Unlocking the Potential of Cancer Immunotherapy with 3D Immune Cell Killing Assays
Whitepaper
Last Updated: November 13, 2023
(+ more)
Published: October 11, 2023
Credit: iStock
CAR T-cell therapies have shown impressive results in treating hematological malignancies. However, developing specialized therapies for the treatment of solid tumors still remains a challenge.
3D cell culture models are essential in researching how therapies can infiltrate and remain effective in solid tumor microenvironments. By optimizing 3D cell killing assays using the latest analysis/analytical technologies, researchers can guide and accelerate the engineering of novel therapeutics.
This literature review explores a range of successful assay setups that leverage different analytical technologies.
Download this literature review to discover:
- Different assay setups for 3D killing assays
- High-content analysis, image cytometry and in vivo imaging for 3D killing assays
- Benefits and challenges of the different technologies
LITERATURE REVIEW
Unlocking the potential of cancer
immunotherapy: Overcoming the challenges
of 3D immune cell killing assays
Immunotherapy harnesses the innate capabilities of the
body’s immune system to prevent, control, and eliminate
cancer. This transformative approach to cancer treatment
encompasses an array of strategies, including chimeric antigen
receptor (CAR) T-cell therapy, immune checkpoint inhibitors
(ICIs), monoclonal antibodies, and adoptive cell transfer.
CAR T-cell therapy involves genetically modifying T cells
to recognize specific antigens expressed by tumor cells,
effectively activating the patient’s immune system to
target cancerous cells. CAR T-cell therapies have shown
impressive results in treating hematological malignancies,
such as leukemia and lymphoma, with the FDA already
approving six CAR T-cell therapies. But developing
specialized therapies for the treatment of solid tumors
has progressed at a much slower pace.
Obstacles include the challenge of identifying tumor-specific
antigens due to tumor heterogeneity, the limited ability
of CAR T-cells to reach and infiltrate tumor tissue, and the
risk of off-target toxicity. Additionally, most solid tumors
are embedded in a hostile microenvironment, with poor
blood supply, inadequate oxygen, and nutrient delivery,
an acidic pH, and a dense extracellular matrix. These
factors can hinder the effector function of CAR T-cells and
inhibit their clinical efficacy. To overcome these challenges,
researchers are attempting to model the tumor environment
in vitro, which is crucial for making informed decisions about
the effectiveness of CAR T-cell and other immunotherapies.
Immune cell killing assays are critical to these pursuits
as they are used to gain insights into the cytotoxic functions
of different immune cell types and evaluate the efficacy
of potential treatments. Traditionally, cells are grown in two
dimensions on a flat surface of a culture dish. More recently,
three-dimensional (3D) cell cultures, which more closely
mimic the complexities of the in vivo microenvironment,
are being used to gain more physiologically relevant data
compared to 2D cell culture assays. Various technologies
are employed for 3D immune cell killing assays, each
offering unique advantages and insights into the cytotoxic
functions of immune cells and the evaluation of treatment
efficacy. Analysis approaches include flow cytometry,
high-content analysis, live-cell imaging and in vivo
bioluminescence imaging in small animal models. Although
3D immune cell killing assays yield more physiologically
relevant results, they come with various technical and
logistical difficulties. Some of the challenges include:
• Difficulties growing consistent and reproducible 3D cultures
• Reliable quantification of target cell killing in 3D cultures
over time
• Reliable quantification of immune cell infiltration into
3D cultures over time
• Distinguishing between different cell types, such as
differentiating dead cancer cells from dead immune cells
• Obtaining kinetic information over a sufficient time
period to assess immune cell persistence
• Identifying suitable fluorescent dyes that can penetrate
the 3D cell model, do not impair immune cell function,
and sufficiently stain over the whole time course.
Unlocking the potential of cancer immunotherapy: Overcoming the challenges of 3D immune cell killing assays
www.revvity.com 2
To address these challenges, this paper presents several
successful assay setups that utilize various analysis
technologies to study the antitumor functions
of immunotherapies in a 3D environment. We also provide
a technology comparison to help researchers understand
the benefits and challenges associated with these
different approaches.
High-content analysis
Modified CAR T-cells targeting membrane-proximal
epitope of mesothelin
Developing high-efficiency CAR T-cells requires appropriate
antigen selection to eliminate tumor cells with minimal
toxicity. A promising antigen for targeted immunotherapy
is mesothelin (MSLN), a cell surface glycoprotein with low
expression in normal tissues and high expression in various
solid tumors. In a recent study, Zhang et al. engineered two
types of CARs targeting either a membrane-distal (meso1
CAR T-cells) or membrane-proximal (meso3 CAR T-cells)
epitope of MSLN using a modified piggyBac transposon
Figure 1: a) The killing activity of meso1 CAR and meso3 CAR T-cells was detected using the 3D cancer spheroid model in gastric cancer. b)
The time effect for the death rate of tumor cells was shown by a histogram in gastric cancer. c) The killing activity of meso1 CAR and meso3
CAR T-cells was detected using the 3D cancer spheroid model in ovarian cancer. d) The time effect of the death rate of tumor cells was
shown by the histogram in ovarian cancer. Image credit: Zhang et al., 2019.1
system.1
The researchers utilized 3D cancer spheroid
models to assess the killing activity of the modified T cells.
Gastric and ovarian cancer cells were stained with Hoechst
and cultured in wells for 48 hours to generate 3D spheroid
cancer cells. Calcein-AM stained CAR T-cells were then
added to the wells at the effector/target (E:T) ratio of 2:1,
along with propidium iodide. To determine the death ratio
of tumor cells the researchers used the Opera Phenix™
high-content screening system to analyze the fluorescence
values at 0, 4, 6, and 24 hours after co-culturation.
Although both CAR T-cells were found to infiltrate into the
tumor sphere, the team observed stronger killing activity
against MSLN-expressing cancer cells from meso3 CAR
T-cells in both cancer models (Figure 1). These findings
showcase how 3D spheroid models and advanced
screening systems can be used to gain valuable insights
into the cytotoxicity of engineered CAR T-cells, while also
underscoring the potential of targeting the membraneproximal epitope of MSLN to treat MSLN-expressing tumors.
Unlocking the potential of cancer immunotherapy: Overcoming the challenges of 3D immune cell killing assays
www.revvity.com 3
Modeling immune cytotoxicity with tumor-derived
organoids and effector T cells
ICIs have demonstrated remarkable efficacy in activating
pre-existing anticancer T-cell responses among patient
subsets affected by various advanced malignancies.
However, it remains difficult to accurately predict which
ICI will be effective for individual patients. Zhou and
colleagues set out to establish a co-culture platform
involving cholangiocarcinoma (CCA) organoids and immune
cells that could potentially serve as an in vitro personalized
model to evaluate the efficacy of ICIs.2
In the co-culture experiment, T cells were stained with
CellTrace Far Red and combined with Hoechst-stained
CCA organoids. To monitor cell death, a green caspase 3/7
detection reagent and Hoechst 33342 were also added to
the medium. Confocal time-course imaging and quantitative
assessments were performed on co-cultures every six hours
for seven days. Time-lapse images presented at 0, 90, and
180 hours (Figure 2a) using the Opera Phenix high-content
screening system indicate a noticeable increase in apoptosis
in co-cultured organoids. Specifically, a higher number
of CCA2 organoid cells were caspase 3/7 probe positive
in the presence of T cells compared to the culture of only
organoids. Quantitative image analysis confirmed that
the apoptotic cell area increased about sevenfold in the
organoid region after seven days of co-culture with T cells
compared to the single culture of organoids (Figure 2b).
As demonstrated in Figure 2c, the number of T cells was
similar in the co-culture and single culture. These findings
underscore the potential of this 3D co-culture system for
quantifying patient-specific cytotoxic effects of immune cells
in CCA organoids. The model could also be a useful tool for
examining the efficacy of new ICIs and predicting which ICI
will be effective for individual patients.
Figure 2: Representative confocal images of Cell Trace Far Red-stained T cells (red), Hoechst 33342-stained CCA2 organoids (blue), and
co-cultures in the presence of a Caspase 3/7-probe (green) at timepoints 0, 90, and 180h (a). Quantification of the dead cell surface (Caspase
3/7 probe-positivity) in the organoid area (Hoechst 33342 labeled) (b) and the number of T cells (c) in nine fields of view of confocal time-lapse
imaging every 6h for 180h. Organoid death is higher in co-cultures with T cells compared to CCA2 organoids alone. The number of T cells
(Cell Trace Far Red labeled) is comparable between solo culture and co-culture. Image credit: Zhou et al., 2022.2
Unlocking the potential of cancer immunotherapy: Overcoming the challenges of 3D immune cell killing assays
www.revvity.com 4
Image cytometry
A high-throughput method to analyze the cytotoxicity
of CAR-modified immune cells in 3D tumor spheroid
models using image cytometry
Image-based cytometry has emerged as a powerful
technique for investigating and characterizing the functions
of CAR-modified immune cells in a high-throughput manner.
Plate-based image cytometry has shown its efficacy
in analyzing various aspects of CAR T-cell therapy, including
transduction efficiency, cell proliferation, and cytotoxicity.
Recent advancements in 3D spheroid models have opened
up new possibilities for image cytometry in the realm
of CAR T-cell therapy, especially for treating solid tumors.
In a recent study conducted by Zurowski and colleagues,
the potential of image cytometry was explored to
characterize the cytotoxicity exhibited by CAR T-cells
targeting the prostate-specific membrane antigen (PSMA)
in a 3D tumor spheroid model.3
PSMA is a transmembrane
glycoprotein known for its expression in healthy prostate
tissue and significant upregulation in cancerous tissue,
making it an attractive target for CAR T-cell therapy.
3D tumor spheroids were formed by seeding antigenexpressing (PC3-PSMA+
GFP+
) or negative control (MCF7-
GFP+
) cells into Nexcelom3D 96-well ultra-low attachment
round-bottom plates. After two days, CAR T-cells and
untransduced (UTD) T cells were introduced at various E:T
ratios (10:1 , 5:1 and 1:1). The co-cultures were then imaged
using a Celigo image cytometer at 0, 24, 48, and 72 hours.
By evaluating the removal of GFP expression, the efficacy
of the effector cells in inducing cytotoxic effects on the
target cells was assessed.
The image analysis revealed that the CAR T-cells exhibited
high cytotoxicity against antigen-expressing spheroids,
while UTD T cells exhibited no noticeable effects. When
the GFP fluorescent intensities were quantified over time,
the researchers observed a time- and E:T ratio-dependent
killing of the spheroids by the CAR T-cells. Notably, high
levels of cytotoxicity were observed at all E:T ratios after
72 hours (10:1 ≅90%, 5:1 ≅89%, and 1:1 ≅78% reduction).
In contrast, no observable cytotoxicity was seen with
UTD T cells or non-antigen-expressing spheroids.
In another study, Sefan Grote and his team investigated
the cytotoxic effects of CAR-modified effector cells that
targeted the immune checkpoint molecule B7-H3 (CD276)
in a 3D tumor spheroid model.4
CD276 is frequently
overexpressed in the majority of solid human tumors, while
its expression is either weak or absent in normal tissues and
cell types, making it an attractive target for CAR-mediated
interventions. Instead of relying on the more conventional
approach of employing T cells as effector cells, the
researchers chose to use the natural killer (NK)-92 cell line
in an attempt to overcome the challenges associated with
producing autologous CAR T-cell products.
After demonstrating the ability of CD276-CAR NK-92 cells
to eliminate NB cells in monolayer cultures, the team seeded
GFP-positive neuroblastoma cells (NB) into Nexcelom
96-well low-attachment U-bottom plates, allowing them
to form spheroids. The study used three high-grade NB
cell lines: Kelly, LAN-1, and LS. After 72 hours, CD276-CAR
NK-92 cells or parental NK-92 cells were introduced into the
3D cultures and fluorescence was measured using a Celigo
image cytometer over a period of 96 hours. CAR-mediated
cytotoxicity was calculated by anaylyzing the fluorescence
intensity of NB spheroids.
Figure 3: CD276-CAR NK-92-mediated lysis of 3D neuroblastoma
spheroids. GFP-transduced neuroblastoma cell lines LAN-1,
Kelly, and LS were grown as 3D spheroids and subsequently
co-incubated with CD276-CAR NK-92 or parental NK-92 cells
for 96 h in at least three individual experiments. Representative
fluorescence images show LAN-1 spheroids (A). Integrated
fluorescence intensity of neuroblastoma (NB) spheroids
(LAN-1, Kelly, LS) was measured regularly using the Celigo S
Imaging Cytometer (Nexcelom), representative fluorescence
images of the NB spheroids are shown after co-incubation
of 96 h (B, C). Image credit: Grote et al. 2020.4
Unlocking the potential of cancer immunotherapy: Overcoming the challenges of 3D immune cell killing assays
www.revvity.com 5
Analysis revealed that the CD276-CAR NK-92 cells were
able to specifically target and eliminate NB cells in the
spheroid model, as shown in Figure 3. Specifically,
all LAN-1 speroids were completely eradicated in less
than 48 hours of CAR exposure. Spheroids established from
the Kelly cell line were almost entirely eliminated within
72 hours, whereas the LS cell line’s spheroids exhibited
partial lysis due to insufficient CAR-mediated cytotoxicity
during the given timeframe. Interestingly, in prior
investigations where the team had screened the cell
lines for expression of known NK cell ligands using flow
cytometry, they found complete absence of the inhibitory
NK ligand HLA-ABC on Kelly cells, mediocre expression on
LAN-1 cells, and elevated HLA-ABC expression on LS cells.
Altogether, these studies highlight the capacity of
plate-based image cytometry as a high-throughput tool
for characterizing CAR-based treatment within a 3D
spheroid model. This approach opens opportunities
to rapidly identify suitable CAR-modified immune cell
candidates for subsequent downstream processes.
In vivo imaging
Evaluating in vivo CAR T-cell toxicity in a mouse model
One of the primary hurdles in CAR T-cell therapies is the
risk of adverse effects caused by on-target, off-tumor
toxicity. This is a concern in patients who exhibit target
antigen expression on both the tumor and healthy tissues.
Although preclinical animal studies serve as useful tools for
testing the efficacy of therapeutic CARs, they are limited in
their ability to accurately identify potential adverse events
in humans, potentially leading to a false sense of safety.
To address this, Castellarin and collaborators established
a mouse model expressing the human Her2 (hHer2) antigen
on both tumor and normal tissue.5
Her2 is an attractive
target for CAR T-cell therapy since it can be overexpressed
40- to 100-fold in human tumors. They then assessed the
antitumor efficacy of high and low-affinity Her2 CAR T-cells
using in vivo imaging.
For the study, mice were engrafted with a tumor xenograft
displaying high Her2 expression and injected with a low
dose of Her2-AAV8 to produce a low Her2-expressing
liver. The Her2+
tumor cells were genetically modified to
emit fluorescence via the IRFP720 fluorescent reporter,
facilitating in vivo imaging. The mice were then infused with
high or low-affinity CAR T-cells and the antitumor efficacy
was assessed. (Figure 4A)
Figure 4: The low-affinity CAR has better tumor control than the
high-affinity CAR when antigen is also expressed in normal tissue.
(A) Overview of the experimental design for comparing Her2+
tumor control between affinity-tuned Her2 CAR T-cells. All mice
received 1.5 × 1010 GCs of Her2-AAV8 and were implanted with
5 × 106
Her2+
SKOV3 tumor cells. Then, three groups were injected
with either 5 × 106
high-affinity (HA) or low-affinity (LA) Her2-CARTs
or no CAR control T cells. (B) The Her2+
tumor cells, SKOV3, were
genetically modified to express the fluorescent reporter, IRFP720,
for in vivo imaging. Tumor xenograft fluorescence is shown
in a yellow-to-red spectrum. Lateral views of fluorescent tumor
imaging. (C) Mean tumor volume ± SEM measured by calipers
in n = 6 mice per group. A 2-way repeated measures ANOVA
with Bonferroni’s multiple comparisons test was used for
statistical analysis. Statistical significance is denoted as *
P < 0.5
and ****P < 0.0001. Image credit: Castellarin et al., 2020.5
Utilizing the IVIS® Spectrum in vivo imaging system, the
researchers found that mice treated with low-affinity CAR
T-cells exhibited significantly better antitumor efficacy
compared to those treated with high-affinity CAR T-cells
(Figure 4B and 4C). To investigate whether differences
in tumor control between the two groups were due to
differences in T-cell abundance and/or trafficking, the
team used in vivo bioluminescent imaging (BLI) to track
the bioluminescence emitted by T cells expressing the
CBR luciferase reporter gene 10 minutes post injection of
IVISbrite™ D-luciferin, using the same IVIS Spectrum. While
there were initial discrepancies in abundance between the
Unlocking the potential of cancer immunotherapy: Overcoming the challenges of 3D immune cell killing assays
www.revvity.com 6
Figure 5: Low-affinity CAR T-cells spend less time off-tumor than high-affinity CAR T-cells. In vivo, CAR T-cell kinetics were captured using
IVIS imaging for n = 6 mice per group. (A) T cells were engineered to express a luciferase gene for in vivo luminescent imaging. The dorsal
views of the mice were kept in the same order as in Figure 4B, and luminescence intensity is shown in a blue-to-red spectrum. In addition
to luciferase expression, the T cells contained either no CAR expression (negative control) or were engineered to express a high-affinity
(HA) or low-affinity (LA) Her2 CAR. (B) Whole body bioluminescent imaging (BLI) of T cell luciferase. Statistical significance for HA-CAR versus
LA-CAR (*) or HA-CAR vs. No CAR (+) was compared by 2-way repeated measures ANOVA with a Tukey’s multiple comparison test. (C)
Spatial luciferase expression was measured along a line that starts in the upper left thorax (point A) and ends in the lower right abdomen
(point B). Luminescence from the spleen, liver, and tumor appears at the beginning (~0–1.5 cm), middle (~1–3 cm), and end (~2.5–4 cm) of the
line, respectively. Mean luminescence along the line was compared between groups by 2-way repeated measures ANOVA with Bonferroni’s
multiple comparisons test. Statistical significance is denoted as **P < 0.01 and ++++P < 0.0001. Image credit: Castellarin et al., 2020.5
high- and low-affinity CAR T-cells, they converged after the
first week and became comparable throughout the rest of
the experiment (Figure 5). Importantly, the low-affinity CAR
T-cells migrated out of the liver to the tumor site faster than
their high-affinity counterparts. This suggests that
low-affinity CAR T-cells exhibit an enhanced ability
to differentiate between low-antigen healthy tissue
and high-antigen tumor tissue, resulting in a better
therapeutic outcome. The study demonstrates the efficacy
of an off-tumor model for evaluating the recognition
of tumors by CAR T-cells. This approach holds promise
for predicting the therapeutic effectiveness of CAR T-cells
compared to tumor xenografts alone.
Unlocking the potential of cancer immunotherapy: Overcoming the challenges of 3D immune cell killing assays
www.revvity.com 7
Conclusion
Immune cell killing assays have emerged as pivotal tools
in immunotherapy research for investigating immune
cell functions and evaluating treatment efficacy. These
assays are not without their challenges, especially
when run in 3D. Technical and logistical hurdles include
consistent and reproducible 3D culture establishment,
accurate quantification of target cell elimination, reliable
measurement of immune cell infiltration, identification
of various cell types, acquisition of kinetic data, and
identification of suitable fluorescent dyes.
These difficulties demand innovative solutions, and this
paper presents a range of successful assay setups that
leverage diverse analytical technologies, including highcontent analysis, flow cytometry, image cytometry, and
in vivo imaging to gain valuable insights into the complex
interplay of immune cells, target tissues, and therapeutic
interventions (Table 1). These innovative techniques open
new avenues for precise assessment, informed therapeutic
design, and the advancement of cancer immunotherapy.
Detection Method Description Benefits Challenges
Image cytometry Quantification of spheroid size
and killing in vitro
• Live cell kinetic assay
• Provides morphology information
• Simple and robust method
• Need GFP recombinant cancer cell
line or appropriate live-cell dyes
High-content analysis Quantification of spheroid size,
killing, and infiltration in vitro
• Live cell kinetic assay
• Multiplex cell type analysis
• Provides morphology and
infiltration information
• Creates movies
• Need appropriate live-cell dyes
• Data volumes
• Imaging depth is limited in large
3D models without tissue clearing
Flow cytometry
Digest tumor samples and measure
the number of viable and apoptotic
cells in the sample
• Quantification of a large
number of cells
• Multiplex cell type analysis
• Tumor infiltration
information available
• No kinetic information
• No morphology
or spatial information
In vivo imaging Quantification of tumor size
in vivo in mouse models
• Non-invasively quantify tumor
size and metastasis
• Analyze on-target cell activity
• Analyze toxicity
to organs/off-target effects
• Mouse models have limited
physiological relevance
• Xenograft models cannot
recapitulate toxicity sufficiently
• Costly models
Table 1. Technology comparison for 3D immune cell killing assays.
Unlocking the potential of cancer immunotherapy: Overcoming the challenges of 3D immune cell killing assays
References
1. Zhang Z, Jiang D, Yang H, He Z, Liu X, Qin W, et al.
Modified car T cells targeting membrane-proximal
epitope of mesothelin enhances the antitumor function
against large solid tumor. Cell Death and Disease.
2019;10(7). doi:10.1038/s41419-019-1711-1
2. Zhou G, Lieshout R, van Tienderen GS, de Ruiter V,
van Royen ME, Boor PP, et al. Modelling immune
cytotoxicity for cholangiocarcinoma with tumour-derived
organoids and effector T cells. British Journal of Cancer.
2022;127(4):649–60. doi:10.1038/s41416-022-01839-x
3. Zurowski D, Patel S, Hui D, Ka M, Hernandez C, Love AC,
et al. High-throughput method to analyze the cytotoxicity
of CAR-T cells in a 3D tumor spheroid model using
image cytometry. SLAS Discovery. 2023;28(3):65–72.
doi:10.1016/j.slasd.2023.01.008
4. Grote S, Chan KC, Baden C, Bösmüller H, Sulyok
M, Frauenfeld L, et al. CD276 as a novel CAR
NK-92 therapeutic target for neuroblastoma.
ADVANCES IN CELL AND GENE THERAPY. 2020;4(1).
doi:10.1002/ acg2.105
5. Castellarin M, Sands C, Da T, Scholler J, Graham K, Buza
E, et al. A rational mouse model to detect on-target,
off-tumor car T cell toxicity. JCI Insight. 2020;5(14).
doi:10.1172/jci.insight.136012
For a complete listing of our global offices, visit www.revvity.com
Copyright ©2023, Revvity, Inc. All rights reserved. 1106671
Revvity, Inc.
940 Winter Street
Waltham, MA 02451 USA
(800) 762-4000
www.revvity.com
Brought to you by
Access this Whitepaper for FREE Now!
Information you provide will be shared with the sponsors for this content. Technology Networks or its sponsors may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.
Experiencing issues viewing the form? Click here to access an alternate version