Abstract
Cellular effector function assays traditionally rely on bulk cell populations that mask complex heterogeneity and rare subpopulations. The Xdrop® droplet technology facilitates high-throughput compartmentalization of viable single cells or single-cell pairs in double-emulsion droplets, enabling the study of single cells or cell–cell interactions at an individual level. Effector cell molecule secretion and target cell killing can be evaluated independently or in combination. Compatibility with a wide range of commercial assay reagents allows for single-cell level readouts using common laboratory techniques such as flow cytometry or microscopy. Moreover, individual cells of interest can be viably isolated for further investigation or expansion. Here we demonstrate the application of the double-emulsion droplet technology with a range of cell types commonly utilized for adoptive cell therapy of cancer: natural killer cells, blood-derived T cells, tumor-infiltrating lymphocytes, and chimeric antigen receptor T cells. Single-cell compartmentalization offers unparalleled resolution, serving as a valuable tool for advancing the development and understanding of cellular therapy products.
Key words: Xdrop, cancer immunotherapy, single-cell analysis, cell therapy, microfluidics, cell characterization
Highlights
-
•
High-throughput single-cell microfluidics assays are routine with Xdrop® technology.
-
•
Single-cell Xdrop® assays are compatible with common effector cell types.
-
•
Viable cells with specific functions can be isolated from Xdrop® single-cell assays.
Background
Adoptive cell therapies (ACT) have demonstrated impressive curative effects against specific cancer types, as illustrated by the success of chimeric antigen receptor (CAR) T cells for hematological malignancies1,2 and expanded tumor-infiltrating lymphocytes (TILs) for advanced melanoma.3,4 Natural killer (NK) and γδ T cells are now also being extensively explored as potential immunotherapies.5,6 While both the utilization and impact of cellular therapies are expected to increase in the future, challenges persist in predicting their potency and improving treatment efficacy.
Our understanding of what drives clinical responses to ACT has evolved over time, in TILs progressing from initial studies highlighting increased numbers of CD8+ T cells in infusion products7 to more nuanced recent studies emphasizing the importance of neoantigen-specific8 and stem-like9 CD8+ T cells instead. Together with the incredible success of CAR-T cell clinical trials,10,11 where much fewer cells are administered, these findings suggest that small subpopulations of ACT products drive the antitumor effects.
Given that ACT products are comprised of effector cells considered to be primary mediators of direct tumor killing, a logical assumption is that highly potent cells will react strongly against their targets and could form an effective ACT product on their own. Such potential is often measured via functional responses to target cells e.g. cytokine secretion or cytotoxic ability. These efforts are dominated by bulk population assays which can often obscure these critical populations of interest. Single-cell RNA sequencing has alleviated this issue to some degree; however, its utility is limited by cost, frequent lack of RNA-to-protein-to-function translatability,12 and inability to select cells of interest for further assays or clinical use. Selection of viable effector cells for further applications is currently achievable via fluorescence and magnetic-activated cell sorting (FACS/MACS). These methodologies are limited to using surface molecules as surrogates of cellular activation and function or secretome-capture methods affected by cytokine crosstalk or requiring complex custom experimental equipment. Accurate identification and selection of rare, potent, and viable effector cells is therefore currently challenging, restricting their subsequent downstream study and potential generation of selectively expanded ACT products.
The Xdrop double-emulsion droplet technology highlighted here offers a solution by providing a standardized and user-friendly high-throughput platform for single-cell study of effector cell functions, such as direct tumor killing or cytokine secretion. The technology is compatible with downstream analysis and selective expansion of viable cells after assay.
The Xdrop technology
Few technologies can accommodate single-cell assessment of cytotoxicity or cytokine secretion,13, 14, 15, 16, 17 often requiring extensive microfluidics know-how and specialized equipment. The Xdrop instrument addresses these issues. The double-emulsion droplet technology facilitates high throughput compartmentalization of viable single cells or cell pairs in droplets, allowing the study of individual cells or cell–cell interactions over time (Figure 1). The technology uses the same droplet encapsulation technology previously employed for microbes and DNA,18,19 instead utilizing a larger 50 μm diameter double-emulsion droplet (DE50) to accommodate eukaryotic cells.
Before encapsulation, cells are stained with fluorescent cell-labelling dyes to facilitate deconvolution during analysis. During the production process, DE50 droplets can also be loaded with standard cell culture media, varied cell types, and supplementary reagents (e.g. viability stains and stimulatory molecules) tailored to specific readout requirements. The droplets are stable after production and can be transferred to common cell culturing plastic containers by pipetting. Droplets are kept in standard CO2 incubators to maintain viability, and we have observed as high as ∼80% viability 24 h after encapsulation. Decreases in viability over time are observable, although the extent of this is highly cell type-dependent. Assay optimization and monitoring of cell viability in samples is recommended to minimize background signals.
Analysis of droplets can be conducted at specific time points of interest using conventional cell analysis methods such as microscopy and flow cytometry. Continued culturing of cells after assay is possible by treating droplets with a break reagent (Figure 1D), similar to the Xdrop DE20 targeted enrichment procedure for recovery of DNA.18,19 The current droplet break procedure results in cell recovery of up to 60% with a viability >95%.
Detection and enrichment of potent cytokine secretors
Cytokine secretion upon stimulation is commonly detected within culture medium through singleplex [e.g. enzyme-linked immunosorbent assay (ELISA), enzyme-linked immunosorbent spot (ELISpot)] or elaborate multiplexed approaches (e.g. bead-based immunoassays). These assays, however, do not permit single-cell deconvolution. An alternative is intracellular cytokine staining which provides single-cell resolution, yet by nature measures intracellular accumulation of cytokines rather than actual secretion. Importantly, none of these approaches allow the viable isolation of specific cells.
Contemporary detection technologies have evolved to capture secreted cytokines on the surface of the secreting cell without impacting viability, potentially providing a solution to the described challenges.20 Such approaches are vulnerable to cytokine crosstalk, however, where an excess of produced cytokine or premature saturation of capture reagents can cause false-positive and high background signals. It has previously been demonstrated that these pitfalls can be avoided by encapsulating assay reagents in microfluidic droplets,15,17 and cytokine secretion assays can similarly be carried out within DE50 droplets by loading detection reagents and single cells with capture antibody. For example, the increased resolution reveals a distinct high tumor necrosis factor-α (TNF-α)-producing subpopulation of T cells previously masked in a parallel bulk assay21 (Figure 2A), highlighting the importance of such single-cell resolution for identification of rare subpopulations.
The translation of such increased resolution into downstream applications, such as deeper analysis or preferential expansion of the most potent secretors during ACT product generation, is often limited by technical constraints. The ability to break DE50 droplets and successfully recover viable cells helps bridge this gap, as demonstrated with cytokine-stimulated NK cells22 (Figure 2B). Following a DE50 interferon-γ (IFN-γ) assay, NK cells were released from droplets and a potent IFN-γ-secreting subpopulation of NK cells was sorted by fluorescence-activated cell sorter (FACS) and cultured in vitro for an additional 2 weeks. Subsequent analysis revealed that potent IFN-γ secretors retained their superior secretory abilities upon re-stimulation.22
Detection of effective target cell killers
Cytotoxicity assays are rational markers of effector cell potency that reflect the in vivo mode of action, a crucial feature for ACT product potency assays as noted in recent proposed Food and Drug Administration guidance documentation.23 Isotopic chromium release, bioluminescence, impedance, and flow cytometry-based assays are all commonly employed; however, the observed effect cannot be linked to specific individual cells. The observed cytotoxic abilities may therefore be due to confounding factors such as highly potent serially killing effectors16,24 masking a lack of cytotoxicity in the bulk of the population. Instead, DE50 droplets provide the single-cell resolution required to appropriately assess the cytotoxic potential of ACT products and potentially isolate this population.
To quantify and identify effective killers, target and effector cells are labelled with distinct dyes and dead cell-staining propidium iodide (PI) is added immediately before encapsulation in DE50 droplets. The use of these dyes enables clear identification of droplet contents and the real-time labelling of newly dead cells throughout the incubation (Figure 3A and B). Calculation of true target cell killing utilizes the observed death in single-cell encapsulations (effector-only and target-only) to account for ‘background cell death’. This single-cell level cytotoxicity assay is compatible with a wide range of ACT-relevant cell types, including NK cells,25 TILs, and CAR-T cells (Figure 3C-E and Supplementary Figure S1, available at https://doi.org/10.1016/j.iotech.2024.100738).
Alternative readouts of cytotoxicity can also be employed in the DE50 droplet setup. Cytotoxic T, CAR-T, and NK cells all utilize rapid release of pre-formed cytotoxic effector proteins (chiefly perforin and granzymes) as their primary target killing mechanism.26 In bulk cytotoxicity assays, target recognition-dependent secretion of granzyme B (GzmB) is utilized as a reliable measure of effector cell reactivity and as a surrogate of cytotoxic ability.27,28 The addition of a cleavage-triggered fluorescent GzmB substrate to the encapsulation process facilitates the rapid detection of its release,29 as exemplified here with CAR-T cells (Figure 4A-C). The granularity of this assay, and subsequent identification of highly potent effectors, can be achieved via combination with the PI-based cytotoxicity assay described above.29 Using CAR-T cells (>98% CAR+), we demonstrate the simultaneous detection of GzmB secretion and cell killing in DE50 droplets (Figure 4D-F). This combination reveals that not all GzmB+ encapsulations result in rapid target-cell killing, emphasizing the value of integrating multiple parameters for enhanced single-cell insights.
Isolation and expansion of cells with indicated killing ability is of great interest for improving ACT products. Our preliminary studies with DE50 NK/K562 cell co-encapsulations demonstrate the potential feasibility of applying the Xdrop technology to this context (Figure 5). Intact PI+ droplets were FACS sorted and the recovered NK cells cultured in vitro. Cytotoxic ability was then retested in another DE50 droplet cytotoxicity assay where the sorted NK cells clearly demonstrated their retained cell-killing ability. The experiment applied adjusted sorting conditions which typically yield sorting efficiencies of 40%-60% and purity >95%; however, sorting can be further optimized to obtain 80%-90% efficiency.
Conclusion and future perspectives
The double-emulsion droplet technology presented here is a robust single-cell level platform that overcomes many current limitations in detecting highly potent effector cells within ACT products and demonstrates significant potential for their isolation and continued expansion.
The highly customizable nature of the assay promotes flexibility across many cell types and potency parameters, and the ability to isolate viable truly potent effectors allows a wide range of downstream in-depth characterizations concerning general phenotype, proliferative ability, response to repeated stimulation, functional profiles, and other such attributes. Findings resulting from these kinds of studies are likely to inform the future development of improved ACT by guiding the production of highly potent infusion products. Revealing the true functional profiles of individual cells in ACT products may also produce advances in linking clinical responses to these traits, and in the future, there is substantial potential for direct downstream therapeutic ACT applications. In particular, the technology presents an exciting prospect for the generation of selective ACT products.
Although promising, the described technology has certain limitations. Droplets with more than two cells can occur and confound analyses, but this can be mitigated by optimizing loading ratios and concentrations as cell encapsulation follows a Poisson distribution.30 Genuine cell-pair interaction assays form a minority of the total droplet population, but this still represents a substantial number due to the high number of generated droplets (750 000 per sample). Importantly, the singlet encapsulating droplets facilitated by the Poisson distribution function are valuable assay controls avoiding the need for multiple parallel assay control productions.
Background cell death does occur throughout the assay, but this can be accounted for when calculating target cell killing and can be significantly reduced by optimizing pre-assay cell handling procedures for each cell type. The cytotoxicity assays presented here cannot definitively confirm which cell of the pair within the droplet has died when analyzed using flow cytometry; however, complementary imaging analyses can confirm the observed/expected trends, as demonstrated in Figure 4D. Alternatively, target cell lines engineered to express specific fluorescent markers (e.g. GFP/YFP/RFP) upon death are likely an effective strategy for clarifying any ambiguity. Continued expansion after assay may be affected by potential cytotoxic effects of the droplet break reagent, although suitable recovery is still possible as demonstrated. This issue may be especially problematic in the case of extremely rare populations, although effector cell expansion protocols often result in significant fold expansions from even minimal starting material. While the data presented here highlight the feasibility of direct sorting of droplets of interest, we believe the effectiveness may be improved through additional optimization.
In conclusion, the double-emulsion droplet technology has the potential to play a sizable role in properly understanding and harnessing highly potent effector cells in an ACT context.
Declaration of generative AI and AI-assisted technologies in the writing process
During the preparation of this work, the authors used ChatGPT3.5 and Grammarly v1.2 in order to improve the readability of the manuscript. After using these tools, the authors reviewed and edited the content as needed and take full responsibility for the content of the publication.
Acknowledgements
The authors thank all patients who donated the clinical material used in this study. Written informed consent was provided by all patients before sample collection. Samples were collected from patients enrolled in clinical protocols at the National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark. We thank the healthcare personnel involved in these studies, in particular Eva Ellebaek, Rikke Andersen, and Troels Holz Borch. All procedures were approved by the Ethics Committee of the Capital Region of Denmark and national regulations for biomedical research (Ethical approval reference: H-20070020; Data Protection approval P-2021-303). The CD19-specific CAR T cell construct31 was generously provided by the Holt group at the BC Cancer Research Institute (Vancouver, Canada). GraphPad Prism v10 (GraphPad Software, Boston, Massachusetts, USA), and FlowJo™ v10.0 Software (BD Life Sciences, Ashland, Oregon, USA) were used for data analysis and figure generation. Samplix® and Xdrop® are registered trademarks of Samplix ApS.
Funding
This work was supported by the European Innovation Council [Project 190144395] to Samplix ApS. Salary for ACKR was supported by the Innovation Fund Denmark (3194-00037B), salary for MD was supported by Independent Research Fund Denmark (2034-00406B), and salary for CAC was supported by Lundbeck Foundation (R307-2018-3636).
Disclosure
ACKR, DLP, MJJ, MM, and PM are employees of Samplix ApS. MD has received access to research data from Bristol Myers Squibb and Genentech, and is an advisor for Achilles Therapeutics (past 2 years). All other authors have declared no conflicts of interest.
Footnotes
Supplementary data to this article can be found online at https://doi.org/10.1016/j.iotech.2024.100738.
Contributor Information
C.A. Chamberlain, Email: christopher.aled.chamberlain@regionh.dk.
P. Mouritzen, Email: pmo@samplix.com.
Supplementary data
References
- 1.Westin J.R., Oluwole O.O., Kersten M.J., et al. Survival with axicabtagene ciloleucel in large B-cell lymphoma. N Engl J Med. 2023;389:148–157. doi: 10.1056/NEJMoa2301665. [DOI] [PubMed] [Google Scholar]
- 2.Maude S.L., Laetsch T.W., Buechner J., et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378:439–448. doi: 10.1056/NEJMoa1709866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Rohaan M.W., Borch T.H., van den Berg J.H., et al. Tumor-infiltrating lymphocyte therapy or ipilimumab in advanced melanoma. N Engl J Med. 2022;387:2113–2125. doi: 10.1056/NEJMoa2210233. [DOI] [PubMed] [Google Scholar]
- 4.Dafni U., Michielin O., Lluesma S.M., et al. Efficacy of adoptive therapy with tumor-infiltrating lymphocytes and recombinant interleukin-2 in advanced cutaneous melanoma: a systematic review and meta-analysis. Ann Oncol. 2019;30:1902–1913. doi: 10.1093/annonc/mdz398. [DOI] [PubMed] [Google Scholar]
- 5.Laskowski T.J., Biederstadt A., Rezvani K. Natural killer cells in antitumour adoptive cell immunotherapy. Nat Rev Cancer. 2022;22:557–575. doi: 10.1038/s41568-022-00491-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Mensurado S., Blanco-Dominguez R., Silva-Santos B. The emerging roles of γδ T cells in cancer immunotherapy. Nat Rev Clin Oncol. 2023;20:178–191. doi: 10.1038/s41571-022-00722-1. [DOI] [PubMed] [Google Scholar]
- 7.Radvanyi L.G., Bernatchez C., Zhang M., et al. Specific lymphocyte subsets predict response to adoptive cell therapy using expanded autologous tumor-infiltrating lymphocytes in metastatic melanoma patients. Clin Cancer Res. 2012;18:6758–6770. doi: 10.1158/1078-0432.CCR-12-1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Kristensen N.P., Heeke C., Tvingsholm S.A., et al. Neoantigen-reactive CD8+ T cells affect clinical outcome of adoptive cell therapy with tumor-infiltrating lymphocytes in melanoma. J Clin Invest. 2022;132 doi: 10.1172/JCI150535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Krishna S., Lowery F.J., Copeland A.R., et al. Stem-like CD8 T cells mediate response of adoptive cell immunotherapy against human cancer. Science. 2020;370:1328–1334. doi: 10.1126/science.abb9847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Cappell K.M., Kochenderfer J.N. Long-term outcomes following CAR T cell therapy: what we know so far. Nat Rev Clin Oncol. 2023;20:359–371. doi: 10.1038/s41571-023-00754-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Park J.H., Geyer M.B., Brentjens R.J. CD19-targeted CAR T-cell therapeutics for hematologic malignancies: interpreting clinical outcomes to date. Blood. 2016;127:3312–3320. doi: 10.1182/blood-2016-02-629063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Reimegard J., Tarbier M., Danielsson M., et al. A combined approach for single-cell mRNA and intracellular protein expression analysis. Commun Biol. 2021;4:624. doi: 10.1038/s42003-021-02142-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Le K., Tan C., Gupta S., et al. A novel mammalian cell line development platform utilizing nanofluidics and optoelectro positioning technology. Biotechnol Prog. 2018;34:1438–1446. doi: 10.1002/btpr.2690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Josephides D., Davoli S., Whitley W., et al. Cyto-Mine: an integrated, picodroplet system for high-throughput single-cell analysis, sorting, dispensing, and monoclonality assurance. SLAS Technol. 2020;25:177–189. doi: 10.1177/2472630319892571. [DOI] [PubMed] [Google Scholar]
- 15.Antona S., Abele T., Jahnke K., et al. Droplet-based combinatorial assay for cell cytotoxicity and cytokine release evaluation. Adv Funct Mater. 2020;30 [Google Scholar]
- 16.Antona S., Platzman I., Spatz J.P. Droplet-based cytotoxicity assay: implementation of time-efficient screening of antitumor activity of natural killer cells. ACS Omega. 2020;5:24674–24683. doi: 10.1021/acsomega.0c03264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Yuan Y., Brouchon J., Calvo-Calle J.M., et al. Droplet encapsulation improves accuracy of immune cell cytokine capture assays. Lab Chip. 2020;20:1513–1520. doi: 10.1039/c9lc01261c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Blondal T., Gamba C., Møller Jagd L., et al. Verification of CRISPR editing and finding transgenic inserts by Xdrop indirect sequence capture followed by short- and long-read sequencing. Methods. 2021;191:68–77. doi: 10.1016/j.ymeth.2021.02.003. [DOI] [PubMed] [Google Scholar]
- 19.Madsen E.B., Hoijer I., Kvist T., Ameur A., Mikkelsen M.J. Xdrop: targeted sequencing of long DNA molecules from low input samples using droplet sorting. Hum Mutat. 2020;41:1671–1679. doi: 10.1002/humu.24063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Deng N., Mosmann T.R. Optimization of the cytokine secretion assay for human IL-2 in single and combination assays. Cytometry A. 2015;87:777–783. doi: 10.1002/cyto.a.22668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Jacobsen MJ, Hussing C, Højmark RS, Frøsig TM, Mouritzen P, Mikkelsen MJ. Application Note: Identifying highly potent TNF-α-secreting T cells using the Xdrop® single-cell format. Samplix ApS; 2022. https://samplix.com/files/samplix/downloads/AppNotes/Identify%20TNFa_ver.01_SX-000699-AN.pdf [Google Scholar]
- 22.Jacobsen MJ, Amasia M, Hussing C, et al. Application Note: Revealing and retrieving highly potent IFN-γ secretors using an Xdrop single-cell format workflow based on double-emulsion droplets. Samplix ApS; 2023. https://samplix.com/files/samplix/downloads/AppNotes/IFN-y%20secretion_ver.01_SX-000701-AN.pdf [Google Scholar]
- 23.Food and Drug Administration; 2023. FDA guidance for industry: potency assurance for cellular and gene therapy products.https://www.fda.gov/media/175132/download [Google Scholar]
- 24.Davenport A.J., Jenkins M.R., Ritchie D.S., et al. CAR-T cells are serial killers. Oncoimmunology. 2015;4 doi: 10.1080/2162402X.2015.1053684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Jacobsen MJ, Peterson DL, Amasia M., et al. Application Note: Rapidly identifying active natural killer cells. Samplix ApS; 2023. https://samplix.com/files/samplix/downloads/AppNotes/Agilent_Samplix_AppNote_Cell_killing_assays_Nov2023.pdf [Google Scholar]
- 26.Voskoboinik I., Whisstock J.C., Trapani J.A. Perforin and granzymes: function, dysfunction and human pathology. Nat Rev Immunol. 2015;15:388–400. doi: 10.1038/nri3839. [DOI] [PubMed] [Google Scholar]
- 27.Packard B.Z., Telford W.G., Komoriya A., Henkart P.A. Granzyme B activity in target cells detects attack by cytotoxic lymphocytes. J Immunol. 2007;179:3812–3820. doi: 10.4049/jimmunol.179.6.3812. [DOI] [PubMed] [Google Scholar]
- 28.Glienke W., Dragon A.C., Zimmermann K., et al. GMP-compliant manufacturing of TRUCKs: CAR T cells targeting GD(2) and releasing inducible IL-18. Front Immunol. 2022;13 doi: 10.3389/fimmu.2022.839783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Petersen DP, Jacobsen MJ, Schlicht B, Mikkelsen MJ, Mouritzen P. Application Note: Revealing granzyme B secretion and cell killing dynamics in a single-cell format. Samplix ApS; 2024. https://samplix.com/files/samplix/downloads/AppNotes/GranzymeB-ver.01_SSX-000649-AN.pdf [Google Scholar]
- 30.Cell distribution calculator. Samplix ApS; 2024. https://samplix.com/products/digital-tools [Google Scholar]
- 31.Kekre N., Hay K.A., Webb J.R., et al. CLIC-01: Manufacture and distribution of non-cryopreserved CAR-T cells for patients with CD19 positive hematologic malignancies. Front Immunol. 2022;13 doi: 10.3389/fimmu.2022.1074740. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.