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:

BEHAV3D: a 3D live imaging platform for comprehensive analysis of engineered T cell behavior and tumor response

Abstract

Modeling immuno-oncology by using patient-derived material and immune cell co-cultures can advance our understanding of immune cell tumor targeting in a patient-specific manner, offering leads to improve cellular immunotherapy. However, fully exploiting these living cultures requires analysis of the dynamic cellular features modeled, for which protocols are currently limited. Here, we describe the application of BEHAV3D, a platform that implements multi-color live 3D imaging and computational tools for: (i) analyzing tumor death dynamics at both single-organoid or cell and population levels, (ii) classifying T cell behavior and (iii) producing data-informed 3D images and videos for visual inspection and further insight into obtained results. Together, this enables a refined assessment of how solid and liquid tumors respond to cellular immunotherapy, critically capturing both inter- and intratumoral heterogeneity in treatment response. In addition, BEHAV3D uncovers T cell behavior involved in tumor targeting, offering insight into their mode of action. Our pipeline thereby has strong implications for comparing, prioritizing and improving immunotherapy products by highlighting the behavioral differences between individual tumor donors, distinct T cell therapy concepts or subpopulations. The protocol describes critical wet lab steps, including co-culture preparations and fast 3D imaging with live cell dyes, a segmentation-based image processing tool to track individual organoids, tumor and immune cells and an analytical pipeline for behavioral profiling. This 1-week protocol, accessible to users with basic cell culture, imaging and programming expertise, can easily be adapted to any type of co-culture to visualize and exploit cell behavior, having far-reaching implications for the immuno-oncology field and beyond.

Key points

  • BEHAV3D is a 3D live imaging platform for the analysis of engineered T cell behavior and tumor response. It enables assessment of inter- and intratumoral heterogeneity in response to cellular immunotherapy and the discovery of T cell modes of action associated with this heterogeneity.

  • This approach is unique because it combines monitoring the response of patient-specific tumor cells or organoids with tracking of large numbers of therapeutic T cell populations.

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 representation of the BEHAV3D protocol and applications.
Fig. 2: BEHAV3D co-culture preparation and imaging.
Fig. 3: BEHAV3D data-processing procedure.
Fig. 4: BEHAV3D brings insight into the behavioral landscape of engineered T cells.
Fig. 5: BEHAV3D refines the evaluation of inter- and intratumoral heterogeneity in response to immunotherapy.
Fig. 6: BEHAV3D identifies the most-potent killer populations.

Similar content being viewed by others

Data availability

All data described in this protocol are available from the corresponding author upon reasonable request. Representative raw imaging datasets used for PDO Behavioral map generation can be found on BioImage Achive under accession no. S-BIAD448, and representative processed imaging datasets can be found on Dryad (https://datadryad.org/stash/dataset/doi:10.5061/dryad.3n5tb2rp6). Analyzed demo data are made publicly available through GitHub (https://github.com/AlievaRios/BEHAV3D/tree/main/demos).

Code availability

All source codes of BEHAV3D are publicly available through GitHub (https://github.com/AlievaRios/BEHAV3D).

References

  1. Labanieh, L. & Mackall, C. L. CAR immune cells: design principles, resistance and the next generation. Nature 614, 635–648 (2023).

    Article  ADS  CAS  PubMed  Google Scholar 

  2. Irvine, D. J., Maus, M. V., Mooney, D. J. & Wong, W. W. The future of engineered immune cell therapies. Science 378, 853–858 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  3. Tuveson, D. & Clevers, H. Cancer modeling meets human organoid technology. Science 364, 952–955 (2019).

    Article  ADS  CAS  PubMed  Google Scholar 

  4. Ooft, S. N. et al. Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients. Sci. Transl. Med. 11, eaay2574 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. Vlachogiannis, G. et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 359, 920–926 (2018).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  6. Dijkstra, K. K. et al. Generation of tumor-reactive T cells by co-culture of peripheral blood lymphocytes and tumor organoids. Cell 174, 1586–1598.e12 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Liu, C., Yang, M., Zhang, D., Chen, M. & Zhu, D. Clinical cancer immunotherapy: current progress and prospects. Front. Immunol. 13, 961805 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Dekkers, J. F. et al. Uncovering the mode of action of engineered T cells in patient cancer organoids. Nat. Biotechnol. 41, 60–69 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Jacob, F., Ming, G. & Song, H. Generation and biobanking of patient-derived glioblastoma organoids and their application in CAR T cell testing. Nat. Protoc. 15, 4000–4033 (2020).

    Article  CAS  PubMed  Google Scholar 

  10. Cattaneo, C. M. et al. Tumor organoid–T-cell coculture systems. Nat. Protoc. 15, 15–39 (2020).

    Article  CAS  PubMed  Google Scholar 

  11. Schnalzger, T. E. et al. 3D model for CAR‐mediated cytotoxicity using patient‐derived colorectal cancer organoids. EMBO J. 38, e100928 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Freckmann, E. C. et al. Traject3d allows label-free identification of distinct co-occurring phenotypes within 3D culture by live imaging. Nat. Commun. 13, 5317 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  13. Matthews, J. M. et al. OrganoID: a versatile deep learning platform for tracking and analysis of single-organoid dynamics. PLoS Comput. Biol. 18, e1010584 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bandey, I. N. et al. Designed improvement to T-cell immunotherapy by multidimensional single cell profiling. J. Immunother. Cancer 9, e001877 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Romain, G. et al. Multidimensional single-cell analysis identifies a role for CD2-CD58 interactions in clinical antitumor T cell responses. J. Clin. Invest. 132, e159402 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Crainiciuc, G. et al. Behavioural immune landscapes of inflammation. Nature 601, 415–421 (2022).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  17. Upadhaya, S., Hubbard-Lucey, V. M. & Yu, J. X. Immuno-oncology drug development forges on despite COVID-19. Nat. Rev. Drug Discov. 19, 751–752 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Voabil, P. et al. An ex vivo tumor fragment platform to dissect response to PD-1 blockade in cancer. Nat. Med. 27, 1250–1261 (2021).

    Article  CAS  PubMed  Google Scholar 

  19. Kantari-Mimoun, C. et al. CAR T-cell entry into tumor islets is a two-step process dependent on IFNγ and ICAM-1. Cancer Immunol. Res. 9, 1425–1438 (2021).

    Article  CAS  PubMed  Google Scholar 

  20. You, R. et al. Active surveillance characterizes human intratumoral T cell exhaustion. J. Clin. Invest. 131, e144353 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wang, Z. et al. Rapid tissue prototyping with micro-organospheres. Stem Cell Rep. 17, 1959–1975 (2022).

    Article  CAS  Google Scholar 

  22. Ding, S. et al. Patient-derived micro-organospheres enable clinical precision oncology. Cell Stem Cell 29, 905–917.e6 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Khazen, R. et al. Functional heterogeneity of cytotoxic T cells and tumor resistance to cytotoxic hits limit anti‐tumor activity in vivo. EMBO J. 40, e106658 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Cazaux, M. et al. Single-cell imaging of CAR T cell activity in vivo reveals extensive functional and anatomical heterogeneity. J. Exp. Med. 216, 1038–1049 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Weigelin, B. et al. Cytotoxic T cells are able to efficiently eliminate cancer cells by additive cytotoxicity. Nat. Commun. 12, 5217 (2021).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ineveld et al. Multispectral confocal 3D imaging of intact healthy and tumor tissue using mLSR-3D. Nat. Protoc. 17, 3028–3055 (2022).

    Article  PubMed  Google Scholar 

  27. Bayarmagnai, B., Perrin, L., Pourfarhangi, K. E. & Gligorijevic, B. Intravital imaging of tumor cell motility in the tumor microenvironment context. Methods Mol. Biol. 1749, 175–193 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kimmel, J. C., Chang, A. Y., Brack, A. S. & Marshall, W. F. Inferring cell state by quantitative motility analysis reveals a dynamic state system and broken detailed balance. PLoS Comput. Biol. 14, e1005927 (2018).

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  29. Tormene, P., Giorgino, T., Quaglini, S. & Stefanelli, M. Matching incomplete time series with dynamic time warping: an algorithm and an application to post-stroke rehabilitation. Artif. Intell. Med. 45, 11–34 (2009).

    Article  PubMed  Google Scholar 

  30. Gründer, C. et al. γ9 and δ2CDR3 domains regulate functional avidity of T cells harboring γ9δ2TCRs. Blood 120, 5153–5162 (2012).

    Article  PubMed  Google Scholar 

  31. Kuball, J. et al. Facilitating matched pairing and expression of TCR chains introduced into human T cells. Blood 109, 2331–2338 (2006).

    Article  PubMed  Google Scholar 

  32. Wallstabe, L. et al. ROR1-CAR T-cells are effective against lung and breast cancer in advanced microphysiologic 3D tumor models. JCI Insight 4, e126345 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Fujii, M., Matano, M., Nanki, K. & Sato, T. Efficient genetic engineering of human intestinal organoids using electroporation. Nat. Protoc. 10, 1474–1485 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Broutier, L. et al. Culture and establishment of self-renewing human and mouse adult liver and pancreas 3D organoids and their genetic manipulation. Nat. Protoc. 11, 1724–1743 (2016).

    Article  CAS  PubMed  Google Scholar 

  35. Drost, J. et al. Organoid culture systems for prostate epithelial and cancer tissue. Nat. Protoc. 11, 347–358 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Dekkers, J. F. et al. High-resolution 3D imaging of fixed and cleared organoids. Nat. Protoc. 14, 1756–1771 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Deben, C. et al. OrBITS: label-free and time-lapse monitoring of patient derived organoids for advanced drug screening. Cell Oncol. (Dordr.) 46, 299–314 (2023).

    Article  CAS  PubMed  Google Scholar 

  38. Dekkers, J. F. et al. Long-term culture, genetic manipulation and xenotransplantation of human normal and breast cancer organoids. Nat. Protoc. 16, 1936–1965 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the Princess Máxima Center for Pediatric Oncology for technical support and the Hubrecht Institute and Zeiss for imaging support and collaboration. All imaging was performed at the Princess Máxima Imaging Center. We thank Hubrecht Organoid Technology for providing BC PDOs and the Princess Máxima Center Organoid Facility for organoid culture support. We thank E. Bokobza and M. Buchholz for providing the healthy breast organoid lines, D. van Vuurden (Princess Máxima Center for Pediatric Oncology) for providing DMG cultures, M. Koomen (Hubrecht Institute) for support with H&N cancer PDO culture and members of the Dream3DLAB (Rios group) for offering critical feedback on the project. This work was financially supported by the Princess Máxima Center for Pediatric Oncology. M.A. is supported by the Comunidad de Madrid (2022-T1/BMD-24021). J.F.D. was supported by a KWF Young Investigator grant (no. 14135). A.C.R. is supported by an ERC-starting grant 2018 project (no. 804412). M.B.R. and N.M.M.D. and the imaging work are supported by the Dutch ODAS Foundation.

Author information

Authors and Affiliations

Authors

Contributions

M.A. designed and performed the computational analysis. J.F.D., M.B.R. and D.P., set up co-cultures and performed imaging experiments, with assistance from A.L.Z. A.L.Z. cultured BC PDOs. M.A. and S.d.B. analyzed data. S.d.B. further adapted the computational workflow. M.B.R. assisted with imaging data processing. J.F.D. optimized experimental conditions for BC-PDO experiments, and M.B.R. optimized experimental conditions for liquid tumor experiments with supervision from J.F.D. A.M.C. and S.N. provided ROR1-CAR T cells. A.M.C., C.v.d.V. and N.M.M.D. generated and cultured CD19 cells. M.L.d.B. provided the ALL cells. R.P., S.N., F.G.J.C. and C.v.d.V. supervised the work with liquid tumors. Z.S. and J.K. provided the TEG cells and supervised work with these cells. H.C. supervised work with organoids. M.A., J.F.D. and A.C.R. designed the study. M.A. wrote the manuscript, with support from J.F.D., M.B.R., A.L.Z., D.P., A.C.R. and E.J.W. All authors critically revised the manuscript.

Corresponding authors

Correspondence to Maria Alieva or Anne C. Rios.

Ethics declarations

Competing interests

H.C. and J.F.D. are named as inventors on (pending) patents related to the organoid technology. For the full disclosure of H.C., see https://www.uu.nl/staff/JCClevers/Additional%20functions. Z.S. and J.K. are inventors on different patents for γδ TCR sequences, recognition mechanisms and isolation strategies. J.K. is a scientific cofounder and shareholder of Gadeta (www.gadeta.nl). The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Protocols thanks Navin Varadarajan and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Related links

Key reference using this protocol

Dekkers, J. F. et al. Nat. Biotechnol. 41, 60–69 (2022): https://doi.org/10.1038/s41587-022-01397-w

Extended data

Extended Data Fig. 1 Organoid harvesting and co-culture setup.

a, Quantification of PDO targeting by using a CellTiter-Glo viability assay upon 24-h co-culture of organoids with TEGs with different cell culture media. b and c, Representative images of confluent BC PDO cultures (b) and BC PDO cultures plated too sparsely, plated too densely, with clear presence of cell debris or with good density (c). Scale bars, 100 μm. Numbers in the left corner of the images in b and c refer to specific BC PDO cultures (described in table 1 of ref. 38). d, Representative images of BME drops in a 12-well plate. Scale bar, 5 mm. Panels b–d reproduced with permission from ref. 38, Springer Nature America, Inc.

Extended Data Fig. 2 Specifics of the different live cell fluorescent dyes.

Representative images of distinct dye specific characteristics as described in Table 1. a, Living cells (yellow) present a dim dye eFluor 450 staining (green arrowheads), whereas dead cells (red) appear brighter (purple arrows). The gray arrowhead indicates an example of a non-cell-specific staining. b, Representative 3D images showing specifics of calcein AM. Viable cells are exclusively stained with this dye (white arrows), whereas dead cells can no longer retain this dye (purple arrows). c, Representative 3D images showing a main property of CellTracker Orange CMRA dye. Dead cells can also retain this dye (purple arrows), which holds true for all Cell Tracker dyes from Table 1. The gray arrowhead indicates debris. d, Representative optical sections showing specifics of CellBrite Steady 650. Cells exhibit a more membranal staining at the start of culture (gray arrow), which transitions into a preponderantly cytoplasmic staining at later time points (white arrow). e, Representative 3D images depicting specifics of Annexin V-CF555. Contrary to the manufacturer’s recommendations, no prelabeling of the dead cells is required, because a bright signal in the Annexin V-CF555 channel can be detected upon direct addition to the imaging medium (white arrowheads). Scale bars, 10 µm.

Extended Data Fig. 3 BEHAV3D imaging on SP8.

a, Representative 3D multispectral images of ALL tumor cells (yellow) in co-culture with CD8+ (blue) and CD4+ (green) CAR19 cells, showing tumor response (red) at the indicated time points. Scale bars, 100 µm. b and c, Single channels showing raw images (b) and corrected images after Step 37 (c) for CD8+ CAR19 (top) and CD4 CAR19 (bottom) channels.

Extended Data Fig. 4 Example of the BEHAV3D image-processing procedure.

a, Representative overlayed and single-channel images of an example dataset consisting of PDOs in co-culture with CD4+ and CD8+ TEGs. These images are used as input for Step 38 explained in b. b and c, Channel arithmetic’s procedure and formulas described in Step 38 (b) that generate new image channels with cell-specific signal (c). d, Schematic representation of T cell segmentation and tracking in Steps 39 and 40. Channels 7 or 8 are taken as input for either CD8+ T cell or CD4+ T cell tracking. An autoregressive motion algorithm is used for tracking. e, Schematic representation of organoid segmentation in Step 41A for population-based death dynamics monitoring. f, Schematic representation of single organoid tracking in Step 41B. ROIs are defined around each individual organoid to facilitate their separation. A connected components algorithm is used for tracking. g, Optionally, distance transformation in Step 42 can be used to generate a new channel that represents the distance to organoid surfaces. h, T cell and tumor cell dynamic statistics are exported. The yellow boxes indicate the statistics that have to be selected in the statistics tab.

Extended Data Fig. 5 BEHAV3D imaging of TEGs targeting H&N and DMG tumors.

a, Representative 3D multispectral images of H&N tumor organoids (yellow) in co-culture with TEG cells (blue), showing the organoids’ response (red) at the indicated time points. b, Representative 3D multispectral images of DMG tumor organoids (yellow) in co-culture with TEG cells (blue), showing the organoids’ response (red) at the indicated time points. Scale bars, 100 µm.

Supplementary information

Supplementary Information

Supplementary Methods and Figs. 1 and 2

Supplementary Video 1

Representative video of an imaging experiment performed on the Zeiss LSM 880. The left panel displays raw imaging data of a co-culture of CD8+ CAR19 cells (blue), CD4+ CAR19 cells (green) and ALL cells (yellow). Dead dye presence is visualized in red. The right panel displays segmented and tracked data for CD8+ CAR19 cells (blue), CD4+ CAR19 cells (green) and ALL cells (gray). The track of the CAR19 cells represents the cell path over the last hour.

Supplementary Video 2

Representative video of an imaging experiment performed on the Leica SP8. The left panel displays raw imaging data of a co-culture of CD8+ CAR19 cells (blue), CD4+ CAR19 cells (green) and ALL cells (yellow). Dead dye presence is visualized in red. The right panel displays segmented and tracked data for CD8+ CAR19 cells (blue), CD4+ CAR19 cells (green) and ALL cells (gray). The track of the CAR19 cells represents the cell path over the last hour.

Supplementary Video 3

Representative video showing one single TEG cell, displaying morphological plasticity and destroying an entire tumor organoid in over 10 h. The left panel displays raw imaging data with TEG cells (blue) and a BC tumor organoid (yellow). Dead dye presence is visualized in red. The right panel displays segmented and tracked TEG cells (blue) and a BC tumor organoid (gray). The full T cell track is shown in blue.

Supplementary Video 4

Representative video showing one single motile CD8+ CAR19 cell (outlined in purple) performing five tumor cell killings in 8 h. CD8+ CAR19 cells (blue), CD4+ CAR19 cells (green) and ALL cells (yellow). Dead dye presence is visualized in red.

Supplementary Table 1

Table to quantify tracking accuracy metrics

Supplementary Table 2

BEHAV3D metadata template for organoid data processing

Supplementary Table 3

BEHAV3D metadata template for T cell data processing

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Alieva, M., Barrera Román, M., de Blank, S. et al. BEHAV3D: a 3D live imaging platform for comprehensive analysis of engineered T cell behavior and tumor response. Nat Protoc (2024). https://doi.org/10.1038/s41596-024-00972-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41596-024-00972-6

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