Best practices recommendations in the application of immunohistochemistry in the prostate: report from the International Society of Urologic Pathology consensus conference. Am. J. Surg. Pathol. 38, e6–e19 (2014).

  3. Falcon, B. L. et al. High-content multiplexed tissue imaging and quantification for cancer drug discovery. Drug Discov. Today 18, 510–522 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  5. Carvajal-Hausdorf, D. E., Schalper, K. A., Neumeister, V. M. & Rimm, D. L. Quantitative measurement of cancer tissue biomarkers in the lab and in the clinic. Lab. Invest. 95, 385–396 (2015).

    Article  CAS  PubMed  Google Scholar 

  6. 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 

  7. 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 

  8. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Decalf, J., Albert, M. L. & Ziai, J. New tools for pathology: a user’s review of a highly multiplexed method for in situ analysis of protein and RNA expression in tissue. J. Pathol. 247, 650–661 (2018).

    Article  Google Scholar 

  10. Glass, G., Papin, J. A. & Mandell, J. W. SIMPLE: a sequential immunoperoxidase labeling and erasing method. J. Histochem. Cytochem. 57, 899–905 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Tóth, Z. E. & Mezey, E. Simultaneous visualization of multiple antigens with tyramide signal amplification using antibodies from the same species. J. Histochem. Cytochem. 55, 545–554 (2007).

    Article  PubMed  CAS  Google Scholar 

  12. Pirici, D. et al. Antibody elution method for multiple immunohistochemistry on primary antibodies raised in the same species and of the same subtype. J. Histochem. Cytochem. 57, 567–575 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Remark, R. et al. In-depth tissue profiling using multiplexed immunohistochemical consecutive staining on single slide. Sci. Immunol. 1, aaf6925 (2016).

    Article  PubMed  Google Scholar 

  14. Goltsev, Y. et al. Deep profiling of mouse splenic architecture with CODEX multiplexed imaging. Preprint at bioRxiv, https://doi.org/10.1101/203166 (2017).

  15. 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 

  16. 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 

  17. 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 

  18. Friedenberger, M., Bode, M., Krusche, A. & Schubert, W. Fluorescence detection of protein clusters in individual cells and tissue sections by using toponome imaging system: sample preparation and measuring procedures. Nat. Protoc. 2, 2285–2294 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. Hillert, R. et al. Large molecular systems landscape uncovers T cell trapping in human skin cancer. Sci. Rep. 6, 19012 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. 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 

  21. 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  CAS  Google Scholar 

  22. Ståhl, P. L. et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science 353, 78–82 (2016).

    Article  PubMed  CAS  Google Scholar 

  23. Lee, J. H. et al. Highly multiplexed subcellular RNA sequencing in situ. Science 343, 1360–1363 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Lee, J. H. et al. Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression profiling in intact cells and tissues. Nat. Protoc. 10, 442–458 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Larsson, C., Grundberg, I., Söderberg, O. & Nilsson, M. In situ detection and genotyping of individual mRNA molecules. Nat. Methods 7, 395–397 (2010).

    Article  CAS  PubMed  Google Scholar 

  26. Chen, K. H., Boettiger, A. N., Moffitt, J. R., Wang, S. & Zhuang, X. RNA imaging. Spatially resolved, highly multiplexed RNA profiling in single cells. Science 348, aaa6090 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Moffitt, J. R. et al. High-performance multiplexed fluorescence in situ hybridization in culture and tissue with matrix imprinting and clearing. Proc. Natl. Acad. Sci. USA 113, 14456–14461 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Stewart, J. P. et al. Standardising RNA profiling based biomarker application in cancer-The need for robust control of technical variables. Biochim. Biophys. Acta 1868, 258–272 (2017).

    CAS  Google Scholar 

  29. True, L. D. Methodological requirements for valid tissue-based biomarker studies that can be used in clinical practice. Virchows Arch. 464, 257–263 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Srivastava, S., Ghosh, S., Kagan, J. & Mazurchuk, R. The making of a PreCancer atlas: promises, challenges, and opportunities. Trends Cancer 4, 523–536 (2018).

    Article  PubMed  Google Scholar 

  32. Coy, S. et al. Multiplexed immunofluorescence reveals potential PD-1/PD-L1 pathway vulnerabilities in craniopharyngioma. Neuro Oncol. 20, 1101–1112 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Dunn, I. F. et al. Mismatch repair deficiency in high-grade meningioma: a rare but recurrent event associated with dramatic immune activation and clinical response to PD-1 blockade. JCO Precis. Oncol. https://doi.org/10.1200/PO.18.00190 (2018).

  34. Ke, R. et al. In situ sequencing for RNA analysis in preserved tissue and cells. Nat. Methods 10, 857–860 (2013).

    Article  CAS  PubMed  Google Scholar 

  35. Krzywkowski, T. & Nilsson, M. Padlock probes to detect single nucleotide polymorphisms. Methods Mol. Biol. 1649, 209–229 (2018).

    Article  CAS  PubMed  Google Scholar 

  36. Krzywkowski, T., Hauling, T. & Nilsson, M. In situ single-molecule RNA genotyping using padlock probes and rolling circle amplification. Methods Mol. Biol. 1492, 59–76 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. Surrette, C. et al. Microfluidic tissue mesodissection in molecular cancer diagnostics. SLAS Technol. 22, 425–430 (2017).

    Article  PubMed  Google Scholar 

  38. Schulz, D. et al. Simultaneous multiplexed imaging of mRNA and proteins with subcellular resolution in breast cancer tissue samples by mass cytometry. Cell Syst. 6, 531 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. 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 

  40. Wei, S. C., Duffy, C. R. & Allison, J. P. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 8, 1069–1086 (2018).

    Article  PubMed  Google Scholar 

  41. Sharma, P. & Allison, J. P. Immune checkpoint targeting in cancer therapy: toward combination strategies with curative potential. Cell 161, 205–214 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Wong, P. F. et al. Multiplex quantitative analysis of tumor-infiltrating lymphocytes and immunotherapy outcome in metastatic melanoma. Clin. Cancer Res. 25, 2442–2449 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Jerby-Arnon, L. et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 175, 984–997.e24 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Socinski, M. A. et al. Atezolizumab for first-line treatment of metastatic nonsquamous NSCLC. N. Engl. J. Med. 378, 2288–2301 (2018).

    Article  CAS  PubMed  Google Scholar 

  45. Carbone, D. P. et al. First-line nivolumab in stage IV or recurrent non-small-cell lung cancer. N. Engl. J. Med. 376, 2415–2426 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Forde, P. M. et al. Neoadjuvant PD-1 blockade in resectable lung cancer. N. Engl. J. Med. 378, 1976–1986 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Topalian, S. L. et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J. Clin. Oncol. 32, 1020–1030 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Topalian, S. L. et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 366, 2443–2454 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ansell, S. M. et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N. Engl. J. Med. 372, 311–319 (2015).

    Article  PubMed  CAS  Google Scholar 

  50. Leach, D. R., Krummel, M. F. & Allison, J. P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 271, 1734–1736 (1996).

    Article  CAS  PubMed  Google Scholar 

  51. Pardoll, D. M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 12, 252–264 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Weber, J. S. et al. Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 16, 375–384 (2015).

    Article  CAS  PubMed  Google Scholar 

  53. Gulley, J. L. et al. Avelumab for patients with previously treated metastatic or recurrent non-small-cell lung cancer (JAVELIN solid tumor): dose-expansion cohort of a multicentre, open-label, phase 1b trial. Lancet Oncol. 18, 599–610 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hollman-Hewgley, D. et al. A single slide multiplex assay for the evaluation of classical Hodgkin lymphoma. Am. J. Surg. Pathol. 38, 1193–1202 (2014).

    Article  PubMed  Google Scholar 

  55. Jiang, Y., Li, Y. & Zhu, B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis. 6, e1792 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Chen, D. S. & Mellman, I. Elements of cancer immunity and the cancer-immune set point. Nature 541, 321–330 (2017).

    Article  CAS  PubMed  Google Scholar 

  57. Pearce, E. L. & Pearce, E. J. Metabolic pathways in immune cell activation and quiescence. Immunity 38, 633–643 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Baumeister, S. H., Freeman, G. J., Dranoff, G. & Sharpe, A. H. Coinhibitory pathways in immunotherapy for cancer. Annu. Rev. Immunol. 34, 539–573 (2016).

    Article  CAS  PubMed  Google Scholar 

  59. Kim, J. E. et al. Combination therapy with anti-PD-1, anti-TIM-3, and focal radiation results in regression of murine gliomas. Clin. Cancer Res. 23, 124–136 (2017).

    Article  CAS  PubMed  Google Scholar 

  60. Bendall, S. C. et al. Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science 332, 687–696 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Chevrier, S. et al. An immune atlas of clear cell renal cell carcinoma. Cell 169, 736–749.e18 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Lavin, Y. et al. Innate immune landscape in early lung adenocarcinoma by paired single-cell analyses. Cell 169, 750–765.e17 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Korin, B., Dubovik, T. & Rolls, A. Mass cytometry analysis of immune cells in the brain. Nat. Protoc. 13, 377–391 (2018).

    Article  CAS  PubMed  Google Scholar 

  64. Fung, E., Esposito, L., Todd, J. A. & Wicker, L. S. Multiplexed immunophenotyping of human antigen-presenting cells in whole blood by polychromatic flow cytometry. Nat. Protoc. 5, 357–370 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Herring, C. A. et al. Unsupervised trajectory analysis of single-cell RNA-seq and imaging data reveals alternative tuft cell origins in the gut. Cell Syst. 6, 37–51.e9 (2018).

    Article  CAS  PubMed  Google Scholar 

  66. Baker, G. J. et al. Systemic lymphoid architecture response assessment (SYLARAS): an approach to multi-organ, discovery-based immunophenotyping implicates a role for CD45R/B220+ CD8T cells in glioblastoma immunology. Preprint at bioRxiv, https://doi.org/10.1101/555854 (2019).

  67. Goltsev, Y. et al. Deep profiling of mouse splenic architecture with CODEX multiplexed imaging. Cell 174, 968–981.e15 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Kedziora, K. M., Prehn, J. H. M., Dobrucki, J. & Bernas, T. Method of calibration of a fluorescence microscope for quantitative studies. J. Microsc. 244, 101–111 (2011).

    Article  CAS  PubMed  Google Scholar 

  69. Waters, J. C. Accuracy and precision in quantitative fluorescence microscopy. J. Cell Biol. 185, 1135–1148 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Simon, R. M., Paik, S. & Hayes, D. F. Use of archived specimens in evaluation of prognostic and predictive biomarkers. J. Natl. Cancer Inst. 101, 1446–1452 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Mandrekar, S. J. & Sargent, D. J. Predictive biomarker validation in practice: lessons from real trials. Clin. Trials 7, 567–573 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  72. Gosho, M., Nagashima, K. & Sato, Y. Study designs and statistical analyses for biomarker research. Sensors 12, 8966–8986 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Du, Z. et al. Meningioma transcription factors link cell lineage with systemic metabolic cues. Neuro Onco. 20, 1331–1343 (2018).

    Article  CAS  Google Scholar 

  74. Koch, B. et al. Generation and validation of homozygous fluorescent knock-in cells using CRISPR-Cas9 genome editing. Nat. Protoc. 13, 1465–1487 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Sood, A. et al. Multiplexed immunofluorescence delineates proteomic cancer cell states associated with metabolism. JCI Insight 1, e87030 (2016).

    Article  PubMed Central  Google Scholar 

  76. 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 

  77. Engel, K. B. & Moore, H. M. Effects of preanalytical variables on the detection of proteins by immunohistochemistry in formalin-fixed, paraffin-embedded tissue. Arch. Pathol. Lab. Med. 135, 537–543 (2011).

    Article  PubMed  Google Scholar 

  78. Vassilakopoulou, M. et al. Preanalytical variables and phosphoepitope expression in FFPE tissue: quantitative epitope assessment after variable cold ischemic time. Lab. Invest. 95, 334–341 (2015).

    Article  CAS  PubMed  Google Scholar 

  79. Masucci, G. V. et al. Validation of biomarkers to predict response to immunotherapy in cancer: Volume I–pre-analytical and analytical validation. J. Immunother. Cancer 4, 76 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  80. Swedlow, J. R., Goldberg, I., Brauner, E. & Sorger, P. K. Informatics and quantitative analysis in biological imaging. Science 300, 100–102 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Goldberg, I. G. et al. The open microscopy environment (OME) data model and XML file: open tools for informatics and quantitative analysis in biological imaging. Genome Biol. 6, R47 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  82. Moore, J. et al. Open tools for storage and management of quantitative image data. Methods Cell Biol. 85, 555–570 (2008).

    Article  PubMed  Google Scholar 

  83. Schiffmann, D. A. et al. Open microscopy environment and findspots: integrating image informatics with quantitative multidimensional image analysis. BioTechniques 41, 199–208 (2006).

    Article  CAS  PubMed  Google Scholar 

  84. Shen, D., Wu, G. & Suk, H.-I. Deep learning in medical image analysis. Annu. Rev. Biomed. Eng. 19, 221–248 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Mika, S., Ratsch, G., Weston, J., Scholkopf, B. & Mullers, K. R. Fisher discriminant analysis with kernels. in Neural Networks for Signal Processing IX: Proceedings of the 1999 IEEE Signal Processing Society Workshop (Cat. No. 98TH8468) 41–48 (IEEE Signal Processing Society with support from the THOR Center for Neuromfomtics; 1999).

  86. Fisher, R. A. The use of multiple measurements in taxonomic problems. Ann. Eugen. 7, 179–188 (1936).

    Article  Google Scholar 

  87. 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 

  88. McKinley, E. T. et al. Optimized multiplex immunofluorescence single-cell analysis reveals tuft cell heterogeneity. JCI Insight 2, 93487 (2017).

    Article  PubMed  Google Scholar 

  89. Grys, B. T. et al. Machine learning and computer vision approaches for phenotypic profiling. J. Cell Biol. 216, 65–71 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Caicedo, J. C. et al. Data-analysis strategies for image-based cell profiling. Nat. Methods 14, 849–863 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Espina, V. et al. Tissue is alive: new technologies are needed to address the problems of protein biomarker pre-analytical variability. Proteom. Clin. Appl. 3, 874–882 (2009).

    Article  CAS  Google Scholar 

  92. Hicks, D. G. & Boyce, B. F. The challenge and importance of standardizing pre-analytical variables in surgical pathology specimens for clinical care and translational research. Biotech. Histochem 87, 14–17 (2012).

    Article  CAS  PubMed  Google Scholar 

  93. McLachlan, G. & Peel, D. Finite Mixture Models 349–393 (Wiley-Blackwell, 2005).

  94. Wilks, S. S. The large-sample distribution of the likelihood ratio for testing composite hypotheses. Ann. Math. Stat. 9, 60–62 (1938).

    Article  Google Scholar 

  95. Schwarz, G. Estimating the dimension of a model. Ann. Stat. 6, 461–464 (1978).

    Article  Google Scholar 

  96. Amir, E. D. et al. viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat. Biotechnol. 31, 545–552 (2013).

    Article  CAS  PubMed Central  Google Scholar 

  97. 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 

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

    Article  CAS  PubMed  Google Scholar 

  99. Dove, A. Agreeable antibodies: antibody validation challenges and solutions. Sci. 357, 1165–1167 (2017).

    Article  Google Scholar 

  100. Roncador, G. et al. The European antibody network’s practical guide to finding and validating suitable antibodies for research. MAbs 8, 27–36 (2016).

    Article  CAS  PubMed  Google Scholar 

  101. Ellington, A. A., Kullo, I. J., Bailey, K. R. & Klee, G. G. Antibody-based protein multiplex platforms: technical and operational challenges. Clin. Chem. 56, 186–193 (2010).

    Article  CAS  PubMed  Google Scholar 

  102. Wang, W., Lilyestrom, W. G., Hu, Z. Y. & Scherer, T. M. Cluster size and quinary structure determine the rheological effects of antibody self-association at high concentrations. J. Phys. Chem. B 122, 2138–2154 (2018).

    Article  CAS  PubMed  Google Scholar 

  103. Gulshan, V. et al. Development and validation of a deep learning algorithm for detection of diabetic retinopathy in retinal fundus photographs. JAMA 316, 2402–2410 (2016).

    Article  PubMed  Google Scholar 

  104. Churlaud, G. et al. Human and mouse CD8(+)CD25(+)FOXP3(+) regulatory T cells at steady state and during interleukin-2 therapy. Front. Immunol. 6, 171 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Kiniwa, Y. et al. CD8+ Foxp3+ regulatory T cells mediate immunosuppression in prostate cancer. Clin. Cancer Res. 13, 6947–6958 (2007).

    Article  CAS  PubMed  Google Scholar 

  106. Yang, Z.-Q. et al. Increased liver-infiltrating CD8+FoxP3+ regulatory T cells are associated with tumor stage in hepatocellular carcinoma patients. Hum. Immunol. 71, 1180–1186 (2010).

    Article  CAS  PubMed  Google Scholar 

  107. Woo, S.-R. et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 72, 917–927 (2012).

    Article  CAS  PubMed  Google Scholar 

  108. Bottai, G. et al. An immune stratification reveals a subset of PD-1/LAG-3 double-positive triple-negative breast cancers. Breast Cancer Res. 18, 121 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  109. Matsuzaki, J. et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc. Natl. Acad. Sci. USA 107, 7875–7880 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Banerjee, A., McKinley, E. T., von Moltke, J., Coffey, R. J. & Lau, K. S. Interpreting heterogeneity in intestinal tuft cell structure and function. J. Clin. Invest 128, 1711–1719 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  111. Wang, C., Hillsamer, P. & Kim, C. H. Phenotype, effector function, and tissue localization of PD-1-expressing human follicular helper T cell subsets. BMC Immunol. 12, 53 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors are members of the Harvard Tissue Atlas Program and the Ludwig Tumor Atlas. The Dana-Farber/Harvard Cancer Center was supported in part by NCI Cancer Center Support grant P30-CA06516. This work was also funded by NIH grants U54-CA225088 and U2C-CA233262 to P.K.S. and S.S., U2C-CA233280 to P.K.S., and R41-CA224503 to P.K.S. as well as by the Ludwig Center at Harvard. S.W. was supported by Molecular Biophysics Training grant T32-GM008313. B.I. was supported by NIH grant K08-CA222663 and funding from the Claudia Adams Barr Program for Innovative Cancer Research. We thank Y.A. Chen, M. P. Wu, G. Baker, Y. Chen, J. Muhlich, S. Mei, and C. Yapp for their expert assistance.

Author information

Authors and Affiliations

Authors

Contributions

S.S. and P.K.S. supervised the project; S.S., J.-R.L., Z.D., B.I., J.C.A., and P.K.S. were involved in planning; Z.D. and J.-R.L. performed the experiments and data analysis; R.R., S.W., and Z.M. contributed to data collection and analysis. All authors wrote and reviewed the paper.

Corresponding authors

Correspondence to Peter K. Sorger or Sandro Santagata.

Ethics declarations

Competing interests

P.K.S. is a member of the Scientific Advisory Board and a holder of equity in RareCyte, which manufactures one of the slide scanners used in this study; he is also a co-founder of Glencoe Software, which contributes to and supports the open-source OME/OMERO software used for image visualization in this work. S.S. has consulted for RareCyte. The remaining authors declare no competing interests.

Additional information

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

Related links

Key references using this protocol

Lin, J.-R. et al. Elife 7, e31657 (2018): https://elifesciences.org/articles/31657

Jerby-Arnon, L. et al. Cell 175, 984–997.e24 (2018): https://www.sciencedirect.com/science/article/pii/S0092867418311784

Dunn, I. F. et al. JCO Precis. Oncol. 2018, 10.1200/PO.18.00190 (2018): https://ascopubs.org/doi/full/10.1200/PO.18.00190

Coy, S. et al. Neuro Oncol. 20, 1101–1112 (2018): https://academic.oup.com/neuro-oncology/article-abstract/20/8/1101/4917565

Integrated supplementary information

Supplementary Fig. 1 Markers and major cell types identified by the t-CyCIF immune panel.

a) Canonical immune cell types and their markers. Seven major immune cell subtypes were assayed using a panel of 16 markers. Three additional markers (Ki-67, α-SMA and pan-keratin) were used to identify cell states (Ki-67 for proliferative cells) or separate immune cells from tumour cells (keratin-positive) or stromal cells (α-SMA-positive). b) Actual immune subpopulation identified from t-CyCIF immune profiling of LUNG-3-PR. 23,079 immune cells (keratin/α-SMA-negative cells) from the sample were used for binary gating of 15 different markers. A total of 1,356 different subpopulations were identified, of which 37 subpopulations represented >0.5% of total immune cells. The four subpopulations highlighted here are CD45+/IBA1+ (macrophage or dendritic cells), CD45+/CD20+ (B cells), CD45+/CD3+/CD8a+ (cytotoxic T cells) and CD45+/CD3+/CD4+ (helper T cells). Asterisks label 19 common immune cell subtypes.

Supplementary Fig. 2 Multi-antibody qualification of PD-L1 and FOXP3 antibodies by t-CyCIF in human FFPE tonsil tissue.

Representative images of immunofluorescence staining from human FFPE tonsil sections using a) three different antibodies for PD-L1: E1L3N, CST; 22C3, DAKO; and 28-8, Abcam (Scale bars: 100 µm). b) Plots of the pixel-by-pixel correlation of the signal intensity generated by the various PD-L1 antibodies. c) Immunofluorescence staining with two different antibodies for FOXP3: 23A/E7, eBioscience; and 206D, BioLegend (Scale bars: 100 µm). d) Plots of the pixel-by-pixel correlation of the signal intensity generated by the various FOXP3 antibodies. The plots in b and d display the correlation of a random sampling of 2,000 pixels. The plots on the lower left (with blue dots) show the original fluorescence intensity at each pixel, and the plots on the upper right (with cyan dots) show the log-transformed fluorescence intensity at each pixel. The Pearson correlation coefficients (R) are shown. DR, dynamic range. The dynamic range and correlation values from 5 different regions are presented in Supplementary Table 1.

Supplementary Fig. 3 Multi-antibody qualification of CD45 antibodies by t-CyCIF in human FFPE tonsil tissue.

Individual and merged a) low (Scale bars: 100 µm) and b) high (Scale bars: 50 µm) magnification images of immunofluorescence from three CD45 antibodies: 2D1, R&D; HI30, BioLegend; and PD7/26, eBioscience. c) Plots of the pixel-by-pixel correlation of the signal intensity generated by these CD45 antibodies. The plots display the correlation of a random sampling of 2,000 pixels. The plots on the lower left (with blue dots) show the original fluorescence intensity at each pixel, and the plots on the upper right (with cyan dots) show the log-transformed fluorescence intensity at each pixel. The Pearson correlation coefficients (R) are shown. DR, dynamic range. The dynamic range and correlation values from 5 different regions are presented in Supplementary Table 1.

Supplementary Fig. 4 Multi-antibody qualification of LAG3 antibodies by t-CyCIF in human FFPE tonsil tissue.

Individual and merged a) low (Scale bars: 100 µm) and b) high (Scale bars: 50 µm) magnification images of immunofluorescence from five LAG3 antibodies: EPR4392, Abcam; Polyclonal, R&D; 17B4, Lifespan; 11E3, Abcam; and T47-530, BD Bioscience. c) Plots of the pixel-by-pixel correlation of the signal intensity generated by the various LAG3 antibodies. The plots display the correlation of a random sampling of 2,000 pixels. The plots on the lower left (with blue dots) are of the original fluorescence intensity at each pixel, and the plots on the upper right (with cyan dots) are of the log-transformed fluorescence intensity at each pixel. The Pearson correlation coefficients (R) are shown. DR, dynamic range. The dynamic range and correlation values from 5 different regions are presented in Supplementary Table 1.

Supplementary Fig. 5 Multi-antibody qualification of CD11b antibodies by t-CyCIF in human FFPE tonsil tissue.

Individual and merged a) low (Scale bars: 100 µm) and b) high (Scale bars: 50 µm) magnification images of immunofluorescence from three CD11b antibodies: EP1345Y, Abcam; C67F154, eBioscience; and EPR1344, Abcam. c) Plots of the pixel-by-pixel correlation of the signal intensity generated by the CD11b antibodies. The plots display the correlation of a random sampling of 2,000 pixels. The plots on the lower left (with blue dots) show the original fluorescence intensity at each pixel, and the plots on the upper right (with cyan dots) show the log-transformed fluorescence intensity at each pixel. The Pearson correlation coefficients (R) are shown. DR, dynamic range. The dynamic range and correlation values from 5 different regions are presented in Supplementary Table 1.

Supplementary Fig. 6 Multi-antibody qualification by t-CyCIF and IHC in human FFPE tonsil tissue.

Representative images of t-CyCIF (left) and IHC staining (right) using antibodies to a) CD8a, b) FOXP3, c) PD-L1, and d) CD68 (Scale bars: 500 µm). e) Bar plot of the percentage of total immune cells in the tonsil section that were positive for the specified antibodies. The estimated number of positive cells was determined by both manual gating and by GMM analysis. The plot includes the percentage of positive cells for each immune antibody by IHC, as counted using Aperio ImageScope software.

Supplementary Fig. 7 Immune profiling in human FFPE tonsil tissue by t-CyCIF.

Merged images of t-CyCIF data (left) and scatter plots (right) from a random sampling of 10,000 cells for a) CD3 and CD45RB, b) CD3 and FOXP3, c) LAG3 and PD-1, and d) IBA1 and CD163. e) Additional merged images for CD3 and CD20, CD4 and CD8a, CD19 and CD20, CD11b and CD14, CD163 and CD68, and IBA1 and CD14 (Scale bars: 50 µm).

Supplementary Fig. 8

Individual t-CyCIF images for 16 different immune markers in human FFPE tonsil tissue. Scale bars: 50 µm.

Supplementary Fig. 9 Evaluation of segmentation accuracy and error composition.

a) Overall error rates of segmentation. Several hundred segmented masks were validated by human review, and the errors versus total counts of segmented cells from four different samples (tonsil, LUNG-1-LN, LUNG-2-BR and LUNG-3-PR) are plotted here. The average error rate was ~20% (S.E. 2%) for each sample. b) Illustration of the types of errors encountered. An example single-channel grey-scale Hoechst image is shown, with segmented masks highlighted in yellow. Three major types of segmentation errors were found: fused (blue arrows), split (red arrows) and missed (green arrows) cells (Scale bar: 20 µm). c) The composition of the different types of segmentation errors from the four samples.

Supplementary Fig. 10 Illustration of manual inspection and gating of intensity data.

a) Histogram of single-cell intensity from CD8a staining. Human inspection of this signal profile was used to set the gate/threshold at 8.5 log. The digital representation of the same intensity data was projected into physical maps of b) the original intensity data in log scale, c) data with all positive cells coloured in red, and d) the relative density of positive cells.

Supplementary Fig. 11 Analysis of effect of t-CyCIF cycle number on antigenicity in human FFPE tonsil tissue.

Histograms of signal intensity (upper left) and representative images of t-CyCIF from eight sequential sections of human FFPE tonsil tissue across different staining cycles for a) CD3, b) CD4, c) CD8a, d) CD20, and e) FOXP3. The same antibody concentration and exposure time for imaging were used for each cycle (see Supplementary Tables 5, 6 for additional details). The histogram plots were made from single cell data to show the intensity distribution from the different populations from different cycles. For the images shown, the same threshold was used for each marker for visualization and comparison. In e, the black arrow indicates the FOXP3+ population (Scale bars: 50 µm).

Supplementary Fig. 12 Analysis of effect of t-CyCIF cycle number on antigenicity in human FFPE tonsil tissue.

Histograms of signal intensity (upper left) and representative images of t-CyCIF from eight sequential sections of human FFPE tonsil tissue across different staining cycles for a) IBA1, b) CD14, c) CD68, d) CD163, and e) keratin. The same antibody concentration and exposure time for imaging were used in each cycle (see Supplementary Table 5, 6 for details). The histogram plots were made from single cell data to show the intensity distribution from the different populations from different cycles. For the images shown, the same threshold was used for each marker for visualization and comparison. In e, the black arrow indicates the keratin+ population (Scale bars: 50 µm).

Supplementary Fig. 13 H&E images of lung cancer specimens.

Whole slide scans of H&E-stained slides (left) and t-CyCIF images (right) from a) lung adenocarcinoma metastasis to the lymph node (LUNG-1-LN), b) lung squamous cell carcinoma metastasis to the brain (LUNG-2-BR), and c) primary lung squamous cell carcinoma (LUNG-3-PR) (Scale bars: 15 mm in H&E images; 1 mm in t-CyCIF images).

Supplementary Fig. 14 Geographic visualization of t-CyCIF data in LUNG-1-LN and LUNG-2-BR.

a) Montage of t-CyCIF images and b) corresponding dot plot for tumour cells (keratin+, blue), fibroblasts (α-SMA+, green) and immune cells (CD45+ or IBA1+, red) in LUNG-1-LN. c) Representative merged images of t-CyCIF data for α-SMA, keratin, CD45RB, IBA1, CD20, and CD3 from LUNG-1-LN (Scale bars: 50 µm). d) Montage of t-CyCIF images and e) corresponding dot plot for tumour cells (keratin+, blue), fibroblasts (α-SMA+, green) and immune cells (CD45+ or IBA1+, red) in LUNG-2-BR. f) Representative merged images of t-CyCIF data for α-SMA, keratin, CD45RB, IBA1, CD20, and CD3 from LUNG-2-BR (Scale bars: 50 µm).

Supplementary Fig. 15 t-SNE analysis of immune cells from lung cancer samples.

t-SNE plots of immune cell markers CD4, CD8a, FOXP3, IBA1, CD68, CD14, PD-1, PD-L1 and LAG3 from a random sampling of 2,000 immune cells for LUNG-1-LN, LUNG-2-BR and LUNG-3-PR. The staining for each of the indicated markers is mapped by colour (red = high, blue = low).

Supplementary Fig. 16 Low-frequency immune cell types detected and confirmed by t-CyCIF in LUNG-1-LN and LUNG-2-BR.

a) Scatter plots of CD4 and FOXP3 expression in LUNG-1-LN, with CD3 and CD8a expression mapped by colour (red = high, blue = low). 2.31% of the immune cells were CD45+/CD3+/FOXP3+/CD4+/CD8a, while 0.031% of the immune cells were CD45+/CD3+/FOXP3+/CD4/CD8a+. b, Representative image of t-CyCIF data for CD4, CD8a and FOXP3 in LUNG-1-LN (Scale bar: 50 µm). c. Scatter plots for CD4 and FOXP3, with CD3 and CD8a mapped as in panel a in LUNG-2-BR. 2.65% of the immune cells were CD45+/CD3+/FOXP3+/CD4+/CD8a while 0.006% of the immune cells were CD45+/CD3+/FOXP3+/CD4/CD8a+. d) Merged image of t-CyCIF data for CD4, CD8a and FOXP3 in LUNG-2-BR (Scale bar: 50 µm). e) Scatter plots for LAG3 and PD-1 expression in LUNG-1-LN, with CD3 and CD8a expression mapped as in panel a. 3.6% of immune cells were CD45+/CD3+/PD-1+, 0.87% were CD45+/CD3+/PD-1+/LAG3+ cells, 0.67% were CD45+/CD3+/ PD-1+/LAG3+/ CD4/CD8a+, and 0.07% were CD45+/CD3+/PD-1+/LAG3+/ CD4+/CD8a. See Supplementary Table 8 for additional details on these data.

Supplementary Fig. 17 Galleries of images of immune markers expressed in rare cell clusters 1 and 2.

Galleries of images of immune markers (LAG3, PD-1, CD45RB, CD3, PD-L1, CD4, CD45, CD8a, CD163, CD68, CD14, CD11b, FOXP3, IBA1, CD20) and DNA stain for 12 individual rare cells identified in cluster 1 using automated methods that putatively express CD45+/CD45RB+/CD3+/CD8a+/PD-1+ (top) or 11 individual rare cells in cluster 2 that putatively express CD45+/CD45RB+/CD3+/CD4+/PD-1+ (bottom) (Scale bar: 25 µm). Supplementary Table 10 indicates whether visual review of these images by a trained pathologist was deemed to be consistent with the automated calls.

Supplementary Fig. 18 Galleries of images of immune markers expressed in rare cell clusters 3 and 4.

Galleries of images of immune markers (LAG3, PD-1, CD45RB, CD3, PD-L1, CD4, CD45, CD8a, CD163, CD68, CD14, CD11b, FOXP3, IBA1, CD20) and DNA stain for 10 individual rare cells in cluster 3 identified using automated methods that putatively express CD45+/CD45RB+/CD3+/CD8a+/PD-1+/LAG3+ (top), or 10 individual rare cells in cluster 4 that putatively express CD45+/CD45RB+/CD3+/CD8a+/PD-1+/LAG3+/PD-L1+ (bottom) (Scale bar: 25 µm). Supplementary Table 10 indicates whether visual review of these images by a trained pathologist was deemed to be consistent with the automated calls.

Supplementary Fig. 19 Galleries of images of immune markers expressed in rare cell clusters 5, 6, and 7.

Galleries of images of immune markers (LAG3, PD-1, CD45RB, CD3, PD-L1, CD4, CD45, CD8a, CD163, CD68, CD14, CD11b, FOXP3, IBA1, CD20) and DNA stain for 10 individual rare cells in cluster 5 identified using automated methods that putatively express CD45+/CD45RB+/CD3+/CD4+/CD20+/PD-1+ (top), 6 individual rare cells in cluster 6 that putatively express CD45+/CD45RB+/IBA1+/CD163+/CD14+/CD68+/CD11b+/PD-1+/LAG3+/PD-L1+ (middle), or 6 individual rare cells in cluster 7 that putatively express CD45+/CD45RB+/CD3+/PD-1+ (Scale bar: 25 µm). Supplementary Table 10 indicates whether visual review of these images by a trained pathologist was deemed to be consistent with the automated calls.

Supplementary Fig. 20 Galleries of images of immune markers expressed in rare cell clusters 8, 9, 10, and 11.

Galleries of images of immune markers (LAG3, PD-1, CD45RB, CD3, PD-L1, CD4, CD45, CD8a, CD163, CD68, CD14, CD11b, FOXP3, IBA1, CD20) and DNA stain for 6 individual rare cells in cluster 8 identified using automated methods that putatively express CD45+/CD45RB+/CD3+/CD8a+/IBA1+/PD-1+, 6 individual rare cells in cluster 9 that putatively express CD45+/CD45RB+/CD3+/CD8a+/CD20+/PD-1+/LAG3+, 5 individual rare cells in cluster 10 that putatively express CD45+/CD45RB+/IBA1+/CD163+/CD14+/CD68+/PD-1+/LAG3+/PD-L1+, or 5 individual rare cells in cluster 11 that putatively express CD45+/CD45RB+/CD3+/CD8a+/FOXP3+/PD-1+/LAG3+ (Scale bar: 25 µm). Supplementary Table 10 indicates whether visual review of these images by a trained pathologist was deemed to be consistent with the automated calls.

Supplementary Fig. 21 Galleries of images of immune markers expressed in rare cell clusters 12, 13, 14, 15, and 16.

Galleries of images of immune markers (LAG3, PD-1, CD45RB, CD3, PD-L1, CD4, CD45, CD8a, CD163, CD68, CD14, CD11b, FOXP3, IBA1, CD20) and DNA stain for 5 individual rare cells in cluster 12 identified using automated methods that putatively express CD45+/CD45RB+/CD3+/CD4+/CD8a+/PD-1+, 4 individual rare cells in cluster 13 that putatively express CD45+/CD45RB+/IBA1+/CD14+/CD11b+/PD-1+/LAG3+/PD-L1+, 4 individual rare cells in cluster 14 that putatively express CD45+/CD45RB+/CD3+/CD20+/PD-1+, 4 individual rare cells in cluster 15 that putatively express CD45+/CD45RB+/CD3+/IBA1+/CD14+/CD11b+/PD-1+/LAG3+/PD-L1+, or 4 individual rare cells in cluster 16 that putatively express CD45+/CD45RB+/CD3+/CD8a+/CD20+/PD-1+ (Scale bar: 25 µm). Supplementary Table 10 indicates whether visual review of these images by a trained pathologist was deemed to be consistent with the automated calls.

Supplementary Fig. 22 Galleries of images of immune markers expressed in rare cell clusters 17, 18, 19, 20, and 21.

Galleries of images of immune markers (LAG3, PD-1, CD45RB, CD3, PD-L1, CD4, CD45, CD8a, CD163, CD68, CD14, CD11b, FOXP3, IBA1, CD20) and DNA stain for 4 individual rare cells in cluster 17 identified using automated methods that putatively express CD45+/CD45RB+/CD3+/CD8a+/FOXP3+/PD-1+, 4 individual rare cells in cluster 18 that putatively express CD45+/CD45RB+/CD3+/CD4+/IBA1+/CD163+/CD14+/PD-1+, 3 individual rare cells in cluster 19 that putatively express CD45+/CD45RB+/CD3+/CD8a+/CD11b+/PD-1+, 3 individual rare cells in cluster 20 that putatively express CD45+/CD45RB+/CD3+/CD8a+/IBA1+/PD1+/PD-L1+/LAG3+, or 3 individual rare cells in cluster 21 that putatively express CD45+/CD45RB+/CD3+/CD8a+/IBA1+/CD14+/CD68+/CD11b+/PD-1+/PD-L1+/LAG3+ (Scale bar: 25 µm). Supplementary Table 10 indicates whether visual review of these images by a trained pathologist was deemed to be consistent with the automated calls.

Supplementary Fig. 23 Galleries of images of immune markers expressed in rare cell cluster 1.

Image galleries of cells identified by automated systematic cell calling that express CD45, CD45RB, CD3, CD8a, and PD-1 (rare cell cluster 1, Fig. 8) (Scale bar: 25 µm) were reviewed by a trained pathologist who then annotated whether the automated calls were consistent with visual review (notes are shown in the right-most column; visual review confirmation for all rare cell populations is presented in Supplementary Table 10). Pseudo-colour images and merged images of the markers are shown here; monochromatic images for this rare cell cluster are shown in Supplementary Fig. 17.

Supplementary information

Supplementary Information

Supplementary Figs. 1–23 and Supplementary Tables 1–10

Reporting Summary

Supplementary Movie 1

t-CyCIF images from FFPE human tonsil tissue.

Supplementary Movie 2

t-CyCIF images from an FFPE human primary lung cancer resection sample, LUNG-3-PR.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Du, Z., Lin, JR., Rashid, R. et al. Qualifying antibodies for image-based immune profiling and multiplexed tissue imaging. Nat Protoc 14, 2900–2930 (2019). https://doi.org/10.1038/s41596-019-0206-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-019-0206-y

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer