3D Tumor Models for More Predictive ADC Drug Development
App Note / Case Study
Published: February 24, 2025

Credit: iStock
Antibody–drug conjugates (ADCs) represent a breakthrough in cancer therapy, combining the precision of monoclonal antibodies with powerful chemotherapeutic drugs.
While ADC development is accelerating rapidly, researchers need more sophisticated tools to evaluate ADC efficacy and tumor penetration in models that better reflect the complex tumor microenvironment.
This application note demonstrates a novel workflow for comprehensive ADC evaluation in 3D tumor spheroid models, offering deeper insights into drug function and cellular response.
Download this application note to learn how to:
- Implement a streamlined workflow that combines temporal analysis and high-throughput cytometry
- Measure critical parameters including antibody internalization, cytotoxicity and spheroid composition
- Obtain more translational data through advanced tumor microenvironment modeling
Application Note
Evaluating Antibody Drug Conjugates (ADCs)
In Vitro Using 3D Tumor Spheroid Models
Kirsty McBain, Kalpana Barnes & Nicola Bevan
Sartorius UK Ltd., Units 2 & 3 The Quadrant, Newark Close, Royston, Hertfordshire, SG8 5HL, UK
Abstract
Antibody-drug conjugates (ADCs) combine highly targeted
delivery of chemotherapeutic drug molecules with an
immunotherapeutic intervention for cancer cell killing. A
rapid increase in ADC development in recent years has
created a need for robust and reliable techniques for
assessing novel candidate drugs. The use of 3D advanced cell
models can improve the translational ability of these in vitro
techniques. Here, we present a combined Incucyte® LiveCell Analysis and iQue® High-Throughput Screening (HTS)
by Cytometry workflow for assessing anti-HER2 ADC activity
in single and multi-spheroid models. Incucyte® Live-Cell
Analysis data demonstrated greater induction of cytotoxicity
by anti-HER2 ADCs (Kadcyla® and Enhertu®) compared to
the monoclonal antibody (mAb) backbone on which they
were based (Trastuzumab). iQue® HTS analysis revealed the
unique Enhertu® bystander activity, by allowing the cellular
composition of a spheroid co-culture to be measured. These
techniques facilitate in-depth analysis of ADC activity and
allow mechanistic differences between ADCs to be unpicked.
February, 2024
Key words or phrases:
Antibody-drug conjugates (ADCs), Incucyte® Live-Cell
Analysis, iQue® High-Throughput Screening (HTS) by
Cytometry, mechanism of action (MoA), spheroids, 3D
advanced cell models, cytotoxicity, internalization
2
Introduction
ADCs are a unity between highly specific mAbs and
cytotoxic chemotherapeutic drugs. This combination
creates a powerful therapeutic for the treatment of several
types of cancer: both solid tumors and hematological
malignancies. Over the last few years, research and
development of ADCs has accelerated, as evidenced by a
35% increase in clinical trials and a 90% increase in phase 1
clinical trials investigating ADCs in 2022 compared to in
2021.1 The two main targets for ADC development at this
time, HER2 and TROP2, are overexpressed on breast
cancers and comprise 20% of all ADC studies.1
ADCs have three main components: the targeting mAb,
the cytotoxic payload, and a stable linker between them.
Each element adds an extra layer of tumor targeting
specificity to the ADC, on top of the highly specific
delivery of the payload to the tumor cell following mAb
binding. For example, the linker is highly stable prior to
internalization of the antibody into the target cell, which
ensures the payload is not released elsewhere in the body.
ADC linkers can be categorized into cleavable or noncleavable types and are either severed by enzymatic
activity of proteases or are degraded in the acidic
lysosome.2 Another factor contributing to ADC specificity
is the mechanism of action (MoA) of the cytotoxic
payload. Since these are typically chemotherapeutic
drugs, their MoAs act preferentially towards rapidly
proliferating cancer cells, for example by inhibiting
microtubule polymerization or inducing DNA damage.3-5
Recent years have seen a shift in the choice of in vitro
models utilized for oncology research. The 2D cell
monolayer has long been used due to its simplicity, costeffectiveness, and scalability, however it can lack some
critical features of the 3D tumor microenvironment (TME).
For this reason, many researchers now use 3D models, such
as spheroids or organoids, to assess drug effects in vitro.
These models provide a much closer reflection of the TME,
with more complex cell-cell interactions and the option to
include extracellular matrix (ECM) proteins. The tumor cells
also grow in a more layered structure, with the outer layer
undergoing fast proliferation and interaction with the TME,
followed by a middle quiescent layer and an inner necrotic
core.6,7 This facilitates a more representative modelling of
drug behavior in vivo, for example by allowing the
comparison of tumor penetration of candidate drugs.
Traditional methods for measuring drug response in 3D
models are often limited because they:
1. Require multiple workflows for quantification of different
parameters, often using multiple instruments
2. Involve lengthy, time-consuming protocols, which
require multiple rounds of optimization, fixation, and
repetitive washes
3. Necessitate correlation of data from several different
assays for each treatment condition, increasing the risk
of data variability
4. Provide bulk measurement of cytotoxicity without a
deeper investigation into spheroid cell type composition.
In this application note, we present a combined Incucyte®
Live-Cell Analysis and iQue® HTS by Cytometry approach
for quantifying the function of two Trastuzumab (antiHER2) based ADCs: Trastuzumab emtansine (Kadcyla®)
and Trastuzumab deruxtecan (Enhertu®), using both single
and multi-spheroid 3D models. The Incucyte® Live-Cell
Analysis System captures temporal information on mAb
internalization and target cell death by quantifying spheroid
size and fluorescence intensity. The iQue® 3 Cytometry
Instrument measures target cell counts and viability after
spheroid dissociation, enabling a closer look at different
populations within a co-culture.
Cell Culture and Maintenance
Antibodies
Three anti-HER2-hIgG1 antibodies were characterized: a
Trastuzumab biosimilar (Absolute Antibody); Kadcyla®
(Trastuzumab emtansine; a therapeutic-grade ADC based on
Trastuzumab and the chemotherapy drug emtansine (also
known as DM1), Midwinter Solutions) and Enhertu®
(Trastuzumab deruxtecan, a therapeutic-grade ADC based
on Trastuzumab and the chemotherapy drug deruxtecan (or
DXd), Midwinter Solutions). An Anti-β-Gal-hIgG1 mAb from
InvivoGen was used as an isotype control.
Cell Lines
BT474 cells (from a breast cancer cell line) or SKOV-3 cells
(from an ovarian cancer cell line) were used as high HER2
expressing cell types. MDA-MB-231 cells, from a breast
cancer cell line, express low levels of HER2 and were used
as a negative control throughout.
Antibody Internalization
Target cells were seeded in an Ultra-Low Attachment (ULA)
96-well microplate (Corning® 7007) for 72 hours to promote
3
spheroid formation. Antibodies, labeled with Incucyte®
Human Fabfluor-pH Orange Antibody Labeling Dye
(Sartorius 4812), were then added to spheroids. Phase and
fluorescence images (10X) were captured every 15 minutes
using the Incucyte® Live-Cell Analysis System.
Internalization was quantified as an increase in Total Orange
Area (µm2 /image).
Single Spheroid ADC Cytotoxicity
Target cells (transduced with Incucyte® Cytolight Green
Lentivirus for stable expression of nuclear restricted GFP)
were seeded in a ULA plate for 72 hours to promote
spheroid formation. Phase and fluorescence images (10X)
were captured using the Incucyte® Live-Cell Analysis
System on a 3-hour repeating scan schedule for 9 days.
Cell death was quantified as a reduction in spheroid area.
Multi-spheroid ADC Cytotoxicity
A flat bottom 96-well plate (Corning® 3595) was coated
with a layer of Matrigel® (5 mg/mL) as per a validated
Incucyte® protocol (Incucyte® Multi-Spheroid Assay for
the Quantification of Multi-Spheroid Growth and Health
on a Layer of Matrigel®).8 Incucyte® Nuclight Green
Lentivirus labeled target cells (2 K/well) were seeded on
top and incubated for 72 hours to promote multi-spheroid
formation. Antibodies were added (2 µg/mL) and cells
were monitored using the Incucyte® Live-Cell Analysis
System via a repeating scan schedule (every 3 hours) for
10 days. Images were quantified for a Brightfield Green
Integrated Intensity (GCU x µm2/image) over time as an
indicator of cell death.
Single Spheroid Antibody-Dependent Cellular
Cytotoxicity (ADCC)
Target cells labeled with Incucyte® Cytolight or Nuclight
Green Lentivirus were seeded in a ULA plate (4 K/well) for
72 hours to promote spheroid formation. As MDA-MB-231
cells require Matrigel® to form tight spheroids, Matrigel®
was added to culture media at a final concentration of 2%.
Matrigel® was added to both target cell types to ensure
differences in diffusion didn’t impact ADCC. Test antibodies
were added at a range of concentrations. Natural killer (NK)
cells were added (16 K/well) alongside IL-12 (10 ng/mL) to
improve their longevity. Phase and fluorescence images (4X
objective) were captured using the Incucyte® Live-Cell
Analysis System on a 3-hour repeating scan schedule for 10
days. Cell death was quantified as a reduction in spheroid
green mean intensity.
Single Spheroid Bystander Activity
Single spheroids were formed by mixing high HER2
expressing BT474 cells (labeled with Incucyte® Cytolight
Green Lentivirus) and unlabeled, low HER2 expressing
MDA-MB-231 cells at a 2:3 ratio. Antibodies were added
after 72 hours and images were captured on a repeat
scanning schedule (every 3 hours) using the Incucyte®
Live-Cell Analysis System. On day 8, the spheroids were
dissociated (using a previously validated protocol, T Cell
Killing in Single Spheroids) and cells were labeled using
the iQue® 3 Cell Membrane Integrity (R/Red) Dye.9 This
enabled live cell counts of each cell type to be quantified
using the iQue® HTS by Cytometry Platform.
Results
The described experiments have utilized both single and
multi-spheroid tumor models to profile the function of
three antibodies: Trastuzumab, Kadcyla®, and Enhertu®.
Trastuzumab is an anti-HER2 mAb therapeutic, whilst
Kadcyla® and Enhertu® are ADCs which contain a
Trastuzumab backbone. Due to this commonality in
structure, we expect the antibodies to share many
functional capabilities, however, they do also include
unique structural features which may distinguish their
anti-tumor function (Figure 1). One difference is in the
cytotoxic payload included on each ADC, with Kadcyla®
including chemotherapy drug emtansine (DM1) whilst
Enhertu® is linked to the payload deruxtecan (Dxd). Each
ADC also differs in the number and positions of the
payload conjugation sites, with DM1 binding to lysine
residues in the Trastuzumab backbone in Kadcyla® at an
average of 3.5 conjugations per antibody, whilst binding of
Dxd in Enhertu® is via thioether bonds with cysteine
residues, with 8 conjugations per molecule. 10,11 The
cytotoxic payload in the ADCs also has different linker
chemistries, with Kadcyla® containing a non-cleavable
amine-to-sulfhydryl crosslinker (SMCC) and Enhertu®
containing a valine-citrulline cleavable linker.12
4
Figure 2. Internalization of ADCs into BT474 spheroids is greater than internalization of Trastuzumab.
BT474 cells (4 K/well) were seeded into ultra-low attachment (ULA) plates for 72 hours to promote spheroid formation. Antibodies were labeled
with Incucyte® Human Fabfluor-pH Orange Antibody Labeling Dye then added to spheroids (n=3). Phase and fluorescence images (10X objective)
were captured every 15 minutes using the Incucyte® Live-Cell Analysis System. (A) Representative Incucyte® images of single spheroids taken 48
hours after antibody addition. (B) Internalization was quantified as an increase in Spheroid Orange Mean Intensity (OCU) and plotted over time.
Results
Figure 1. Structures of anti-HER2 antibodies and ADCs
Trastuzumab is an anti-HER2-IgG1 mAb. Kadcyla® is a modified version of Trastuzumab which includes non-cleavable linkers to the chemotherapy
drug emtansine (DM1). Enhertu® is also an adapted version of Trastuzumab, with cleavable linkers to attach the cytotoxic payload, deruxtecan (Dxd).
Trastuzumab Kadcyla® Enhertu®
Variable region
Trastuzumab
(monoclonal
antibody)
Trastuzumab Trastuzumab
Fc region
Non-cleavable
linker
DM1
(cytotoxic agent)
Cleavable linker
Dxd
(cytotoxic agent)
Antibody Internalization
Efficient antibody internalization is critical for the delivery of
the ADC cytotoxic payload into target cells. Initial
experiments aimed to quantify the relative level of
internalization of each of the antibodies into single
spheroids formed from high HER2 expressing cells
(BT474s). The antibodies were pre-labeled with Incucyte®
Human Fabfluor-pH Orange Antibody Labeling Dye, which
fluoresces upon internalization into the acidic lysosomal
and endosomal pathways. Incucyte® images were captured
over time and an increase in internalization was quantified
as an increase in spheroid orange mean intensity.
Phase and fluorescence images (Figure 2A) clearly show
that fluorescence, and therefore internalization, in the
presence of all three anti-HER2 antibodies was much
higher than the IgG control. The time course graph (Figure
2B) provides a closer examination of the differences in
internalization between the anti-HER2 antibodies and
indicates considerably higher internalization of the two
ADCs (Kadcyla® and Enhertu®) compared to the backbone
antibody (Trastuzumab), with endpoint Orange Calibrated
Units (OCU) intensity values of 31.8, 27.0 and 17.7,
respectively. These data are in line with results generated
previously for internalization of these antibodies in a 2D
monolayer assay format (data available in Application Note:
Cross-Platform Analysis of the Binding and Function of
Anti-HER2 Antibody-Drug Conjugates (ADCs)).13 Spheroid Orange Mean Intensity (OCU)
40
10
20
30
0
0 12 24 36 48
A. B.
Time (h)
No Antibody
IgG Control
Trastuzumab
Enhertu®
Kadcyla®
Phase and
Fluorescence
Fluorescence
Only
No Antibody IgG Control Trastuzumab Kadcyla® Enhertu®
5
Figure 3. ADCs induced a concentration dependent increase in cytotoxicity of BT474 cells in a 3D single spheroid model.
BT474 cells (1 K/well) were seeded in ULA plates for 72 hours to promote spheroid formation. Antibodies were added to spheroids at a range of
concentrations (n=3). Incucyte® images were captured on a 3-hour repeating scan schedule for 9 days. Time-course data for change in Largest
Object Area (µm2) for spheroids incubated with (A) IgG control, (B) Trastuzumab, (C) Kadcyla® and (D) Enhertu®.
Single Spheroid ADC Cytotoxicity
Following internalization of ADCs into target antigenexpressing cells, the chemotherapeutic drug payload is
released, either through proteolytic degradation of the
linker or by its metabolism in the acidic lysosome. This
means the drug molecule can exert a cytotoxic effect,
specifically towards the target cell. This reduces off-target
cytotoxicity compared to administration of the
chemotherapeutic drug alone. The mechanism of this
cytotoxic effect can vary depending on the drug used. For
example, the DM1 in Kadcyla® acts to inhibit microtubule
assembly, whilst the Dxd payload in Enhertu® disrupts DNA
replication through inhibition of topoisomerase I.14,15 To
explore this cytotoxicity in vitro, a range of concentrations
of antibody were tested with a monoculture of BT474 cells
in a 3D single spheroid format. Reduction in spheroid size
over time was quantified using the Incucyte® Live-Cell
Analysis System and was used as a measure of cell death.
Both ADCs induced a concentration dependent reduction
in spheroid area over time, indicating induction of
cytotoxicity (Figures 3C and 3D). The effect of Kadcyla®
varied much more across the concentration range tested,
with average spheroid size at endpoint of 0.92 x 105 µm2 at
the highest concentration and 1.91 x 105 µm2 at the lowest
concentration. Contrastingly, the sensitivity to varying
concentrations of Enhertu® was much lower with the
spheroid area ranging only from 1.15 x 105 µm2 ¬(25 µg/mL)
to 1.68 x 105 µm2 (0.39 µg/mL). Only the highest
concentration of Trastuzumab (25 µg/mL) had an impact on
spheroid size (Figure 3B). Clearly this is not down to the
action of chemotherapeutic drugs and instead may be the
result of Trastuzumab’s other mechanisms of action,
including inhibition of cell signaling, for example in the
PI3K-AKT pathway.17 Maintenance of this activity in the
Kadcyla® ADC may explain the much larger reduction in
spheroid area at the highest Kadcyla® concentration
compared to the second highest concentration tested (13
µg/mL). Across the concentration range tested, the IgG
control had no effect on spheroid size (Figure 3A).
Comparing the data in Figures 2 and 3, there is a clear
difference in the time taken for internalization compared to
the cytotoxic response. Internalization begins rapidly within
the first 24 hours and starts to plateau between 24-48
hours. In contrast, with the cytotoxic effect, there is minimal
difference between the conditions until after the 48 hours
when the concentration dependent response starts to
materialize. This time course could reflect the MoA of the
ADCs in that they need to be internalized into the target
cells first before the cytotoxic payload can be cleaved and
start to induce cytotoxicity.
0 0
0 0
96 96
96 96
144 144
144 144
192 192
192 192
240 240
240 240
48 48
48 48
Time (h) Time (h)
Time (h) Time (h)
Spheroid Area x105(µm2
)
Spheroid Area x105(µm2
)
Spheroid Area x105(µm2
)
Spheroid Area x105(µm2
)
2.5 2.5
2.5 2.5
1.5 1.5
1.5 1.5
2.0 2.0
2.0 2.0
1.0 1.0
1.0 1.0
A. B.
C. D.
0.39 µg/mL
0.78
1.6
3.1
6.3
13
25
IgG Control
Enhertu® Kadcyla®
Trastuzumab
6
Figure 4. ADCs induce death of HER2 expressing SKOV-3 cells in a multi-spheroid model.
Incucyte® Nuclight Green Lentivirus labeled SKOV-3 and MDA-MB-231 cells (2 K/well) were seeded on a layer of Matrigel® (5 mg/mL) to promote
multi-spheroid formation. Antibodies were added (2 µg/mL) and cells were monitored using the Incucyte® Live-Cell Analysis System via a
repeating scan schedule (every 3 hours) for 10 days. (A) Representative images of SKOV-3 multi-spheroids from day 9. Cell death in (B) SKOV-3
and (C) MDA-MB-231 spheroids was quantified as a loss of fluorescence intensity of spheroids over time.
Multi-spheroid ADC Cytotoxicity
Multi-spheroid tumour models can also be utilized to test
the activity of ADCs in conditions more representative of
the in vivo tumor environment. Adding further 3D
complexity with increased cellular and ECM interactions,
this model may bring us closer to a translational model for
oncology drug research. This involved coating plates with
a layer of Matrigel® before seeding the target cells (high
HER2 expressing SKOV-3 or low HER2 expressing MDAMB-231 cells) on top. After 72 hours of multi-spheroid
formation, antibodies were added at a single
concentration (2 µg/mL). Images were captured using the
Incucyte® Live-Cell Analysis System with the multispheroid scan type and analysis.
The images in Figure 4A show the difference between
SKOV-3 spheroids in the presence of each antibody on day
9. Compared to the IgG control, there was a visible
reduction in the number and size of spheroids in the
presence of each of the antibody treatments. A mask was
applied which allowed quantification of death as a loss of
fluorescence intensity within the spheroid brightfield
object. Figure 4B shows that the ADCs induced a high level
of death in the SKOV-3 spheroids, with a 63.9 and 73.7 %
reduction in spheroid intensity at assay endpoint
compared to the IgG control for Kadcyla® and Enhertu®,
respectively. Trastuzumab also induced considerable cell
death, but at a slightly reduced level compared to the
ADCs (49.7% reduction from IgG control). Overall, the cell
death at this concentration of antibody is considerably
higher than was seen in the single spheroid model, which is
likely due to differences in cell type and spheroid size.
Figure 4C shows that this is an antigen positive cell type
specific effect as there was no impact on HER2-low MDAMB-231 spheroids.
Brightfield Green Integrated Intensity
x 106(GCU x µm2/Image)
Brightfield Green Integrated Intensity
x 106(GCU x µm2/Image)
2.5 8
0.5
2
1.0
4
1.5
2.0
6
0.0 0
0 48 96 144 192 240 0 48 96 144 192 240
B. C.
A.
Time (h) Time (h)
IgG Control
Trastuzumab
Enhertu®
Kadcyla®
IgG Control
SKOV-3 MDA-MB-231
Enhertu® Kadcyla® Trastuzumab
7
Figure 5. Induction of NK-mediated ADCC activity is much greater with high HER2 expressing targets in the presence of Kadcyla®.
Target cells (4 K/well) were MDA-MB-231 (HER2-low) or BT474 (HER2-high) labeled with Incucyte® Nuclight or Cytolight Green Lentivirus,
respectively. Matrigel® was added (2%) to aid tight spheroid formation. After 72 hours, cells were mixed with test antibodies and natural killer (NK)
cells (20 K/well). Phase and fluorescence images (4X) were captured using the Incucyte® Live-Cell Analysis System on a 3-hour repeating scan
schedule for 10 days for spheroid green mean intensity (OCU). (A) Temporal microplate graph of intensity of BT474 spheroids. Outliers are
removed in grey. (B) and (C) Bar charts for intensity of spheroids formed from BT474 and MDA-MB-231 targets. Spheroid Green Mean Intensity (OCU) Spheroid Green Mean Intensity (OCU)
150 15
50 5
100 10
0 0
0.08 0.31 1.3 5 0.08 0.31 1.3 5
B. C.
Time (h) Time (h)
BT474 MDA-MB-231
Single Spheroid ADCC
A key MoA of Trastuzumab in the killing of HER2 expressing
cancers is ADCC.17 It is expected that ADCs based on
Trastuzumab would retain this killing function on top of the
payload-induced cytotoxicity. To investigate this, NK cells,
which are the main cell type involved in ADCC, were added
to the single spheroid model. Incucyte® images were
captured on a repeating schedule and cell death was
quantified as a reduction in spheroid intensity over time.
This metric was chosen as it allowed us to distinguish
differences between both antibodies and concentrations
in terms of induction of spheroid death.
The temporal microplate graph (Figure 5A) shows the
green intensity of spheroids formed from high HER2
expressing, green-labeled BT474 spheroids over time. The
data highlights a much larger decrease in spheroid green
intensity over time in the presence of the anti-HER2
antibodies, compared with the IgG control. This, combined
with the bar chart in Figure 5B, shows that Kadcyla®
induced the most cell death, followed by Trastuzumab,
then Enhertu®. This contrasts with what was observed in
the absence of NK cells (Figure 3), when the Enhertu®
induced greater cytotoxicity than Trastuzumab. This may
suggest that NK-mediated ADCC has a greater overall
cytotoxic effect than ADC-induced cytotoxicity in this
model, and that this effect is slightly attenuated in the
Enhertu® antibody, perhaps due to the addition of the
payload molecules to the Fc region. The antibodies had
minimal effect on the intensity of the HER2-low MDAMB-231 spheroids (Figure 5C).
A.
No Antibody
IgG Control
189.78
A
0
189.78
B
0
189.78
C
0
189.78
D
0
189.78
E
0
189.78
F
0
189.78
G
0
189.78
H
0
10d 10d 10d 10d 10d 10d10d 10d 10d 10d 10d 10d 10d 10d
1 2 3 4 5 6 7 8 9 10 11 12
Trastuzumab Kadcyla® Enhertu®
Antibody
Green Mean Intensity
IgG Control
Trastuzumab
Kadcyla®
Enhertu®
8
Single Spheroid Bystander Activity
In 2022, Enhertu® was the first HER2-directed therapy to
be approved for treatment of patients with low-HER2
expressing breast cancer.18 The reason it has been given this
additional indication (alongside treatment for high HER2-
expressing breast cancers) is its potential to exert a unique
‘bystander’ effect. This activity is thought to be facilitated
by the high membrane permeability of the Dxd payload,
meaning that once the ADC has killed a HER2 expressing
cell, the payload can then be released and diffuse easily
into neighboring cells, regardless of their HER2 expression
and result in their death.19
To examine this in vitro using a 3D single spheroid model,
spheroids were first formed from a mixture of of high
HER2 expressing BT474 cells (labeled with Incucyte®
Cytolight Green Lentivirus) and low HER2 expressing,
unlabeled MDA-MB-231 cells. The cells were mixed at a
2:3 BT474 to MDA-MB-231 ratio. Incucyte® images were
captured every 3 hours for 8 days and a reduction in
spheroid area used to quantify cell death (Figure 6A). This
showed that spheroid area was reduced only in the
presence of Enhertu® and remained comparable in the
presence of the other three antibodies.
However, the Incucyte® Live-Cell Analysis data alone
lacked information on the growth of the individual cell
types in the co-culture. To determine this, at assay
endpoint, the spheroids were dissociated to create a single
cell suspension. Cells were then labeled using iQue® 3 Cell
Membrane Integrity (R/Red) Dye and analyzed for the
green (BT474) and unlabeled (MDA-MB-231) live cell
counts using the iQue® HTS by Cytometry Platform. The
data in Figure 6B show that, as in each of the previous
assays, all three of the anti-HER2 antibodies caused a
reduction in the number of high HER2-expressing BT474
cells. The percentage cell death compared to IgG control
was again greater for the two ADCs, at 96.4% and 97.2%,
respectively, compared to Trastuzumab, which induced
46.4% cell death.
Figure 6B shows that, unlike in the monoculture assays,
there has been considerable death of the HER2-low cell
type in the presence of Enhertu® when the cells are cocultured with the high HER2 expressing cells. This
provides evidence that the bystander activity of Enhertu®
can be observed using this in vitro model. These data also
show that there has been an increase in the number of
MDA-MB-231 cells compared to the IgG control in the
wells treated with Kadcyla®. This suggests that as Kadcyla®
induces cytotoxicity of the BT474 cells, the MDA-MB-231
cells have more space and/or nutrients to grow. This could
explain why the Incucyte® Live-Cell Analysis
quantification of spheroid area remains constant unless
the antibody has induced death of both cell types in the
co-culture (as seen with Enhertu®).
This demonstrates that, although we can measure the
bulk reduction in spheroid area using the Incucyte®
Live-Cell Analysis System (as in Figure 6A), we get little
information on the cellular composition of the spheroid.
Using the Incucyte® Live-Cell Analysis System to gather
temporal information and then dissociating spheroids
for iQue® HTS Cytometry analysis allows quantification
of the different cell types comprising the spheroid. This
in turn can provide information on differential MoAs of
test antibodies.
Figure 6. Bystander killing of low HER2 expressing cells is only seen with the Enhertu® ADC.
Single spheroids were formed from a 2:3 ratio of high HER2 expressing BT474 cells (labeled with Incucyte® Cytolight Green Lentivirus) to low HER2
expressing, unlabeled MDA-MB-231 cells. Antibodies were added after 72 hours. Images were captured on a repeat scanning schedule (every 3
hours) using the Incucyte® Live-Cell Analysis System. On day 8, spheroids were dissociated and cells labeled using the iQue® 3 Cell Membrane
Integrity (R/Red) Dye. Live cell counts were quantified using the iQue® 3 Platform. (A) Cell death was quantified from Incucyte® images as a
reduction in spheroid area over time. Bar graphs show viable cell counts from the iQue® 3 Platform of (B) BT474 cells and (C) MDA-MB-231 cells. Spheroid Area [x106] (µm2
)
1.0
0.4
0.2
0.8
0.6
0.0
0 48 96 144 192
A.
Time (h)
IgG Control
Enhertu®
Kadcyla®
Trastuzumab
Live BT474 Counts
1500
500
1000
0
B.
IgG
Control
Enhertu® Kadcyla® Trastuzumab
Live MDA-MB-231 Counts
4000
1000
2000
3000
0
C.
IgG
Control
Enhertu® Kadcyla® Trastuzumab
9
Conclusions
These data describe the use of the Incucyte® Live-Cell
Analysis System and iQue® HTS by Cytometry Platform
to profile the in vitro function of ADCs. Incucyte® assays
quantified antibody internalization and cytotoxicity over
time, whilst dissociated spheroids could be assessed
using the iQue® 3 Cytometry Instrument to reveal
differences in spheroid composition. The advantages of
this workflow include:
1. 3D single and multi-spheroid models provide a closer
reflection of the TME, for example due to more
complex cell-cell and cell-ECM interactions. This can
mean a more translational model for drug
development.
2. The Incucyte® Live-Cell Analysis System provides visual
and temporal analysis of spheroids over time, with easy
quantification of metrics such as spheroid size and
fluorescence intensity allowing antibody internalization
and cytotoxicity to be measured over time.
3. The iQue® HTS by Cytometry facilitates highthroughput analysis of dissociated cells, with the ability
to distinguish individual cell types within a mixture. This
can reveal mechanistic information on a drug’s activity.
4. Mix and read reagents combined with validated
protocols make experimentation simple and
streamlined.
5. This workflow and the described advantages allow a
comprehensive assessment of the function of ADCs in
3D models and can enhance drug discovery and
biological research applications.
This workflow and the described advantages allow a
comprehensive assessment of the function of ADCs in
3D models and can enhance drug discovery and
biological research applications.
References
1. The Clinical Landscape Of ADCs In 2023 Diverse Technologies Narrow Target. (2023) Available at: https://www.
clinicalleader.com/doc/the-clinical-landscape-of-adcs-in-diverse-technologies-narrow-target-0001.
2. Hafeez, U. et al. (2020)“Antibody–drug conjugates for cancer therapy,” Molecules, 25(20), p. 4764.
3. Ponziani, S. et al. (2020) “Antibody-drug conjugates: The New Frontier of chemotherapy,” International Journal of
Molecular Sciences, 21(15), p. 5510.
4. Dumontet, C. and Jordan, M.A. (2010) “Microtubule-binding agents: A dynamic field of cancer therapeutics,” Nature
Reviews Drug Discovery, 9(10), p. 790–803.
5. Reuvers, T.G., Kanaar, R. and Nonnekens, J. (2020) “DNA damage-inducing anticancer therapies: From global to precision
damage,” Cancers, 12(8), p. 2098.
6. Habanjar, O. et al. (2021) ‘3D cell culture systems: Tumor application, advantages, and disadvantages’, International Journal
of Molecular Sciences, 22(22), p. 12200.
7. Zanoni, M. et al. (2016) ‘3D tumor spheroid models for in vitro therapeutic screening: A systematic approach to enhance
the biological relevance of data obtained’, Scientific Reports, 6(1). doi:10.1038/srep19103.
8. Sartorius. (2020) Incucyte® Multi-Spheroid Assay for the Quantification of Multi-Spheroid Growth and Health on a Layer
of Matrigel®
9. Sartorius. T Cell Killing in Single Spheroids Protocol
10. Chen, L. et al. (2016) “In-depth structural characterization of Kadcyla® (ADO-trastuzumab emtansine) and its biosimilar
candidate,” mAbs, 8(7), p. 1210–1223.
11. Agency Committee for Medicinal Products for Human Use (CHMP), E.M. (2020) “CHMP assessment report: Enhertu,”.
12. Kenney, D.J. (2022) Taking aim with antibody-drug conjugates, Taking Aim with Antibody-Drug Conjugates. Available at:
https://www.antibody.com/news/taking-aim-with-antibody-drug-conjugates.
13. Sartorius. (2023) Cross-Platform Analysis of the Binding and Function of Anti-HER2 Antibody- Drug Conjugates (ADCs)
14. Peddi, P.F. and Hurvitz, S.A. (2013) ‘Trastuzumab emtansine: The first targeted chemotherapy for treatment of breast
cancer’, Future Oncology, 9(3), pp. 319–326.
(References continue on next page)
10
Specifications subject to change without notice. ©2024 All rights reserved. All names of Sartorius products are registered trademarks and the property of Sartorius AG and/or one of its affiliated companies.
AB-Drug Conjugates-Using-Spheroids-App-Note-2402-en-L-Sartorius Status: 02|2024
References (continued from previous page)
15. Iwata, T.N. et al. (2023) Data from a HER2-targeting antibody–drug conjugate, trastuzumab Deruxtecan (DS-8201A),
enhances antitumor immunity in a mouse model [Preprint].
16. Ewesuedo, R.B. and Ratain, M.J. (1997) ‘Topoisomerase I inhibitors’, The Oncologist, 2(6), pp. 359–364.
17. Valabrega, G., Montemurro, F. and Aglietta, M. (2007) ‘Trastuzumab: Mechanism of action, resistance and future
perspectives in her2-overexpressing breast cancer’, Annals of Oncology, 18(6), pp. 977–984.
18. Center for Drug Evaluation and Research (no date) FDA approves fam-trastuzumab deruxtecan-nxki for HER2-low breast
cancer, U.S. Food and Drug Administration. FDA. Available at: https://www.fda.gov/drugs/resources-information-approveddrugs/fda-approves-fam-trastuzumab-deruxtecan-nxki-her2-low-breast-cancer.
19. Ogitani, Y. et al. (2016) “Bystander killing effect of ds-8201A, a novel anti-human epidermal growth factor receptor 2
antibody–drug conjugate, in tumors with human epidermal growth factor receptor 2 heterogeneity,” Cancer Science,
107(7), p. 1039–1046.
North America
Phone +1 734 769 1600
Europe
Phone +44 1763 227400
Email:
Japan
Phone +81 3 6478 5202
China
Phone +86 21 6878 2300
Rest of Asia Pacific and other
countries around the world:
Phone +65 6872 3966
Brought to you by

Download the Application Note for FREE Now!
*This also covers the processing of my personal data provided above for these purposes, including profile building to better tailor the communication to my personal interests based on my previous and predicted choices. Learn More
You can withdraw your consent at any time with effect for the future. This does not affect the lawfulness of the processing up to the time of withdrawal.
Further information on the processing of personal data can be found in the Sartorius Privacy Notice.
Technology Networks may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.
You can withdraw your consent at any time with effect for the future. This does not affect the lawfulness of the processing up to the time of withdrawal.
Further information on the processing of personal data can be found in the Sartorius Privacy Notice.
Technology Networks 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