Immuno-Phenotyping: Methods and Protocols
Immuno-Phenotyping: Methods and Protocols
J. Philip McCoy, Jr
Editor
Immuno-
phenotyping
Methods and Protocols
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, UK
Edited by
J. Philip McCoy, Jr
Frederick, MD, USA
Editor
J. Philip McCoy, Jr
Frederick, MD, USA
This Humana imprint is published by the registered company Springer Science+Business Media, LLC, part of Springer
Nature.
The registered company address is: 233 Spring Street, New York, NY 10013, U.S.A.
Preface
Immunophenotyping has become more complex in recent years due to the increasing
availability of new fluorochromes with novel spectral characteristics and the development
of instrumentation with expanded capabilities. Propelled by technological advances in
cytometry as well as the development of commercially available monoclonal antibodies,
immunophenotyping of a multitude of cell types using flow cytometry has become com-
monplace in virtually all academic research centers, pharmaceutical companies, and clinical
laboratories. Thirty-five to forty years ago, immunophenotyping was limited to one or two
colors while today it is possible to construct panels consisting of several dozen fluorescent-
conjugated markers. Similarly, analysis of flow cytometric immunophenotyping has devel-
oped beyond real time, direct observation of electronic signals represented by one- or
two-parameter histograms viewed at the cytometer. Today, data acquired in list mode fcs
format files allows for remote, retrospective, highly complex, and sometimes automated,
data analysis performed independent of the cytometer and shared beyond the laboratory.
Additionally, newer cytometric technologies such as imaging flow cytometry and mass
cytometry permit immunophenotyping in conjunction with morphologic studies or with
an expanded number of parameters, respectively. The following chapters will present a
number of these topics as well as newer developments in the field of immunophenotyping.
Today, it is virtually impossible to present a complete and comprehensive compendium
of all immunophenotyping methods and applications in one volume. Therefore, this text will
present a representative collection of immunophenotypic methods and applications with
examples of newer technologies and reagents used in the research and clinical environments.
Basic methods in immunophenotyping such as construction of high-dimensional fluores-
cence and mass cytometry panels; fluorescence barcoding; and use of dried or lyophilized
reagents will be presented first, followed by immunophenotyping examples of specific cell
types often studied in research laboratories. The final chapters present immunophenotyping
topics useful in the clinical laboratory, including a chapter on the critical role of quality
control and immunophenotyping in the clinical environment.
I would like to thank each of the authors for volunteering to take the time to share their
expertise for this volume. Without their hard work and dedication, it would not have been
possible to share these methods with you.
v
Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
1 High-Dimensional Immunophenotyping with Fluorescence-Based
Cytometry: A Practical Guidebook . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Florian Mair and Aaron J. Tyznik
2 Immunophenotyping by Mass Cytometry. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
Gregory K. Behbehani
3 Fluorescent Cell Barcoding for Immunophenotyping. . . . . . . . . . . . . . . . . . . . . . . . 53
Valentina Giudice, Giovanna Fantoni, and Angélique Biancotto
4 Immunophenotyping Using Dried and Lyophilized Reagents . . . . . . . . . . . . . . . . 69
Marc Langweiler
5 Guidelines for Gating Flow Cytometry Data for Immunological Assays. . . . . . . . 81
Janet Staats, Anagha Divekar, J. Philip McCoy, Jr, and Holden T. Maecker
6 Strategies and Techniques for NK Cell Phenotyping. . . . . . . . . . . . . . . . . . . . . . . . . 105
Chen Ziqing, Andreas Lundqvist, and Kristina Witt
7 Immunophenotyping of Human B Lymphocytes in Blood
and in Adipose Tissue . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 115
Alain Diaz, Maria Romero, Daniela Frasca, and Bonnie B. Blomberg
8 Multiparametric Flow Cytometry Analysis of Naı̈ve, Memory,
and Effector T Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 129
Ankit Saxena, Pradeep K. Dagur, and Angélique Biancotto
9 Immunophenotyping of Human Regulatory T Cells . . . . . . . . . . . . . . . . . . . . . . . . 141
Janet Staats
10 Immunophenotyping of Human Innate Lymphoid Cells . . . . . . . . . . . . . . . . . . . . 179
Sara Trabanelli, Alejandra Gomez-Cadena, and Camilla Jandus
11 Enumeration of Plasmacytoid Dendritic Cells in Peripheral Blood
and Bone Marrow by Flow-Cytometric Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . 193
Abdullah Alsuwaidan, Franklin Fuda, Weina Chen, and Mingyi Chen
12 Immunophenotyping of Circulating Endothelial Cells
and Endothelial Microparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 203
Nicholas Wanner and Kewal Asosingh
13 Rare Event Phenotyping and Molecular Characterization:
Circulating Tumor Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 213
Moen Sen, Ling Wang, Liping Yu, and Erica L. Carpenter
14 Quality Control of Immunophenotyping . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
Bruce Greig
15 Immunophenotyping of Acute Myeloid Leukemia . . . . . . . . . . . . . . . . . . . . . . . . . . 281
Pallavi Kanwar Galera, Chunjie Jiang, and Raul Braylan
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 355
Contributors
ix
x Contributors
Abstract
Recent technological advances have greatly diversified the platforms that are available for high-dimensional
single-cell immunophenotyping, including mass cytometry, single-cell RNA sequencing, and fluorescent-
based flow cytometry. The latter is currently the most commonly used approach, and modern instrumenta-
tion allows for the measurement of up to 30 parameters, revealing deep insights into the complexity of the
immune system.
Here, we provide a practical guidebook for the successful design and execution of complex fluorescence-
based immunophenotyping panels. We address common misconceptions and caveats, and also discuss
challenges that are associated with the quality control and analysis of these data sets.
1 Introduction
Since its invention more than 50 years ago, flow cytometry has been
a key tool for novel discoveries in the field of immunology
[1, 2]. This has been driven primarily by the ability of flow cyto-
metry to qualitatively and (within certain constraints) quantitatively
measure multiple features on a single cell level. Given the diversity
of immune cell subsets and their functional complexity, there is an
increasing need to measure more targets per cell in order to accu-
rately classify a given subtype while still being able to assess addi-
tional features of interest.
This need has fueled the development of novel cytometry plat-
forms during the past decade, most notably mass cytometry (cyto-
metry by time-of-flight, CyTOF) [3–5] and single-cell RNA
sequencing (sc-RNAseq) [6–9]. Both of these approaches can pro-
vide previously unprecedented insight into the immune system, but
come with their own set of limitations. Mass cytometry currently
allows the measurement of up to 50 protein (or RNA) targets on
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_1, © Springer Science+Business Media, LLC, part of Springer Nature 2019
1
2 Florian Mair and Aaron J. Tyznik
Fig. 1 Overview of the typical steps required for a high-dimensional immunophenotyping experiment. Each of
the six steps is described in detail in Subheadings 3.1 through 3.7
2 Materials
3 Methods
3.1 Generating Your Any immunophenotyping experiment should start with a biological
Hypothesis and List of hypothesis and a list of targets that are to be included in the
Biological Targets experiment. Typically, this will comprise two types of target anti-
gens: the so-called lineage markers, which are used to delineate
canonical immune cell populations (such as CD3 for T cells), and
the markers of interest, which can either have a known expression
pattern that changes under experimental conditions (such as CD69
as an early activation marker for T cells), or a potentially unknown
expression pattern.
As a first step, we recommend compiling a table that lists all
target antigens, their expected expression pattern (continual or
variable), expression type (bimodal, continuum), and expression
level (low to high) as well as expected coexpression patterns, that
is, which markers are expected to be expressed on the same cell
types. An example for such a table with some human T cell-centric
markers is given in Fig. 2. Furthermore, it is helpful to include
reagent availability in this table, that is, which fluorophore–anti-
body conjugates are available from which vendor for a given target.
Having this information at hand simplifies the actual panel design
process, which will be discussed in detail in Subheading 3.4. The
antigen targets listed within this chapter are based on human
leukocytes, but the same principles apply for any other cell type
such as murine leukocytes.
1. Define the cellular populations that are relevant for the planned
experiment (e.g., CD4+ T cells, CD8+ T cells, and CD56+ NK
cells).
High-Dimensional Immunophenotyping 5
Fig. 2 Example table with target antigen information. This table lists a selection of antigens for a human T cell
centric panel and the anticipated coexpression patterns. Researchers should adopt this table to fit their own
needs. Alternatively, one can draw a gating tree instead of using a table. If available, it is helpful to assess
common expression patterns in preliminary experiments or in public data sets such as those included in
published OMIPs
stems from the fact that photon multiplier tubes (PMTs), which are
still the most commonly used detector type, show significant
model-to-model variation, and thus each detector will require a
different voltage gain to deliver optimal resolution (i.e., optimal
signal-to-noise ratio). A more extensive discussion on the factors
describing the performance of a given PMT can be found in the
following reference [25]. Of note, avalanche photodiodes (APDs)
which are better suited for the detection of long-wavelength signals
[26] are becoming more popular but are beyond the scope of this
chapter.
There are several methods available to perform detector opti-
mization: for instruments from BD Biosciences, the Cytometry
Setup and Tracking (CS&T) module is commonly used to generate
consistent settings and track cytometer performance (see vendor
specific CS&T application guide). This system relies on a set of
hard-dyed dim, medium, and bright beads (CS&T beads). During
initial setup, voltages are determined such that the signal of the dim
beads is placed 10 times above the robust standard deviation (rSD)
of the electronic noise. Later on, the signal of the bright beads is
used to set target values, which are being matched during daily
quality control (QC) by modification of the detector voltages.
Often times, users utilize PMT voltage settings from the CS&T
application for setting and tracking detector gains. However, as
CS&T was developed at a time when the selection of available
fluorophores was limiting, the CS&T routine might deliver unnec-
essarily high baseline voltages for some detectors excited off the
violet, blue, and ultraviolet laser, thereby pushing signals from cells
off scale. Additionally, the rSD of the dim bead may not be the same
as the autofluorescence of a biological sample and correlation to
optimal PMT voltage settings may not be accurate. As a result,
researchers may need to lower or, in some instances, increase their
detector gains manually, making the CS&T method of setting
baseline voltages suboptimal.
Another approach is based on using unstained cells and setting
the voltages in such a way that the rSD of the cells equals 2.5 times
the rSD of the electronic noise, which ensures that dim signals are
pulled out of the electronic noise [27]. Finally, there are two
approaches that utilize a voltage titration experiment using either
the peak 2 of 8-peak rainbow beads (called the second peak
method, mimicking a dim signal) or single-stained samples (using
cells or antibody capture beads).
Here, we will describe a simplified voltage titration (voltration)
experiment using cells stained at saturating levels with a single
antibody clone (e.g., anti-CD4 or anti-CD8) conjugated to all
fluorophores that can be measured on a given instrument
[28]. Of note, the same data can be used to generate a relative
fluorochrome brightness chart, which will be useful in Subheading
3.4 for panel design. This experiment needs to be carried out only
High-Dimensional Immunophenotyping 7
once for any cytometer unless the optical configuration, that is,
lasers, filters, or detectors are changed. A representative analysis of
two typical voltage titrations is shown in Fig. 3a, b, and two
fluorochrome brightness charts with a classification of fluoro-
chromes into four broad categories is shown in Fig. 3c.
Performing the voltage titration experiment (unique to
instrument):
1. Obtain antibodies for a given clone (typically targeting a line-
age marker such as CD4) conjugated to all different fluoro-
phores that can be measured on the available instrument.
2. Obtain cells (typically cryopreserved PBMCs or murine
splenocytes).
3. Prepare antibody dilutions of every conjugate at a saturating
titer in stain buffer (see Subheading 3.5).
4. Aliquot 500,000 cells in as many wells as required into a
96-well plate.
5. Spin plate at 400 g for 5 min. Flick supernatant, dry the top
of the plate by carefully placing it on a paper towel and draining
remaining liquid.
6. Add 100 μl of a single antibody dilution per well, resuspend,
and incubate for 20 min at RT in the dark.
7. Add 200 μl of stain buffer to all wells, centrifuge plate at
400 g for 5 min, flick supernatant, and dry the top of the
plate by carefully placing it on a paper towel. Repeat this wash
for a total of two washes.
8. Optional: add 100 μl of fixative to each well, resuspend, and
incubate for 20 min at RT in the dark (see Note 1).
9. Add 200 μl of stain buffer to all wells, centrifuge plate at
400 g for 5 min, flick supernatant, and dry the top of the
plate by carefully placing it on a paper towel.
10. Resuspend in 200 μl of stain buffer and store in the dark at 4 C
until acquisition.
11. Perform flow cytometer startup and daily QC as required per
instrument-specific instructions.
12. Load one of the single-stained samples, and adjust the voltage
of the respective detector in 30 V increments from 200 V to
700 V. Record 10,000 events for every given detector voltage
setting and name the tubes accordingly (BV421_200V,
BV421_230V, BV421_260V, etc.).
13. Disregard samples where the MFI of the positive signal has
exceeded detector linearity or is no longer on scale as errone-
ous values may be reported.
14. Repeat this step for all single-stained samples.
Fig. 3 Examples for a voltage titration experiment and a fluorochrome brightness chart. (a) and (b) Two
representative voltage titration experiments are shown as concatenated pseudocolor plots (left) and as
histogram overlays (right). Staining indices have been calculated as described in Subheading 3.2. For
CD4-PE the plateau of the stain index is reached around 420 V, indicating that this is the minimal voltage
required to deliver optimal resolution. If the positive population exceeds the upper end of the scale as is the
case for CD4-BB515, see Note 2. (c) Two representative fluorochrome brightness charts generated on two
independent commercially available high dimensional flow cytometers. As each instrument is unique in laser
power and detection optics, it is best to characterize the relative fluorochrome brightness for each individual
instrument. However, assigning fluorochromes to four broad categories will allow some generalization for
relative brightness (e.g., APC-H7 tandems being at the lower end of the scale, or BV421 and PE-tandem dyes
being at the upper end of the scale)
High-Dimensional Immunophenotyping 9
15. Export data in FCS3.1 file format and load into a flow cyto-
metry data analysis program.
16. For a given single-stained control, draw gates delineating sing-
lets, and subsequently the positive peak and the negative peak
for the corresponding fluorophore.
17. Optional: concatenate the individual FCS files for a given
single-stained control into a single FCS-file, or generate histo-
gram overlays to obtain a visual representation of the voltration
experiment.
18. Repeat this step for all single-stained samples.
19. Calculate the median fluorescence intensity (MFI) of both the
positive and the negative peak, as well as the rSD for the
negative peak.
20. Calculate the staining index (SI) for each sample by using the
following formula: SI ¼ MFI(pos) MFI(neg)/(2 rSD).
21. Using a spreadsheet program, generate a plot showing the SI
on the y-axis relative to the voltage on the x-axis.
22. For each detector, pick the voltage on which the SI starts to
reach a plateau, which is the minimal voltage delivering the best
possible resolution for a given detector. As an alternative visu-
alization, plot the SI as a percentage of maximum SI on the y-
axis relative to the voltage on the x-axis (also see Note 2).
23. Once the optimal baseline PMT voltages have been deter-
mined, record a sample of appropriate reference beads (e.g.,
Spherotech Ultra Rainbow Beads) as described in steps 29–34.
See Note 3 for examples of reference particles.
Create relative fluorochrome brightness ranking chart unique to
the instrument
24. Identify the single-stained samples acquired to reach the opti-
mal PMT voltage for any given detector.
25. Calculate the median fluorescence intensity (MFI) of both the
positive and the negative peak, as well as the rSD for the
negative peak.
26. Calculate the staining index (SI) for each fluorophore by
using the following formula: SI ¼ MFI(pos) MFI(neg)/
(2 rSD).
27. Using a spreadsheet program, generate a plot showing the SI
for each fluorophore and arrange them by decreasing values.
Alternatively, plot the SI as percentage of maximum SI.
28. Assign the fluorochromes into broad categories based on their
SI, that is, bright, intermediate-high, intermediate-low,
and dim.
10 Florian Mair and Aaron J. Tyznik
Fig. 4 Fluorochrome overlap, compensation, and spreading error (SE). (a) Fluorochrome spectra depicting
excitation (dotted curve) and emission (solid filled curve) of Brilliant Violet 650 (BV650, left) and Phycoerythrin
12 Florian Mair and Aaron J. Tyznik
Fig. 5 The spillover spreading matrix (SSM). (a) A representative spillover spreading matrix (SSM) calculated
from a 28-color experiment is shown. Color coding is from no SE (white) to high SE (red). Note that the SSM is
instrument specific and does not provide generalizable conclusions about the extent of SE, and SE is a
unitless, relative number. For this particular instrument, the three fluorophores contributing the least SE are
BB515, AF647 and BUV395. Thus, these three dyes would be optimal to assign for lineage markers as they are
contributing minimal SE to all the other detectors. In turn, the three detectors receiving the least SE are B-515,
V-510 and V-450. Since BB515 as well as BV421 are typically two very bright fluorophores (see Fig. 3c), on
this particular instrument these two detectors would be optimal for dimly expressed targets. Note that when
assessing the row-sums, that is, the total SE contributed by a given fluorophore, the case can be that this sum
is driven mostly by one particular value, for example BUV661, where SE into R-660 is the major contributor to
the sum. In this case, BUV661 can still readily be used without major impact to all other detectors except
R-660
Fig. 5. The use of the SSM is more extensively discussed in the next
section.
1. Obtain antibodies carrying all fluorochromes that are to be
included in the new panel (the antigen targets do not necessar-
ily matter at this point in time, but should match the fluoro-
chromes that will be used) (also see Note 4).
2. Aliquot antibody-capture beads (i.e., compensation particles)
into as many wells as required into a 96-well plate.
3. Resuspend in 100 μl of staining buffer.
4. Add 1 μl of a single antibody into each well and resuspend
using a 100 μl pipette. Incubate for a minimum of 15 min at RT
in the dark.
5. Add 200 μl of stain buffer to all wells, centrifuge plate at
400 g for 5 min, flick supernatant, and dry the top of the
plate by carefully placing it on a paper towel. Repeat this
wash once.
6. Optional: add 100 μl of fixative to each well, resuspend, and
incubate for 20 min at RT in the dark.
7. Add 200 μl of stain buffer to all wells, centrifuge plate at
400 g for 5 min, flick supernatant, and dry the top of the
plate by carefully placing it on a paper towel.
High-Dimensional Immunophenotyping 15
3.4 In Silico Panel The two key considerations for successful panel design are (1) fluo-
Design Using the rochrome brightness, and (2) dealing with spreading error (SE),
Spillover Spreading whereby the latter is the more relevant issue. There are several ways
Matrix (SSM) that SE and the resulting loss of resolution can be controlled. The
most straightforward way is to use fluorophore–detector pairs with
high SE for mutually exclusive markers (e.g., CD3 and CD19)
because in this case the SE will not interfere with detection of either
signal. However, this tactic is only applicable to a limited extent as
most researchers’ experimental aim is to analyze as many target
antigens as possible on one or two cell types.
Thus, if fluorophore–detector pairs with significant SE are to be
used for molecules expressed on the same cell type, one has to assess
the expression level of the target antigens. This can be done either
by an internet search, or by preliminary experiments assessing
antigen density on the target population. An excellent resource in
this context are optimized multicolor immunophenotyping panels
(OMIPs) [33, 34], which typically show raw data in their supple-
ments and cover many of the commonly used targets in immunol-
ogy [13, 35, 36]. If the target expression level is known, one can
use a fluorophore generating a large SE for low-expression anti-
gens, which will thus generate only weak signals, which in turn will
reduce the amount of SE (as SE is proportional to the signal
intensity). Alternatively, one can use a highly expressed antigen in
the secondary detector, and assess whether the signal of the target
16 Florian Mair and Aaron J. Tyznik
3.5 Titration of After selection of reagents for the panel run, titration is an essential
Reagents step that is often omitted. The primary reason for titrating anti-
bodies is twofold: first, to minimize background from nonspecific
binding and second, to ensure that staining is performed at satur-
ating concentration (if necessary) [37]. For a typical titration exper-
iment, it is recommended to do 8–12 twofold serial dilutions
starting from a highest concentration of 2 μg per staining reaction.
After titration, the stain index (SI) can be plotted relative to the
dilution factor. This will reveal the saturating titer, which is the
concentration at which further addition of antibody does not
increase the signal intensity, that is, all target binding sites have
been occupied. If any quantitative measurements (i.e., extracting
MFI as a means of estimating target antigen density) are intended,
antibodies have to be used at the saturating concentration. How-
ever, for some lineage markers (e.g., CD3 or CD4) it is sufficient to
work at nonsaturating concentration. The benefits are that it saves
reagent and also minimizes spreading error into other detectors. If
one chooses to do so, it is important to note that signal intensity at
nonsaturating concentration can change with the number of cells
stained, incubation length, and temperature. Thus, one has to work
with standardized cell numbers and staining conditions. For differ-
ent examples of antibody titrations, we direct the reader to the
supplementary materials of recent OMIPs [13, 35] and the infor-
mation found in the “Guidelines” [23].
1. Obtain cells (typically cryopreserved PBMCs or murine sple-
nocytes). See Note 5 for targets that are not readily detected on
steady-state cells.
2. Aliquot cells in as many wells as required into a separate 96-well
plate.
3. Spin plate at 400 g for 5 min. Flick supernatant and dry the
top of the plate by carefully placing it on a paper towel and
draining remaining liquid.
4. For a given titration experiment, add 96 μl of stain buffer to the
empty well 1 of a 96-well plate, and 60 μl of stain buffer to wells
2–12.
5. Add 24 μl of antibody stock to well 1 and mix. If the specific
antibody concentration is known, use 2 μg of antibody stock
solution and add it to well 1 with additional stain buffer as
required to achieve a final volume of 120 μl.
18 Florian Mair and Aaron J. Tyznik
3.6 Single-Stained Once all reagents have been titrated, the full panel can be tested.
Controls and Testing of The most essential aspect of this validation experiment is not nec-
Full Panel essarily the full stain but rather the inclusion of the appropriate
controls, specifically fluorescence-minus-one controls (FMOs)
[38]. An FMO contains the full antibody mix except one antibody,
and thus allows assessing problems that can potentially arise from
unforeseen spillover or unexpected interaction between dyes
[39]. While it is beneficial to prepare FMOs for every single reagent
in a panel, this can be tedious and might not always be necessary.
Instead, one should include FMO controls for every target where
the expected signal is either not known or includes potentially
problematic fluorochrome–detector pairs (also see Note 6).
The quintessential prerequisite (and most common source of
error) for the success of any polychromatic experiment is the quality
of single-stained controls [30]. This is an area where several mis-
conceptions are commonly found as to what constitutes a “good”
single-stained control, in particular whether one should use cells or
antibody-capture beads (compensation particles). There are four
generalizable rules that apply for single-stained controls, and as
long as these are met one can use either cells or beads (see also
Note 7): (1) Within one single-stained control, the positive and
negative particles must be of the same kind, that is, have the same
autofluorescence. This means that both populations must be beads
(of the same batch and type), or both populations must be the same
type of cells (and not one population comprising T cells and the
other one monocytes, as would be the case if using human PBMCs
and staining them with CD3). The reason for this is that during
compensation, the compensation value is calculated such that MFIs
of positive and negative populations are equal [30]. If the auto-
fluorescence, that is, the background fluorescence of these two
populations are different, this will introduce an error into the
compensation matrix. (2) The positive signal of the single-stained
control must be as bright as or brighter than the experimental
sample, but remain within the linear range of a given detector.
The reason for this rule is that the error that is associated with a
measurement is proportional to the square root of the signal inten-
sity, such that a dimmer signal will carry a proportionally larger
measurement error. Therefore, it is better if the single-stained
control is bright but does not exceed the linear range of the
detector (instrument-dependent), which would again introduce
errors into the compensation matrix. (3) The single-stained control
must use the exact same fluorophore as the real experimental sam-
ple. Though this is an obvious rule, it is sometimes not followed
because researchers use different antibodies for their single-stained
control relative to their experiment. However, this can be detri-
mental in particular for tandem dyes because different product lots
will show different conjugation efficiencies. Therefore, it is required
that the antibody–fluorochrome molecule used to prepare the
20 Florian Mair and Aaron J. Tyznik
single-stained control is the same type and lot as the one used to
stain the actual sample. (4) The single-stained control should be
treated exactly the same way as the real experimental sample,
including fixation. The reason for this is that some fixation proto-
cols can impact fluorophore characteristics, and similar to the pre-
vious point, the single-stained control should carry the exact same
fluorophore as is present in the sample.
1. Prepare a spreadsheet showing antibody volumes to be
pipetted in the planned experiment.
2. Obtain cells (typically cryopreserved PBMCs or murine
splenocytes).
3. Resuspend cells in the appropriate volume and dispense into a
96-well plate.
4. Centrifuge plate at 400 g for 5 min. Flick supernatant and
dry the top of the plate by carefully placing it on a paper towel.
5. Add 100 μl of freshly prepared Fc-Block/Viability dye solution
and incubate for 15 min at room temperature (RT) in the dark
(see Note 8). In the meantime, start preparing the antibody
master mix (and the required FMO mixes) using Brilliant
Staining Buffer (see Note 9).
6. Wash by adding 200 μl of stain buffer, centrifuge plate at
400 g for 5 min, flick supernatant, and dry the top of the
plate by carefully placing it on a paper towel.
7. Add 50 μl of antibody staining mix containing the correct final
dilutions of all antibodies, resuspend, and incubate for 20 min
at RT in the dark.
8. In parallel, dilute 7–15 drops of compensation beads with stain
buffer to a total volume of 1.5 ml, aliquot 50 μl into the wells of
the 96-well plate, add 0.7 μl of a single antibody into each well.
Incubate for a minimum of 15 min at RT in the dark.
9. Add 200 μl of stain buffer to all wells, centrifuge plate at
400 g for 5 min, flick supernatant, and dry the top of the
plate by carefully placing it on a paper towel. Repeat this wash
once for a total of two washes.
10. Optional: add 100 μl of fixative to each well, resuspend, and
incubate for 20 min at RT in the dark.
11. Add 200 μl of stain buffer to all wells, centrifuge plate at
400 g for 5 min, flick supernatant, and dry the top of the
plate by carefully placing it on a paper towel.
12. Resuspend the cells in 100–200 μl of stain buffer and store at
4 C in the dark until acquisition.
13. Startup flow cytometer and perform daily QC as required per
manufacturer’s instructions.
High-Dimensional Immunophenotyping 21
14. Adjust the PMT voltages to match your MFI target values
of your reference particles as described in Subheading 3.2,
steps 29–34.
15. Record 10,000 events from each single-stained control, and
perform compensation using instrument-specific acquisition
software.
16. Record as many events as desired from the full stain as well as
the FMO controls.
17. Export data in FCS3.1 file format and load into a flow cyto-
metry data analysis program. Follow the analysis steps outlined
in Subheading 3.7.
3.7 Analysis and Within this section we focus on manual analysis and quality control
Troubleshooting of the newly established panel. This can be performed using any
software that is able to read and visualize FCS files. Among the
commonly used stand-alone packages are FlowJo (BD Biosciences),
and a powerful cloud-based solution is offered by Cytobank (www.
cytobank.com), but there are several other options available on the
market (e.g., FCS Express from De Novo Software).
For fluorescent data an often-undervalued aspect is the appro-
priate transformation and visualization of the data, which is essen-
tial for successful manual analysis. Historically, flow cytometric data
has often been displayed using a purely logarithmic scale. This can
introduce visual artifacts at the lower end of the scale, which is why
the logicle/biexponential transformation was developed in the
mid-2000s [40–42]. These so-called hybrid scales will be logarith-
mic at the high end of the scale and gradually approach a linear scale
around 0, after which they would transition back to a logarithmic
scale below 0. In the biexponential implementation, the “width”
basis will determine how much space around 0 is displayed in a
linear domain, thus either “compressing” or “widening” the
appearance of the negative populations. Currently, there is no
reliable automated solution available, and thus researchers need to
adjust the width basis as well as the amount of negative decades
manually. This should be done in a way that (1) all events are visible
on a plot and (2) the negative population appears as a normally
distributed (i.e., round) population. In particular for complex
polychromatic data sets, it is essential to use logicle/biexponential
(or any other appropriate version such as arcsin) transformation of
the data in order to visualize spreading error (SE) accordingly.
After the first run of a newly established panel, validation and
troubleshooting is critical, as even with the best possible planning,
unexpected issues can arise from SE or improper compensation. To
assess this, two different strategies should be employed: (1) Prepare
an N N view of all parameters against each other and visually
screen for erroneous patterns. This include either “leaning,” trian-
gular populations or the so-called “super-negative” populations
that appear below 0 (Fig. 6a). Of note, the negative population
22 Florian Mair and Aaron J. Tyznik
Fig. 6 Examples of erroneous staining patterns and (SE) spreading error. Example data are from the
development of a 28-color immunophenotyping panel centered on myeloid cells (published in OMIP-044).
High-Dimensional Immunophenotyping 23
Fig. 6 (continued) (a) Upper panel shows correctly compensated data. If overcompensated (middle panel), this
would be visible either as a population of “super-negative” events (if displaying two unrelated detectors) or as
a leaning, banana-shaped population pointing below 0 (if displaying the fluorophore–detector pair that is
wrongly compensated). If the population is undercompensated, this would be very hard to diagnose solely
from the full stain, but the FMO control clearly identifies the issue by showing a leaning, banana-shaped
population pointing toward the upper right. (b) Two examples where spreading error (SE) was dealt with by
assigning the corresponding fluorophores to mutually exclusive antigens. (c) Example for improving a
suboptimal panel after initial testing. Left plot shows CX3CR1 on PE-Cy7, which will show significant SE
into the B-780 detector (excitation laser: 488 nm, detection window: 780/60) and makes it hard to discern
CD38+ events in this detector. To improve this, CX3CR1 was moved to BV421, a fluorophore that does not
show any SE into the B-780 detector. PE-Cy7 should instead be used for a target that delivers a dimmer signal,
which will in turn diminish the SE generated
24 Florian Mair and Aaron J. Tyznik
4 Notes
panels, and will likely become even more prevalent in the future
[45]. Due to their chemistry, these polymer-based dyes can
potentially interact with each other, causing artifacts in flow
cytometry experiments that appear as inappropriately compen-
sated data. To prevent this, it is advisable to use commercially
available staining buffers preventing this interaction every time
one is using more than two polymer-based dyes in an experi-
ment. Since different vendors use different proprietary formu-
lations for these buffers, users might need to test which buffer
works best for their experimental setup.
Acknowledgments
The authors thank all members of the Prlic lab and Dr. Sabine Spath
for critical reading of the manuscript. F.M. and A.J.T. are supported
by a Marylou scholarship from the International Society for
Advancement of Cytometry (ISAC).
References
1. Fulwyler MJ (1965) Electronic separation of 8. Klein AM, Mazutis L, Akartuna I et al (2015)
biological cells by volume. Science Droplet barcoding for single-cell transcrip-
150:910–911 tomics applied to embryonic stem cells. Cell
2. Robinson JP, Roederer M (2015) History of 161:1187–1201. https://doi.org/10.1016/j.
science flow cytometry strikes gold. Science cell.2015.04.044
350:739–740. https://doi.org/10.1126/sci 9. Zheng GXY, Terry JM, Belgrader P et al
ence.aad6770 (2017) Massively parallel digital transcriptional
3. Bandura DR, Baranov VI, Ornatsky OI et al profiling of single cells. Nat Commun
(2009) Mass cytometry: technique for real time 8:14049. https://doi.org/10.1038/
single cell multitarget immunoassay based on ncomms14049
inductively coupled plasma time-of-flight mass 10. Brodie TM, Tosevski V (2017) High-
spectrometry. Anal Chem 81:6813–6822. dimensional single-cell analysis with mass cyto-
https://doi.org/10.1021/ac901049w metry. Curr Protoc Immunol
4. Bendall SC, Simonds EF, Qiu P et al (2011) 118:5.11.1–5.11.25. https://doi.org/10.
Single-cell mass cytometry of differential 1002/cpim.31
immune and drug responses across a human 11. Papalexi E, Satija R (2017) Single-cell RNA
hematopoietic continuum. Science sequencing to explore immune cell heteroge-
332:687–696. https://doi.org/10.1126/sci neity. Nat Rev Immunol 510:363. https://doi.
ence.1198704 org/10.1038/nri.2017.76
5. Spitzer MH, Nolan GP (2016) Mass cytome- 12. Chattopadhyay PK, Roederer M (2012) Cyto-
try: single cells, many features. Cell metry: today’s technology and tomorrow’s
165:780–791. https://doi.org/10.1016/j. horizons. Methods 57:251–258. https://doi.
cell.2016.04.019 org/10.1016/j.ymeth.2012.02.009
6. Tang F, Barbacioru C, Wang Y et al (2009) 13. Mair F, Prlic M (2018) OMIP-044: 28-color
mRNA-Seq whole-transcriptome analysis of a immunophenotyping of the human dendritic
single cell. Nat Methods 6:377–382. https:// cell compartment. Cytometry A 106:255.
doi.org/10.1038/nmeth.1315 https://doi.org/10.1002/cyto.a.23331
7. Shalek AK, Satija R, Adiconis X et al (2013) 14. Grégori G, Patsekin V, Rajwa B et al (2012)
Single-cell transcriptomics reveals bimodality Hyperspectral cytometry at the single-cell level
in expression and splicing in immune cells. using a 32-channel photodetector. Cytometry
Nature 498:236–240. https://doi.org/10. A 81:35–44. https://doi.org/10.1002/cyto.
1038/nature12172 a.21120
28 Florian Mair and Aaron J. Tyznik
39. Mahnke YD, Roederer M (2007) Optimizing a 43. Roederer M (2016) Distributions of autofluor-
multicolor immunophenotyping assay. Clin escence after compensation: be panglossian,
Lab Med 27:469–485. v. https://doi.org/10. fret not. Cytometry A 89:398–402. https://
1016/j.cll.2007.05.002 doi.org/10.1002/cyto.a.22820
40. Parks DR, Roederer M, Moore WA (2006) A 44. Andersen MN, Al-Karradi SNH, Kragstrup
new “Logicle” display method avoids deceptive TW, Hokland M (2016) Elimination of erro-
effects of logarithmic scaling for low signals neous results in flow cytometry caused by anti-
and compensated data. Cytometry A body binding to Fc receptors on human
69:541–551. https://doi.org/10.1002/cyto. monocytes and macrophages. Cytometry A
a.20258 89:1001–1009. https://doi.org/10.1002/
41. Trotter J (2007) Alternatives to log-scale data cyto.a.22995
display. Curr Protoc Cytom Chapter 10:Unit 45. Chattopadhyay PK, Gaylord B, Palmer A et al
10.16–10.16.11. https://doi.org/10.1002/ (2012) Brilliant violet fluorophores: a new class
0471142956.cy1016s42 of ultrabright fluorescent compounds for
42. Herzenberg LA, Tung J, Moore WA et al immunofluorescence experiments. Cytometry
(2006) Interpreting flow cytometry data: a A 81A:456–466. https://doi.org/10.1002/
guide for the perplexed. Nat Immunol cyto.a.22043
7:681–685. https://doi.org/10.1038/
ni0706-681
Chapter 2
Abstract
Mass cytometry is a novel technology similar to flow cytometry in which antibodies are tagged with heavy
metal molecules rather than fluorophores and then detected with time-of-flight mass spectrometry. This
enables measurement of up to 50 simultaneous parameters with no autofluorescent background and little or
no spillover or required compensation. Mass cytometry has tremendous potential for the analysis of highly
complex research or clinical samples and can measure 40–50 immunophenotypic markers at a time. This
chapter describes most of the commonly used methods for performing basic immunophenotyping experi-
ments by mass cytometry, and how this can be combined with measurement of cellular functional
properties.
Key words Immunophenotyping, Mass cytometry, CyTOF, Flow cytometry, Minimal residual dis-
ease, Acute leukemia, Aberrant marker expression
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_2, © Springer Science+Business Media, LLC, part of Springer Nature 2019
31
32 Gregory K. Behbehani
1.1 Mass Cytometry Mass cytometry (MC) is a cytometry technique similar to fluores-
Versus Fluorescent cent flow cytometry that detects cellular antigens through the use
Flow Cytometry of mass spectrometry to detect binding of heavy-metal conjugated
antibodies to the cells being studied. Mass cytometry experiments
are typically performed using the same antigen-specific antibodies
used in conventional flow cytometry, but the binding of these
antibodies is measured by vaporizing and ionizing the cells (with
the bound metal-conjugated antibodies) followed by the
subsequent detection of the molecules of isotopically purified
heavy metal atoms by time-of-flight mass spectrometry
(ICP-MS). The primary advantage of this approach stems from
the ability of ICP-MS to distinguish ions of different atomic weight
with less than 0.5% signal spillover between adjacent masses (which
are each conjugated to a particular antibody). The data generated
from the analysis of each antigen is very similar to the data gener-
ated when the same antigen is analyzed by flow cytometry, and
currently up to 40–50 parameters can be simultaneously recorded
per cell with minimal or no signal compensation and no back-
ground due to autofluorescence. The greatest advantage of mass
cytometry is the large increase in the number of simultaneous
parameters that can be recorded. This is particularly useful in the
analysis of highly complex or heterogeneous cell populations in
which the additional parameters enable simultaneous identification
of numerous cell types or functional states to be uniquely identified
and characterized, even when some of these cell types are very rare.
There are now numerous publications utilizing this technology and
many excellent reviews [2–6]; this chapter will provide a basic
protocol for immunophenotyping by mass cytometry, how this
can be combined with measurement of intracellular antigens for
functional studies, and focus on the unique practical considerations
that are important in designing mass cytometry experiments.
Since mass cytometry experiments can typically be performed
using the same exact antibody clones as fluorescent flow cytometry
experiments, the immunophenotypes derived from the two techni-
ques should be nearly identical. However, labeling of antibodies
with chelated heavy metal and its detection by time-of-flight mass
spectrometry creates several unique considerations that must be
accounted for in the design of a mass cytometry experiment.
These can be grouped into issues related to the antibody labeling
polymer and the ion detector.
Antibody considerations: Mass cytometry antibodies are pro-
duced by conjugation of polymer of DTPA chelators (functiona-
lized using a maleimide group) to available sulfhydryl groups of the
Immunophenotyping by Mass Cytometry 33
1.2 General Method In this chapter the most common approaches to mass cytometry
Design cell processing will be described. However, if converting a previ-
ously established fluorescent flow cytometry protocol to a mass
cytometry protocol, the same sample processing and antibody
staining conditions can typically be used. The only absolutely
required modifications needed to convert a flow cytometry immu-
nophenotyping protocol to mass cytometry protocol are as
follows: (1) use of metal-conjugated antibodies instead of the
fluorochrome-conjugated ones, (2) the use of a fixative-containing
metal intercalator solution after the completion of antibody stain-
ing (Subheading 3.6, steps 1–3), and (3) running the sample into
the mass cytometer after dilution in either pure water or a
specialized running buffer free from nonorganic salts (Subheading
3.6, steps 4–9). If using commercially prepared solutions for fixa-
tion and permeabilization, all solutions should be checked for heavy
metal contamination on a regular basis (unless the solutions are
specifically made for mass cytometry).
The minimum number of cells required for mass cytometry
analysis varies based on the exact number of centrifugation steps,
the type of centrifuge tubes employed, and the number of washes
performed. For the protocol described here (utilizing standard
5 mL polystyrene FACS tubes), 500,000 cells is a good minimum
number as the mass cytometer generates data from 30% to 50% of
the cells that are injected (after staining and washing), necessitating
a proportional increase in the number of starting cells (as compared
Immunophenotyping by Mass Cytometry 35
2 Materials
2.1 Precautions To avoid background due to heavy metal contamination, all solu-
tions for mass cytometry should be prepared using ultrapure water
and all reagent solutions (when first prepared from new lots of stock
reagents) should be tested for heavy metal contamination (see
Note 2). Note that cisplatin, paraformaldehyde, and the Smart
Tube buffer are all toxic and potentially mutagenic. They should
be handled with appropriate protections (gloves, eye and respira-
tory protection as appropriate). It is also important to mention that
standard laboratory dishwashers, autoclaves, and dishwashing
detergents are commonly contaminated with heavy metals (partic-
ularly lead and barium) that can disrupt experiments or damage the
mass cytometer. We use either disposable plastic containers or
glassware that we wash by hand for producing and storing all
mass cytometry solutions and reagents.
3 Methods
3.1 Viability Staining If a significant fraction of the cells of interest could be dead at the
(Optional) time of collection, it is generally advisable to perform a viability
(live–dead) stain prior to antibody staining or cell fixation. Dead or
dying cells typically display different size and surface marker expres-
sion characteristics and almost universally will have altered levels of
intracellular functional markers. It is thus critical to have a means of
excluding dead cells (if expected to be present) as part of most
experimental designs. This is particularly important for analysis of
cryopreserved cells, in which the freezing and thawing process will
reliably kill a fraction of cells by necrosis. The simplest method of
live–dead staining is use of cisplatin (which nonspecifically and
covalently binds to protein), based on the publication from Fien-
berg et al. [14], which will be described here. Alternative
approaches include staining with bifunctional chelators loaded
with heavy metal, such as barcoding reagents [15], or use of
metal-containing DNA intercalators such as rhodium [16]
(although in our experience the latter stain is generally not stable
enough to be suitable if intracellular antigens will be measured). In
all cases, the basic concept is to incubate cells in a reagent that does
not rapidly cross the cell membrane of live cells but can rapidly
enter the disrupted membrane of a dead cell where it will bind to
intracellular DNA or protein, resulting in higher signal in the dead
cells.
1. Fresh cells (from primary tissue or in vitro culture media, prior
to any fixation) should first be washed one or two times with
serum/protein-free PBS (see Notes 7 and 8).
Immunophenotyping by Mass Cytometry 39
3.2 Cell Stimulation While beyond the scope of this chapter, if the planned experiment
(Optional) calls for a cell stimulation prior to antibody staining (e.g., cytokine
stimulation [17] or IdU incorporation for cell cycle analysis [18])
this should be performed after viability staining and before anti-
body surface staining. Typically, if a stimulation is performed, the
cells will need to be fixed with PFA or STPS (or another fixative
product) before antibody staining to preserve the intracellular
response to the stimulation.
3.4 Fixed Sample 1. Fixation: If the goal of the experiment is to study the intracel-
Surface Staining lular properties of the cells of interest, it is typically advanta-
geous to gently fix the cells prior to antibody staining.
Commonly used fixation methods utilize PFA at 1.5–4% for
10–15 min at room temperature. For analysis of fresh primary
human cell suspensions (typically, human peripheral blood or
bone marrow aspirate), we will typically utilize Smart Tube
proteomic stabilizer (STPS) solution in accordance with the
manufacturer’s instructions. A variety of other commercial
fixative solutions are available and would likely be compatible
with this protocol. This solution utilizes a proprietary fixative
cocktail, but is sufficiently gentle to allow subsequent lysis of
red blood cells as well as detection of antigens known to be
disrupted by standard PFA fixation. An alternative fixation
procedure developed by Chow et al. [21] utilizing PFA fixation
followed by Triton X-100 can also work well for ex vivo sample
fixation; however, this method tends to be slightly more dis-
ruptive to fixation-sensitive antigens in our experience. With
either approach, cells can be frozen at 80 C after fixation but
before red cell lysis. If freezing other cell types after fixation
with PFA, cells should be washed twice in CSM and then
resuspended in CSM + 10% DMSO before snap-freezing at
80 C (see Note 13). As previously noted, some surface
antigens can be disrupted by fixation, so the effect of fixation
should be empirically tested for each antigen–antibody combi-
nation of interest.
2. Red cell lysis (optional): If blood is present in the sample, it
should be lysed before proceeding with antibody staining. If
using STPS, we lyse the red cells in accordance with the man-
ufacturer’s instructions, if using the protocol of Chow et al.
[21] lysis is performed with a detergent containing lysis buffer.
After the completion of red cell lysis the cells should be washed
twice in CSM.
42 Gregory K. Behbehani
3.6 DNA 1. After the final wash following the surface or intracellular stain-
Intercalation and Data ing reaction, resuspend the cell pellet by vortexing thoroughly.
Acquisition 2. Poststaining fixation (optional): If the cells of interest are par-
ticularly fragile, or if clogging of the mass cytometer has been
experienced when running similar cell samples, it may be help-
ful to perform an additional fixation step, by resuspending the
cell pellet in PBS plus 1.5% PFA and incubating for 10–15 min
at room temperature. This additional fixation will reduce lysis
of particularly fragile cells during sample acquisition, which can
lead to clogging of the mass cytometer’s fluidics. Ensure that
that the PFA is fresh. After this fixation, pellet the cells by
centrifugation and resuspend the cell pellet by vortexing
thoroughly.
3. Add at least 100–200 μL of intercalator solution for each
million cells in the cell pellet. This can also be titrated for the
specific cell types being stained. Mix or vortex cells gently to
resuspend them evenly in the intercalator solution. Place the
cells at 4 C for at least 20 min. The cells will be stable in the
intercalator solution at 4 C for at least a week.
4. After intercalation, cells should be washed twice in either ultra-
pure (heavy metal free) water or in a cell acquisition solution
(see Note 19). Cells should then be resuspended at a density of
approximately one million cells per mL in either water or cell
acquisition solution (if used). Four element EQ beads should
44 Gregory K. Behbehani
3.7 Initial Data There are now a large number of data analysis approaches for high
Processing dimensional mass cytometry data ranging from large scale analysis
of hundreds of standard biaxial plots to complex high-dimensional
analysis strategies. A thorough discussion of the pros and cons of
each approach is beyond the scope of this chapter, but some guid-
ance can be found in several recent publications [1, 3, 6]. Regardless
of the final analysis approach, several steps should always be per-
formed to ensure the data quality before more complex analysis is
undertaken. It is always ideal to still view the data in the relevant key
biaxial plots that have been traditionally used to define relevant cell
Immunophenotyping by Mass Cytometry 45
4 Notes
20. Note that in regular data collection mode, only signal within
the time-of-flight windows of the masses set in the panel will be
recorded or displayed on the preview window of the computer.
As a result, if the sample is contaminated with a metal not being
measured in the experiment’s antibody panel, this contamina-
tion (even if at relatively high levels) can be invisible to the user.
A quick preview in TOF mode will show all of the ion species
striking the detector, and if the preview shows cell events
without significant amounts of metal contamination, the
chance of problematic contamination is quite low.
21. When calculating the number of required cell events, it is
important to remember that up to 10% of events will be from
EQ beads and will not be present in the final data file. Addi-
tionally, if significant amounts of debris are present in the
sample, these debris particles will often be counted as cell
events. Thus, if debris is observable in preview screen of the
cytometer during sample acquisition, additional events will
need to be collected in proportion with the approximate fre-
quency of debris relative to true cell events. It is thus not
uncommon to need to collect 10–50% more events than
desired final number of cell events.
22. If running wash buffer (which contains acid) or nitric acid
between samples, it is important to ensure that all acid has
been washed away before starting the next sample. Persistent
acid will strip metal from the antibodies attached to the cells in
subsequent samples until the acid is neutralized.
23. If samples have not been adequately fixed before being placed
into pure water the antibodies can disassociate from their anti-
gens and/or cells may lyse during prolonged cell acquisition.
This effect is directly dependent on the time cells are in water
and cannot be normalized by the EQ bead-based normaliza-
tion algorithms. This problem can be minimized by ensuring
that the PFA used in initial sample fixation and in the inter-
calator solution is sufficiently fresh, by minimizing the time
cells are kept in water prior to being loaded in the cytometer,
and by keeping cells cold between water washes and analyses.
Additionally, use of the optional poststaining fixation steps
(Subheading 3.5, step 1; Subheading 3.6, step 2), or use of a
cell acquisition solution can minimize this problem.
References
1. Bendall SC, Nolan GP, Roederer M, Chatto- applications. Cancer Immunol Immunother
padhyay PK (2012) A deep profiler’s guide to 62(5):955–965
cytometry. Trends Immunol 33(7):323–332 3. Spitzer MH, Nolan GP (2016) Mass cytome-
2. Tanner SD, Baranov VI, Ornatsky OI, Bandura try: single cells, many features. Cell 165
DR, George TC (2013) An introduction to (4):780–791
mass cytometry: fundamentals and
Immunophenotyping by Mass Cytometry 51
4. Bjornson ZB, Nolan GP, Fantl WJ (2013) 14. Fienberg HG, Simonds EF, Fantl WJ, Nolan
Single-cell mass cytometry for analysis of GP, Bodenmiller B (2012) A platinum-based
immune system functional states. Curr Opin covalent viability reagent for single-cell mass
Immunol 25(4):484–494 cytometry. Cytometry A 81(6):467–475
5. Di Palma S, Bodenmiller B (2015) Unraveling 15. Zunder ER, Finck R, Behbehani GK et al
cell populations in tumors by single-cell mass (2015) Palladium-based mass tag cell barcod-
cytometry. Curr Opin Biotechnol 31:122–129 ing with a doublet-filtering scheme and single-
6. Behbehani GK (2017) Applications of mass cell deconvolution algorithm. Nat Protocols
cytometry in clinical medicine: the promise 10(2):316–333
and perils of clinical CyTOF. Clin Lab Med 16. Ornatsky OI, Lou X, Nitz M et al (2008) Study
37(4):945–964 of cell antigens and intracellular DNA by iden-
7. Rahman AH, Tordesillas L, Berin MC (2016) tification of element-containing labels and
Heparin reduces nonspecific eosinophil stain- metallointercalators using inductively coupled
ing artifacts in mass cytometry experiments. plasma mass spectrometry. Anal Chem 80
Cytometry A 89(6):601–607 (7):2539–2547
8. Frei AP, Bava F-A, Zunder ER et al (2016) 17. Bendall SC, Simonds EF, Qiu P et al (2011)
Highly multiplexed simultaneous detection of Single-cell mass cytometry of differential
RNAs and proteins in single cells. Nat Methods immune and drug responses across a human
13:269 hematopoietic continuum. Science 332
9. Behbehani GK, Bendall SC, Clutter MR, Fantl (6030):687–696
WJ, Nolan GP (2012) Single-cell mass cytome- 18. Behbehani GK (2018) Cell cycle analysis by
try adapted to measurements of the cell cycle. mass cytometry. In: Lacorazza HD
Cytometry A 81(7):552–566 (ed) Cellular quiescence: methods and proto-
10. Edgar LJ, Vellanki RN, Halupa A, Hedley D, cols. Springer New York, New York, NY, pp
Wouters BG, Nitz M (2014) Identification of 105–124
hypoxic cells using an organotellurium tag 19. Behbehani GK, Thom C, Zunder ER et al
compatible with mass cytometry. Angew (2014) Transient partial permeabilization with
Chem Int Ed Engl 53(43):11473–11477 saponin enables cellular barcoding prior to sur-
11. Han G, Spitzer MH, Bendall SC, Fantl WJ, face marker staining. Cytometry A 85
Nolan GP (2018) Metal-isotope-tagged (12):1011–1019
monoclonal antibodies for high-dimensional 20. Hartmann FJ, Simonds EF, Bendall SC (2018)
mass cytometry. Nat Protoc 13 A universal live cell barcoding-platform for
(10):2121–2148 multiplexed human single cell analysis. Sci
12. Chang Q, Ornatsky OI, Koch CJ et al (2015) Rep 8(1):10770
Single-cell measurement of the uptake, intratu- 21. Chow S, Hedley D, Grom P, Magari R, Jacob-
moral distribution and cell cycle effects of cis- berger JW, Shankey TV (2005) Whole blood
platin using mass cytometry. Int J Cancer 136 fixation and permeabilization protocol with red
(5):1202–1209 blood cell lysis for flow cytometry of intracellu-
13. Chevrier S, Crowell HL, Zanotelli VRT, lar phosphorylated epitopes in leukocyte sub-
Engler S, Robinson MD, Bodenmiller B populations. Cytometry A 67(1):4–17
(2018) Compensation of signal spillover in sus- 22. Finck R, Simonds EF, Jager A et al (2013)
pension and imaging mass cytometry. Cell Syst Normalization of mass cytometry data with
6(5):612–620 e615 bead standards. Cytometry A 83(5):483–494
Chapter 3
Abstract
Immunophenotyping using flow cytometry highly benefits from multiplexing samples for generation of
more robust data, because of reduction of antibody consumption, batch effect and technical variations. One
way to multiplex is via fluorescent cell barcoding (FCB) prior to staining procedure.
FCB is a high-throughput multiplexed assay using various concentrations of different fluorescent dyes.
Individual samples are uniquely labeled, then mixed together, stained and analyzed as a single sample,
decreasing technical variations and increasing throughput and speed of acquisition. In addition, FCB
simplifies implementation of normalization using a bridge control sample.
In this chapter, we illustrate the protocol for FCB and recommendations for choosing barcoding dyes and
concentrations among other technical considerations.
1 Introduction
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_3, © Springer Science+Business Media, LLC, part of Springer Nature 2019
53
54 Valentina Giudice et al.
Perm Buffer II
DyLight 800
0 13 250
cells/30μL
DyLight 800
15-25 min incubation on ice
Fig. 1 Illustration of FCB protocol for a 3 3 matrix using DyLight 350 and DyLight 800. In (1) FCB dye
preparation is shown how to perform dye dilution (Subheading 3.1); then, 5 μL of each dye at various
Fluorescent Cell Barcoding 55
2 Materials
2.1 FCB Buffers Barcoding dyes from Thermo Fisher Scientific (Waltham, MA,
and Dyes USA): DyLight 350—NHS; DyLight 800—NHS; and Pacific
Orange—NHS (see Note 1).
Barcoding buffers from BD Biosciences (San Jose, CA, USA):
BD Perm Buffer II; 5 BD Phosflow Lyse/Fix Buffer; 4 BD
Phosflow Barcoding Wash Buffer.
Other reagents: Ficoll-Paque gradient centrifugation (MP Bio-
medicals, LLC, Santa Ana, CA, USA), according to the manufac-
turer’s instructions for PBMC isolation; Ack Lysing buffer (Quality
Biological, Gaithersburg, MD, USA) for red blood cell lysis; Phos-
phate buffer saline (PBS) and dimethylsulfoxide (DMSO) Sigma-
Aldrich (St. Louis, MO, USA).
Fig. 1 (continued) concentrations are pipetted in appropriate wells (according to colors). In two separate
wells, 10 μL of DyLight 350 or DyLight 800 are added and will be used as single-color controls. In another well
no dye is added and 40 μL of unstained sample will be pipetted, will be considered unstained control for
cytometer setting. Once prepared, dyes can be kept on ice until use. In (2) Sample preparation,
0.6–1 106 cells/well per sample of fresh PBMCs or frozen PBMCs or fresh whole blood after RBC lysing
are incubated for 15 min at RT with 3 mL of BD Lyse/Fix Buffer. After centrifugation, supernatants are removed
and cell pellets loosen; then, 3 mL of cold 1 Perm Buffer II are added and samples incubated for 20 min on
ice. After incubation, cells are washed with 2 mL of PBS and centrifuge at 400 g for 5 min. Cells are
subsequently resuspended in cold 1 Perm Buffer II to have 0.6–1 106 cells/30 μL. In (3) FCB, samples are
added in appropriate wells prepared with FCB dyes and incubated on ice for 15–25 min. Samples from the
3 3 matrix are combined together, while single-color controls and unstained sample are transferred to
separate tube. Then cells are washed with 3 mL of 1 Barcoding Wash Buffer and centrifuge at 400 g for
5 min. In (4) Antibody staining, after centrifugation, cells are resuspended in 300 μL of Wash Buffer and
antibodies are added according to the manufacturer’s instructions. After 20–30 min incubation at RT, samples
are washed with 3 mL of Barcoding Wash Buffer, centrifuged, and resuspended in 300 μL of the same buffer
for cytometer acquisition
56 Valentina Giudice et al.
2.2 Buffer and Dye All buffers and dyes are prepared using PBS and stored as indicated.
Preparation
1. Prepare 1 BD Perm Buffer II by diluting 1:2. Add 10 mL of
BD Perm Buffer II at room temperature (RT) and 10 mL of
cold PBS to a 50 mL Falcon Conical tube (Fisher Scientific,
Loughborough, UK), mix by inversion and store at +4 C.
2. Prepare 1 BD Phosflow Lyse/Fix Buffer from the 5 stock
solution. Add 10 mL of 5 BD Phosflow Lyse/Fix Buffer and
40 mL of PBS at RT to a 50 mL Falcon Conical tube, mix by
inversion and store at RT.
3. Prepare 1 Barcoding Wash Buffer by diluting 1:4. Add 5 mL
of cold 4 Barcoding Wash Buffer and 35 mL of PBS at RT to a
50 mL Falcon Conical tube, gently mix by inversion and store
at RT (use the solution within 1–2 days).
4. Dyes used in this protocol are DyLight 350 and DyLight 800.
Upon arrival dry powder dyes are reconstituted at 500 μg/mL
concentration in DMSO in 15 mL Falcon Conical tubes
(Fisher Scientific, Loughborough, UK). For 1 mg of dry pow-
der, add 1 mL of DMSO to the tube containing the powder
and transfer to a 15 mL Falcon Conical tube; then rinse with
1 mL of DMSO the tube and transfer to the 15 mL Falcon
tube. Mix well by pipetting and aliquot in 1.5 mL Eppendorf
tubes. Store at 80 C protected from light (see Note 2).
3 Methods
3.1 Sample FCB for lymphocyte phenotyping can be performed on fresh sam-
Preparation ples, as whole blood or peripheral blood mononuclear cells
(PBMCs) or cryopreserved PBMCs. If frozen PBMCs are used,
thaw cells in a water bath at +37 C for 1–2 min. Add 2 mL of PBS
and centrifuge at 400 g for 5 min. Discard supernatant and
vortex to loosen cell pellets. If fresh PBMCs are used for barcoding,
start from step 5.
If whole fresh blood is employed in the experiment:
1. Collect at least 0.2 mL of whole blood in the presence of
anticoagulant (EDTA or sodium heparin).
2. Lyse with 1:10 Ack Lysing buffer. For 0.2 mL of fresh blood,
add 2 mL of Ack Lysing buffer and incubate for 10 min on ice
(see Note 3).
3. Centrifuge at 400 g for 5 min and discard supernatant.
4. Repeat steps 2 and 3.
5. Add 3 mL of 1 Lyse/Fix Buffer, gently mix by pipetting and
incubate at RT for 15 min.
6. Centrifuge at 400 g for 5 min. Discard supernatant and
vortex to loosen the cell pellets.
Fluorescent Cell Barcoding 57
3.2 Nine-Sample Prepare dye working concentrations with DMSO from stock con-
Matrix Preparation centration immediately before starting the experiment, as DMSO
(3 3) tends to precipitate.
1. Thaw dye stock solution (500 μg/mL) at RT protected from
light. After thawing, vortex vigorously.
2. Prepare dye working concentrations with DMSO in round
bottom 96-well plate and label wells as “DyL350-0”,
“DyL350-13”, “DyL350-250” and “DyL800-0”, “DyL800-
13”, “DyL800-250” (see Note 5).
For each dye, prepare the following dilutions:
250-dilution ¼ 25 μL of 500 μg/mL dilution + 25 μL of
DMSO
50-dilution ¼ 10 μL of 250-dilution + 40 μL of DMSO
13-dilution ¼ 13 μL of 50-dilution + 37 μL of DMSO
0-dilution ¼ 30 μL of DMSO
The 50-dilution is used only for preparing the 13-dilution
and not for FCB matrix preparation (see Note 6).
3. Add 5 μL of diluted dye to appropriate well in a round bottom
96-well plate as shown in Fig. 1. For a final volume of 40 μL/
well (10 μL of dyes +30 μL of sample), final concentrations will
be 0, 3.25, and 62.5 μg/mL.
4. Seal or cover the plate and keep at RT protected from light.
5. Proceed to Subheading 3.4.
3.3 Twelve-Sample Prepare dye working concentrations from stock concentration right
Matrix Preparation before starting the experiment, as DMSO tends to precipitate.
(4 3)
1. Thaw dye stock solution (500 μg/mL) at RT protected from
light. After thawing, vortex vigorously. In a round bottom
96-well plates, prepare dye working concentrations with
DMSO as follows (Fig. 2).
2. For DyLight 350, name tubes/wells as follows: “DyL350-0”,
“DyL350-30”, “DyL350-150” and “DyL350-500”. Prepare
dilutions as follows:
58 Valentina Giudice et al.
Pacific Orange
500
50
25 μL 40 μL 40 μL
DMSO DMSO DMSO
0
500 250 50 0
DyLight 350
DyLight 350
0 30 150 500
15 μ
μL 10 μL
μ
35 μL 40 μL 40 μL
DMSO DMSO DMSO
500 150 30 0
3.4 Fluorescent Cell Once matrix and samples are prepared, proceed to FCB as follows.
Barcoding Perform all steps at RT unless otherwise specified.
1. Add 30 μL of each sample into appropriate well, according to
their matrix position. Mix well the solution as dyes tend to
precipitate.
2. Add 30 μL of samples into one well with 10 μL of DyLight
350 and other 30 μL of sample into a different well with 10 μL
of DyLight 800. These samples were prepared in step 9 of
Subheading 3.1 and will be used as single-color controls for
cytometer setting.
3. Seal or cover the plate and incubate for 15–25 min on ice.
Longer incubation time is preferable for whole blood barcod-
ing (see Note 7).
4. Transfer all nine barcoded samples into one FACS tube named
“Combo”. Transfer single-color controls prepared in step 2 in
two separate FACS tubes labeled “DyL350” and “DyL800.”
5. Wash with 3 mL of 1 Barcoding Wash Buffer (see Note 8).
Centrifuge at 400 g for 5 min and discard supernatant.
Vortex to loosen cell pellets. Repeat step 5 (see Note 9).
6. Resuspend “Combo”, “DyL350”, and “DyL800” in 300 μL of
1 Barcoding Wash Buffer. Keep “DyL350” and “DyL800”
on ice until cytometer setting.
7. Add antibodies according to the manufacturer’s instructions to
the appropriate tube and incubate at RT in the dark for
20–30 min.
8. Wash with 3 mL of 1 Barcoding Wash Buffer. Centrifuge at
400 g for 5 min, then discard supernatant and vortex to
loosen cell pellet. Repeat washing step one more time.
9. Resuspend cells in 300–500 μL of 1 Barcoding Wash Buffer
and keep on ice or +4 C until acquisition.
10. Acquisition can be carried out using appropriate lasers
(355, 407, or 630 nm) on cytometers. Preacquisition compen-
sation can be calculated using bead standards for each fluoro-
chrome (anti-mouse Ig, κ/Negative Control Compensation
Particles Set, BD Biosciences) and prepared barcoded cells
with the highest concentration of each dye. For each fluoro-
chrome, cells as well as single-color controls can be used.
A minimum of 50,000 lymphocytes needs to be recorded.
3.5 Control Samples During protocol optimization, a bridge sample within one matrix is
for Protocol used in order to measure intra-assay variability. Indeed, percent of
Optimization positive cells and MFI values should not significantly differ across
all 9/12 barcoded populations within the same matrix.
Control samples are (1) “No fix/perm” control, without fixa-
tion, permeabilization, and barcoding; and (2) “No FCB” control,
60 Valentina Giudice et al.
3.5.1 “No fix/perm” 1. Resuspend cells from step 4 of Subheading 3.1 in PBS.
Control Preparation 2. Add antibodies according to the manufacturer’s instructions
and incubate at RT for 20–30 min in the dark.
3. Wash with 3 mL of PBS, centrifuge at 400 g for 5 min and
discard supernatant. Vortex to loosen cell pellets.
4. If intracellular staining is required, add 200 μL of CytoFix/
CytoPerm BD buffer and incubate at +4 C for 30 min. Other-
wise skip to step 7.
5. Wash with 2 mL of 1 Perm Wash, centrifuge at 400 g for
5 min, discard supernatant, and vortex to loosen cell pellets.
6. Resuspend cells in 175 μL of 1 Perm Wash and add intracel-
lular markers antibodies according to the manufacturer’s
instructions. Incubate at +4 C for 30–40 min in the dark.
7. Wash with 2 mL of 1 Perm Wash. Centrifuge at 400 g for
5 min, discard supernatant and vortex to loosen cell pellets.
8. Resuspend cells in 300 μL of 1 Perm Wash for acquisition.
3.5.2 “No FCB” Control 1. Prepare cells as in Subheading 3.1 (steps 1–4).
Preparation 2. After centrifugation, add 3 mL of 1 Lyse/Fix Buffer and
incubate at RT for 15 min.
3. Centrifuge at 400 g for 5 min. Discard supernatant and
vortex to loosen the cell pellets.
4. Add 3 mL of cold 1 Perm Buffer II. Incubate on ice for
20 min.
5. Add 2 mL of PBS and centrifuge at 400 g for 5 min. Discard
supernatant and vortex to loosen cell pellets.
6. Resuspend cells in 200 μL of 1 Barcoding Wash Buffer.
7. Add antibodies according to the manufacturer’s instructions to
the appropriate tube and incubate at RT in the dark for
20–30 min.
8. Wash with 3 mL of 1 Barcoding Wash Buffer. Centrifuge at
400 g for 5 min, then discard supernatant and vortex to
loosen cell pellet. Repeat washing step one more time.
9. Resuspend cells in 300–500 μL of 1 Barcoding Wash Buffer
and keep on ice or +4 C until acquisition.
Fluorescent Cell Barcoding 61
3.6 Post-Acquisition Post-acquisition analysis can be carried out using FlowJo or RStu-
Analysis dio software. Recommended analysis steps using both software are
described below.
3.6.2 RStudio RStudio also can be used using the following packages (flowCore,
flowClust, flowViz, flowWorkspace, ggcyto, and flowType)
(Fig. 4a). In RStudio, antibodies are identified by cytometer chan-
nel used during acquisition (e.g., on LSR Fortessa cytometer,
DyLight 350 was acquired using the channel BUV396-A and
DyLight 800 with APC-H7-A channel).
1. Remove debris by filtering the data using rectangleGate func-
tion and FSC-A and SSC-A parameters. Events with FSC values
lower than 20 k are removed by using the Subset method.
2. Compensate data using the matrix made in step 1 of FlowJo
analysis and compensate function of flowWorkspace.
3. Transform compensated data using arcsinh function.
4. Filter data for FCB dye channels (e.g., BUV.396.A and APC.
Cy7.A) by rectangleGate function.
5. Identify barcoded populations using flowClust function. Clus-
tering is performed by setting varNames parameter on FCB dye
channels (e.g., BUV.396.A and APC.Cy7.A) and K is set based
on the number of barcoded samples in the matrix (9 or 12).
62 Valentina Giudice et al.
250K
105
200K
4
10
150K
SSC-A
FSC-H
103 100K
0
50K
0
0 50K 100K 150K 200K 250K 0 50K 100K 150K 200K 250K
FSC-A FSC-A
105
250
DyLight 350
DyLight 350
104
13
103
0
102
DyLight 800 0
Fig. 3 Manual deconvolution using FlowJo. In order to gate the nine barcoded populations using FlowJo
software, first lymphocytes are identified based on linear parameters (FSC-A vs SSC-A) and double cells
excluded (FSC-H vs FSC-A). On single cells, barcoded populations are shown using FCB dye parameters
(DyLight 350 vs DyLight 800) and samples are visualized on a dot plot according to dye concentrations (bottom
left panel)
3.7.1 MFI Fold Increase 1. Make post-acquisition compensation matrix in FlowJo soft-
Calculation ware and apply it to barcoded samples.
2. Identify lymphocytes and/or monocytes based on forward
scatter area (FSC-A) and side scatter area (SSC-A).
3. Exclude doublets based on FSC-A vs FSC-H.
4. On single cells, shown each FCB dye as histogram and identify
each dye concentration used and name populations accordingly
(e.g., “DyL350-0”, “DyL350-13”, “DyL350-250” and
“DyL800-0”, “DyL800-13”, “DyL800-250” when a nine-
sample matrix is made).
5. Calculate MFI and CV for each FCB dye dilution. Export data
as .csv or .xls file.
6. In a spreadsheet, calculate MFI fold change
as follows: MFI
fold increase ¼ MFIpeak2 CV peak2 = MFIpeak1 þ CV peak1 :
Fluorescent Cell Barcoding 65
100 100
MFI fold increase
13μg/ml / 0μg/ml
Normalized Barcoded
80 80
[MFIpeak2 – CVpeak2]
Cell Number
60 60
[MFIpeak1 + CVpeak1]
40 40
MFI fold increase
20 20 250μg/ml / 13μg/ml
[MFIpeak2 – CVpeak2]
0 0
0 102 103 104 105 0 102 103 104 105
[MFIpeak1 + CVpeak1]
DyLight 350
Control
Mean SD M+2SD M-2SD
w/o FCB
95.5 2.4 100.3 90.8 95.7
Variability Ratio
LOW Mean
M-2D < Control w/o FCB < M+2D 0.8 < < 1.2
Control w/o FCB
HIGH
Control w/o FCB < M-2D
or
Control w/o FCB > M+2D
Fig. 5 FCB efficacy and inter-assay/intra-assay variability. (a) FCB efficacy is calculated using the MFI fold
change. For each dye, MFI values are calculated for each of the three concentrations and MFI fold
change
is determined
according to the following formula: MFI fold increase ¼ MFI peak 2
CV peak 2
=
MFIpeak1 þ CVpeak1 : (b) Inter-assay/intra-assay variability is assessed for each fluorochrome and percent
of cell population (Subheading 3.7). For intra-assay variability, using a 3 3 matrix with the same donor in all
nine positions and matched No-FCB controls, range of variability is calculated as < mean in controls –2SD or
> mean in controls 2SD and percent of barcoded populations after deconvolution should be within this
range. For inter-assay variability, the average of all barcoded samples across all experiments is calculated and
compared to the average of all matched control samples used. Values should be within mean of controls
2SD. The ratio of variability is calculated as the ratio between the mean of barcoded samples and the mean
of matched No-FCB controls. The ratio should be within 0.8–1.2
66 Valentina Giudice et al.
For example, MFI fold increase ¼ MFIDyL350‐13 CV DyL350‐
13= MFIDyL350‐0 þ CV DyL350‐0 MFI fold increase ¼ MFIDyL
350‐250 CV DyL350‐250 = MFIDyL350‐13 þ CV DyL350‐13 .
3.7.2 Intra-assay/Inter- 1. Calculate percent of positive cells (e.g., CD3+ cells, CD4+
assay Variability of Percent cells) in control samples using FlowJo software and export
of Positive Cells Calculation data as .csv or .xls file.
2. Calculate the range of variability as <mean in controls –2SD or
>mean in controls +2SD (Fig. 5b).
3. Compare percent of barcoded populations after deconvolution
to matched control samples. Values should be within 2SD.
4. For intra-assay variability, for each cell population (e.g., CD3+
cells), calculate the average across all barcoded samples within
one matrix and compare to the average of all matched control
samples used in that matrix. Values should be within mean of
controls 2SD.
5. For inter-assay variability, of each cell population (e.g., CD3+
cells), calculate the average of all barcoded samples with the
same FCB dye dilution (e.g., 0 μg/mL) across all matrix and
compare to the average of all matched control samples used.
Values should be within mean of controls 2SD.
3.7.3 Intra-assay/Inter- 1. Calculate MFI for each antibody used in barcoded samples
assay Variability of MFI using FlowJo software and export data as .csv or .xls file.
Values for Each 2. Calculate the range of variability as <mean in controls –2SD or
Fluorochrome Calculation >mean in controls +2SD.
3. Compare MFI values of barcoded populations after deconvolu-
tion to population barcoded with lowest concentration (e.g.,
0 μg/mL) (see Notes 10 and 11). Values should be within 2SD.
4. For intra-assay variability, for each cell population (e.g., CD3+
cells), calculate the average across all barcoded samples within
one matrix and compare to the average of all matched control
samples used in that matrix. Values should be within mean of
controls 2SD.
5. For inter-assay variability, for each cell population (e.g., CD3+
cells), calculate the average of all barcoded samples with the
same FCB dye dilution (e.g., 0 μg/mL) across all matrix and
compare to the average of all matched control samples used.
Values should be within mean of controls 2SD.
4 Notes
References
1. Finak G, Langweiler M, Jaimes M et al (2016) throughput drug screening and signaling
Standardizing flow cytometry immunopheno- profiling. Nat Methods 3:361–368
typing analysis from the human immunopheno- 5. Giudice V, Feng X, Kajigaya S, Young NS, Bian-
typing consortium. Sci Rep 6:20686 cotto A (2017) Optimization and standardiza-
2. Maecker HT, JP MC Jr, FOCIS Human Immu- tion of fluorescent cell barcoding for
nophenotyping Consortium et al (2010) A multiplexed flow cytometric phenotyping. Cyto-
model for harmonizing flow cytometry in clini- metry A 91:694–703
cal trials. Nat Immunol 11(11):975–978 6. Lim CY, Owens NA, Wampler RD et al (2014)
3. Maecker HT, McCoy JP, Nussenblatt R (2012) Succinimidyl ester surface chemistry: implica-
Standardizing immunophenotyping for the tions of the competition between aminolysis
human immunology project. Nat Rev Immunol and hydrolysis on covalent protein immobiliza-
12(3):191–200 tion. Langmuir 30(43):12868–12878
4. Krutzik PO, Nolan GP (2006) Fluorescent cell
barcoding in flow cytometry allows high-
Chapter 4
Abstract
Antibody reagents that are used for flow cytometry immunophenotyping have traditionally been prepared
by combining individual liquid antibody conjugates into mixtures. These cocktails have limited shelf-life,
and their preparation is time-consuming and prone to laboratory error. Manufacturers of these reagents, in
collaboration with several clinical and research centers, have made advances in constructing dried antibody
cocktails which have addressed many of the problems inherent in preparing the liquid cocktails on the lab
bench. This chapter discusses methods for the use of dried reagents.
1 Introduction
From the outset, one of the fundamental uses for flow cytometry
has been immunophenotyping of cell suspensions using antibody
cocktails. Throughout the evolution of this methodology, the par-
adigm for the procedure has been and continues to consist of
(1) preanalytical steps including processing of a cellular substrate
to be stained and preparation of antibody cocktails, (2) analytical
steps including cell staining and data acquisition using cytometers,
and (3) postanalytical steps including off-line data analyses. There
have been marked advances in each of these steps, including the
number of markers contained in cocktails, the variety of fluoro-
chromes available to be conjugated with these markers, the number
of parameters available on cytometers that are manufactured with
various technologies for creating flow streams, and the progression
of the data analysis strategies.
Throughout this process, there have always been the goals of
maximizing accuracy, precision, and reproducibility, while minimiz-
ing variability and cost. These goals are essential in performing
good science, but they also have a financial impact on the opera-
tions in the research and clinical laboratory realm [1]. One of, if not
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_4, © Springer Science+Business Media, LLC, part of Springer Nature 2019
69
70 Marc Langweiler
2 Materials
2.1 Assay Lyoplate, Each of the five rows contains 12 replicates of the five cocktails (see
BD Biosciences Note 1).
Designate 175 (Fig. 1)
A: T-cell.
B: Treg.
C: B-cell.
Dried Antibody Cocktails 71
Fig. 1 Assay Lyoplate (number 175) from BD Biosciences is a 96-well plate with rows A through F and columns
1 through 12 containing lyophilized antibody cocktail buttons. Each row includes 12 replicates of the complete
cocktail as follows: T-cell (A), Treg (B), B-cell (C), DC-Mono-NK (D), and T-helper (E)
D: DC-Mono-NK.
E: T-helper.
2.2 Compensation Each row contains wells with individual antibodies that are consti-
Lyoplate, BD tuents of one of the five cocktails, as well as negative and positive
Biosciences Designate compensation bead particles. Beads are not used to incorporate
177 (Fig. 2) viability dyes into the compensation matrix. Instead, cellular mate-
rial is stained separately, in tubes with cells, and these are acquired
on the cytometer at the same time as bead acquisition. Several
strategies are available for calculating the compensation matrices
for each cocktail (see Note 2).
2.3 Fluorescence- Each row contains antibodies specific for each of the five cocktails in
Minus-One (FMO) a similar fashion as the compensation Lyoplate. Columns A and B
Lyoplate, BD include all the cocktail antibodies. Cells that have been stained with
Biosciences Designate viability dye are added to column A which represents the “com-
176 (Fig. 3) plete” cocktail. Cells that have not been stained with viability dye
are added to column B which represents the viability dye FMO. The
remaining columns contain the antibody cocktail minus one of the
antibody conjugate constituents (see Note 3).
2.5 Supplemented Staining Buffer: PBS supplemented with 1% fetal calf serum, 1%
PBS normal mouse serum, 0.02% sodium azide (see Note 4).
72 Marc Langweiler
Fig. 2 Compensation Lyoplate (number 177) from BD Biosciences is a 96-well plate with rows A through F and
columns 2 through 10 containing lyophilized buttons of a single antibody from each cocktail per well along
with both negative and positive beads that are included in BD Biosciences Anti-Mouse Ig Compensation
Particles Kit (part number 552843). Each row contains the constituents of one of the five cocktails as follows:
T-cell (A), Treg (B), B-cell (C), DC-Mono-NK (D), and T-helper (E). The antibody in well E3 is a rat monoclonal
anti-CXCR5 conjugate; therefore, the compensation bead set for this well is derived from the BD Biosciences
Anti-Rat Ig Compensation Particle Kit (catalog number 552844)
Fig. 3 FMO Lyoplate (number 176) from BD Biosciences is a 96-well plate with rows A through F and columns
1 through 11 containing lyophilized antibody buttons. Columns 1 and 2 are complete cocktails as follows:
T-cell (A), Treg (B), B-cell (C), DC-Mono-NK (D), and T-helper (E). Columns 3 through 11 are “minus-one” of
the constituents of the cocktail for that row
Dried Antibody Cocktails 73
2.6 Fixation Medium 2% formalin solution in PBS, pH 7.4: Prepared fresh from a 10%
methanol-free buffered formalin stock (Polysciences, catalog num-
ber 04018-1).
2.7 Viability Dye Fixable Blue Live/Dead Cell Stain Kit (ThermoFisher Scientific,
catalog number L23105): The kit includes four vials of frozen dye
and one vial of dimethyl sulfoxide (DMSO) diluent. Stock solutions
are prepared by addition of 50 μl of DMSO to each of the four vials
of dye, which are pooled. Aliquots of 16 μl are placed in screw-
capped freezing vials which are stored at 80 C. One of these vials
is sufficient to stain cells from five different samples.
2.8 Cell Samples PBMC: peripheral blood mononuclear cells that had been prepared
using density gradient centrifugation and preservation in liquid
nitrogen for long-term storage (see Note 5).
3 Methods
3.1 Processing 1. PBS is added to each well in 100 μl volumes, starting in well
the Compensation A1, proceeding down the column then moving to the top of
Lyoplate successive columns.
2. After addition of PBS to all wells, the plate is incubated for
30 min at the ambient temperature, protected from light.
There is a 5-min period during which the added buffer dis-
solves the lyophilized button; the plate is placed on a rotary
plate mixer at low speed for the remaining 25 min.
3. At the end of this interval, the plate is centrifuged (ambient
temperature, 400 g, for 5 min) and flicked to remove super-
natants, followed by addition of 100 μl of Staining Buffer. The
contents of each well are transferred into labeled polystyrene
tubes and held at 4 C until cytometer acquisition, which is
completed within 30 min of staining. Beyond this interval, the
beads will begin to deteriorate, which may compromise the
validity of the compensation matrices.
3.2 Processing Cell 1. After cells have been thawed and washed, 1 ml of PBS is added
Samples to the residual volume remaining in each tube.
2. Before the addition of 1 μl of the viability dye working solution,
a 50 μl aliquot of cell suspension is removed and added to
another labeled tube containing 100 μl of Staining Buffer, to
be acquired on the cytometer as unstained samples.
3. The tubes with cells and viability dye are incubated, at the
ambient temperature, protected from light for 5 min, followed
by the remaining 25 min on a rotating plate mixer.
4. At the end of this interval, the tubes are centrifuged (ambient
temperature, 400 g, for 5 min), decanted one-by-one, and
the pellets resuspended by gentle repipetting in their residual
supernatant with addition of 450 μl of Staining Buffer, to
ensure sufficient volume for staining the five wells for each
sample (100 μl per well).
3.3 Processing 1. For either plate, once removed from its pouch, and peeling off
the FMO and Assay of the foil cover, 100 μl of cells from Subheading 3.2 is added
Lyoplates into rows A through E in the column assigned to each sample.
Pipetting of sample starts in column 1 and proceeds through
column 10, using a new pipette tip for each well.
2. Once all wells are filled, a 30-min incubation at the ambient
temperature ensues, with the plate protected from light. After
the first 5 min, the cells are resuspended, column-by-column,
using an eight-channel multipipettor set at 80 μl. The suspen-
sions are mixed by gentle repetitive pipetting. After addition to
each column, the tips are discarded and replaced with new tips
Dried Antibody Cocktails 75
4 Notes
CCR7∗PE/CD45RA∗PE-Cy7/CD38∗APC/
CD8∗APC-H7>
Row B (Treg) <viability dye/CD45∗BUV/CD3∗V450/
HLA-DR∗V500/CD39∗FITC/CD4∗PerCP-Cy5.5/
CD25∗PE/CCR4∗PE-Cy7/CD127∗AF647/
CD45RO∗APC-H7>
Row C (B-cell) <viability dye/CD45∗BUV/CD3∗V450/
IgD∗V500/CD10∗BB515/CD19∗PerCP-Cy5.5/
CD24∗PE/CD27∗PE-Cy7/CD38∗APC/
CD20∗APC-H7>
Row D (DC-Mono-NK) <viability dye/CD45∗BUV/
CD14∗V450/HLA-DR∗V500/CD163∗FITC/
CD123∗PerCP-Cy5.5/CD56∗PE/CD11c∗PE-Cy7/
CD16∗APC/CD3 + CD19 + CD20∗APC-H7>
Row E (T-helper) <viability dye/CD45∗BUV/CD3∗V450/
HLA-DR∗V500/CXCR5∗BB515/CD4∗PerCP-
Cy5.5/CXCR3∗PE/CCR6∗PE-Cy7/CD38∗APC/
CD8∗APC-H7>
2. The original iteration of the Lyoplate product for the HIPC
project [11] used a single plate that contained the compensa-
tion beads and the assay cocktails. Combining the assay and
compensation wells in one plate results in the compensation
beads being washed more aggressively and incubating longer
than necessary, leading to inconsistencies when calculating
matrices. An alternative strategy is to use separate plates for
compensation and assay. Using a separate compensation
plate—with more wells available—allows the use of the full
complement of the conjugates for each cocktail. This offers
the opportunity for two strategies to construct the matrices.
In the first scenario, separate matrices are constructed for each
cocktail, using the full complement of wells in each row. In the
second scenario, only one well of conjugates with replicates
present in more than one cocktail is acquired. Following data
acquisition, the experiment in the cytometer software is repeat-
edly duplicated resulting in five copies, one for each cocktail.
The data files that are not relevant for a given cocktail are then
deleted before the matrix is constructed. With regards to the
frequency of preparation/data acquisition/data analysis of the
Compensation Lyoplate, the expectation at the outset, when
the compensation wells and antibody conjugate wells were
included on a single plate, was that new matrices would be
constructed for every plate. In the protocol described herein,
matrices were constructed with much less frequency—only
when there were significant changes to the cytometer instru-
ment optics/fluidics/electronics during servicing. The rela-
tively low number of parameters associated with the panels
Dried Antibody Cocktails 77
References
1. Hammerling JA (2012) A review of medical 3. van Dongen JJM, Lhermitte L, Böttcher S et al
errors in laboratory diagnostics and where we (2012) EuroFlow antibody panels for standar-
are today. Lab Med 43(2):41–44 dized n-dimensional flow cytometric immuno-
2. Jamin C, Lann L, Alvarez-Errico D et al (2016) phenotyping of normal, reactive and malignant
Multi-center harmonization of flow cytometers leukocytes. Leukemia 26(9):1908–1975
in the context of the European “PRECI- 4. Van der Velden VHJ, Flores-Montero J, Perez-
SEADS” project. Autoimmun Rev 15 Andres M et al (2017) Optimization and test-
(11):1038–1045 ing of dried antibody tube: the EuroFlow LST
and PIDOT tubes as examples. J Immunol
Dried Antibody Cocktails 79
Abstract
“Gating” refers to the selection of successive subpopulations of cells for analysis in flow cytometry. It is
usually performed manually, based on expert knowledge of cell characteristics. However, there can be
considerable disagreement in how gates should be applied, even between individuals experienced in the
field. While clinical software often automates gating, and some guidelines do exist (especially for clinical
assays), there are no comprehensive guidelines across the various types of immunological assays performed
using flow cytometry. Here we attempt to provide such guidelines, focused on the most general and
pervasive types of gates, why they are important, and what recommendations can be made regarding
their use. We do so through the display of example data, collected by academic, government, and industry
representatives. These guidelines should be of value to both novice and experienced flow cytometrists
analyzing a wide variety of immunological assays.
1 Introduction
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_5, © Springer Science+Business Media, LLC, part of Springer Nature 2019
81
82 Janet Staats et al.
Table 1
Factors affecting population resolution in flow cytometry
2 Guidelines
2.1 Acquisition Definition: When acquiring samples using a flow cytometer, the
Threshold or cells of interest are usually in a suspension that contains many other
Trigger Gate cells or particles. Each cell or particle is measured by the cytometer
as an event in the form of a pulse; some events represent signals of
Flow Cytometry Gating Guidelines 85
interest and some are noise. An operator may set a limit for the type
of pulse that is acceptable to be included in the resulting data file;
this is called an acquisition (detection) threshold or trigger gate.
Trigger gates are set prior to acquisition as a means of excluding
events of noninterest or noise. The most common type of trigger
gate is used to exclude debris based on size, measured using for-
ward scatter. Using forward scatter, the trigger gate is set to exclude
events that are too small to be considered cells. Another type of
commonly used trigger gate is based on CD45 expression, where
CD45 is a panleukocyte marker (see Fig. 1). Setting the trigger gate
on CD45+ cells will essentially exclude all nonleukocyte events.
Note that on some cytometers, a storage gate can be set in
addition to an acquisition threshold or trigger. Care should be
taken that the storage gate as well as the acquisition gate includes
all populations of potential interest for later analysis. For example, a
storage gate set on lymphocytes (using forward vs. side scatter)
would preclude downstream analysis of monocytes.
Fig. 1 A CD45 trigger gate (threshold) is used to exclude nonleukocytes. In a sample heavily contaminated with
erythrocytes, a CD45 threshold can help to exclude these cells and “clean up” the downstream gates. In this
example, the same erythrocyte-contaminated sample was run with a high, medium, or low threshold on CD45.
Left panels: With no further gating on CD45, the initial FSC vs. SSC gate for lymphocytes (first column) is
uncontaminated with a high CD45 threshold (top row), but progressively more contaminated as the CD45
threshold is reduced. This results in an artificially low observed percentage of B cells (second column), due to
dilution of lymphocytes by contaminating erythrocytes. Right panels: The same data can be effectively rescued
by applying a postacquisition gate on CD45high events (first column), prior to lymphocyte gating (second
column). This results in similar observed percentages of B cells (third column), albeit at the expense of greatly
diminished cell numbers when there was a low CD45 threshold on acquisition
86 Janet Staats et al.
2.2 Time Gate Definition: Time was first introduced as a flow cytometry parameter
to measure kinetic responses [30]. Subsequently, in 1987, James
Watson first described the concept of using time as a quality control
(QC) parameter [8]. The principle of flow cytometry is based on
regulating the differential pressure between sample and sheath fluid
to establish a laminar flow, enabling a single cell to pass in front of
the laser for interrogation. The steps involved with acquiring a
sample influence the fluid dynamics throughout the course of
acquisition. When a sample is first introduced into the cytometer,
a burst of pressure may cause a high density of events to pass in
front of the laser during the first few seconds. After the initial
sample introduction, the flow rate stabilizes and becomes laminar.
Nearing the end of acquisition, the sample volume is greatly
reduced, which may influence the fluid dynamics again. During
acquisition, fluidic changes may be observed as either an increase
or decrease in the event rate. The most frequent culprit is air
bubbles or clogs. For these reasons, the time parameter may be
used during analysis to exclude those events that were acquired
during periods of nonlaminar flow. This is called a time gate. For
manual analysis, a time gate is created by visualizing time versus
scatter or fluorescence. Alternatively, there are automated software
options, such as FlowClean [9], that may be used to effectively
remove anomalies, such as those caused by changes in the event
rate, from an FCS file. Of note, the time parameter may be dis-
played differently based on the software being used for acquisition
or analysis.
How data is affected: Anomalies in the flow rate result in wider
CVs and/or shifts in scatter and fluorescence parameters (see
Fig. 2). The effect of nonlaminar flow on the resulting data may
be modest or severe, depending upon the degree of fluidic instabil-
ity and the integrity of any subsequent quality control gates, such as
singlet and fluorescence gates. In the most severe case, an entire
data file may be unacceptable for analysis due to unstable fluidics
that occurred throughout the time of acquisition.
Conclusion: The time parameter is an effective QC tool for
identifying temporal changes in fluid dynamics that occurred
Flow Cytometry Gating Guidelines 87
Fig. 2 Use of time gates. (a) Time as a QC parameter. PBMCs were thawed, permeabilized, stained, and
acquired. Time versus SSC-A is used to visualize the temporal changes in fluid dynamics that occur
throughout the sample acquisition. Left plot illustrates an ideal time parameter plot. The burst of sample at
the beginning, observed as the red or higher density lower scatter population, has an initially wider SSC CV
that rapidly resolves into a tight SSC CV. The SSC CV remains the same throughout the remainder of
acquisition and slightly decreases near the end of acquisition, as the sample rate decreases (red turns to
orange). There are no skips or jumps in the number of events relative to time and no shifting of SSC over time.
Middle plot: moderate nonlaminar flow. A more noticeable sample burst at the beginning of acquisition with
wide SSC-A CV. SSC-A CV changes throughout the acquisition in an oscillating pattern, caused by swirling
fluidics. The time gate, indicated in pink, is set to include the most stable portion of the events acquired. Right
plot: illustrates nonstable fluidics that occurred throughout the entire sample acquisition. Clogs are indicated
by skips or jumps in time, where the event rate is greatly reduced followed by bursts of events when the clog
breaks. Changes in SSC-A CV and shifting of SSC-A are prominent throughout the acquisition. The time gate,
in pink, is placed around the most stable area, although no area in this example is ideal. While a significant
portion of events are excluded due to unstable fluidics, the data included in the time gate represents those
events with the highest integrity. (b) Time gate used to exclude shifting scatter created by nonlaminar flow.
LyoCell PBMC control product was stained with CD14. The gating strategy shown was used to identify CD14+
monocytes by first parsing through time, then viability, singlets (FSC and SSC), and scatter gates. Both rows
represent analyses using the same FCS file. Top row: the time gate is set around the region with best laminar
flow, identifying approximately 71% of monocytes as CD14+. Bottom row: using a time gate set on the region
with nonlaminar flow there are no CD14+ monocytes. (c) Time gating affects CV of downstream gates. Human
PBMC were stained with CD4 PE-Cy7. The leftmost plot is ungated, while the middle and the right histograms
are gated on FSC/SSC Singlets. If placement of time gate includes sections of the plot where nonlaminar flow
exists then the CV as well as median of the CD4+ cell peak and the frequency of CD4+ cells is affected.
Inclusion of events from the unstable fluidics results in a wider CV and higher percent positive cells and
exclusion of these events results in a tighter peak with lower CV and higher fluorescence intensity
88 Janet Staats et al.
2.3 Singlet Gates Definition: Aggregates consist of one or more cells or particles; they
form for many reasons, including cells clumping together due to
dying/death, adherent properties, antigen presentation, and also
high sample concentration, high flow rate, and even dye–dye aggre-
gation. When running clumped samples with many aggregates, it is
important to filter the samples prior to acquisition to remove as
many aggregates as possible. When filtering is insufficient to
remove all aggregates, they may be further removed using singlet
gating. To understand how singlet gates work, it is important to
understand how signals are detected by a flow cytometer. When a
particle passes in front of the laser, the cytometer measures the
resulting photons of light in the form of a pulse. Digital cytometers
are capable of collecting three values that define a pulse: Width is a
measurement that represents the time a cell passes in front of the
laser. Width is proportional to the size of the cell or particle and is
unaffected by PMT voltage settings [31]. Height is the measure-
ment of the intensity of the signal (maximum fluorescence, size, or
scatter) and is affected by PMT voltages. Area is not measured; it is
derived using both width and height measurements, and is only
slightly affected by PMT voltage, owing to its relationship to both
width and height. Area scaling is used in some digital instruments
to equalize height and area. Pulses can be distorted by coincidence,
when two events (a doublet) pass in front of the laser simulta-
neously. This distortion results in a twofold stretching of the area
and width, while the height remains unchanged. During analysis,
singlet gating uses the distortion aggregates impose on pulse width
and area to distinguish singlets from aggregates. Singlet gating is
performed using either forward scatter (FSC) or side scatter (SSC)
parameters. It is important to remember that FSC is collected using
a diode, unless you have an instrument specially equipped with FSC
PMT, and SSC is collected using a PMT. Since PMTs are more
sensitive than diodes, a better resolution between singlets and
aggregates may be visualized using the SSC over FSC parameter.
As a best practice, ideally both FSC and SSC singlet gates would be
used sequentially to remove coincidental events occurring in differ-
ent orientations. There are two common approaches to singlet
gating; one is dependent upon area scaling and one is not: area
versus height and width versus height. For area versus height singlet
gating, coincidence is identified by the events for which there is an
increase in area without a corresponding increase in height. Thus,
cells along the diagonal are singlets and should be included in the
singlet gate. Use of area versus height singlet gating requires the
area scaling to be properly adjusted on your cytometer using cells;
otherwise, there is a risk for gating errors. Maximum resolution
between singlets and aggregates is observed using width versus
Flow Cytometry Gating Guidelines 89
2.4 Exclusion Gates Definition: In the most general sense, an exclusion gate is any gate
that removes unwanted cell populations, before positive gating on
markers of interest. Practically, this is often useful to prevent the
interference of these excluded populations on the downstream
analysis. Besides time and singlet gates already discussed above,
two common variations of such exclusion gates are viability gates
and lineage marker cocktails (often called a “dump channel”).
Viability is most frequently assessed by the fact that live cells have
intact membranes and thus exclude dyes that would otherwise
enter the cell and bind, for example, DNA. Propidium iodide and
7-AAD are examples of such dyes. Alternately, agents that react
with free amine [4] or thiol groups can be used, taking advantage of
the fact that relatively few such reactive groups are present on the
cell surface, but many more are available in the interior of cells.
Many dyes are available in this category, with different fluorescent
properties, such that they can be combined with almost any com-
bination of fluorochrome-labeled antibodies. A major advantage of
these dyes is that they form a covalent bond with cellular proteins,
and thus do not diffuse away if cells are fixed, permeabilized, or
otherwise processed after application of the viability stain. For this
reason, they are often referred to as “fixable” viability dyes.
90 Janet Staats et al.
a WvH AvH
250K
200K
150K
FSC-H
FSC-H
Forward
scatter 75.7
100K
50K
0
FSC-W FSC-A
250K
Side SSC-H
scatter 100K
50K
0
SSC-W SSC-A
104 104
FoxP3 PE
FoxP3 PE
103 103
0 0
Q4 Q3 Q4 Q3
103 26.7 40.6 103 20.4 42.6
Fig. 3 Singlet gating. (a) Width vs height and area versus height singlet gates using FSC and SSC. Whole blood
was subjected to erythrocyte lysis, stained, and acquired by flow cytometry. Top row: Forward scatter
parameter is used to create singlet gates. Bottom row: side scatter is used to create singlet gates. Left
column: width (-W) versus height (-H) plots are used for singlet gating and to exclude debris in the lower
corner. Aggregates are those events for which there is an increase in -W relative to -H. Right column: area (-A)
versus -H is used for gating singlets; aggregates are those events for which -A increases disproportionately to
-H. (b) Exclusion of doublets can alter the frequency of gated populations. Whole blood was stained with a
cocktail containing CD3, CD4, CD127, CD25, and Foxp3. Left image depicts frequency of Treg cells without
doublet exclusion and right panel shows frequency of Treg cells with singlet gating or doublet exclusion. Note
increase in frequency of CD25+ Foxp3+ cells following gating on singlets. Singlet gating was performed using
SSC-A vs SSC-H followed by FSC-A vs FSC-H
Flow Cytometry Gating Guidelines 91
2.5 Scatter Gates Definition: Fluorescence flow cytometry typically measures forward
and right-angle (side) scatter of the laser light used to excite fluor-
ochromes on the particles of interest. These two parameters contain
information on cell size and granularity, respectively. As such, they
are frequently used as initial gates on immune cell populations, such
as lymphocytes, monocytes, and granulocytes.
How data is affected: While a population such as lymphocytes
may appear to have a relatively homogeneous scatter profile, there
are in fact subtle differences in the scatter profile of different
lymphocyte subsets. Most dramatic is the increased forward scatter
of proliferating lymphocytes (blasts); a tight lymphocyte gate thus
misses most of the proliferating cells. But even populations such as
B cells or CD8+ T cells can have subtle shifts in their scatter relative
to the rest of the lymphocytes. The result is that the placement of
the lymphocyte gate can have small but potentially significant
92 Janet Staats et al.
Fig. 4 Excluding dying/dead cells reduces noise. PBMCs were thawed, stimu-
lated with either no specific antigen or CEF peptide mix (optimal 8–9mers for
CMV, EBV, and influenza) in the presence of protein transport inhibitors, per-
meabilized, stained, and acquired. For all plots, CD3 versus IFN-γ + IL-2 are
shown after gating on CD8+ T cells. For this ICS assay, a positive response is
defined as a response that is at least 2 background and greater than 0.10%
and values are reported after background subtraction. Left column: unstimu-
lated. Right column: CEF stimulated. Top row: The background subtracted CEF
response in the absence of a dead cell exclusion gate is 0.058% or no response.
Bottom row: When the same FCS file as was used in the top row is analyzed
using a dead cell exclusion gate, the background is reduced, and the net
response is 0.109% and thus positive
2.6 Specific Definition: A specific population gate defines a cell population such
Population Gates as CD4+ T cells, B cells, or monocytes, focusing further subset
identification within those populations. The size, placement, and
Flow Cytometry Gating Guidelines 93
250 250
200 200
150 150
SSC-H
SSC-H
100 100
NK cells B cells
50 50
0 0
0 50 100 150 200 250 0 50 100 150 200 250
FSC-H FSC-H
Fig. 5 Differential scatter profiles of specific lymphocyte subsets. NK cells (left panel) and B cells (right panel)
were gated separately using specific markers from the green, liberal scatter gate for lymphocytes and then
projected back into the scatter dimensions as red dots. Note that the pink (tight) scatter gate for lymphocytes
would exclude a subset of both NK and B cells because of their biased and/or diffuse scatter profile relative to
most lymphocytes
order of such gates can influence the reported results in ways that
may not be obvious to an inexperienced operator.
How data is affected: Knowledge of the expression properties of
specific subsets of interest can be useful for setting a parental
population gate. For example, CD3 may be used as a parental
gate prior to analysis of T cell subsets such as ɣδC5; T cells. By
examining a dot plot of CD3 vs. SSC, one can identify the ɣδC5; T
cells as CD3high relative to the rest of the CD3+ population
(Fig. 6). Knowing this, care should be taken not to exclude
CD3high cells when enumerating ɣδC5; T cells as a subset thereof.
Another example in T cell gating concerns the simple discrimi-
nation of CD4+ and CD8+ populations. Since these are largely
mutually exclusive populations, it may seem irrelevant whether
one gates on CD3+ CD4+ and CD3+ CD8+ populations, or rather
sequentially gates on CD3+ and then CD4 vs. CD8. However, the
presence of a small but variable population of CD4+ CD8+ T cells
means that these two strategies will not give identical results. In
fact, the former strategy will double-count these CD4+ CD8+
events as belonging to both populations, while the latter method
allows their true discrimination.
Still another example with regard to T cell gating is the down-
modulation of CD3, CD4, and CD8 on activated T cells. When
performing stimulation assays, the gates for these markers need to
be sufficiently “loose” to avoid cutting off potentially small sub-
populations of activated (and thus responding) T cells that have
partially down-modulated these markers.
94 Janet Staats et al.
Specimen_001-Tube_004 Specimen_001-Tube_004
(x 1.000)
150 200 250
105
104
APC-A
SSC-A
P3
103
100
50
102
TCR γδ
Fig. 6 ɣǖC5;T cells are CD3high. Human lysed wash blood was stained with CD3 and TCRɣǖC5; antibodies.
Orange population indicates TCRɣǖC5; cells on a plot of CD3 vs TCRɣǖC5; (left) and CD3 bright staining on
the TCRɣǖC5; population depicted in CD3 vs SSc plot (right image)
2.7 Data Display Definition: The premise of flow cytometry is that one marker is
tagged with one fluorescent dye and the photons generated by the
2.7.1 Compensation
dye are measured in a single detector. Spectral overlap occurs when
the fluorescence emission from one dye emits photons into the
fluorescence emission spectrum of a neighboring detector. In a
multicolor panel, spectral overlap from multiple dyes may be
observed in a single detector. Electronic subtraction is applied to
correct the interference caused by the spectral overlap in each
channel; this is called compensation. A compensation matrix is
generated to correct the spectral overlap for each dye versus all of
Flow Cytometry Gating Guidelines 95
Fig. 7 Undercompensation leads to a false positive response in an ICS assay. Undercompensation between PE
(IFNγ + IL-2) and FITC (CD4) created the appearance of CD4+ IFNγ + IL-2+ cells, when in fact with correct
compensation these cells were CD4 . In this case, the operator used software to create a compensation
matrix but neglected to verify the accuracy of the compensation matrix by visual inspection the data (e.g.,
using an N N plot matrix as described in the text)
2.7.2 Transformation Definition: The range of signals in flow cytometry can span very
negative values (particularly after compensation) to very high values
(104 or greater). Given the approximately log-normal distribution
of many populations in flow data, a linear scale will severely distort
populations and make inefficient use of the display space. However,
a simple logarithmic scale is unable to display zero or negative
values. Therefore, transformations have emerged that preserve the
ability to display negative and zero values while approximating a
logarithmic scale in the positive space. These have variably been
referred to as logicle, biexponential, or VLog scales [15, 35, 36].
Some version of these is available in all modern flow cytometry
software, often with automated application in a data-dependent
fashion.
How data is affected: Choosing an optimal transformation is
important for a number of reasons. First, visualizing the effects of
compensation requires a scale that smoothly transitions through
zero and displays negative data. Undercompensation or overcom-
pensation can then be accurately judged based on how orthogo-
nally the edges of populations align in the negative to low positive
range. Second, the choice of transformation is important to effi-
cient use of the graphical display space. If a majority of the axis
length is devoted to high values (e.g., >102), and if the data
consists entirely of dim events, then the transformation is not
appropriate. Conversely, if a majority of the axis length is devoted
to negative values, and high positive populations are compressed
and difficult to distinguish, the transformation is not appropriate.
Finally, an inappropriate transformation can create artifacts such
as “splitting” of a negative population into what appears to be
two populations (“valley artifact”). This particular artifact is a
result of displaying too many decades of logarithmic scaling at
the low end of the log scale (usually below 10 or 1). Since
judgement of positive and negative expression is often a goal of
flow cytometry, such artifacts can clearly be an impediment to
interpretation.
Conclusion: Choosing the right transformation is critical
for accurate visualization of compensation, for efficient use of
display space, and for complete visualization of negative popula-
tions without introducing the artificial appearance of breaks in
the data.
Flow Cytometry Gating Guidelines 97
2.8 Analytical An unstained cell sample is often used for calculation of compensa-
Controls tion, relative to single positive samples for each fluorochrome.
However, this is not strictly required for performing compensation,
2.8.1 Unstained Cells
if negative populations are included in each single-color control. An
unstained control is, however, still useful for judging the PMT
voltage gain in each sample, since it represents the position of the
most negative possible population. It is generally not useful for
determining gate placement on stained samples, since it does not
account for either spillover from other fluorescent channels, or
nonspecific binding in the channel of interest.
2.8.2 Isotype Controls Isotype controls are sometimes useful when deciding gate place-
ment in flow cytometry. These controls are made of the same
isotype as the antibody of interest and have the same fluorescent
dye attached to them. In specific instances, such as when monocytes
are being stained with an antibody tagged with tandem dyes such as
PE/Cy7, APC/Cy7, APC/Fire 750, APC-H7, or PE-CF594, the
use of an isotype control antibody is extremely helpful. Monocytes
tend to nonspecifically bind to dyes such as Cy7, Fire 750, and H7.
Figure 8 shows the effect on tandem dye binding to isotype con-
trols. Some manufacturers formulate their antibodies with reagents
to reduce this background and others may sell buffers to overcome
these issues (e.g., Monocyte Blocker from BioLegend).
2.8.3 Fluorescence In the absence of situations like those shown in Fig. 8, the major
Minus One Controls (FMOs) background signals in high-dimensional flow cytometry tend to
come from spillover rather than nonspecific binding. As such, an
FMO control, which leaves out a single antibody conjugate but
maintains all other stains in the panel, is often more useful for
setting gates than an isotype control in the absence of other stains.
2.8.4 Combined Isotype A control which leaves out one antibody conjugate, but replaces it
and FMO Controls with an isotype control antibody, can be considered a “combined”
control. With such a control, one attempts to combine the benefits
of FMO (accounting for spillover) and isotype controls (accounting
for nonspecific binding). While this in theory may be the “best of
both worlds,” it is worth mentioning that there are still drawbacks
to the use of isotype controls that are not alleviated by combining it
with FMO. Specifically, the particular isotype control antibody may
or may not have similar nonspecific binding properties as the test
antibody [37]; and it will likely require titration of the isotype
control to at least approximate those properties.
2.8.5 Internal Controls For functional readouts like cytokines in intracellular cytokine
(e.g., Unstimulated Cells) staining (ICS), the best gating control may be an internal control
(e.g., unstimulated cells from the same donor). Even so, it can be
difficult to decide exactly where to place the cytokine or functional
98 Janet Staats et al.
41 27 31
Count
Count
Count
27 18 21
M3 M4 M3 M4
98.58% 0.00% 95.78% 0.29%
14 9 10
0 0 0
-101 102 103 104 -101 102 103 104 -101 102 103 104
PE-DZL-A PE-DZL-A PE-DZL-A
unstained Isotype APC-Cy7 CD14 APC-Cy7
30 41 79
M4 M3 M4
99.21% 0.79% 26.94%
23 31 59
M1 M1 M1
Count
Count
Count
15 0.00% 21 M2 61.66% 40 M2 82.13%
40.03% 18.20%
8 10 20
0 0 0
-102 103 104 105 -102 103 104 105 -102 103 104 105
APC-Cy7-A APC-Cy7-A APC-Cy7-A
Fig. 8 Tandem dyes nonspecifically stain monocytes and thus isotype should be used to set marker gate
(s) appropriately. Top row: Human lysed washed blood was not stained or stained with isotype control
PE/Dazzle 594 or CD3 PE/Dazzle 594. Histograms are gated on FSC and SSC Singlets followed by monocyte
gate (scatter based). Red marker gates (M1 and M2) indicate gate placement using unstained cells (leftmost
panel) and blue marker gates (M3 and M4) depict frequency of CD3 positive cells based on gate placement
with isotype control (middle panel). If care is not taken interpreting data based on unstained cells, then users
may report monocytes to be CD3+. Bottom row: A similar case using APC/Cy7. Here we show that gates based
on the isotype control are more appropriate for the positive monocyte stain, CD14
2.9 Flow Cytometry It is worth noting that the readout of a gated population can be
Units expressed in different units. Most gating software by default reports
percent-of-parent statistics, that is, the percentage of cells in the
parent population that are contained in the gate of interest. How-
ever, this may not always be the best readout to report.
Consider the case of lymphocytes, which have major subpopu-
lations of B, T, and NK cells. If an individual’s T cell count
increases, the percentage of lymphocytes that are B and NK cells
will necessarily decrease, even if their absolute frequency is
unchanged. In order to avoid the effect of such reciprocal relation-
ships, one can report absolute cell counts (per microliter of blood).
However, this requires a volumetric measurement or use of count-
ing beads. It can be done most accurately by single-platform meth-
ods (where the counting beads or volumetric measurement is done
in the same flow cytometry assay as the cell population staining).
Alternately, a dual platform method can be used, relating the results
of a counting assay to the flow cytometry staining. For example,
total lymphocyte count can be measured by a complete blood count
with differential (CBCD). The total lymphocyte count can then be
multiplied by the percentage of lymphocytes gated as B, T, and NK
cells, to derive absolute counts of these three populations. Because
of variations in the way lymphocytes are defined in the CBCD
versus flow cytometry platform, however, this will necessarily be
less accurate than single platform counting. Still, both methods can
be made relatively reproducible [39].
Short of reporting absolute counts, one can also report popu-
lation percentages but using a higher-level ancestor gate. For exam-
ple, each gated population in a PBMC sample could be reported as
a percent of all live cells (after exclusion of aggregates). While this is
not the same as absolute counting, it does diminish the effect of
changes in reciprocal populations. It is also easily done using the
reported percent-of-parent statistics, by simply multiplying percen-
tages of each ancestor gate up to the top level gate being reported.
Some software programs can provide such reporting automatically
by choosing the desired ancestral gate from which to report
percentages.
Finally, it is possible to report not the frequency or percentage
of cells in a gate, but rather their mean or median fluorescence
intensity (MFI). This is particularly useful when reporting data on
markers that may not have distinct positive and negative popula-
tions, but whose distribution represents a continuum (“smear”) or
for which shifts in expression level are the biologically important
readout. Examples include CD80 and CD86 expression on den-
dritic cells, whose intensity relates to cell maturation/activation.
Rather than measuring the percent of “positive” cells using a
potentially arbitrary cutoff, it makes more sense to report MFI of
the entire population. However, when comparing MFI across
100 Janet Staats et al.
2.10 Special Flow cytometry experiments and therefore associated results can be
Considerations affected by artifacts in the assay, which are not considered or known
prior to performance of a particular assay. These artifacts can
include samples with too few events or difficult-to-discern rare
populations; or shifts in a cell population from one sample to
another. Visual inspection of gates on all samples is therefore critical
before finalizing the gating. In determining whether to move a gate
for a specific sample, one should attempt to maintain a fair and
unbiased approach, that is, keeping gates as similar as possible
except where clear shifts in a population warrant changing the
gate. Samples to be directly compared, such as stimulated and
unstimulated conditions from the same donor, need to have the
exact same gating applied.
When gating a large study, or when multiple operators and/or
sites are involved, it becomes necessary to create a gating template.
This is a document with default gates provided, that is the starting
point for analysis of each batch of samples. It represents not only
the hierarchy of all gates to be applied but also a “best guess” of
where those gates should lie. Movement of gates from the template
position should only be undertaken when necessary, and with the
caveats described above in mind. In general, central analysis by a
single operator (or at least verified by a single operator) is more
consistent than separate analysis by different individuals or sites
[27, 41].
Some types of artifacts can be anticipated and possibly avoided,
including many of the factors listed in Table 1. Some variables that
should be controlled to avoid excessive population shifts include:
reagent lot control, consistent washing between wells or tubes, and
avoiding degradation of problematic tandem dyes that can be idio-
syncratic between samples. It is of course always preferable to avoid
artifacts that would complicate gating in the first place, but this may
not always be perfect. An example of a “preventable” population
shift is that caused by brilliant dye interaction, as shown in Fig. 9.
Finally, a piece of idiosyncratic advice regarding gate placement
relative to X and Y axes. Even with appropriate transformation and
data display, there are almost always events that are below the axis
for negative populations. As such, gates on the negative population
need to be placed over the relevant axis to include events that are
below that axis. Depending on the plot size and visual resolution,
this difference may be insidious; two gates may look essentially
identical, but one includes events below the axis and one does
not. Experienced cytometrists therefore typically draw a gate and
then drag it well beyond the axis in order to avoid this issue.
Flow Cytometry Gating Guidelines 101
Fig. 9 Interaction between fluorophores may affect populations. Human lysed whole blood was stained with
CD4 Brilliant Violet 421™ and CD8 Brilliant Violet 711™ in the presence (left) or absence (right) of Brilliant
Stain Buffer™. Note loss of mutually exclusive expression of CD4 and CD8 in the left plot
3 Discussion/Conclusions
creating gates that can be applied across samples while still allowing
for donor-specific corrections if needed.
Much of the advice presented here can be broadly summarized
as follows.
1. All relevant parameters in the data file should be used to most
purely and completely define populations of interest. These para-
meters include time, which can give clues to artifacts such as
clogs or bubbles in the data acquisition. They also include the
height, area, and/or width of the light scatter parameters,
which can be used in combination to exclude cell aggregates.
Expression of one or more parameters may be used to exclude
irrelevant events (exclusion gating), in addition to parameters
used to include relevant events. And backgating can be useful
to judge the adequacy of individual gates in a hierarchy, includ-
ing light scatter gates, with regard to their unbiased selection of
the population of interest. Importantly, the fact that a parame-
ter is not used in the gating scheme does not mean that it may
not affect the gated results; so visual inspection of all para-
meters in the data is recommended.
2. Once defined for a single sample, gates should be checked to ensure
fair application across all samples. This may require adjusting
gates between donors, but it should generally not entail
changes in the gate among samples or conditions to be com-
pared within the same donor. Attempts should be made to
avoid gates that slice arbitrarily through an obvious cluster of
cells, including cases where part of a cell cluster lies below the
axis of a histogram or dot plot. In some instances, an otherwise
homogeneous cell population will display a continuous distri-
bution of intensity for a given parameter. It may then be more
sensible to report the mean (or median) intensity of the popu-
lation rather than arbitrarily gating a positive and negative
subset for that parameter and reporting percent positive or
negative.
The eventual goal of these guidelines is not only accuracy in
defining populations of interest but also reproducibility between
experiments, between operators, and between donors and cohorts.
This is particularly important in longitudinal and/or multisite
studies. In these cases, it is best to have all gating verified by the
same personnel, even if initially performed by different personnel.
Further, it is advisable to continually refer to historically gated data,
to ensure reproducible treatment relative to contemporary samples,
for studies that are analyzed over a period of time.
In this document, we have tried to illustrate the most overarch-
ing principles that apply to gating flow cytometry data from immu-
nological assays. There may be exceptions to these principles, but
they are likely to be few. There are also undoubtedly situations
where one can avoid some of these recommendations without
Flow Cytometry Gating Guidelines 103
peril. But it is also likely that following most or all of the recom-
mendations increases the confidence that artifacts and false conclu-
sions will be avoided. Furthermore, there are certainly additional
and more detailed recommendations that could be added to these
guidelines. But with increasingly detailed recommendations comes
the likelihood that they will not apply as broadly across different
situations and assays. So we have mostly avoided too much specific-
ity in these guidelines, in favor of exposing the high-level concepts
behind the gating strategies discussed. We hope that wide disper-
sion of these concepts leads to an overall higher level of accuracy
and reproducibility of flow cytometry gating.
Acknowledgments
References
1. Burel JG, Qian Y, Lindestam Arlehamn C et al 9. Fletez-Brant K, Špidlen J, Brinkman RR et al
(2017) An integrated workflow to assess tech- (2016) flowClean: automated identification
nical and biological variability of cell popula- and removal of fluorescence anomalies in flow
tion frequencies in human peripheral blood by cytometry data. Cytometry A 89:461–471
flow cytometry. J Immunol 198:1748–1758 10. Maecker HT, McCoy JP, Nussenblatt R (2012)
2. Disis ML, dela Rosa C, Goodell V et al (2006) Standardizing immunophenotyping for the
Maximizing the retention of antigen specific Human Immunology Project. Nat Rev Immu-
lymphocyte function after cryopreservation. J nol 12:191–200
Immunol Methods 308:13–18 11. Lamoreaux L, Roederer M, Koup R (2006)
3. Posevitz-Fejfár A, Posevitz V, Gross CC et al Intracellular cytokine optimization and stan-
(2014) Effects of blood transportation on dard operating procedure. Nat Protoc
human peripheral mononuclear cell yield, phe- 1:1507–1516
notype and function: implications for immune 12. Maecker HT, Frey T, Nomura LE et al (2004)
cell biobanking. PLoS One 9:e115920 Selecting fluorochrome conjugates for maxi-
4. Perfetto SP, Chattopadhyay PK, Lamoreaux L mum sensitivity. Cytometry A 62:169–173
et al (2006) Amine reactive dyes: an effective 13. Roederer M (2001) Spectral compensation for
tool to discriminate live and dead cells in poly- flow cytometry: visualization artifacts, limita-
chromatic flow cytometry. J Immunol Meth- tions, and caveats. Cytometry A 45:194–205
ods 313:199–208 14. Chevrier S, Crowell HL, Zanotelli VRT et al
5. Shankey TV, Rabinovitch PS, Bagwell B et al (2018) Compensation of signal spillover in sus-
(1993) Guidelines for implementation of clini- pension and imaging mass cytometry. Cell Syst
cal DNA cytometry. International Society for 6:612–620.e5
Analytical Cytology. Cytometry 14:472–477 15. Finak G, Perez J-M, Weng A et al (2010) Opti-
6. Perfetto SP, Roederer M (2007) Increased mizing transformations for automated, high
immunofluorescence sensitivity using 532 nm throughput analysis of flow cytometry data.
laser excitation. Cytometry A 71:73–79 BMC Bioinformatics 11:546
7. Maecker HT, Trotter J (2006) Flow cytometry 16. Novo D, Wood J (2008) Flow cytometry his-
controls, instrument setup, and the determina- tograms: transformations, resolution, and dis-
tion of positivity. Cytometry A 69:1037–1042 play. Cytometry A 73:685–692
8. Watson JV (1987) Time, a quality-control 17. (1992) Guidelines for the performance of
parameter in flow cytometry. Cytometry CD4+ T-cell determinations in persons with
8:646–649
104 Janet Staats et al.
human immunodeficiency virus infection. 28. Verschoor CP, Lelic A, Bramson JL et al (2015)
MMWR Recomm Rep 41:1–17 An introduction to automated flow cytometry
18. (1994) 1994 revised guidelines for the gating tools and their implementation. Front
performance of CD4+ T-cell determinations Immunol 6:380
in persons with human immunodeficiency 29. Mair F, Hartmann FJ, Mrdjen D et al (2016)
virus (HIV) infections. Centers for Disease The end of gating? An introduction to auto-
Control and Prevention. MMWR Recomm mated analysis of high dimensional cytometry
Rep 43:1–21 data. Eur J Immunol 46:34–43
19. (1997) 1997 revised guidelines for performing 30. Martin JC, Swartzendruber DE (1980) Time:
CD4+ T-cell determinations in persons a new parameter for kinetic measurements in
infected with human immunodeficiency virus flow cytometry. Science 207:199–201
(HIV). Centers for Disease Control and Pre- 31. Hoffman RA (2009) Pulse width for particle
vention. MMWR Recomm Rep 46:1–29 sizing. Curr Protoc Cytom Chapter 1:Unit
20. Mandy FF, Nicholson JKA, McDougal JS et al 1.23
(2003) Guidelines for performing single- 32. Wersto RP, Chrest FJ, Leary JF et al (2001)
platform absolute CD4+ T-cell determinations Doublet discrimination in DNA cell-cycle anal-
with CD45 gating for persons infected with ysis. Cytometry 46:296–306
human immunodeficiency virus. Centers for 33. Furman MI, Barnard MR, Krueger LA et al
Disease Control and Prevention. MMWR (2001) Circulating monocyte-platelet aggre-
Recomm Rep 52:1–13 gates are an early marker of acute myocardial
21. O.W. Health (2007) Laboratory guidelines for infarction. J Am Coll Cardiol 38:1002–1006
enumerating CD4 T lymphocytes in the con- 34. Nomura LE, Walker JM, Maecker HT (2000)
text of HIV/AIDS. World Health Organiza- Optimization of whole blood antigen-specific
tion Regional Office for South-East Asia, New cytokine assays for CD4(+) T cells. Cytometry
Delhi 40:60–68
22. Sutherland DR, Anderson L, Keeney M et al 35. Parks DR, Roederer M, Moore WA (2006) A
(1996) The ISHAGE guidelines for CD34+ new “Logicle” display method avoids deceptive
cell determination by flow cytometry. Interna- effects of logarithmic scaling for low signals
tional Society of Hematotherapy and Graft and compensated data. Cytometry A
Engineering. J Hematother 5:213–226 69:541–551
23. Illingworth A, Marinov I, Sutherland DR et al 36. Bagwell CB, Hill BL, Herbert DJ et al (2016)
(2018) ICCS/ESCCA consensus guidelines to Sometimes simpler is better: VLog, a general
detect GPI-deficient cells in paroxysmal noc- but easy-to-implement log-like transform for
turnal hemoglobinuria (PNH) and related dis- cytometry. Cytometry A 89:1097–1105
orders part 3—data analysis, reporting and case
studies. Cytometry B Clin Cytom 94:49–66 37. Andersen MN, Al-Karradi SNH, Kragstrup TW
et al (2016) Elimination of erroneous results in
24. McNeil LK, Price L, Britten CM et al (2013) A flow cytometry caused by antibody binding to
harmonized approach to intracellular cytokine Fc receptors on human monocytes and macro-
staining gating: results from an international phages. Cytometry A 89:1001–1009
multiconsortia proficiency panel conducted by
the Cancer Immunotherapy Consortium 38. Richards AJ, Staats J, Enzor J et al (2014)
(CIC/CRI). Cytometry A 83:728–738 Setting objective thresholds for rare event
detection in flow cytometry. J Immunol Meth-
25. Britten CM, Janetzki S, Ben-Porat L et al ods 409:54–61
(2009) Harmonization guidelines for
HLA-peptide multimer assays derived from 39. Hultin LE, Chow M, Jamieson BD et al (2010)
results of a large scale international proficiency Comparison of interlaboratory variation in
panel of the Cancer Vaccine Consortium. Can- absolute T-cell counts by single-platform and
cer Immunol Immunother 58:1701–1713 optimized dual-platform methods. Cytometry
B Clin Cytom 78:194–200
26. Nomura L, Maino VC, Maecker HT (2008)
Standardization and optimization of multipa- 40. Vogt RF, Cross GD, Henderson LO et al
rameter intracellular cytokine staining. Cyto- (1989) Model system evaluating fluorescein-
metry A 73:984–991 labeled microbeads as internal standards to cal-
ibrate fluorescence intensity on flow cyt-
27. Finak G, Langweiler M, Jaimes M et al (2016) ometers. Cytometry 10:294–302
Standardizing flow cytometry immunopheno-
typing analysis from the human immunophe- 41. Maecker HT, Rinfret A, D’Souza P et al (2005)
notyping consortium. Sci Rep 6:20686 Standardization of cytokine flow cytometry
assays. BMC Immunol 6:13
Chapter 6
Abstract
Therapies based on activating the immune system, that is, immunotherapy, are now widely implemented in
clinical praxis in patients with advanced cancer. Although cancer immunotherapy can result in long-lasting
clinical responses, the majority of patients do not respond or develop resistance. Furthermore, cancer
immunotherapy is being increasingly combined with other forms of immunotherapy or conventional cancer
therapies. It is therefore much needed to identify biomarkers that can precisely classify what patients will
benefit from the treatment without any major adverse events and to further develop the efficacy of cancer
immunotherapy. While much attention has been focused on monitoring T cell responses in cancer immu-
notherapy, recent reports have shown that NK cells also play a major role in the response to cancer
immunotherapy. The gold standard for immunoprofiling of NK cells is flow cytometry, but other technol-
ogies have emerged and include mass cytometry, multiplex immunohistochemistry, and single-cell RNA--
sequencing. In this chapter we provide a detailed protocol to profile NK cells using flow cytometry, and a
brief introduction to other techniques.
1 Introduction
NK cells are innate lymphocytes that can kill virus infected and
malignant cells without any prior sensitization of antigens. They
play a major role in cancer immunosurveillance, and high frequency
and activity of NK cells is correlated with improved prognosis in
several different solid tumors and hematological malignancies. In
recent years, immunotherapy based on activating NK cells has been
explored in several clinical trials including bispecific antibodies or
adoptive transfer of unmodified or genetically modified NK cells
[1, 2]. Cancer immunotherapy and in particular treatment with
immunomodulatory antibodies has now established itself as one of
the pillars in cancer care. While this treatment can result in long-
lasting clinical responses in patients with advanced cancers, many
patients do not respond or develop resistance to the treatment.
Recently, reports have demonstrated that NK cells may play an impor-
tant role in the response to immunomodulatory antibodies
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_6, © Springer Science+Business Media, LLC, part of Springer Nature 2019
105
106 Chen Ziqing et al.
2 Materials
3 Methods
3.1 Flow Cytometry 1. NK cell phenotyping using flow cytometric analysis requires
single cell suspensions of viable cells. Possible starting materials
are whole blood (see Note 1), fresh polymorphonuclear cells
(PBMC) (see Note 2), frozen PBMC (see Note 3), fine needle
aspiration biopsy (see Note 4), and fresh organ or tumor mate-
rial (see Note 5).
2. The panel to study NK cells in the blood or tissue should be
designed according to the study interest and the available flow
cytometer. To simply identify human NK cells in blood or
tissue the following markers are widely used and accepted:
CD45, CD3, CD56, and CD16. Additional makers that are
NK Cell Phenotyping 107
Fig. 1 Identification of NK cells in human PBMC. Human PBMC are stained and acquired on flow cytometry. NK
cells are identified with the following gating strategy. (1) Gate on lymphocyte population in FSC–SSC plot.
(2) Next gate on linear population in FSC-A–FSC-H plot. (3) Gate on live cells in dead cell marker–FSC plot.
(4) Within the live cells gate on CD19neg cell population in CD19–FSC plot. (5) From the CD19neg population
gate on CD56+CD3 cells in CD56–CD3 plot. In this plot the CD56dim and CD56bright cells appear as two
separate populations
9. Vortex each sample for 3 s to loosen cell pellet (see Note 11).
10. Repeat steps 7 and 8.
11. Vortex each sample for 3 s to loosen cell pellet and resuspend
cells in leftover DPBS.
12. Add fixable dead cell marker to samples according to the
manufacturer’s protocol (see Notes 12 and 13).
13. Incubate according to the manufacturer’s protocol.
14. Add 1 ml of FACS buffer and centrifuge tubes at 700 g for
2 min.
15. Discard supernatant by inverting tube and blotting on tissue
(see Note 10).
16. Vortex each sample for 3 s to loosen cell pellet (see Note 11).
17. Repeat steps 14–16 two times.
18. Resuspend cells to single cell suspension in leftover FACS
buffer.
19. Add FcBlock to each sample at a concentration of 2.5 μg for
1 106 cells.
20. Prepare master mix containing surface staining antibodies.
Mixes should be filled up with FACS buffer to a volume of
10–20 μl added to each sample (see Note 14).
21. Incubate samples for 20 min at 4 C in the dark (see Note 15).
22. Repeat steps 14–16 two times.
For intracellular or intranuclear staining continue with
step 26.
23. Vortex each sample for 3 s to loosen cell pellet and resuspend
cells in 200 μl DPBS.
24. If no fixable dead cell marker was used, add nonfixable dead cell
marker to all cells and incubate for 5 min at 4 C in the dark (see
Note 16).
25. Keep samples on 4 C until acquisition on flow cytometer (see
Note 17).
26. Resuspend cells in fixation solution (see Note 18).
27. Incubate samples for 30 min at 4 C in fixation solution or
according to the manufacturer’s protocol.
28. Prepare permeabilization buffer (see Note 19).
29. Add 1 ml of permeabilization buffer (Perm buffer) to each
sample centrifuge tubes at 700 g for 2 min.
30. Discard supernatant by inverting tube and blotting on tissue
(see Note 10).
31. Vortex each sample for 3 s to loosen cell pellet (see Note 11).
32. Repeat steps 29–31.
NK Cell Phenotyping 109
3.2 Mass Cytometry Mass cytometry, also called cytometry by time of flight (CyTOF) is
a novel platform to perform even deeper analysis of immune cell
subsets compared with flow cytometry [4]. Immunophenotyping
by mass spectrometry provides the ability to measure >40 proteins
at a rate of 1000 cells/s, compared with 18 proteins per cell, at
>10,000 cells/s for fluorescence-based flow cytometry. Theoreti-
cally, mass cytometry is capable to measure up to 100 different
stable isotope tags, but the high-purity requirement of these tags
limits the usage down to around 40 rare earth metal tags [5]. The
sample preparation is similar to flow cytometry. In contrast to the
acquisition on a flow cytometer, the single cells are analyzed in a
mass spectrometer. The antibodies are not coupled to fluoro-
chromes but instead to heavy metals. Each heavy metal conjugated
to an antibody binding an NK cell will give a unique peak at an exact
molecular weight. The advantage is that there is no overlap
between different heavy metals; thus, no compensation needs to
be performed. More than 100 heavy metals are available for mass
cytometry, giving the opportunity to analyze in depth NK cell
phenotypes and subsets. Due to the high degree of customization,
the library of preconjugated commercially available antibodies is
limited. This requires self-conjugation of antibodies but at the same
time allows for the design of large individualized and specific
panels. Details on the design and optimization of an NK cell-
specific mass spectrometry panel have been described by Kay
et al. [6].
110 Chen Ziqing et al.
3.3 Multiplex Flow cytometry and CyTOF can identify the presence of rare
Immunohisto- immune cell subsets in single-cell suspensions. While these tech-
chemistry nologies are powerful, no information regarding spatial distribu-
tion can be obtained. The localization of immune cells and their
local environment indeed differ between different areas in the
tumor (stromal, intratumoral, and tumor margin). The under-
standing of cell localization can be of great prognostic value
[7]. Multiplex immunohistochemistry has the ability to provide
this missing information. Sequential staining of a single section
allows for the understanding of spatial cell type distribution within
a tumor and to draw conclusions on cell-specific protein expression.
The sequential stainings are individually acquired on a fluorescent
microscope and are overlapped upon image analysis. Since the
majority of antibodies are produced in mice, rats, and rabbits,
antibody stripping is required after individual rounds of staining.
Stripping of antibodies always have the risk to remove or damage
antigens. The order in which the antigens are stained requires
therefore a lot of optimization. The technology has further been
developed to allow for standardization and clinical application. For
example, the whole-slide eight-plex mIHC platform is set up to
analyze six antigens and cell nuclei in parallel in whole FFPE sec-
tions [8]. The staining procedure is standardized, and while the
secondary antibodies are not changeable, the primary antibodies
can be freely selected.
There are certainly developments to perform deeper analysis
using multiplex immunohistochemistry. Imaging mass cytometry is
one such development, combining mass cytometry with tissue
localization [9]. In this application, fresh-frozen paraffin-embed-
ded tissue or frozen sections are stained with mass spectrometry
antibodies following standard immunofluorescence protocols.
With a precisely directed laser beam, tissue pieces of 1 μm2 are
removed one after the other from the section and acquired in the
mass spectrometer. This allows to analyze the protein expression on
the tissue section to a resolution of 1 μm2. Another development is
the CO-Detection by indEXing (CODEX) technology to enable
multiplexing of antibody-tagged target epitopes. In CODEX, anti-
body binding events are rendered iteratively using DNA barcodes,
fluorescent dNTP analogs, and an in situ polymerization-based
indexing procedure [10].
4 Notes
References
1. Davis ZB, Vallera DA, Miller JS, Felices M margin of colorectal cancer liver metastases
(2017) Natural killer cells unleashed: check- associated with improved survival. Oncoimmu-
point receptor blockade and BiKE/TriKE uti- nology 6(3):e1286436. https://doi.org/10.
lization in NK-mediated anti-tumor 1080/2162402X.2017.1286436
immunotherapy. Semin Immunol 31:64–75. 8. Blom S, Paavolainen L, Bychkov D, Turkki R,
https://doi.org/10.1016/j.smim.2017.07. Maki-Teeri P, Hemmes A, Valimaki K,
011 Lundin J, Kallioniemi O, Pellinen T (2017)
2. Fang F, Xiao W, Tian Z (2017) NK cell-based Systems pathology by multiplexed immunohis-
immunotherapy for cancer. Semin Immunol tochemistry and whole-slide digital image anal-
31:37–54. https://doi.org/10.1016/j.smim. ysis. Sci Rep 7(1):15580. https://doi.org/10.
2017.07.009 1038/s41598-017-15798-4
3. Hsu J, Hodgins JJ, Marathe M, Nicolai CJ, 9. Chang Q, Ornatsky O, Hedley D (2017) Stain-
Bourgeois-Daigneault MC, Trevino TN, ing of frozen and formalin-fixed, paraffin-
Azimi CS, Scheer AK, Randolph HE, Thomp- embedded tissues with metal-labeled antibo-
son TW, Zhang L, Iannello A, Mathur N, Jar- dies for imaging mass cytometry analysis. Curr
dine KE, Kirn GA, Bell JC, McBurney MW, Protoc Cytom 82:12.47.11–12.47.18.
Raulet DH, Ardolino M (2018) Contribution https://doi.org/10.1002/cpcy.29
of NK cells to immunotherapy mediated by 10. Goltsev Y, Samusik N, Kennedy-Darling J,
PD-1/PD-L1 blockade. J Clin Invest 128 Bhate S, Hale M, Vazquez G, Black S, Nolan
(10):4654–4668. https://doi.org/10.1172/ GP (2018) Deep profiling of mouse splenic
JCI99317 architecture with CODEX multiplexed imag-
4. Ornatsky O, Bandura D, Baranov V, Nitz M, ing. Cell 174(4):968–981 e915. https://doi.
Winnik MA, Tanner S (2010) Highly multi- org/10.1016/j.cell.2018.07.010
parametric analysis by mass cytometry. J Immu- 11. Naranbhai V, Bartman P, Ndlovu D,
nol Methods 361(1–2):1–20. https://doi. Ramkalawon P, Ndung’u T, Wilson D,
org/10.1016/j.jim.2010.07.002 Altfeld M, Carr WH (2011) Impact of blood
5. Atkuri KR, Stevens JC, Neubert H (2015) processing variations on natural killer cell fre-
Mass cytometry: a highly multiplexed single- quency, activation, chemokine receptor expres-
cell technology for advancing drug develop- sion and function. J Immunol Methods 366
ment. Drug Metab Dispos 43(2):227–233. (1–2):28–35. https://doi.org/10.1016/j.jim.
https://doi.org/10.1124/dmd.114.060798 2011.01.001
6. Kay AW, Strauss-Albee DM, Blish CA (2016) 12. Florek M, Schneidawind D, Pierini A, Baker J,
Application of mass cytometry (CyTOF) for Armstrong R, Pan Y, Leveson-Gower D,
functional and phenotypic analysis of natural Negrin R, Meyer E (2015) Freeze and thaw
killer cells. Methods Mol Biol 1441:13–26. of CD4+CD25+Foxp3+ regulatory T cells
https://doi.org/10.1007/978-1-4939-3684- results in loss of CD62L expression and a
7_2 reduced capacity to protect against graft-
7. Berthel A, Zoernig I, Valous NA, Kahlert C, versus-host disease. PLoS One 10(12):
Klupp F, Ulrich A, Weitz J, Jaeger D, Halama e0145763. https://doi.org/10.1371/journal.
N (2017) Detailed resolution analysis reveals pone.0145763
spatial T cell heterogeneity in the invasive
Chapter 7
Abstract
The human obese subcutaneous adipose tissue (SAT) contributes to systemic and B cell intrinsic inflamma-
tion, reduced B cell responses, and increased secretion of autoimmune antibodies. Immune cells are
recruited to the SAT by chemokines released by both adipocytes and infiltrating immune cells. We describe
here the characterization of B lymphocytes from the SAT and blood (control) of obese females undergoing
weight reduction surgeries (breast reduction or panniculectomy). We show how to isolate the immune cells
from the blood and SAT, how to characterize B cells and their subsets, and how to measure markers of
activation and/or transcription factors in SAT-derived B cells and B cell subsets. We also show how to
evaluate other immune cell types infiltrating the SAT, including T cells, NK cells, monocyte/macrophages,
in order to measure relative proportions of these cell types as compared to the blood.
Key words Adipose tissue (AT), Body mass index (BMI), Cardiovascular (CV), Dulbecco’s modified
Eagle’s medium (DMEM), Fetal calf serum (FCS), Free fatty acids (FFAs), Germinal center (GC),
Hanks’ balanced salt solution (HBSS), Insulin resistance (IR), Insulin sensitivity (IS), Peripheral blood
mononuclear cells (PBMC), Red blood cells (RBC), Reactive oxygen species (ROS), Room tempera-
ture (RT), Subcutaneous adipose tissue (SAT), Stromal vascular fraction (SVF), Type-2 diabetes
(T2D), Toll-like receptor (TLR)
1 Introduction
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_7, © Springer Science+Business Media, LLC, part of Springer Nature 2019
115
116 Alain Diaz et al.
2 Materials
2.1 Isolation 1. Fresh SAT specimen (100–200 g of tissue) and blood as control
of Lymphocytes are from females undergoing breast reduction surgery or pan-
niculectomy surgery (removal of lower abdominal fat) at the
Division of Plastic and Reconstructive Surgery at the Univer-
sity of Miami Hospital.
2. Supplemented HBSS: 1 Hanks’ balanced salt solution
supplemented with 200 nM adenosine and 1
penicillin–streptomycin.
3. Supplemented DMEM (s-DMEM): 1 High Glucose Dulbec-
co’s modified Eagle’s medium supplemented with 15 mM
HEPES, 1 mM sodium pyruvate, 1 penicillin–streptomycin,
1% BSA, and 200 nM adenosine.
4. Complete RPMI: RPMI 1640, supplemented with 10% FCS,
10 μg/mL gentamicin, 2 10 5 M 2-mercaptoethanol, and
2 mM L-glutamine.
5. Collagenase Type V (Sigma).
6. Sharp scissors and blender (for mincing adipose tissue).
7. Forceps.
8. Siliconized glass beakers.
9. Stainless steel 316 syringe needle, blunt tip point (gauge 18, L
6 in).
10. 300 μm Mesh (Spectra/Mesh Nylon, 30 30 cm square).
11. 50 mL polypropylene centrifuge tubes (BD Falcon).
12. 20 cc syringe.
13. Sigmacote (Sigma).
3 Methods
3.1 Isolation of SAT- 1. Weigh the AT. The sample should be collected in HBSS or
Derived Blood and SVF similar buffer.
2. Remove connective tissue with scissors. Proceed to cut the
tissue in smaller pieces and place them in 50 mL tubes until
approximately half-filled. Remove connective tissue with
scissors.
3. Wash tissue three times by filling the tubes with supplemented
HBSS and carefully removing the buffer below the floating
tissue by aspiration with syringe and blunt needle. Use at least
2 new 50 mL tubes to collect the first wash from this step that
contains most of the blood surrounding the tissue. This will be
used for blood cell staining subsequently.
4. After the last wash, add one and a half volumes of supplemen-
ted DMEM and transfer to a food blender.
5. Blend the washed tissue in the blender, using very short strokes
to avoid damaging the cells.
6. Transfer tissue to new 50 mL tubes and centrifuge for 3 min at
1200 g.
7. Weigh 1 mg/g of tissue of collagenase type V and dissolve in
s-DMEM at 1 mg/mL.
8. After centrifugation (step 6), remove the precipitated debris
and most of the liquid with the syringe and transfer the tissue
to a siliconized beaker containing about an equal volume of
s-DMEM containing 1 mg/mL of collagenase.
9. Incubate for 1 h in a 37 C water bath, gently mixing the
reaction mix every 10 min to help the digestion of the tissue.
10. After incubation, filter the reaction mix through the 300 μm
Mesh into 50 mL tubes to eliminate undigested larger tissue
pieces. This step will require several pieces of mesh since they
get clogged rapidly, as well as squeezing the mesh by gloved
hand in order to recover most of the digested sample.
120 Alain Diaz et al.
3.2 Staining 1. Distribute 500,000 to 106 SVF cells in five 5 mL FACS tubes
of the SVF for Flow for antibody staining. Add 1 mL of FACS buffer and centrifuge
Cytometry Analyses for 5 min at 500 g.
2. Discard the supernatant and briefly vortex to resuspend the
pellet.
3. Add the adequate amount of the following combination of
antibodies for membrane staining in a volume of at least
50 μL of staining buffer:
Tube 1 (B cell phenotyping and GC B cells): LIVE/
DEAD™ Fixable Aqua Dead Cell Stain, Pacific Blue-CD45,
FITC-IgD, PE-CD27, APC-CD19 and Brilliant Violet
(BV) 711-CD10.
Tube 2 (T/NK-T cells phenotyping): LIVE/DEAD™ Fix-
able Aqua Dead Cell Stain, Pacific Blue-CD45, FITC-CD3,
PE-CD56, PE/Cy7-TCRγ/δ, PerCP-CD4, APC-TCRα/β,
and APC/H7-CD8.
Tube 3 (Tγδ subsets): LIVE/DEAD™ Fixable Aqua Dead
Cell Stain, Pacific Blue-CD45, Alexa Fluor 700-CD3, FITC-
TCRγδ1, PE-TCRγδ2, PerCP-CD4, APC-TCRαβ, and
APC/H7-CD8.
Tube 4 (GC T cells): LIVE/DEAD™ Fixable Aqua Dead
Cell Stain, Pacific Blue-CD45, Alexa Fluor 700-CD3, PerCP-
CD4, APC-PD1, APC/Cy7-CXCR5.
Tube 5 (Monocytes/NK cells): LIVE/DEAD™ Fixable
Aqua Dead Cell Stain, Pacific Blue-CD45, FITC-CD3,
PE-CD56, PE/Cy7-CD19, APC-CD14, APC/Cy7-CD16.
Immunophenotyping of Human B Cells 121
3.3 Staining of SAT- 1. Use the blood collected in Subheading 3.1, step 3 (“Isolation
Derived Blood for Flow of the blood and the SVF from the AT”). Centrifuge the 50 mL
Cytometry Analyses tubes containing the first wash of the AT sample for 5 min at
500 g. Carefully remove the supernatant by slowly inverting
the tube without disturbing the more viscous bloody fluid and
pellet that remain at the bottom of the tube. A volume of
approximately 500 μL of blood fluid and pellet should remain
in the tube.
2. Vortex briefly to resuspend the pellet and transfer 100 μL of
blood to four 5 mL FACS tubes for antibody staining.
3. Add the same volume and combination of antibodies for mem-
brane staining used in step 3 in Subheading 3.2.
4. Incubate for 20 min at RT in the dark.
5. Add 2 mL of BD FACS™ lysing solution to each tube, vortex
to mix and incubate for 15 min at RT in the dark.
6. Centrifuge the tubes for 5 min at 500 g. Discard the super-
natant and briefly vortex.
7. Add 2 mL of staining buffer to each tube and centrifuge for
5 min at 500 g.
8. Perform steps 6–10 from Subheading 3.2 for intracellular
staining of blood.
122 Alain Diaz et al.
SSC
CD45
CD19
CD27
20±2 6±2
8±1
BLOOD
17±1 57±5
SSC FSC Live/Dead FSC IgD
CD19
CD45
CD27
31±3 3±2
12±2
SVF
49±4 17±4
FSC Live/Dead FSC IgD
Fig. 1 Gating strategies and frequencies of B cells in the obese SAT versus blood. The blood and the SVF from
the SAT of the same individual were stained to evaluate the frequencies of B cells. Gating strategies and a
representative dot plot from one individual are shown. Means SE from 20 individuals are shown in red. The
figure is adapted from Frasca D et al., PLOS One, 2018
3.4 Acquisition 1. Single color controls should be prepared for each experiment
and Analysis to assist with compensation. We routinely use cryopreserved
of Lymphocyte normal PBMC from healthy donors for single color staining.
Populations in SAT- 2. Follow your instrument guidelines to perform automatic com-
Derived Blood and SVF pensation using the single color controls as well as the
(see Note 1) unstained cells.
3. Acquire at least 50,000 (or as many as possible) events in the
Live/CD45+ gate.
4. After acquisition we export the files and perform the analyses at
a later time with the FlowJo 10.0.6 software. Figure 1 shows
our gating strategy and analysis of B cell subsets in blood versus
SVF from the same obese individual.
T-bet
Unstimulated: 3% CL097: 12%
BLOOD
SVF
FSC
4 Notes
Acknowledgments
References
1. Apovian CM, Gokce N (2012) Obesity and study II. Arch Intern Med 167
cardiovascular disease. Circulation 125 (15):1670–1675. https://doi.org/10.1001/
(9):1178–1182. https://doi.org/10.1161/ archinte.167.15.1670
CIRCULATIONAHA.111.022541 7. Casas R, Sacanella E, Estruch R (2014) The
2. Hotamisligil GS (2006) Inflammation and immune protective effect of the Mediterranean
metabolic disorders. Nature 444 diet against chronic low-grade inflammatory
(7121):860–867. https://doi.org/10.1038/ diseases. Endocr Metab Immune Disord Drug
nature05485 Targets 14(4):245–254
3. Johnson AM, Olefsky JM (2013) The origins 8. Hass DJ, Brensinger CM, Lewis JD, Lichten-
and drivers of insulin resistance. Cell 152 stein GR (2006) The impact of increased body
(4):673–684. https://doi.org/10.1016/j.cell. mass index on the clinical course of Crohn’s
2013.01.041 disease. Clin Gastroenterol Hepatol 4
4. Shoelson SE, Lee J, Goldfine AB (2006) (4):482–488. https://doi.org/10.1016/j.
Inflammation and insulin resistance. J Clin cgh.2005.12.015
Invest 116(7):1793–1801. https://doi.org/ 9. Collaborators GBDO, Afshin A, Forouzanfar
10.1172/JCI29069 MH, Reitsma MB, Sur P, Estep K, Lee A,
5. Renehan AG, Tyson M, Egger M, Heller RF, Marczak L, Mokdad AH, Moradi-Lakeh M,
Zwahlen M (2008) Body-mass index and inci- Naghavi M, Salama JS, Vos T, Abate KH,
dence of cancer: a systematic review and meta- Abbafati C, Ahmed MB, Al-Aly Z, Alkerwi A,
analysis of prospective observational studies. Al-Raddadi R, Amare AT, Amberbir A, Ame-
Lancet 371(9612):569–578. https://doi.org/ gah AK, Amini E, Amrock SM, Anjana RM,
10.1016/S0140-6736(08)60269-X Arnlov J, Asayesh H, Banerjee A, Barac A,
6. Setty AR, Curhan G, Choi HK (2007) Obesity, Baye E, Bennett DA, Beyene AS, Biadgilign S,
waist circumference, weight change, and the Biryukov S, Bjertness E, Boneya DJ, Campos-
risk of psoriasis in women: nurses’ health Nonato I, Carrero JJ, Cecilio P, Cercy K, Cio-
banu LG, Cornaby L, Damtew SA,
126 Alain Diaz et al.
Abstract
Polychromatic flow cytometry enables the detection and characterization of markers which are helpful in
defining phenotype of various cell subsets. Here we describe flow cytometry-based method to characterize
phenotype of naı̈ve, memory, and effector T cells. Being able to differentiate these cells is crucial in
understanding immune response, and immune profiling. Naı̈ve T cells enable the body to fight off new,
unrecognized infections and diseases, and memory T cells are enriched for response to recall antigens.
Furthermore, the antigen-experienced T cell populations can be broadly divided into effector and memory
cell compartments, both of which are needed for sustaining a responsive immune system. Simplistically, the
effector T cells require active antigenic stimulation to eliminate pathogens. On the other hand, memory T
cells are described as cells which remain present in the absence of antigenic stimulation and have the capacity
to expand rapidly upon secondary challenges. Recently, with the identification of central and effector
memory T cell subsets, tremendous efforts have been devoted to characterize markers on the surfaces of
these cells. Though, various markers have been used to identify the subsets, no single marker that segregates
one subset from the other has been described. Thus, multiple markers are needed to subset the cells in order
to characterize them. Here we report the verification of a nine-color panel (CD3, CD4, CD8, CD45RO,
CD28, CD95, CCR7, Live/Dead Aqua, dump channel-CD19, CD14, CD56, CD16) that can successfully
identify six distinct CD4 and CD8 T cell populations within the naı̈ve and effector cell subsets from human
donors.
Key words Immunophenotyping, Multicolor flow cytometry, Whole blood, T cells, Memory T cells,
Naı̈ve T cells
1 Introduction
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_8, © Springer Science+Business Media, LLC, part of Springer Nature 2019
129
130 Ankit Saxena et al.
2 Materials
Fig. 1 T cell Heterogeneity among naı̈ve and memory subsets. CD45R0, CCR7, CD28, and CD95 helps in
identifying six major subsets of T cells. While differentiating (through activation) from TSCM to TCM, TTM, TEM
and culminating in TTE cells, memory T cells progressively lose or acquire specific surface protein markers.
Following encounter with antigen, these naive T cells develop into effectors, for which phenotype is highly
dynamic and unpredictable. When the antigen is cleared, surviving effector T cells that survive return to
memory state. Bottom-Multiparametric flow cytometry identification of multiple T cell subsets among naı̈ve
and memory compartment in the peripheral blood of healthy individuals according to expression of CD45RO,
CCR7, CD28, and CD95
Optical filters
Table 2
Antibody panel details: antigen specific mAb used for various cell surface antigens
3 Methods
3.1 Blood Sample 1. Whole Blood Lysis: collect blood in sodium heparin, sodium
Collection and Cell citrate, or EDTA tube and keep it at RT on a nutator. Transfer
Preparation 5–10 mL blood with a disposable Pasteur pipette into a 50 mL
falcon tube and add 40–45 mL of RBC lysis buffer. Place the
50 mL falcon tube on a nutator for 2 min. Centrifuge tube at
300 g for 5 min at 25 C. Carefully remove the lysis buffer
and break the cell pellet by slow vortex or gentle tapping.
Repeat addition of 45–50 mL of RBC lysis buffer and keep
cells on nutator for 1 min. Centrifuge tube at 300 g for 5 min
at 25 C followed by lysis buffer removal and pellet breaking.
Now add 10 mL of staining buffer (RT) to the cells and give a
final wash to replace remnant RBC lysis buffer with the staining
buffer. Pellet the cells (5 min, 300 g) and aspirate the superna-
tant. Centrifuge tube at 300 g for 5 min at 25 C to make a
final cell concentration of 1–2 106cells/mL (see Note 4).
2. Add fresh staining buffer and adjust cell suspension to a con-
centration of 1–2 106 cells/mL in ice-cold PBS, 10% FCS. If
large clumps seen the sample may be passed through the
40–100 μm mesh to get rid of clumps and obtain single cell
suspension. Cells are usually stained in polystyrene round-
bottom 12 75 mm2 Falcon tubes. However, they can be
stained in any container for which you have an appropriate
centrifuge (e.g., test tubes, microcentrifuge tubes, and
96-well round-bottomed microtiter plates) (see Note 5).
3. Add titrated amount of fluorochrome-conjugated antibodies
and live/dead aqua stain along with FcR blocker or in the
presence of 2 μL of normal mouse serum to reduce background
staining.
4. Incubate cells at 4 C for 15 min (see Note 6).
5. Add 2 mL of staining buffer and wash the cells (5 min, 300 g).
Resuspend the pellet in 0.4–0.5 mL of staining buffer.
6. If sample acquisition is to be done after 24 h, then fix cells using
fixation buffer. Add 0.5 mL of fixation buffer to the cells and
incubate at RT for 15–20 min in dark (see Note 7).
134 Ankit Saxena et al.
7. Pellet the cells (5 min, 500 g) and decant the fixation buffer-
containing supernatant (paraformaldehyde is toxic: handle with
care; dispose of according to material safety regulations) (see
Note 8).
8. Resuspend cells with 0.5 mL of staining buffer keep it in dark
and cold till samples are acquired on cytometer. (It is advised to
run processed samples as soon as possible or within 72 h of
staining.)
3.2 Data Collection Specific methods depend on the available make and model of flow
on Flow Cytometer cytometer. It is important to appropriately establish forward and
side scatter gates to exclude debris and cellular aggregates from
analysis. If cells are stained with multiple fluorescent labels, atten-
tion must be paid to balance the signal amplification among photo-
multiplier tubes (PMTs) in a way that particular fluorophore is
brighter in its respective PMT in order to minimize potential
spillover between fluorochromes and detectors.
1. Single-color controls for each fluorochrome is prepared and
unstained cells are kept as negative control. Single-color con-
trols can be prepared both by using cells or compensation
beads as long as bright signal is observed in each respective
detector PMT (see Note 9).
2. Here a BD LSR-II was used to acquire stained samples
(Table 3) using BD FACS DIVA v8.0 (BD) acquisition and
analysis software.
Table 3
Rationale for panel development. The table highlights important points to be considered in order to
successfully design and execute a multicolor panel. Points to be considered are ordered under
priority rating followed by rationale and reagents concerned
13. Enter a compensation setup name, click Link and Save, and
click OK. To apply compensation values to that experiment’s
cytometer settings, click “Apply Only.”
14. Toggle back to global work sheet (in case gating strategy
changes in different tube then use “sheet” instead of “Global
work sheet”).
15. Use menu commands or plot buttons to create dot plots,
contour plots on worksheet.
16. To create a plot using a plot button, click the appropriate
button on the worksheet toolbar, and click once on the work-
sheet to draw a plot of default size.
17. Acquire cells at an event rate of 3000–5000 events per second
and record data.
18. Use a series of plots and establish a gating hierarchy to ensure
recording of debris and doublet free live T cells (at least
250,000 single live T cells to ensure sufficient number of rare
subsets to analyze in recorded data file, if needed).
19. Postacquisition, the fcs files should be transferred as follows:
Right-click on the Experiment and select Export fcs Files. Save
files in the desired folders. This will transfer all your compensa-
tion files as well as sample fcs files.
3.3 Data Analysis 1. On Flow-jo software open a new workspace and add compen-
sation fcs files and samples fcs files in separate group folders.
2. If required, compensation can be recalculated using compensa-
tion fcs files and newly calculated compensation can be applied
to the sample fcs files for further data analysis (see Note 11).
3. Open the fcs files and start creating gates as follows.
4. Define the gated populations as shown in (Fig. 2a, b).
5. Lymphocyte gate: Create bivariant polychromatic plot and gate
click FSC-A on X-axis and SSC-A on Y axis. Make a polygonal
gate around lymphocyte population as shown. Please ensure in
active gate check box gate events inside is checked on the flow-
jo plot.
6. Singlet gating: select gate and right-click, select drill down
bivariant polychromatic plot with FSC-A on X axis and
FSC-H on Y axis for preselected lymphocytes. Make a gate
around singlets. If the area scaling for FSC detector and Blue
LASER is properly adjusted while data recording, then singlets
will be visible on a diagonal line when FSC-H and FSC-A
parameters are plotted as X and Y axis of bivariate plot. Doub-
lets will fall toward FSC-A but not FSC-H and can be gated out
to identify single cells.
7. Live cell gating: Gate Aqua LD negative subsets as live cells.
Immunophenotyping Naı̈ve, Memory, and Effector T Cells 137
Fig. 2 Gating strategy for identifying naı̈ve and memory T cell subsets. (a)The sequential gating strategy
defines gated total lymphocytes from whole blood lysed sample. (b) Lymphocytes were gated for singlets, live
and dump channel which excluded B, T, NK, and Monocytes. CD3 gated cells were further gated for CD4, CD8
followed by CD45RO and CCR7. On the basis of CD45RO and CCR7 the memory and naı̈ve T cells were defined
as follows. From CD45RO–CCR7+ subset gate on CD95 and CD28. TSCM– CD45RO–CCR7+ CD28 bright CD95+,
TN– CD45RO–CCR7+ CD95–CD28+, TCM– CD45RO+CCR7– CD95+CD28+, TTM– CD45RO+CCR7– CD95
+CD28+, TEM– CD45RO+CCR7– CD95+CD28–, TTE– CD45RO–CCR7–CD28– and CD95–
138 Ankit Saxena et al.
Fig. 2 (continued)
4 Notes
Also, unfixed cells kept long in azide buffer may show more
death than normal.
2. To avoid nonspecific binding of antibodies/reagents each
fluorochrome-conjugated antibody/reagent should be opti-
mally titrated before use.
3. High-throughput screen (HTS) should be set according to the
manufacturer’s instructions; please refer to the manual. It is
Important to note that there is a dead volume of 30 μL for
plates of all kind. The samples should preferably be run at a low
flow rate between 1 and 2 μL/s for up to 106 cells.
4. Avoid prolonged (>5 min) incubation with ACK lysis buffer as
it will significantly affect the lymphocyte viability.
5. Supplementing the medium with 20 μg/mL DNase helps to
dissociate cell aggregates that result from DNA released by
dead cells. This is especially helpful if working with cryopre-
served cells as freeze–thaw may cause cell lysis.
6. Cell staining at 4 C in the dark is a preferred method as it
avoids antibody capping as well as photobleaching.
7. If there are any questions about the pathogenicity of the cells
involved, then fixation of cells after staining and before running
them through a flow cytometer is highly recommended for
immunophenotyping purposes, not for live cell sorting.
8. Fixative solution should be removed after incubation by wash-
ing cells with FACS buffer. Prolonged fixation may alter fluo-
rochrome properties and may lead to spillover in other
channels.
9. Compensation beads should not be washed—as it may lead to
loss of beads.
10. Optimize PMT voltages using unstained and single colors in
order to obtain minimal spectral overlap.
11. Compensation matrix may still require to be adjusted during
analysis. After adjusting the compensation electronically
update biexponential transformation. On FlowJo—Select Plat-
form<Custom transformation<Compensation matrix. Select
appropriate compensation matrix. And appropriately adjust
Biexponential scales in order to visualize the population clus-
ters on scales.
References
1. Maecker HT, McCoy JP, Nussenblatt R (2012) 2. De Rosa SC, Herzenberg LA, Herzenberg LA
Standardizing immunophenotyping for the et al (2001) 11-color, 13-parameter flow cyto-
human immunology project. Nat Rev Immunol metry: identification of human naive T cells by
12(3):191–200. https://doi.org/10.1038/ phenotype, function, and T-cell receptor diver-
nri3158 sity. Nat Med 7(2):245–248. https://doi.org/
10.1038/84701
140 Ankit Saxena et al.
3. Picker LJ, Singh MK, Zdraveski Z et al (1995) 6. Zhang N, Bevan MJ (2011) CD8(+) T cells: foot
Direct demonstration of cytokine synthesis het- soldiers of the immune system. Immunity 35
erogeneity among human memory/effector T (2):161–168. https://doi.org/10.1016/j.
cells by flow cytometry. Blood 86 immuni.2011.07.010
(4):1408–1419 7. Mahnke YD, Brodie TM, Sallusto F et al (2013)
4. Sprent J, Cho JH, Boyman O et al (2008) T cell The who’s who of T-cell differentiation: human
homeostasis. Immunol Cell Biol 86 memory T-cell subsets. Eur J Immunol 43
(4):312–319. https://doi.org/10.1038/icb. (11):2797–2809. https://doi.org/10.1002/
2008.12 eji.201343751
5. Sprent J, Tough DF (1994) Lymphocyte life- 8. Golubovskaya V, Wu L (2016) Different subsets
span and memory. Science 265 of T cells, memory, effector functions, and
(5177):1395–1400 CAR-T immunotherapy. Cancers (Basel) 8(3).
https://doi.org/10.3390/cancers8030036
Chapter 9
Abstract
Regulatory T cells, also known as Tregs, play a pivotal role in maintaining homeostasis of the immune
system and self-tolerance. Tregs express CD3, CD4, CD25, and FOXP3 but lack CD127. CD4 and CD3
identify helper T lymphocytes, of which Tregs are a subset. CD25 is IL-2Rα, an essential activation marker
that is expressed in high levels on Tregs. FOXP3 is the canonical transcription factor, important in the
development, maintenance, and identification of Tregs. CD127 is IL-7 receptor, expressed inversely with
suppression, and is therefore downregulated on Tregs. Flow cytometry is a powerful tool that is capable of
simultaneously measuring Tregs along with several markers associated with subpopulations of Tregs,
activation, maturation, proliferation, and surrogates of functional suppression. This chapter describes a
multicolor flow cytometry-based approach to measure human Tregs, including details for surface staining,
fixation/permeabilization, intracellular/intranuclear staining, acquisition of samples on a flow cytometer,
plus analysis and interpretation of resulting FCS files.
Key words Tregs, nTregs, iTregs, Effector tregs, Immunophenotyping, Flow cytometry, FoxP3,
CD127, Suppressor T cells
1 Introduction
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_9, © Springer Science+Business Media, LLC, part of Springer Nature 2019
141
142 Janet Staats
2 Materials
(a) Basic panel with CD127 includes CD3, CD4, CD25, and
CD127. Requires surface stain only.
(b) Basic panel with FOXP3 includes CD3, CD4, CD25, and
FOXP3. Requires surface and intranuclear stain. CD4,
CD25, CD127, FOXP3, Ki67, and CD45RA.
(c) HIPC panel CD3, CD4, CD25, CD127, CD194,
CD45RO, and HLA-DR. Requires surface stain only.
(d) CIP panel includes CD3, CD4, CD25, CD127, FOXP3,
CD45RA, and Ki67. Requires surface stain and intracel-
lular/intranuclear stain.
11. Nuclear fixation/permeabilizing solution (Fix/Perm): com-
mercially available, usually as a concentrate, prepare and store
according to the manufacturer’s instructions (see Note 6).
12. Permeabilization buffer (Perm buffer): commercially available,
usually comes with the Fix/Perm as a concentrate, prepare and
store according to the manufacturer’s instructions.
13. Compensation beads (see Note 7).
14. 1% Fixative: PBS with 1% formalin. Add 5 mL 10% formalin to
a 45 mL PBS in a 50 mL conical tube, cap and mix by inverting
several times, store at ambient temperature for up to 1 month
after preparation (see Note 8).
15. Vacuum manifold, if staining in plates (see Note 9).
3 Methods
1. Read all steps and Notes prior to performing the assay, as much
advanced preparation is required to perform this assay.
2. Perform all steps on ice or at 4 C unless otherwise noted,
including centrifugation.
3. Store buffers on ice until use unless otherwise noted.
4. Vital dye stain.
(a) Add 100 μL whole blood or viable PBMC viable cells to
the staining tubes or wells (see Note 10).
Note, for the remainder of the assay, through sample
acquisition, the samples must be protected from light and
kept on ice, unless otherwise noted (see Note 11).
(b) Wash cells by adding FACSWash buffer, 2 mL for tubes or
100 μL for wells.
(c) Centrifuge tubes for 5 min or plates for 3 min at 350 g
and 4 C (see Note 12).
(d) Decant tubes or aspirate wells to remove supernatant (see
Note 13).
Immunophenotyping of Human Regulatory T Cells 149
Fig. 1 N N compensation plot. Dot plots in Fig. 1 represent an N N plot for using to visualize compensation
errors. Overcompensation and undercompensation errors are noted. The over compensation of Zombie from
the HLA-DR creates a subnegative HLA-DR population that is visible in the HLA-DR versus CD45RO dot plot, as
well as other Y axes markers paired with HLA-DR. Subnegative values are usually caused by
overcompensation
Fig. 2 Treg gating scheme. Cells obtained from a normal donor were surface stained with the HIPC panel using
lyoplates and acquired on a BD LSRFortessa. CD25 versus CD127 is used to identify Tregs. The gates are
drawn as described for Time, Zombie-, Singlets (FSC and SSC), Lymphocytes, CD3+CD4+ Helper T cells, and
CD25+CD127low Tregs. The proportion of Tregs is 8.23% of Helper T cells, in the normal range of
approximately 5–9%. The gates are listed above each respective dot plot and green arrows indicate the
gating hierarchy from one gate to the next. The number of events are shown for the whole FCS file (925428)
and CD3+CD4+ (labeled CD4+) (177801); 8.23% of 177801 is 14626 Treg events collected from a total of
2 106 viable PBMCs added to the staining well. This is a sufficient number of events to continue with
analyzing Treg states of being, if desired
Fig. 3 CD25 versus FOXP3 Treg analysis region. Cells obtained from an HIV+
patient were stained for surface CD25 and intranuclear FOXP3 (PCH101 clone)
using the optimized panel in OMIP 006 and acquired on an LSRII [56]. The
sample in the left plot is stained with FOXP3 (Y axis). The frequency of Tregs in
the HIV+ sample are low, but close to the lower limit of the normal range. The
control on the right plot is a gating control. It contains all markers except FOXP3,
like an FMO control. However, unlike an FMO control, the gating control includes
an isotype control for PE, used at the same concentration of the FOXP3 mAb. In
this instance the gating control was not helpful due to a shift in the PE negative.
The shift in the PE negative population is an artifact with two possible causes.
Negative drag-up created by improper washing that leaves unbound conjugated
mAbs in solution that is excited by the LASER. Staining fixed/permeabilized cells
with intracellular and intranuclear markers. This example is likely a combination
of the two artifacts. In this example, the PE gate was placed between the visually
discrete populations of FOXP3 and FOXP3+ populations in the sample, called
an internal negative control. If a dimmer fluorophore or different clone for FOXP3
had been used, the resulting FOXP3 staining might not have been so clearly
separated, making the analysis much more challenging. Proper washing and
blocking the intracellular or intranuclear mAb helps to reduce these artifacts
Fig. 4 HIPC panel gating scheme. Cells obtained from a normal donor were surface stained with the HIPC panel
using lyoplates and acquired on a BD LSRFortessa. CD25 versus CD127 is used to identify Tregs. The gates
are drawn as described for Time, Zombie-, Singlets (FSC and SSC), Lymphocytes, CD3+CD4+ Helper T cells,
and CD25+CD127low Tregs. In addition to the Treg markers, the HIPC panel includes markers to measure the
Treg states of being, namely maturation and activation. From the Treg gate, eTregs are identified as CD194+
and from the eTregs memory (CD45RO+) and activated memory (HLA-DR+CD45RO+) Tregs are also
identified. CD25, CD127, CD194, CD45RO, and HLA-DR gates and regions were set using FMO controls.
Figures 2 and 4 are created from the same data file
Fig. 5 FMO control for HLA-DR. Cells obtained from a normal donor were surface
stained with the HIPC panel using lyoplates and acquired on a BD LSRFortessa.
CD25 versus CD127 is used to identify Tregs. Markers for maturation and
activation, namely, CD194, CD45RO, and HLA-DR, were included in the panel.
The plot on the left is a sample stained with all markers in the panel (a vital dye,
CD3, CD4, CD25, CD127, CD194, CD45RO, and HLA-DR). The plot on the left is
an FMO control for HLA-DR, stained with all markers in the panel except for
HLA-DR. The X quadrant was placed using the HLA-DR FMO control
4 Notes
from the tubes with the pellet positioned on the top of the
tube. Follow these recommendations for all decanting steps.
14. Practice using pipettes prior to using them to pipette small
volumes of vital dye or antibodies. When pipetting smaller
volumes check the pipette tips to verify that the volume is
accurate. Quality tips are graduated and have lines for verify
the volumes. After dispensing the liquid verify that the liquid is
no longer in the tip. If pipettes are not used properly the
appropriate amount of liquid is not added stain the cells,
resulting in variable results, usually missing or lower staining
than expected. Refer to manufacturer instructions for more
details. Calibrate pipettes every 6 months to ensure accuracy.
Follow these recommendations for all steps that involve pipet-
ting small volumes.
15. Fluorescent reagents, including vital dyes and mAbs, should be
pretitered, under the conditions of the assay, to determine the
optimal concentration for each reagent [101, 102]. Optimal
staining of vital dyes and fluorescent conjugated mAbs is
achieved by using saturating amounts. Saturation is deter-
mined through titrations based on mass, usually performed
using serial twofold dilutions. Reagents to be used for surface
stain must be titered as a surface stain using the same tempera-
tures, wash buffer and fixative as will be used in the final assay.
Reagents to be used for intracellular or intranuclear stain must
be titered as intracellular or intranuclear stain using the same
temperatures, Fix/Perm, Perm buffer, and fixative as will be
used in the final assay.
16. Mix cells thoroughly after adding each reagent added. When
mixing cells avoid creating air bubbles as these increase surface
tension, leading to cell death. After mixing cells, visually
inspect the tubes or wells to see if a pellet remains. If a pellet
is observed, cells were not mixed properly and should be
remixed until no pellet remains. If cells are not mixed properly,
the stain results will be poor quality. In addition, cells that are
not resuspended properly are left on a pellet, causing increased
cell death and assay variability. Avoid overmixing cells, they
only need to be mixed enough to resuspend the cell pellet in
the added solution. Overmixing might also result in poor
quality results.
17. Create mAb mixes immediately prior to use. Do not allow
them to set longer than 30 min prior to use. Use the appropri-
ate buffer to create mAb mixes, FACSWash buffer for surface
stain mix and Perm buffer for intracellular/intranuclear stain
mix. If more than one brilliant dye is used to stain cells, then a
commercially available stabilization buffer that is specific for
brilliant dyes should be used to prepare the mAb mix. The mAb
168 Janet Staats
References
10. Saito T et al (2016) Two FOXP3(+)CD4(+) T autoimmune and inflammatory diseases. Nat
cell subpopulations distinctly control the Rev Immunol 15(5):283–294
prognosis of colorectal cancers. Nat Med 22 25. Reissig S et al (2017) Elevated levels of Bcl-3
(6):679–684 inhibits Treg development and function
11. Figueiredo AS, Schumacher A (2016) The T resulting in spontaneous colitis. Nat Com-
helper type 17/regulatory T cell paradigm in mun 8:15069
pregnancy. Immunology 148(1):13–21 26. Tan HL et al (2013) T regulatory lympho-
12. Engler JB et al (2017) Glucocorticoid recep- cytes and endothelial function in pediatric
tor in T cells mediates protection from auto- obstructive sleep apnea. PLoS One 8(7):
immunity in pregnancy. Proc Natl Acad Sci U e69710
S A 114(2):E181–e190 27. Ni K et al (2015) Th17/Treg balance in chil-
13. Feyaerts D et al (2017) Human uterine lym- dren with obstructive sleep apnea syndrome
phocytes acquire a more experienced and tol- and the relationship with allergic rhinitis. Int J
erogenic phenotype during pregnancy. Sci Pediatr Otorhinolaryngol 79(9):1448–1454
Rep 7(1):2884 28. Grant J et al (2009) Validated protocol for
14. Guzman-Genuino RM, Diener KR, Regu- FoxP3 reveals increased expression in type
latory B (2017) Cells in pregnancy: lessons 1 diabetes patients. Cytometry B Clin
from autoimmunity, graft tolerance, and can- Cytom 76(2):69–78
cer. Front Immunol 8:172 29. Hori S (2014) Lineage stability and pheno-
15. Cipolletta D et al (2012) PPAR-gamma is a typic plasticity of Foxp3(+) regulatory T cells.
major driver of the accumulation and pheno- Immunol Rev 259(1):159–172
type of adipose tissue Treg cells. Nature 486 30. Wing JB et al (2017) A distinct subpopulation
(7404):549–553 of CD25( ) T-follicular regulatory cells loca-
16. Qiao YC et al (2016) Changes of regulatory T lizes in the germinal centers. Proc Natl Acad
cells and of proinflammatory and immuno- Sci U S A 114(31):E6400–e6409
suppressive cytokines in patients with type 31. Weinstein JS et al (2016) TFH cells progres-
2 diabetes mellitus: a systematic review and sively differentiate to regulate the germinal
meta-analysis. J Diabetes Res 2016:3694957 center response. Nat Immunol 17
17. Serr I et al (2016) Type 1 diabetes vaccine (10):1197–1205
candidates promote human Foxp3(+)Treg 32. Sage PT, Sharpe AH (2016) T follicular regu-
induction in humanized mice. Nat Commun latory cells. Immunol Rev 271(1):246–259
7:10991 33. Sugiyama D et al (2013) Anti-CCR4 mAb
18. Visperas A, Vignali DA (2016) Are regulatory selectively depletes effector-type FoxP3
T cells defective in type 1 diabetes and can we +CD4+ regulatory T cells, evoking antitumor
fix them? J Immunol 197(10):3762–3770 immune responses in humans. Proc Natl Acad
19. Joly AL et al (2018) Alternative splicing of Sci U S A 110(44):17945–17950
FOXP3 controls regulatory T cell effector 34. Steinborn A et al (2012) Pregnancy-
functions and is associated with human ath- associated diseases are characterized by the
erosclerotic plaque stability. Circ Res 122 composition of the systemic regulatory T cell
(10):1385–1394 (Treg) pool with distinct subsets of Tregs.
20. Noval Rivas M et al (2015) Regulatory T cell Clin Exp Immunol 167(1):84–98
reprogramming toward a Th2-cell-like line- 35. Maecker HT, McCoy JP, Nussenblatt R
age impairs oral tolerance and promotes food (2012) Standardizing immunophenotyping
allergy. Immunity 42(3):512–523 for the human immunology project. Nat Rev
21. Bacher P et al (2016) Regulatory T cell speci- Immunol 12(3):191–200
ficity directs tolerance versus allergy against 36. Li Z et al (2015) FOXP3+ regulatory T cells
aeroantigens in humans. Cell 167 and their functional regulation. Cell Mol
(4):1067–1078.e16 Immunol 12(5):558–565
22. Palomares O et al (2017) Mechanisms of 37. Li N et al (2015) The abnormal expression of
immune regulation in allergic diseases: the CCR4 and CCR6 on Tregs in rheumatoid
role of regulatory T and B cells. Immunol arthritis. Int J Clin Exp Med 8
Rev 278(1):219–236 (9):15043–15053
23. Miyara M, Ito Y, Sakaguchi S (2014) TREG- 38. Iellem A et al (2001) Unique chemotactic
cell therapies for autoimmune rheumatic dis- response profile and specific expression of che-
eases. Nat Rev Rheumatol 10(9):543–551 mokine receptors CCR4 and CCR8 by CD4
24. Klatzmann D, Abbas AK (2015) The promise (+)CD25(+) regulatory T cells. J Exp Med
of low-dose interleukin-2 therapy for 194(6):847–853
Immunophenotyping of Human Regulatory T Cells 175
39. Donnelly C et al (2018) Optimizing human 54. McKinnon KM (2018) Multiparameter con-
Treg immunotherapy by Treg subset selection ventional flow cytometry. Methods Mol Biol
and E-selectin ligand expression. Sci Rep 8 1678:139–150
(1):420 55. Wingender G, Kronenberg M (2015) OMIP-
40. Liu W et al (2006) CD127 expression 030: characterization of human T cell subsets
inversely correlates with FoxP3 and suppres- via surface markers. Cytometry A 87
sive function of human CD4+ T reg cells. J (12):1067–1069
Exp Med 203(7):1701–1711 56. Murdoch DM, Staats JS, Weinhold KJ (2012)
41. Seddiki N et al (2006) Expression of interleu- OMIP-006: phenotypic subset analysis of
kin (IL)-2 and IL-7 receptors discriminates human T regulatory cells via polychromatic
between human regulatory and activated T flow cytometry. Cytometry A 81(4):281–283
cells. J Exp Med 203(7):1693–1700 57. Moncunill G et al (2014) OMIP-024: pan--
42. Allan SE et al (2007) Activation-induced leukocyte immunophenotypic characteriza-
FOXP3 in human T effector cells does not tion of PBMC subsets in human samples.
suppress proliferation or cytokine production. Cytometry A 85(12):995–998
Int Immunol 19(4):345–354 58. Mahnke YD, Beddall MH, Roederer M
43. Tran DQ, Ramsey H, Shevach EM (2007) (2013) OMIP-015: human regulatory and
Induction of FOXP3 expression in naive activated T-cells without intracellular staining.
human CD4+FOXP3 T cells by T-cell recep- Cytometry A 83(2):179–181
tor stimulation is transforming growth factor- 59. Biancotto A et al (2012) OMIP-004:
beta dependent but does not confer a regu- in-depth characterization of human T regu-
latory phenotype. Blood 110(8):2983–2990 latory cells. Cytometry A 81(1):15–16
44. Mailer RKW (2018) Alternative splicing of 60. Givan AL (2011) Flow cytometry: an intro-
FOXP3-virtue and vice. Front Immunol duction. Methods Mol Biol 699:1–29
9:530 61. Snow C (2004) Flow cytometer electronics.
45. Sambucci M et al (2018) FoxP3 isoforms and Cytometry A 57(2):63–69
PD-1 expression by T regulatory cells in mul- 62. Data file standard for flow cytometry (1990)
tiple sclerosis. Sci Rep 8(1):3674 Data file standards committee of the society
46. Hartigan-O’Connor DJ et al (2007) Human for analytical cytology. Cytometry 11
CD4+ regulatory T cells express lower levels (3):323–332
of the IL-7 receptor alpha chain (CD127), 63. Seamer L (2001) Data file standard for flow
allowing consistent identification and sorting cytometry, FCS 3.0. Curr Protoc Cytom.
of live cells. J Immunol Methods 319 Chapter 10: p. Unit 10.2
(1–2):41–52
64. Spidlen J et al (2011) Flow cytometry data
47. McKinnon KM (2018) Flow cytometry: an standards. BMC Res Notes 4:50
overview. Curr Protoc Immunol
120:5.1.1–5.1.11 65. Feher K et al (2016) Multispectral flow cyto-
metry: the consequences of increased light
48. Shapiro HM (2018) Flow cytometry: the collection. Cytometry A 89(7):681–689
glass is half full. Methods Mol Biol 1678:1–10
66. Bagwell CB, Adams EG (1993) Fluorescence
49. Perfetto SP, Chattopadhyay PK, Roederer M spectral overlap compensation for any number
(2004) Seventeen-colour flow cytometry: of flow cytometry parameters. Ann N Y Acad
unravelling the immune system. Nat Rev Sci 677:167–184
Immunol 4(8):648–655
67. Roederer M (2002) Compensation in flow
50. Ashhurst TM, Smith AL, King NJC (2017) cytometry. Curr Protoc Cytom. Chapter 1:
High-dimensional fluorescence cytometry. p. Unit 1.14
Curr Protoc Immunol 119:5.8.1–5.8.38
68. Szaloki G, Goda K (2015) Compensation in
51. McLaughlin BE et al (2008) Nine-color flow multicolor flow cytometry. Cytometry A 87
cytometry for accurate measurement of T cell (11):982–985
subsets and cytokine responses. Part I: panel
design by an empiric approach. Cytometry A 69. McNeil LK et al (2013) A harmonized
73(5):400–410 approach to intracellular cytokine staining
gating: results from an international multi-
52. Mahnke Y, Chattopadhyay P, Roederer M consortia proficiency panel conducted by the
(2010) Publication of optimized multicolor cancer immunotherapy consortium (CIC/-
immunofluorescence panels. Cytometry A 77 CRI). Cytometry A 83(8):728–738
(9):814–818
70. Sun YL et al (2013) Application and effects of
53. Mahnke YD, Roederer M (2007) Optimizing mouse Foxp3 antibody and fixation/permea-
a multicolor immunophenotyping assay. Clin bilization buffer on the detection of CD4+
Lab Med 27(3):469–85, v
176 Janet Staats
98. Byrd T et al (2015) Polystyrene microspheres 103. Bagwell CB (2005) Hyperlog-a flexible
enable 10-color compensation for immuno- log-like transform for negative, zero, and pos-
phenotyping of primary human leukocytes. itive valued data. Cytometry A 64(1):34–42
Cytometry A 87(11):1038–1046 104. Herzenberg LA et al (2006) Interpreting flow
99. Staats JS et al (2014) Toward development of cytometry data: a guide for the perplexed. Nat
a comprehensive external quality assurance Immunol 7(7):681–685
program for polyfunctional intracellular cyto- 105. Tung JW et al (2004) New approaches to fluo-
kine staining assays. J Immunol Methods rescence compensation and visualization of
409:44–53 FACS data. Clin Immunol 110(3):277–283
100. Jaimes MC et al (2011) Quality assurance of 106. Novo D, Wood J (2008) Flow cytometry his-
intracellular cytokine staining assays: analysis tograms: transformations, resolution, and dis-
of multiple rounds of proficiency testing. J play. Cytometry A 73(8):685–692
Immunol Methods 363(2):143–157 107. Richards AJ et al (2014) Setting objective
101. Lugli E et al (2013) Identification, isolation thresholds for rare event detection in flow
and in vitro expansion of human and nonhu- cytometry. J Immunol Methods 409:54–61
man primate T stem cell memory cells. Nat 108. Chan C et al (2008) Statistical mixture mod-
Protoc 8(1):33–42 eling for cell subtype identification in flow
102. Lamoreaux L, Roederer M, Koup R (2006) cytometry. Cytometry A 73(8):693–701
Intracellular cytokine optimization and stan- 109. Maecker HT et al (2005) Standardization of
dard operating procedure. Nat Protoc 1 cytokine flow cytometry assays. BMC Immu-
(3):1507–1516 nol 6:13
Chapter 10
Abstract
In the last years, the family of innate lymphocytes has been growing following the discovery of innate
lymphoid cells (ILCs). ILCs are lymphocytes able to rapidly produce a wide range of soluble mediators in an
antigen-independent fashion. So far, three main subsets of ILCs have been discovered, ILC1, ILC2, and
ILC3, expressing respectively the transcription factors T-bet, GATA3, and Rorγt and secreting distinct
types of cytokines. After their discovery, several studies showed that different pathologies, such as allergic
airway diseases and inflammatory disorders, are sustained by dysfunctional ILCs before adaptive immune
sets in. In this regard, considerable efforts are currently performed to harmonize the identification and
monitoring of ILCs in healthy and pathologic conditions to streamline a uniform immunophenotyping.
Standardized ILC monitoring techniques will accelerate our understanding of these effector innate immune
cells and ultimately facilitate their targeting in the context of infection, cancer, autoimmune disease, and
transplantation.
Key words Human, ILCs, Flow cytometry, Staining, Isolation, Cytokines, Transcription factors,
Expansion
1 Introduction
Innate lymphoid cells (ILCs) are the most recently identified family
of lymphocytes that belong to the innate immune system [1].
Indeed, they lack rearranged antigen-specific receptors, such as
the T-cell receptor (TCR, specifically expressed by T lymphocytes)
or the B-cell receptor (BCR, specifically expressed by B lympho-
cytes), on their cell surface. As a consequence, ILCs do not recog-
nize, bind, and respond to specific antigens but to soluble factors or
to molecules expressed on other immune and nonimmune cell
types. Therefore, they rapidly respond to signals coming from the
extracellular environment and constitute one of the first-line
defense of our body. For this reason, ILCs are considered “early
sentinels” enriched at mucosal and barrier surfaces. They are also
present in primary and secondary lymphoid organs, and they can be
found circulating in the peripheral blood.
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_10, © Springer Science+Business Media, LLC, part of Springer Nature 2019
179
180 Sara Trabanelli et al.
2 Materials
2.3 Extracellular 1. FACS buffer: PBS supplemented with 5 mM EDTA, 0.2% BSA,
Staining of Ex Vivo and 0.2% NaN3. Store at 4 C.
Human ILCs 2. LIVE/DEAD™ fixable green dead cell stain kit for 488 nm
excitation (Invitrogen) (see Note 1).
3. Monoclonal antibodies:
(a) Lineage (Lin): CD4 (clone RPA-T4); CD8 (clone
MEM-31) (ImmunoTools). CD3 (clone UCTH1);
CD14 (clone HCD14); CD15 (clone HI98); CD19
(clone H1B19); CD20 (clone 2H7); CD33 (clone
HIM3-4); CD34 (clone 561); CD203c (clone NP406);
FcεRI (clone AER-37) (Biolegend). CD16 (clone
RMO52) (Beckman Coulter), all FITC conjugated.
(b) CD127 BV421 (clone AO19D5); CD56 Alexa
700 (clone HCD56); CD117 (c-Kit) APC-Fire (clone
104D2); CRTH2 PE (clone BM16); CD161 (clone
HP-3G10) PE-Cy7; PerCPCy5.5 NKp44 (clone P44-8)
or NKp46 (clone 9E2) (Biolegend) (see Notes 2 and 3).
4. 5 mL V-bottom tubes or 96-well plates.
5. 5 mL U-bottom tubes.
182 Sara Trabanelli et al.
3 Methods
Fig. 1 Representative example of human ILC subset identification by multiparametric flow cytometry. ILCs
were identified within the peripheral blood lymphocyte region on the basis of their forward (FSC) and side
scatter (SSC) profiles (FSC low and SSC low) and by excluding from the analysis doublets (FSC H/FSC W dot
plot, followed by SSC A/SSC W dot plot). Total ILCs were gated as Lin CD127+ cells. Then, the different ILC
subsets were determined according to the expression of CRTH2 vs cKit: ILC1 CRTH2 cKit , ILC2
CRTH2+cKit+/ , and ILC3 CRTH2 cKit+. The dot plots shown are the result of a minimum of 106 MNCs
acquired on a flow cytometer. Data were analyzed using FlowJo™ software (TreeStar)
Fig. 2 Representative example of specific transcription factor expression by human ILCs. Total ILCs and ILC
subsets were stained as in Fig. 1, then the cells were fixed and permeabilized and stained for T-bet, GATA3,
and RORγt expression
3.6 Functional 1. After obtaining the total ILC suspension or isolating ILC sub-
Analysis sets, ILCs can be stimulated to assess their intracellular cyto-
of Human ILCs kine production. To this purpose, different ILC subsets must
be stimulated with their specific cytokine mix. ILC1 mix: hrIL-
3.6.1 Intracellular 2 (100 U/mL); hrIL-7 (5 ng/mL); hrIL-12; hrIL-18; hrIL-15
Cytokine Staining (50 ng/mL). ILC2 mix: hrIL-2 (100 U/mL); hrIL-7 (5 ng/
for Evaluation of Cytokine mL); hrIL-33; hrIL-25 and TSLP (50 ng/mL). ILC3 mix:
Production by Human ILCs hrIL-2 (100 U/mL); hrIL-7 (5 ng/mL); hrIL-1β; hrIL-23
(50 ng/mL) (see Note 19).
2. Incubate at 37 C with 5% of CO2 overnight with 2 μg/mL of
Brefeldin A (see Note 20).
3. 1 h before starting the staining, stimulate with 50 ng/mL PMA
and 500 ng/mL ionomycin.
4. At the end of the stimulation, harvest the cells and transfer
them to V-bottom tubes or 96-well plates and proceed with the
extracellular staining (see Subheading 3.3).
5. Wash with FACS buffer and centrifuge for 5 min at 528 g.
6. Resuspend the pellet in 100 μL of fixation buffer (see Subhead-
ing 2.6.1). Incubate for 30 min at room temperature.
7. Add 100 μL, for the 96-well plates, or 200 μL, for the
V-bottom tubes, of a 0.1% saponin solution (see Subheading
2.6.1) and centrifuge for 5 min at 528 g.
8. Prepare your cytokine antibody mix in the 0.1% saponin solu-
tion (see Note 21).
9. Incubate for 30 min at 4 C.
10. Finally wash the cells with the 0.1% saponin solution, centri-
fuge for 5 min at 528 g and resuspend your cells in 200 μL of
FACS buffer. Transfer the stained cells in U-bottom tubes
compatible with your flow cytometer (Fig. 3).
3.7 In Vitro 1. Culture the sorted ILC subsets in 96-well plates, with ILC
Expansion culture medium supplemented with rhIL-2 (100 U/mL) and
and Cryopreservation rhIL-7 (5 ng/mL) for all the subsets.
of Human ILCs 2. Place the plates in an incubator at 37 C, with 5% CO2.
3. Refresh the medium every 48 or 72 h, and split the cells if
necessary (see Note 24).
188 Sara Trabanelli et al.
Fig. 3 Representative example of cytokine production evaluated by intracellular staining of human ILCs. ILCs
were cultured with different cytokine cocktails overnight in the presence of brefeldin A. As negative control,
PBMCs were cultured in absence of stimulating cytokines for the same period of time. After the culture, total
ILCs and ILC subsets were stained as in Fig. 1, then the cells were fixed and permeabilized. Intracellular
expression of IFN-γ, IL-5, and IL-22 was evaluated in each subset, respectively
Immunophenotyping of Human Innate Lymphoid Cells 189
4 Notes
References
1. Eberl G, Colonna M, Di Santo JP et al (2015) 2. Diefenbach A, Colonna M, Koyasu S et al
Innate lymphoid cells: a new paradigm in immu- (2014) Development, differentiation, and
nology. Science 348(6237):aaa6566
192 Sara Trabanelli et al.
Abstract
Dendritic cells (DCs) are powerful antigen presenting cells that are involved in regulating immune
response. Plasmacytoid dendritic cells (pDCs) are subtype of DCs that present in small quantity in the
bone marrow, peripheral blood, and lymph nodes. They are important component of our immune system in
normal condition and diseases. They activate T cells and play a critical role in immune tolerance. In this
chapter we review the immunophenotypic features of pDCs and provide a practical protocol for pDCs
enumeration in the peripheral blood and bone marrow samples.
Key words Flow cytometry, Dendritic cells, Plasmacytoid dendritic cells, Immunophenotyping
1 Introduction
Dendritic cells (DCs) are highly specialized cells that link innate and
adaptive immune response [1]. DCs are a heterogeneous popula-
tion of cells that regulate the immune response through their
function as potent antigen presenting cells. They have a special
capacity to present the antigen to the naı̈ve T cells which lead to
T cell activation, and maturation. Additionally, depending on the
inducing stimulus, they play a critical role in immune tolerance [2].
Two subtypes of DCs have been well described, conventional
dendritic cells (cDCs) and plasmacytoid dendritic cells (pDCs)
both derived from a common DC precursor arising from the
CD34+ hematopoietic stem cell (HSC). Precursor pDC
(pre-pDC) differentiate into pDCs in the bone marrow then
released into peripheral blood where they reside in lymphoid tissues
such as lymph nodes, tonsils, spleen, thymus, bone marrow, and
Peyer’s patches.
Mature pDCs stimulate distinct types of CD4 T helper (Th-1)
cells in response to endogenous antigens. Likewise, pDCs can be
stimulated by exogenous antigens like viruses [3]. Activated pDCs
secret type I interferon that activate and enhance T cells and natural
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_11, © Springer Science+Business Media, LLC, part of Springer Nature 2019
193
194 Abdullah Alsuwaidan et al.
killer (NK) cells [4,–6]. Thus, pDCs play a critical role in variety of
immune-mediated pathophysiological process such as graft-versus-
host disease (GvHD), allograft rejection, and autoimmune disor-
ders [2, 7].
Following hematopoietic stem cell transplantation (HSCT),
pDC facilitate HSC engraftment and induce tolerance to prevent
GvHD [5]. Several studies reveal the roles of pDC in immune
reconstitution after HSCT for hematolymphoid malignancies and
disease relapse, patient survival and risk of GvHD [1, 8]. The
higher percentages of pDCs, after allogeneic HSCT for leukemia,
are associated with successful bone marrow engraftment. On the
other hand, lower pDCs count predicts relapse, death, and acute
graft-versus-host disease [1]. In fact, it has been reported that the
percentage of pDCs following HSCT is an independent indicator
for adverse clinical outcomes [1, 8].
pDCs population are present in low quantity in bone marrow,
peripheral blood and lymph nodes. They show plasmacytoid mor-
phology and can be defined by their immunophenotype. In general,
normal mature pDCs in the bone marrow express CD4, CD22dim,
CD33, CD36, CD38, CD45, CD123, CD303, and HLA-DR,
and they are negative for CD5, CD11b, CD13, CD15, CD34,
CD56, and CD64 [8, 9]. Precursors of pDC often express CD34.
Of note, subtle immunophenotypic variations have been described
in pDCs in lymph nodes compared to bone marrow [9]. Further-
more, neoplastic pDCs may show immunophenotypic aberrancy as
in cases of blastic plasmacytoid dendritic cell neoplasm (e.g., CD56
expression [10]).
Given the critical role of pDCs in the immune system and
particularly in allogeneic HSCT, this chapter provides a practical
approach to identify and quantify pDCs using multiparametric flow
cytometry. Our protocol is designed to distinguish pDCs from
other hematopoietic cells present in bone marrow.
2 Materials
2.5 Monoclonal The listed antibody panels (Tables 1, 2, and 3) are suggested
Antibodies monoclonal antibodies combinations [11] to allow the enumera-
tion of the pDCs among other myeloid and lymphoid cells using
four- or ten-color flow cytometry, (see Notes 1–3).
Table 1
Suggested monoclonal antibodies for 10-color flow cytometry panels
Table 2
Antibodies panel for four-color flow cytometry
Table 3
Antibodies panel for 10-color flow cytometry
APC-
Tube FITC PE PerCP-Cy5.5 PE-Cy7 APC R700 APC-H7 BV421 V500-C BV605
1 CD15 CD13 CD4 CD38 CD11b CD64 HLA-DR CD123 CD45 –
2 CD36 CD34 CD4 CD303 CD22 – HLA-DR CD123 CD45 CD56
3 Methods
3.3 Data Acquisition 1. Sequential gating strategies are recommended to identify and
quantify the pDCs (see Notes 1, 7 and 8), (Fig. 1).
(a) Gate out all doublets and nonviable cells (see Notes 9
and 10).
(b) Gate all HLA-DR+/CD45+ and all CD4+ cells
population.
(c) Exclude T lymphocytes (CD3+).
(d) Exclude monocytes (CD14+/CD64+/CD36+).
(e) Exclude all events that are CD123– and/or HLA-DR–.
(f) Report the percentage of pDCs as pDC% of total viable
events.
3.4 Data 1. Nonneoplastic pDCs in the bone marrow and peripheral blood
Interpretation frequently express CD4, CD22dim, CD33, CD36, CD38,
CD45, CD123, CD303, and HLA-DR [12, 13], and lack
198 Abdullah Alsuwaidan et al.
Fig. 1 Multiparametric flow cytometry analysis of a normal bone marrow specimen. The multicolor flow
cytometry panel includes several monoclonal antibodies such as CD2, CD4, CD5, CD11b, CD11c, CD13, CD15,
CD22, CD33, CD38, CD45, CD56, CD64, CD123, CD303, HLA-DR. pDCs (red population) are found just below
monocytes (green population), that is, same size to monocytes but less SSC, in the FSC/SSC plot. Additionally,
pDCs are slightly dimmer for CD45 with less SSC compared to monocytes in the CD45/SSC plot. In CD123/
HLA-DR plot, pDCs are double positive for CD123/HLA-DR compared to basophils (blue population) that are
CD123+/HLA-DR–. In this case, the nonneoplastic pDCs are characteristically positive for CD4, CD22, CD38,
CD45, CD123, CD303, and HLA-DR, and largely negative for CD2, CD5, CD11c, CD13, CD15, CD33, CD56, and
CD64
CD45 dimmer than mature pDCs which are CD34 [14], (see
Note 14).
4 Notes
FSC-A vs. SSC-A plot. The other option to remove the nonvi-
able cells is to use a DNA-binding dye, such as 7-amino-acti-
nomycin D (i.e., 7-AAD) which can provide a good assessment
of sample integrity. However, it is essential to ensure that
excluding nonviable cells do not interfere with the analysis of
cell of interest.
11. Our protocol can be incorporated into routine evaluation of all
bone marrow and peripheral blood samples for pDCs, particu-
larly if there is increase in nonmonocytic CD4+/CD3
population.
12. The use of isotype controls is essential to exclude false-positive
events (nonspecific staining).
13. It is advised to include Fluorescence Minus One (FMO) con-
trols in multiparametric flow cytometry which help in identify-
ing the proper gating boundaries.
14. Based on our experience, the early immature pDC may down-
regulate CD303 expression. This is mainly seen in bone mar-
row sample.
References
1. Reddy V, Iturraspe JA, Tzolas AC (2004) Low 7. Elze MC, Ciocarlie O, Heinze A (2015) Den-
dendritic cell count after allogeneic hemato- dritic cell reconstitution is associated with
poietic stem cell transplantation predicts relapse-free survival and acute GVHD severity
relapse, death, and acute graft-versus-host dis- in children after allogeneic stem cell transplan-
ease. Blood 103(11):4330–4335. Epub 2004 tation. Bone Marrow Transplant 50
Feb 12 (2):266–273
2. Abe M, Wang Z, de Creus A (2005) Plasmacy- 8. Su RJ, Green R, Chen M (2018) Enumeration
toid dendritic cell precursors induce allogeneic of bone marrow plasmacytoid dendritic cells by
T-cell hyporesponsiveness and prolong heart multiparameter flow cytometry as a prognostic
graft survival. Am J Transplant 5 marker following allogeneic hematopoietic
(8):1808–1819 stem cell transplantation. Blood Cells Mol Dis
3. McKenna K, Beignon AS, Bhardwaj N (2005) 69:107–112
Plasmacytoid dendritic cells: linking innate and 9. Pilley J, Chen W, Fuda F et al (2017) The
adaptive immunity. J Virol 79(1):17–27. immunophenotypic appearance of plasmacy-
Review toid dendritic cells in various specimens. Mod
4. Tomasello E, Naciri K, Chelbi R (2018) Pathol 30(Suppl):370A
Molecular dissection of plasmacytoid dendritic 10. Swerdlow SH, Campo E, Jaffe ES et al (2017)
cell activation in vivo during a viral infection. WHO classification of tumours of haemato-
EMBO J 37(19):e98836 poietic and lymphoid tissues, 4th edn. IARC
5. Auletta JJ, Devine SM, Waller EK (2016) Plas- Press, Lyon, pp 174–177
macytoid dendritic cells in allogeneic hemato- 11. Chen W, Luu HS (2017) Immunophenotyping
poietic cell transplantation: benefit or burden? by multiparameter flow cytometry. Methods
Bone Marrow Transplant 51(3):333–343 Mol Biol 1633:51–73
6. Swiecki M, Colonna M (2015) The multiface- 12. Alculumbre S, Pattarini L (2016) Purification
ted biology of plasmacytoid dendritic cells. Nat of human dendritic cell subsets from peripheral
Rev Immunol 15(8):471–485 blood. Methods Mol Biol 1423:153–167
Enumeration of Plasmacytoid Dendritic Cells in Peripheral Blood and Bone. . . 201
13. Setoodeh S, Fuda F, Chen W et al (2017) Flow 14. Martı́n-Martı́n L, Almeida J, Hernández-
cytometric immunophenotypic features of Campo PM (2009) Immunophenotypical,
blastic plasmacytoid dendritic cell neoplasm: a morphologic, and functional characterization
single institution experience. Mod Pathol 30 of maturation-associated plasmacytoid den-
(Suppl):376A–377A dritic cell subsets in normal adult human bone
marrow. Transfusion 49(8):1692–1708
Chapter 12
Abstract
Flow-cytometric detection of circulating endothelial cells and endothelial microparticles is an essential tool
in studies of vascular diseases. Here we describe the principles and detailed methods for human blood
sample processing, storage, labeling, and gating of circulating endothelial elements.
1 Introduction
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_12, © Springer Science+Business Media, LLC, part of Springer Nature 2019
203
204 Nicholas Wanner and Kewal Asosingh
2 Materials
2.1 Blood Sample 1. 10% Formaldehyde: 16% formaldehyde, 0.9% saline. A new
Processing 10 mL ampule of 16% electron microscopy-grade formalde-
hyde is diluted with 6 mL of saline to create a 10% solution.
Store at room temperature for up to 2 weeks.
2. 10% Triton X-100: Triton X-100 detergent, 0.9% saline. Add
100 mL of 0.9% saline to a glass jar. Add 20 mL of Triton
X-100 to the saline. Measure out an additional 80 mL of saline
and with the pipet used to add the Triton X-100, add the saline.
Add a magnetic stirring flea and allow all particles to dissolve
while stirring on a magnetic stirrer. Aliquot 932 μL into cryo-
vials and store in liquid nitrogen.
3. Lyse/perm Buffer: A vial of 10% Triton X-100 is thawed then
added to 39 mL of 0.9% saline. The buffer is prewarmed to
37 C before using.
4. Phosphate Buffered Saline (PBS): 137 mM sodium chloride,
2.7 mM potassium chloride, 9.8 mM phosphate buffer. Does
not contain calcium or potassium.
5. Wash Buffer: 4% fetal bovine serum (FBS), PBS. 20 mL of FBS
is added to 480 mL of PBS. The solution is filtered through a
0.22 μm filter. Store at 4 C.
6. Freezing Medium: RPMI 1640, 20% FBS, 10% glycerol. Com-
bine 35 mL of RPMI 1640 with 10 mL of FBS and 5 mL of
glycerol. Store at 20 C.
Endothelial Flow Cytometry 205
2.2 Cell Surface 1. Antibody diluting Buffer: 1% bovine serum albumin (BSA),
Staining for Flow PBS. Weigh and add 5 g of BSA to 500 mL of PBS with a
Cytometry stirring flea and stir until dissolved. Filter the solution through
a 0.22 μm filter. Store at 4 C.
2. Blocking Buffer: antibody diluting buffer, 10% normal goat
serum (NGS) Abcam, Human Trustain Fc-block, Biolegend.
Amount made is based on number of samples being stained.
250 μL made per sample; 25 μL of NGS and 6.25 μL of
Fc-block added to 218.75 μL of diluting buffer.
3. 5 mL Polystyrene Round-Bottom Tubes.
4. Antibodies: CD3 PE-Cy7, Biolegend, use at 1/1600. CD19
PE-Cy5, Biolegend, use at 1/27. CD45 Alexa Fluor 700, Bio-
legend, use at 1/800. CD34 FITC, BD Bioscience, use at 1/5.
All antibodies are diluted in antibody diluting buffer.
5. 40 ,6-Diamidino-2-phenylindole (DAPI), Life Technologies,
use at 1/400.
6. AbC Total Antibody Compensation Bead Kit, Life
Technologies.
7. Ultra Rainbow Calibration Kit, Spherotech.
8. CS&T Research Bead, Becton Dickinson.
3 Methods
3.1 Blood Processing 1. Pipette 4 mL of whole blood per 50 mL tube. If less than 4 mL
for White Blood Cells of blood is available, add saline to bring the volume to 4 mL.
2. Add 2.6 mL of 10% formaldehyde to the blood and incubate
for 10 min at room temperature (RT).
206 Nicholas Wanner and Kewal Asosingh
3.2 Cell Surface 1. Thaw aliquots of cells in cryovials at room temperature for
Staining for Flow 15 min.
Cytometry 2. Wash cells of freezing medium by diluting suspended cells with
4 mL of PBS in 5 mL polystyrene round bottom tubes.
3. Centrifuge samples at 900 g for 5 min (all subsequent spins
will be at 900 g for 5 min), then aspirate the supernatant.
4. Resuspend each sample in 1 mL of blocking buffer by pipetting
up and down. Incubate cells at RT in the dark for 20 min.
5. Remove an aliquot for the unstained sample.
6. Centrifuge all samples and aspirate.
7. The cells that are stained with DAPI are resuspended in 75 μL
of diluted DAPI (1/400 in PBS). Vortex the cells and incubate
for 15 min at RT in the dark.
8. Wash with 1 mL of diluting buffer then centrifuge.
9. Resuspend the DAPI stained cells in 300 μL of diluting buffer
and divide into the DAPI comp, FMOs, and sample tubes
(75 μL per tube) (see Note 2).
10. Add 200 μL of diluting buffer to all tubes except the DAPI
comp tube. Put the DAPI comp tube aside and keep at 4 C in
the dark.
11. Centrifuge remaining samples and aspirate.
12. Add 100 μL of appropriate antibody dilutions to respective
tubes and vortex (see Note 3). Incubate at room temperature
on the platform lab shaker for 30 min.
13. Wash samples with 500 μL of diluting buffer and centrifuge.
14. Aspirate the supernatant and resuspend in 100 μL of BD
FACSFlow, Becton Dickinson and keep at 4 C until acquisi-
tion on the flow cytometer (see Note 4). See Fig. 1 for gating
strategy of endothelial cells.
Endothelial Flow Cytometry 207
Fig. 1 Circulating endothelial cell gating strategy. Time gating (a), and aggregate exclusion (b) corrected for
fluidic disturbances and cell doublets. DAPI was used to select cells in G0/G1 (c). CD19+ (d) and CD3+ (e) cells
were excluded, then CD45 cells were selected (f) using the CD45 FMO. Finally CECs were gated using CD34
(g). The CD34 FMO was used to set gate boundaries
3.3 Isolation 1. Blood is collected into cell preparation tubes with sodium
of Microparticles heparin to prevent coagulation and platelet activation. Samples
for Flow Cytometry are processed as soon as possible.
2. Platelet-rich plasma is isolated by centrifuging whole blood at
150 g for 20 min with the centrifuge brake off (see Note 5).
3. The supernatant is platelet-rich plasma (PRP) and it is collected
from the tube and placed in a 15 mL tube. For each 1 mL of
PRP, 1 μL of prostaglandin is added to prevent platelet
activation.
4. Centrifuge PRP at 150 g for 10 min. The same brake
instructions from step 2 are followed in this step.
5. The supernatant is again collected and placed in a separate
15 mL tube, then centrifuged at 1500 g for 15 min with
the acceleration and brake of the centrifuge at max.
6. Collect the supernatant and centrifuge at 2500 g for 15 min
(see Note 6).
7. Collect the supernatant again and centrifuge a second time at
2500 g for 15 min.
8. The supernatant is collected then aliquoted in 250 μL portions
before being frozen at 80 C.
3.4 Staining 1. Platelet free plasma (PFP) is thawed at room temperature and
of Microparticles 18 μL of PFP per sample is transferred to an Eppendorf tube.
for Flow Cytometry 2. A reference sample containing 25 μL of PFP is also thawed and
24 μL of PFP is transferred to an Eppendorf tube.
3. AV binding buffer is prepared by adding r-hirudin to the
buffer at a 1 to 1000 dilution then filtering the buffer twice
through a 0.1 μm filter. R-hirudin will prevent aggregation of
microparticles.
4. 400 μL of AV binding buffer is added to all tubes.
5. The tubes are centrifuged at 15,000 g for 5 min (all
subsequent spins are at this speed and length of time) in
order to pellet microparticles (MPs).
208 Nicholas Wanner and Kewal Asosingh
4 Notes
Fig. 2 Gating of endothelial-derived microvesicles in plasma. Small Angle Light Scatter (SALS) and Large
Angle Light Scatter (LALS) were used to gate the microvesicle population based on size (a). Annexin-V+ events
were selected on a LALS/Green fluorescence channel plot (b). Annexin-V+ region was gated based on the
unstained sample (d). CD144 (VE-cadherin)+ events in the annexin-V+ gate were selcted on an LALS/orange
fluorescence channel plot (c) based on an Annexin-V only control (e) Samples were acquired at a flow rate of
6.01 μL/min for 2 min. At least 5000 annexin-V+ CD144-PE+ microparticles were acquired
References
1. Goon PK, Lip GY, Boos CJ, Stonelake PS, 5. Curtis AM, Edelberg J, Jonas R, Rogers WT,
Blann AD (2006) Circulating endothelial Moore JS, Syed W, Mohler ER 3rd. (2013)
cells, endothelial progenitor cells, and endo- Endothelial microparticles: sophisticated vesi-
thelial microparticles in cancer. Neoplasia cles modulating vascular function. Vasc Med
8:79–88 18:204–214
2. Del Papa N, Colombo G, Fracchiolla N, Mor- 6. Zimmerlin L, Donnenberg VS, Donnenberg
onetti LM, Ingegnoli F, Maglione W, Comina AD (2011) Rare event detection and analysis
DP, Vitali C, Fantini F, Cortelezzi A (2004) in flow cytometry: bone marrow mesenchymal
Circulating endothelial cells as a marker of stem cells, breast cancer stem/progenitor cells
ongoing vascular disease in systemic sclerosis. in malignant effusions, and pericytes in disag-
Arthritis Rheum 50:1296–1304 gregated adipose tissue. Methods Mol Biol
3. Bertolini F, Shaked Y, Mancuso P, Kerbel RS 699:251–273
(2006) The multifaceted circulating endothe- 7. Donnenberg AD, Donnenberg VS (2007)
lial cell in cancer: towards marker and target Rare-event analysis in flow cytometry. Clin
identification. Nat Rev Cancer 6:835–845 Lab Med 27:627–652. viii
4. Dignat-George F, Boulanger CM (2011) The 8. Roederer M (2008) How many events is
many faces of endothelial microparticles. Arter- enough? Are you positive? Cytometry A
ioscler Thromb Vasc Biol 31:27–33 73:384–385
Endothelial Flow Cytometry 211
9. Tibbe AG, Miller MC, Terstappen LW (2007) 16. Chow S, Hedley D, Grom P, Magari R, Jacob-
Statistical considerations for enumeration of berger JW, Shankey TV (2005) Whole blood
circulating tumor cells. Cytometry A fixation and permeabilization protocol with red
71:154–162 blood cell lysis for flow cytometry of intracellu-
10. Hedley BD, Keeney M (2013) Technical issues: lar phosphorylated epitopes in leukocyte sub-
flow cytometry and rare event analysis. Int J populations. Cytometry A 67:4–17
Lab Hematol 35:344–350 17. Chow S, Hedley D, Shankey TV (2008) Whole
11. Nolan JP (2015) Flow cytometry of extracellu- blood processing for measurement of signaling
lar vesicles: potential, pitfalls, and prospects. proteins by flow cytometry. Curr Protoc
Curr Protoc Cytom 73(13.14):11–16 Cytom. Chapter 9: Unit 9 27
12. Erdbrugger U, Lannigan J (2016) Analytical 18. de Rond L, van der Pol E, Hau CM, Varga Z,
challenges of extracellular vesicle detection: a Sturk A, van Leeuwen TG, Nieuwland R, Cou-
comparison of different techniques. Cytometry mans FAW (2018) Comparison of generic fluo-
A 89:123–134 rescent markers for detection of extracellular
13. Stoner SA, Duggan E, Condello D, vesicles by flow cytometry. Clin Chem
Guerrero A, Turk JR, Narayanan PK, Nolan 64:680–689
JP (2016) High sensitivity flow cytometry of 19. Rose JA, Wanner N, Cheong HI, Queisser K,
membrane vesicles. Cytometry A 89:196–206 Barrett P, Park M, Hite C, Naga Prasad SV,
14. Nolan JP, Moore J (2016) Extracellular vesi- Erzurum S, Asosingh K (2016) Flow cyto-
cles: great potential, many challenges. Cytome- metric quantification of peripheral blood cell
try B Clin Cytom 90:324–325 beta-adrenergic receptor density and urinary
endothelial cell-derived microparticles in pul-
15. Nolan JP, Duggan E (2018) Analysis of indi- monary arterial hypertension. PLoS One 11:
vidual extracellular vesicles by flow cytometry. e0156940
Methods Mol Biol 1678:79–92
Chapter 13
Abstract
Noninvasive isolation of circulating tumor cells (CTCs) from patient blood samples allows for interrogation
of valuable molecular and phenotypic information useful for disease diagnosis and monitoring response to
therapy. However, CTCs are extremely rare relative to red and white blood cells (R/WBC), thus making
CTC isolation from unmanipulated whole blood very time-consuming. Moreover, single CTC analysis
often requires hand-picking, a step that can result in more CTC loss and compromised cell integrity. Here
we describe an automated flow cytometry-based approach for isolation and analysis of single, viable CTCs
that combines gentle RBC lysis and magnetic, no-wash negative-depletion of WBCs, followed by a highly
adaptable sorting protocol for rare cells of interest. Multiparametric flow-cytometric panels allow probing
of numerous extracellular markers for immunophenotyping, while whole transcriptome analysis contributes
to molecular characterization of individual CTCs. Index sorting links single CTC proteogenomics
information.
Key words Circulating tumor cells, Acoustic cell enrichment, Magnetic depletion, Flow-cytometric
analysis, Molecular indexing, Phenotyping
1 Introduction
Circulating tumor cells (CTCs) are rare cells shed from the primary
tumor into blood. The enumeration and characterization of CTCs
isolated from the blood, also known as liquid biopsy, can serve as a
valuable alternative to repeated invasive tumor biopsies for diagnos-
ing and monitoring disease. However, isolation and characterization
of CTCs has been challenging due to the fact that CTCs are extremely
rare, present at 1–10 cells per ten billion blood cells, and also hetero-
geneous [1]. CellSearch®, the only US Food and Drug Administra-
tion (FDA) approved CTC characterization device has been used for
the enumeration of CTCs in patients with metastatic breast, colorec-
tal or prostate cancer. However, CellSearch-based phenotyping is
restricted to one open channel, and additional steps are necessary
for single cell isolation and molecular analysis [2–6].
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_13, © Springer Science+Business Media, LLC, part of Springer Nature 2019
213
214 Moen Sen et al.
analysis at the single cell level affords truly high dimensional char-
acterization of CTCs [16, 17]. This protocol increases sensitivity
and provides better rare cell characterization by combining enrich-
ment and index sorting of rare cells with downstream single cell
whole transcriptome analysis [18–20].
2 Materials
2.3 Single Cell 1. BD™ Precise WTA Single Cell Encoding Plate, 96 Well
Sorting (BD Biosciences).
2. Flow-cytometric cell sorter (also used in Subheading 2.2,
item 5).
216 Moen Sen et al.
3 Methods
3.1 Mouse Blood All steps involving preparation of blood sample should be per-
Sample Preparation formed at room temperature
1. Coat the syringe and 15 mL conical tube with an anticoagulant
like heparin.
2. Euthanize the mouse according to appropriate ethical
guidelines.
3. Spray 70% ethanol to disinfect the mouse.
4. Insert the 1 mL syringe right below the rib cage at a 45 angle
and slowly draw blood out by cardiac puncture (see Note 1).
Transfer the blood to a heparin coated 15 mL conical tube.
5. Set out lysis buffer (4 the volume of blood collected) at room
temperature for use in step 8.
CTC Phenotypic and Molecular Characterization 217
3.2 Set Up BD Focus The BD Focus should be set up while the blood sample is being
processed. In our lab, the BD Focus set up and mouse blood sample
preparation is conducted simultaneously by two people.
1. Prepare wash buffer for the BD Focus washing step by making
20% Ficoll® Paque in BD FACSFlow™ sheath solution.
2. Turn on the BD Focus.
3. Start the LabVIEW GUI (BD Biosciences, California) that
monitors flow rates read from flow meters (Sensirion, Switzer-
land) on the sorter.
4. Push the magnet tubing into the magnetic field to engage
magnetic separation.
5. Place a 50 mL Falcon conical tube containing 50 mL fresh wash
buffer in the “wash port,” and empty 50 mL Falcon conical
tube on the “waste” port.
6. Connect the sample line of BD Focus to the sample line of the
FACS system. Turn on to backflush BD Focus sample line with
sheath fluid. The fluidic lines are primed when the wash buffer
is pushed to the waste and sample tube.
7. Monitor air in the chip via MicroViewer, to make sure all air is
pushed from the chip by the flow of saline from the sheath tank.
8. Place a 50 mL Falcon conical tube containing 50 mL fresh wash
buffer in the “wash port,” an empty 50 mL Falcon conical tube
on the “waste port,” and a 5 mL polypropylene test tube with
FACS buffer on BD Focus (Fig. 1).
218 Moen Sen et al.
3.3 Run Sample on 1. Start pressure regulators and let fluidics get to equilibrium
BD Focus state, which typically takes a minute. Target rare cells are
enriched as the sample undergoes magnetic depletion of
unwanted cells and acoustic focusing that washes RBC lysate/
debris away.
2. Monitor event rate on flow cytometer and adjust pressure
regulators to control sample flow rate.
3.4 Flow-Cytometric 1. Draw dot plots in FACS software and employ appropriate gating
Analysis strategies to identify and characterize rare events. Gate out
debris based on size (FSC v SSC). This is followed by gating
on “live cells” that are DAPI negative. Following live/dead
selection, live YFP+ CD45 rare CTCs are gated on for sorting.
Gene expression profiling by whole transcriptome analysis is
routinely performed by us to verify cancer cell identity in sorted
CTCs [16]. As shown in the representative plots in Fig. 2, in one
experiment 16 (0.11% of all events, 0.16% of live cells) YFP+
CTCs were detected in the blood of a tumor-bearing KPCY
mouse (We have observed CTCs in the range of 1–254 cells/
mL in KPCY mice with high metastatic burden) [16]. To better
visualize the rare YFP+ events, the properties of the YFP gate
were edited on FACS analysis software to show up as larger dots
on the FACS plots (Fig. 2). Panels of multiple markers can be
optimized to minimize spectral overlap to phenotype CTCs.
3.5 Single Cell 1. Set up the flow sorter to sort per the manufacturer’s
Sorting instructions.
2. Chill the 96 well plate holder of the sorter at 4 C for 10 min.
3. Thaw the sealed 96 well Precise WTA plate at room tempera-
ture and spin at 377 g for 10 s to collect the reagents in each
CTC Phenotypic and Molecular Characterization 219
Fig. 2 Gating strategy for the detection of YFP+ CD45 circulating tumor cells
well. Place the plate on ice, slowly peel the seal from the plate
and place it in the chilled plate holder of the sorter.
4. Sort single cells into the 96 well encoding plate and leave 3–6
wells empty for nontemplate controls. A few pools of 10 or
more cells per well can also be included as sample input con-
trols (see Note 8).
5. Immediately place the plate on ice, seal the plate with a plate
cover, vortex and spin down at 4 C, 377 g for 10 s and
freeze at 80 C (see Note 9).
3.6 Whole Before conducting the procedure for WTA library prep, dedicate
Transcriptome two isolated workspaces in the laboratory as preamplification and
Analysis postamplification workplaces. In our laboratory, we used a PCR
workstation as the preamplification workspace, and a separate clean
3.6.1 Perform Reverse space on the bench as the postamplification workspace.
Transcription, Pool and In the preamplification workspace:
Purify the Product
1. Briefly spin the WTA plate for 10 s at RT on the PCR plate
spinner and incubate the plate at 65 C for 3 min (Program 1 in
the preamplification thermal cycler). Cool on ice.
2. Prepare the RT enzyme master mix sufficient for a 96-well
plate: 324 μL of Nuclease Free Water, 240 μL of Precise RT
Buffer, 12 μL of RNase Inhibitor, and 24 μL Reverse Tran-
scriptase, for a total volume of 600 μL. Use a multichannel
pipette to dispense 5 μL of the mix into each of the 96 wells.
3. Seal the plate followed by a brief vortex and 10 s spin at
377 g at room temperature (RT). Run Program 2 in the
preamplification thermal cycler: 42 C for 30 min ! 80 C for
5 min ! hold at 4 C.
4. Spin the plate for 10 s at 671 g at RT and pool all reactions into
one single 2 mL tube by using a multichannel pipette and then a
single channel pipette. The final volume is approximately 900 μL.
5. Purify the pooled samples using 900 μL of AMPure XP beads.
6. Vortex to mix well and incubate at room temperature for
5 min. Place tube on magnet and wait 5 min until the liquid
is clear and separating from the beads. Keep the tube on the
220 Moen Sen et al.
3.6.5 Perform Quality Store the purified WTA product at 20 C for 6 months or
Control on the WTA Product perform QC on the WTA product before proceeding to Subhead-
ing 3.6.6.
In the postamplification workspace:
1. Use 2 μL of the WTA product to measure the concentration
with the Qubit dsDNA HS Assay according to the
222 Moen Sen et al.
3.6.9 Sequence the Use an Illumina platform to sequence the libraries and generate
Amplified Libraries and 75 75 paired-end reads for a targeted range of 250,000–350,000
Analyze the Data paired reads per cell (see Note 10). Use the BD™ Precise Whole
Transcriptome Assay Analysis Pipeline on the Seven Bridges Geno-
mics platform to demultiplex and map reads from the sequencing
files, and calculate unique Molecular Indexes for each target with
built-in correction algorithms. Use the BD Genomics Data View
software (see Note 11) or open-source R statistical software for
secondary analysis such as dimensionality reduction, differentiated
gene expression, pathway analysis, and correlation of the Molecular
Indexes measured by the BD Precise™ assay with the fluorescence
intensity of the protein measured by flow cytometry at the single
cell level.
224 Moen Sen et al.
4 Notes
sealed WTA Single Cell Encoding Plate with sorted cells can be
stored at 80 C for 6 weeks.
10. The Precise WTA Library Index Primers 1 or 2 are used to
multiplex multiple libraries for sequencing. The plate indexes
for the Precise WTA assay are equivalent to Illumina indexed
adapters D701 (ATTACTCG) and D702 (TCCGGAGA). If
running multiple plates, normalize all libraries to an equivalent
concentration and pool them to prepare one sequencing run
per Illumina guidelines.
11. External release of the BD Genomics Data View software is
available at http://bitbucket.org/CRSwDev/dataview. Instal-
lation of the version 9.3 (R2017b) of the MATLAB Runtime is
required for running the Data View v1.2.2.
References
1. Ting DT, Wittner BS, Ligorio M et al (2014) 7. Rhim AD, Mirek ET, Aiello NM et al (2012)
Single-cell RNA sequencing identifies extracel- EMT and dissemination precede pancreatic
lular matrix gene expression by pancreatic cir- tumor formation. Cell 148(1-2):349–361.
culating tumor cells. Cell Rep 8 https://doi.org/10.1016/j.cell.2011.11.025
(6):1905–1918. https://doi.org/10.1016/j. 8. Gorges TM, Kuske A, Röck K et al (2016)
celrep.2014.08.029 Accession of tumor heterogeneity by multiplex
2. Yee SS, Carpenter EL (2017) Enumeration, transcriptome profiling of single circulating
dielectrophoretic capture, and molecular anal- tumor cells. Clin Chem 62(11):1504–1515.
ysis of circulating tumor cells BT - circulating https://doi.org/10.1373/clinchem.2016.
tumor cells: methods and protocols. In: 260299
Magbanua M, Jesus M, Park JW (eds) Circulat- 9. Carpenter EL, Rader J, Ruden J et al (2014)
ing tumor cells. Springer, New York, NY, pp Dielectrophoretic capture and genetic analysis
193–202 of single neuroblastoma tumor cells. Front
3. Cohen SJ, Punt CJA, Iannotti N et al (2008) Oncol 4:201. https://doi.org/10.3389/fonc.
Relationship of circulating tumor cells to 2014.00201
tumor response, progression-free survival, and 10. Peeters DJE, De Laere B, Van Den Eynden GG
overall survival in patients with metastatic colo- et al (2013) Semiautomated isolation and
rectal cancer. J Clin Oncol 26(19):3213–3221. molecular characterisation of single or highly
https://doi.org/10.1200/JCO.2007.15. purified tumour cells from CellSearch enriched
8923 blood samples using dielectrophoretic cell sort-
4. De Bono JS, Scher HI, Montgomery RB et al ing. Br J Cancer 108(6):1358–1367. https://
(2008) Circulating tumor cells predict survival doi.org/10.1038/bjc.2013.92
benefit from treatment in metastatic castration- 11. Karabacak NM, Spuhler PS, Fachin F et al
resistant prostate cancer. Clin Cancer Res 14 (2014) Microfluidic, marker-free isolation of
(19):6302–6309. https://doi.org/10.1158/ circulating tumor cells from blood samples.
1078-0432.CCR-08-0872 Nat Protoc 9:694–710. https://doi.org/10.
5. Cristofanilli M, Budd GT, Ellis MJ et al (2004) 1038/nprot.2014.044
Circulating tumor cells, disease progression, 12. Issadore D, Chung J, Shao H et al (2012)
and survival in metastatic breast cancer. N Ultrasensitive clinical enumeration of rare cells
Engl J Med 351:781–791. https://doi.org/ ex vivo using a micro-hall detector. Sci Transl
10.1056/NEJMoa040766 Med 4(141):141ra92. https://doi.org/10.
6. Allard WJ, Matera J, Miller MC et al (2004) 1126/scitranslmed.3003747
Tumor cells circulate in the peripheral blood of 13. Harb W, Fan A, Tran T et al (2013) Mutational
all major carcinomas but not in healthy subjects analysis of circulating tumor cells using a novel
or patients with nonmalignant diseases. Clin microfluidic collection device and qPCR assay.
Cancer Res 10(20):6897–6904. https://doi. Transl Oncol 6(5):528–538. https://doi.org/
org/10.1158/1078-0432.CCR-04-0378 10.1593/tlo.13367
226 Moen Sen et al.
14. Rhim AD, Thege FI, Santana SM et al (2014) precise and absolute gene expression measure-
Detection of circulating pancreas epithelial cells ment by single-molecule counting. Anal Chem
in patients with pancreatic cystic lesions. Gas- 86(6):2867–2870. https://doi.org/10.1021/
troenterology 146(3):647–651. https://doi. ac500459p
org/10.1053/j.gastro.2013.12.007 19. Fu GK, Xu W, Wilhelmy J et al (2014) Molec-
15. Stott SL, Hsu C-H, Tsukrov DI et al (2010) ular indexing enables quantitative targeted
Isolation of circulating tumor cells using a RNA sequencing and reveals poor efficiencies
microvortex-generating herringbone-chip. in standard library preparations. Proc Natl
Proc Natl Acad Sci 107(43):18392–18397. Acad Sci 111(5):1891–1896. https://doi.
https://doi.org/10.1073/pnas.1012539107 org/10.1073/pnas.1323732111
16. Bhagwat N, Dulmage K, Pletcher CH et al 20. Yu L, Sa S, Wang L, Dulmage K, Bhagwat N,
(2018) An integrated flow cytometry-based Yee SS, Sen M, Pletcher CH, Moore JS,
platform for isolation and molecular character- Saksena S, Dixon EP, Carpenter EL (2018)
ization of circulating tumor single cells and An integrated enrichment system to facilitate
clusters. Sci Rep 8. https://doi.org/10. isolation and molecular characterization of sin-
1038/s41598-018-23217-5 gle cancer cells from whole blood. Cytometry
17. Lang JE, Scott JH, Wolf DM et al (2015) A 93(12):1226–1233. https://doi.org/10.
Expression profiling of circulating tumor cells 1002/cyto.a.23599
in metastatic breast cancer. Breast Cancer Res 21. Mahnke YD, Roederer M (2007) Optimizing a
Treat 149:121–131. https://doi.org/10. multi-colour immunophenotyping assay. Clin
1007/s10549-014-3215-0 Lab Med 27:469–46v. https://doi.org/10.
18. Fu GK, Wilhelmy J, Stern D et al (2014) Digi- 1016/j.cll.2007.05.002
tal encoding of cellular mRNAs enabling
Chapter 14
Abstract
Applications of immunophenotyping using flow cytometry offer precise and accurate means for providing
information used to both diagnose and monitor disease; they serve as a standard platform for many research
endeavors that study discrete populations of biological entities. The proper use of this highly sophisticated
technology requires daily and ongoing monitoring of both the instrument and the methodology. Best
practices for this begin with quality control (QC) procedures designed to set up and monitor the
instrument performance, the reagents, and the results to ensure that they are working properly both on
the day of use and over time. If the results of those QC procedures are outside of acceptable then recording
the corrective action taken must also be included in the quality control records. Quality assurance (QA) is a
way to know that the three phases of testing, namely, preanalytic, analytic, and postanalytic procedures, are
being followed. This chapter describes the procedures used to assess quality control as it pertains to flow
cytometry and immunophenotyping in all three phases of testing.
Key words Quality control, Quality assurance, Flow cytometry, Levey–Jennings, Immunophenotyp-
ing, Monoclonal antibodies, Immunofluorescence, Color compensation, FMO controls, Proficiency
testing, Method validation, Accuracy, Precision
1 Introduction
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_14, © Springer Science+Business Media, LLC, part of Springer Nature 2019
227
228 Bruce Greig
2 Materials
3 Methods
3.1 Preanalytic Proper specimen collection and timely processing are critical pre-
Quality Control analytical quality control steps required for performing immuno-
Procedures phenotyping procedures. Specimens used in flow cytometry testing
can be obtained from several sources and in the clinical laboratory
3.1.1 Specimen these may include peripheral blood, bone marrow aspirates, solid
Requirements tissue biopsies, fine needle aspirates, and several types of body fluids
such as CSF, pleural fluid, synovial fluid, and vitreous fluid. In a
research setting this list may also include frozen tissue, animal
samples, plant samples, sea water, and a variety of other sources.
Each specimen has specific handling requirements that should be
fully described in the procedures of the laboratory.
The following list includes the most widely used specimen types
and their specific collection requirements for immunophenotyping.
3.1.2 Peripheral Blood All blood or bone marrow samples must be in an anticoagulant if
and Bone Marrow Aspirates the assay is for testing either white blood cells (WBCs) and/or red
blood cells (RBCs) [25]. The most commonly used anticoagulants
for immunophenotyping include EDTA, (sodium) heparin, and
ACD (acid citrate dextrose). All are designed to prevent clotting
while preserving the WBCs however, there are advantages and
disadvantages to each and is discussed below. Another important
consideration is the hold time between specimen collection and
testing. This can be critical depending on the anticoagulant used.
For the times listed in this section the laboratory should do their
own validation of each anticoagulant to determine what is
acceptable.
1. EDTA (ethylenediaminetetraacetic acid) preserves lymphocyte
viability up to 72 h if refrigerated, 48 h at room temperature.
This is the most common anticoagulant used in the clinical
laboratory for testing lymphocyte subsets since it can also be
used for performing white blood counts (WBCs) and differen-
tials necessary for calculating the absolute lymphocyte counts.
Granulocytes, however, may begin to deteriorate in less than
8 h due to the chelation of the calcium in the plasma by the
EDTA which makes this anticoagulant less desirable for flow
assays that focus on myeloid lineage populations. These tubes
usually have a purple stopper [2].
2. Sodium heparin prevents the formation of fibrin from fibrino-
gen and thrombin activation thus preventing clotting of the
sample. It may cause some RBCs to clump and, in some cases,
impart a blue background on Wright-Giemsa stains so it is not
suitable for hematology assays [3]; however, it works quite well
on bone marrow aspirates and is the anticoagulant of choice for
these samples. Samples in heparin are generally good for up to
Quality Control of Immunophenotyping 231
3.1.3 Solid Tissue Tissue samples submitted for flow cytometry testing may be from
Biopsies and Fine Needle many different sources. In a Hematopathology setting this would
Aspirates likely include a lymph node biopsy, soft tissue from an organ such as
the gut, and even skin punches submitted by a dermatologist. If the
sample is submitted from the same place as the lab doing the testing
in most cases it will be reasonably fresh and intact. Care should be
taken to insure good viability by using a transport media with the
tissue such as RPMI with or without albumin that has nutrients and
buffers that greatly improve the chances of maintaining viability.
Using other media like plain PBS is discouraged because the sam-
ples will deteriorate much more quickly compared to a transport
media like RPMI. The use of any fixative in the media is not
acceptable since this will be toxic to the tissue and prohibit any
possibility of doing a flow cytometry assay which requires viable
cells. Some laboratories use frozen tissue banks that allow the lab to
save rare or study samples in a way that they are not damaged by the
subfreezing temperature. Previously frozen samples will need to be
thawed and washed before processed to a single cell suspension.
Follow published procedures that describe the steps necessary to
gently bring the sample back to room temperature prior to making
single cell suspensions. Making a single cell suspension of the solid
tissue can be achieved several different ways including:
1. Grinding the sample with a mortar and pestle and rinsing
with RPMI.
2. Using a tuberculin-sized needle that is filled with RPMI and
repeatedly injecting it into the tissue in a petri dish to extract
the cells of interest and leave behind the sample connective
tissue. The cell suspension can then be washed with RPMI and
concentrated prior to staining.
232 Bruce Greig
3.1.4 Fine Needle FNAs are frequently submitted for flow cytometry testing in the
Aspirates (FNAs) context of hematopathology procedures. Tissue samples collected
with this technique are a much less invasive procedure compared to
obtaining a tissue biopsy. The samples are collected by using a very
thin needle (22 gauge or smaller) that is inserted into an area of
questionable tissue like a lymph node or other soft tissue. The cells
are then aspirated into the syringe and added to a tube containing
transport media such as RPMI. Since the cells are already in sus-
pension very little manipulation of the sample is necessary prior to
using it in an immunophenotyping procedure. It can then be
washed, concentrated, and resuspended in RPMI prior to staining.
These samples may have some peripheral blood contamination that
happens during collection and thus will need to include a RBC
lysing step as part of the staining procedure.
3.1.5 Body Fluids Body fluids submitted for flow cytometry immunophenotyping can
and Cerebrospinal Fluid be collected from several different sources. These may include
(CSF) peritoneal, bronchial, pleural, synovial, pericardial, vitreous, and
cerebrospinal fluids. In most situations the question for testing is
to find and identify an underlying malignancy such as lymphoma or
leukemia. Usually the body fluid itself if collected the same day will
maintain the cell’s viability if transported quickly and refrigerated.
CSF however, is collected using a laparoscopic procedure and even
though these specimens are usually sent to the lab with no addi-
tional media in the tube it is highly recommended that the sample
be resuspended in fresh RPMI as soon as possible to prevent the
rapid deterioration of any cells present. Studies have shown that
CSF with RPMI added may have a much higher rate of recovery
compared to samples that do not include a transport media [10].
Quality Control of Immunophenotyping 233
3.2.2 Antibody Quality Primary reagents like monoclonal antibodies require a more thor-
Control ough control process than other test reagents [9]. Quality control
of antibodies should include the following procedures:
1. Antibody specificity: does the antibody react with the intended
antigen [12]? This aspect of antibody QC requires specific
specimens to prove efficacy of the reagent. For example, if
CD33 is being tested, the control material should include
myeloid lineage cells such as granulocytes and monocytes,
since that antigen is fully expressed on those cells. Lympho-
cytes present in those samples can serve as a negative control.
Other antibody control materials include the following:
(a) Commercial quality controls: These are samples that
include a list of expected results for several antibodies in
the package insert. These controls are generally used for
daily testing of an antibody cocktail panel (a cocktail is two
or more antibodies in the same tube), which is required
for clinical applications like peripheral blood lymphocyte
subsets or stem cell quantitation assays. These commercial
controls will generally have known qualitative and quanti-
tative values for most of the antibodies used for immuno-
phenotyping in a clinical laboratory. See Fig. 2:
Commercial control package insert taken from a commer-
cial whole blood control listing.
(b) In-house controls: Other control samples that can be used
to check antibody specificity may be sourced from previ-
ously tested material that has been confirmed to be posi-
tive or negative for the antibody depending on the
population of interest. If alternate testing is performed
in the laboratory, such as immunohistochemistry (IHC),
the flow cytometry antibody results can be compared to
the results of the alternate assay such as a tissue section
stained for CD20. However, this choice may only offer
234 Bruce Greig
Phenotype3,a WBC Type4 %Positive Cell1,b Expected Ranges6 Absolute Number7,c Expected Range4
+
CD2 Lymphocytes 85.0 73.0 - 97.0 – –
CD3+a Lymphocytes 78.4 68.4 - 88.4 1625 1275 - 1975
CD3+/CD4+a Lymphocytes 51.1 45.1 - 57.1 1059 879 1239
CD5+ Lymphocytes 74.6 64.6 - 84.6 - -
CD3+/CD8+a Lymphocytes 25.3 19.3 - 31.3 524 354 - 694
CD3-/CD16+ Lymphocytes 9.2 4.2 - 14.2 – –
CD3-/CD16+.CD56+ Lymphocytes 10.5 5.5 - 15.5 218 73 - 363
CD19+ Lymphocytes 11.2 6.2 - 16.2 232 82 - 382
CD20+ Lymphocytes 11.1 6.1 - 16.1 – –
CD3-/CD56+ Lymphocytes 8.6 3.6 - 13.6 – –
HLA . DR+ Lymphocytes 15.8 9.8 - 21.8 – –
CD14+ Monocytes 85.0 70.0 - 100.0 – –
CD45+ Leukocyte Gate ................. CD45 is to be used for gating purposes only.8 ............
Fig. 2 Example of a package insert from a whole blood commercial control listing reactivity and expected
quantitative values for antibodies used in Immunophenotyping [28]
3.2.3 Antibody Most antibody manufacturers provide directions that specify the
Concentration amount of antibody to use for samples typically submitted for
and the Stain Index Immunophenotyping. Knowing the concentration of the sample
is also necessary to insure saturation staining [11, 25]. For example,
if a sample has a high concentration of cells, then the recommended
amount of antibody per test may not be enough to insure adequate
staining. In other cases, the amount of antibody listed may be
Quality Control of Immunophenotyping 235
Fig. 3 The stain index (SI) calculation [22, 33]. The SI is the ratio of the difference
between the positive mean intensity (MFI) and negative mean intensity (MFI)
populations divided by two times the standard deviation (SD) of the negative
population. The SI is used to compare fluorescence intensity of one antibody
versus another or when comparing the concentration of one antibody versus a
different dilution of the same antibody
several times more than is necessary and the excess antibody may
negatively impact the staining by increasing the background signal.
The best antibody concentration to use can be determined by
calculating the stain index (SI). The SI is the ratio of the difference
between the positive mean channel result and the negative mean
channel result divided by two times the standard deviation (SD) of
the negative population. See Fig. 3: Stain index calculation.
The stain index can be used to compare different antibodies
and it can also be used to compare different concentrations of the
same antibody. In some cases, the SI will improve using a lower
concentration than what is recommended since the background
signal width may be smaller than it is using a higher concentration.
The higher the SI, the better the resolution between the positive
and negative populations. Different SIs can often be seen with
different concentrations. The difference can be due to one of two
reasons: a lower intensity of the positive peak or a wider SD of the
negative signal due to excess unbound antibody. Titrating the
antibody is advantageous for determining the optimal
236 Bruce Greig
Fig. 4 Antibody concentration versus CD19 PEcy7 expression. Over the course of
different titrations or dilutions the stain index (SI) improves using lower
concentrations of the antibody. There is no noticeable loss in CD19
intensity (Y-axis); however, the background signal is much greater at higher
concentrations of the antibody and thus has a lower stain index. Ideally, the
standard deviation of the negative signal should be somewhere near that of
unstained autofluorescence
3.2.4 Antibody Titration 1. Make serial dilutions of the antibody beginning with the
Procedure recommended volume followed by dilutions of 1:2, 1:4, 1:8,
etc. The diluent used can be simple PBS buffer with or without
calf serum which is reliable for antibodies that have a high
expression such as CD45 and CD8. In some instances, espe-
cially very weak expressing antibodies should be diluted with
unlabeled or unconjugated antibody to insure adequate anti-
body saturation staining.
Quality Control of Immunophenotyping 237
3.2.5 Multiantibody An antibody cocktail is whenever two or more antibodies are com-
Cocktails bined to make a multiuse reagent. As part of panel design the
antibody cocktail is constructed using combinations that are used
for immunophenotyping applications. There are several advantages
to using cocktails. They can save time when setting up multiple
experiments and are likely to lower or prevent pipetting errors that
could happen if the single cocktail was made up each time the
experiment was performed. Another advantage to using cocktails
is that you can standardize delivery volumes. This makes test setups
238 Bruce Greig
3.2.6 Antibody Cocktail The full cocktail procedure can be found in Notes section. The
Procedure (Example) example shown in the recipe is for a 10 test, 4-color cocktail for
T-lymphocyte subset testing using titrated antibodies and buffer
that is aliquoted using 100 μl cocktail/test. Use a previously deter-
mined titer amount for each antibody in the cocktail and add
enough buffer to create a standardized aliquot for testing such as
100 μl/test. The additional volume of the buffer will also allow
better mixing.
3.3.2 Instrument Quality 1. Fluidics system startup: The flow cytometer fluidics system is
Control: Flow Cytometer truly the lifeblood of the instrument. Proper care and cleaning
Startup and Maintenance of the fluidics system is synonymous with good quality control
and helps ensure high quality results. Prior to turning on the
instrument check the levels of the fluidics containers. They are
usually found either on a fluidics cart or other storage system
near the instrument. This includes the sheath fluid, cleaning
solutions, or DI water, etc. that may be a part of the whole
fluidics system. If the instrument has a waste container dispose
of anything from the last time it was used. Be sure to add bleach
to the container to minimize the danger of any hazardous waste
that may result from using biological samples. Most flow cyt-
ometers have built-in programs that provide a means for the
sheath fluid and other solutions to be primed and distributed
throughout the system including the flow cell and ultimately
into the waste container. As part of the instrument startup a
series of priming and flushing cycles takes place to be sure that
everything is working and that all the fluidics lines are free of air
bubbles. Sometimes when the instrument is shut down for
240 Bruce Greig
several hours bubbles can come out of the sheath fluid solution
still in the instrument much like what is observed on the side of
a glass of water left standing overnight. If those bubbles are not
adequately removed as part of the fluidics startup they may
cling to the inside of the flow cell and create focusing issues
much like any other obstruction in the fluidics system.
2. Fluidics clean cycle: Samples routinely run on the instrument
such as tissue cell suspensions and bone marrow specimens may
have high amounts of debris or fibrin or other nonintact cell
residue that can build up on the inside of the sample insertion
rod, fluidics tubing, or flow cell. After starting up the instru-
ment the fluidics system should be put through a short cleaning
cycle that includes running dilute bleach (10%) for 5 min fol-
lowed by a DI water sample for 5 min. Running a series of
cleaning samples helps to remove the buildup and prevent clogs
or other obstructions that might be present in the insertion rod
or fluidics tubing. Some labs prefer to do this cleaning
sequence one or more times a day. A good rule of thumb is
that for every 1 min you run dilute bleach it should be followed
for the same amount of time by running a tube of DI water.
This is to adequately remove any residual bleach from the
fluidics system. A full system clean of 60 min (30/30) or
more should be performed at least once a month to more
thoroughly clean the fluidics lines and flow cell chamber.
3. Fluidics function checks as part of bead QC.
As mentioned before, issues with fluidics are probably the
most common cause of instrument-related problems. Most
every instrument has a quality control program that uses latex
beads to measure and standardize each parameter of the instru-
ment operation. Usually performance issues are first indicated
by out of range bead QC results including diminished resolu-
tion of either or both light scatter parameters, forward scatter
(FSC) and side-scatter (SSC). A well-maintained instrument
that has been set up and optimized should demonstrate that
the FSC/SSC dot plot of a lysed blood sample has at least three
distinct WBC populations corresponding to the lymphocytes,
monocytes, and granulocytes. On the other hand, if the instru-
ment has population resolution issues seen by light scatter due
to fluidics problems the populations tend to drift downward
toward the threshold on the FSC/SSC dot plot (see Fig. 6:
Good light scatter resolution versus poor light scatter
resolution).
If the flow cytometer has debris buildup over time in the
fluidics lines or the sample insertion rod or if the stream is
obstructed in some way the Forward Scatter Channel (FSC)
signal is usually the first to show this problem. There will either
be a gradual loss of the signal resulting in a lower location on
forward scatter of a population or an increase in the detector
Quality Control of Immunophenotyping 241
800 800
SS INT
SS INT
600 600
400 400
200 200
0 0
0 200 400 600 800 1000 0 500 1000
FS INT FS INT
Fig. 6 Good light scatter resolution versus poor light scatter resolution: Left histogram: RBC-lysed peripheral
blood sample with optimized and finely tuned FSC/SSC signals shows granulocyte (blue) populations. Right
Histogram: FSC/SSC dot plot with poor optical alignment, possibly due to fluidics issues. Notice the loss of FSC
output versus SSC resolution
gain if you are using settings that are autoadjusted by the bead
QC software to meet a target range for each peak channel.
Eventually these changes to the detector settings will exceed
the defined limits of the bead QC program and the results will
have one or more parameter failures.
4. Laser performance: The solid-state lasers found on most flow
cytometers are considerably more dependable and less prone to
failure than the high-power water-cooled lasers that were used
on many of the research-type instruments years ago. However,
they still need time to warm up and equilibrate. Usually the
time required to run the fluidics start-up cycle and cleaning
steps is typically long enough for the lasers to warm up. In most
cases the instrument will have an indication of when it is ready
to be used following the startup time for the laser.
5. Flow cell, optics, and electronics QC using commercial beads.
(a) Introduction to the Bead QC procedure: Following
instrument and fluidics startup, initial cleaning, and laser
warm-up the flow cytometer must then be checked for
proper operation. This is typically accomplished using
latex beads or other materials, usually sourced from an
instrument vendor, to ascertain the overall function of
the fluidics, electronics, and optical alignment. These
quality controls checks are where the “quality” of Quality
Assurance begins in flow cytometry. The CAP checklist
requires that performance of function checks on all instru-
ments follow a defined schedule [1]. For results from a
flow cytometer to be reliable and accurate, the setup and
monitoring of the flow cytometer performance should
always be checked when the instrument is started up
each day. Some labs may require this as often as once
each shift if the instrument is used continuously.
242 Bruce Greig
Fig. 7 QC bead signal. QC Beads run each day are placed in a specific channel of
each detector by the software automatically adjusting the detector gain or
voltage to match the baseline target value
Fig. 8 Optical alignment and fine focusing. Good optical alignment equates to a
fine focus which can provide a sharper, clearer signal that gives better resolution
and higher sensitivity (right image) compared to an instrument with poor optical
alignment (left image) that is blurred or out of focus
150
Count
100
50
Fig. 10 Levey–Jennings graph of daily PMT QC settings over time. The voltage of the FL8 detector required to
meet the baseline specifications is gradually rising over time which may indicate an impending failure of that
PMT
Quality Control of Immunophenotyping 247
Fig. 11 Optimizing PMT settings. In Histogram a, WBCs gated on lymphocytes and monocytes stained with
CD4, the PMT setting is too low and as a result the intermediate population (monocytes) is difficult to resolve.
In Histogram b, the default PMT setting is too high so CD3+ T cells gated on lymphocytes appears to be
off-scale when in fact are still on scale; however, the brightest events are piled up against the right axis
and may be visually underestimated. In Fig. c, a lysed whole blood sample, the PMT setting is optimal. CD33
expression shows clear resolution between the bright (monocytes), intermediate (granulocytes), and negative
(lymphocytes) WBC populations
800
600
SS INT
400
200
0
0 200 400 600 800 1000
FS INT
Fig. 12 FSC versus SSC with good separation and resolution. Forward scatter
versus side scatter of a lysed whole blood sample demonstrating good
separation between all 3 WBC populations with the noise threshold set just
below the lymphocytes to allow visualization of intact cells versus debris
using the default settings from the bead QC. The first differ-
ence you will notice is that the light scatter settings and possi-
bly the threshold will need to be adjusted since beads are often
much smaller than cells. Adjust the FSC/SSC light scatter
settings until you can see good separation between the lym-
phocyte, monocyte, and granulocyte populations. Be sure to
set the threshold to just below the lymphocyte population to
minimize the noise and debris events but still show the
entire population of lymphocytes present. Keep the granulo-
cyte population on scale however, take advantage of the entire
FSC/SSC area to see each population clearly like that seen in
Fig. 12: FSC versus SSC with good separation and resolution.
PMT Optimization: Optimizing the fluorescence PMTs
can be performed several different ways and each has advan-
tages and disadvantages, but they should all allow the user to
see an improvement in signal separation between the bright
positive, intermediate, and negative populations present. Two
of the most popular methods used to optimize the PMT set-
tings are the Unstained Sample Method and the Single-stained
Sample method. Note: optimization should be performed with
the compensation settings turned off. That step follows
optimizing the instrument settings.
(a) Unstained sample method: This method has been used
for several years and is relatively easy to perform and not
very expensive since no antibodies are required. Set up the
instrument starting with the default PMT settings from
250 Bruce Greig
Fig. 13 Unstained sample method for optimizing PMT settings. Unstained sample optimization method gated
on lymphocytes from a lysed whole blood specimen. The PMT settings are adjusted so that the leading edge of
the negative signal is just beyond the background signal in each detector
Fig. 14 The Stained cells method for optimizing PMTs. In Histogram a, WBCs gated on lymphocytes and
monocytes stained with CD4, three separate populations can be clearly visualized: T-Helper cells stain bright,
monocytes have an intermediate intensity, and the T-cytotoxic suppressor lymphocytes are negative. In
Histogram b, CD33 is used to optimize the PMT. Ungated WBCs stained with CD33 show monocytes are bright,
granulocytes intermediate, and lymphocytes are negative. In Histogram c, WBCs gated on lymphocytes
express bright CD8, intermediate positive for NK cells, and are negative for T-helper lymphocytes
252 Bruce Greig
5 P1
10
8C COMP QC 10-15-2018-Lymphoma
4
10
150
CD4 PE PE-A
Count
3
100
10
2
50
10
0
0
-76
2 2 3 4 5 2 3 4 5
-10 0 10 10 10 10 0 10 10 10 10
-327 -185
CD2 PC7 PE-Cy7-A CD4 PE PE-A
Fig. 15 PE versus PC7 undercompensated and showing a weak PE positive population. PE and PC7-stained
cells appear to express both PC7 and two populations of PE + PC7 dual-positive populations incorrectly due to
undercompensation of PE-PC7 in PE
104
104
A B
103
103
HLA-DR APC-H7
HLA-DR APC-H7
102
102
101
101
100
100
100 101 102 103 104 100 101 102 103 104
CD34 APC CD34 APC
Fig. 16 Different compensation settings of APC-H7 versus APC. Histogram a: Overcompensation between
APC-H7 and APC appears to show two separate populations and Histogram b: APC-H7 and APC, correctly
compensated, shows a single population that is positive for both CD34 and HLA-DR
Signal into FITC into PE into PerCP-cy5.5 into PE-cy7 into APC into APC-H7 into BV421 into BV 510
Fig. 17 Compensation matrix for an eight-color experiment. Each fluorochrome in the experiment is shown
with its respective compensation setting versus the other fluorochromes being used. The diagonal values are
all 100% because a color cannot be compensated against itself. The higher the value the more overlap each
signal has with the opposing signal. When designing an experiment, the use of color-conjugates with very high
spectral overlap should be avoided for those markers that are used to find very discrete populations since they
might be lost due to the high compensation correction value
Fig. 18 Compensation setup using capture/ comp beads which are beads coated
with anti-mouse IgG can be used to set up a compensation panel. The beads
react with any antibody as long as it is IgG-class. The advantages are that they
will work almost universally, be prepared in advance, and can be used with rare
antibodies. However, the bead staining may show a different intensity compared
to stained cells, and autofluorescence may be an issue with certain lasers.
Beads cannot be used for nonantibody stains like 7-AAD or Syto-6
the brightest color signals that can spill over into the
adjacent channels. One of the best sample types and anti-
bodies to use in the compensation panel is CD8-stained
lymphocytes. CD8 is a high-density antigen on lympho-
cytes and thus whatever conjugate is used CD8 will be as
bright or even more bright than other common lympho-
cyte antigens. It is also negative on CD4+ T-lymphocytes
so now there is both a positive and negative population
present for calculating the compensation. If CD8 is not
available in every conjugate of your panel, choose a differ-
ent one that will have a positive signal intensity of at least
the third decade on a log scale. Other antibodies to con-
sider using in a stained cell compensation panel would be
either/or both CD33 and CD14 gated on monocytes,
and CD16 gated on granulocytes. CD33 gated on all
WBCs in LWB should demonstrate a bright positive
monocyte population, intermediate positive granulocytes,
and be negative on lymphocytes. CD14 will be very bright
on monocytes and negative on lymphocytes. CD16 will be
very bright on neutrophils, moderately positive on NK
lymphocytes, and negative on monocytes. Avoid using
cell cultures in the comp panel unless your experiment
uses these cell sources, due to the increased background
or noise or autofluorescent signal they often have com-
pared to normal lymphocytes.
258 Bruce Greig
104
104
103
103
CD4 PE
CD4 PE
102
102
1
10
1
10
0
0
10
0
10 101 102 103 104 0 101 102 103 104
CD3 APC CD3 APC
Fig. 19 Log versus offset/biexponential dot plots. The dot plot on the left shows the origin of each axis at the
bottom. The dot plot on the right is the same dot plot with the axis offset/biexponential setting turned on
allowing you to see events both above and below the origin (blue arrow) for demonstrating compensation
Fig. 20 Compensation examples using FMO controls. The positive CD4 result is shown versus an unlabeled
detector as a way of demonstrating compensation between the two PMTs. The ideal compensation setting (dot
plot a) shows the mean fluorescence intensity (MFI) of the positive peak to be very close to the MFI of the
negative peak. Undercompensation in the middle figure (b) shows the MFI of the positive peak to be
significantly greater than the negative peak MFI. The MFI of the right dot plot (c) shows the positive MFI to
be significantly less than the negative peak MFI and thus is overcompensated
Fig. 21 FMX CD8 FITC control versus Each PMT. CD8 FITC versus all other colors. In this display, CD8 FITC
versus each other color shows good compensation settings except for the CD8 FITC versus BV421 which is
slightly overcompensated and the same versus BV510 which requires additional compensation
Fig. 22 Example of eight-color compensation. Multiple dot plots of every combination in an eight-color
experiment using a cocktail that includes CD8/4/5/2/3/7/56 or 16/19 + 8. CD19 and CD8 are paired with the
same conjugate to observe both coexpression and single staining. This example only shows two of the
possible combinations, FITC versus X and PE versus X. Instrument compensation settings can now be
monitored daily using a normal whole blood control with the multicolor cocktail
3.4.1 Analytic Quality Daily monitoring of the flow cytometer performance including
Control Conclusion alignment checks, background and sensitivity limits, fluidics, PMT
performance, and laser output, is a standard of best practice in flow
cytometry laboratories and may also be required for regulatory
compliance. It helps insure consistency and reliability on a day-to-
day basis. Tracking the results of QC over time may reveal trends
that could predict future instrument problems. Understanding
how each component of the instrument is performing: fluidics,
optics, alignment, and electronics helps insure that results are reli-
able, accurate, and precise. Besides the flow cytometer, keeping
maintenance and operation records of other miscellaneous equip-
ment in the laboratory such as refrigerators, freezers, and centri-
fuges is also important for meeting quality assurance best practice
standards [30].
3.5 Postanalytic Once the instrument has successfully passed the analytic quality
Quality Control control procedures the final step of verification is to run sample
Procedures controls. This includes both quantitative and qualitative controls
[18]. For example, clinical laboratories that report quantitative
3.5.1 Quantitative Assay results such as lymphocyte subset percent and absolute counts, or
Quality Controls stem cell percent and absolute counts, are required to run at least
two levels of commercial or verified controls each day the test is
performed. The controls should include both a normal control as
well as an abnormal control so that the full range of reportable
results is represented. Quantitative commercial controls come with
package insert values that must also be verified by the laboratory
264 Bruce Greig
prior to being put into use. Oftentimes these control values have a
wide range of acceptable limits and therefore the laboratory should
not only verify those limits but calculate ranges of their own based
on multiple runs. Using the mean 2 standard deviations after
10 times will usually be enough to generate statistical ranges that
satisfy the requirement. The laboratory calculated control ranges
should now reflect what the reports on both normal and abnormal
results will be and should not be below the lower limit of detection
(LLOD).
For those assays in which a commercial control is unavailable
the use of a known positive sample verified by other means or by
another laboratory that performs similar testing can be an accept-
able substitute. Records of the controls and how the results were
calculated are required for verification and acceptability for use.
3.5.2 Qualitative Assay Qualitative controls, also known as method controls, test not only
Quality Controls the instrument but the staining and processing procedures from
setup to acquisition to analysis. Every antibody in an experiment
should be tested for accuracy prior to being put into use whether as
part of a cocktail or as a standalone reagent. This quality testing was
mentioned previously in the preanalytic quality control section.
Some commercial controls used for quantitative testing also include
a list of additional antibodies that will react with the control for
qualitative purposes. Method controls on the other hand are those
controls used in an experiment that prove that the setup and stain-
ing procedures are working as indicated. As was also mentioned in
the preanalytic QC section, material tested using alternate methods
such as histology and hematology can be used as method controls.
If proficiency testing material is available from a recent survey, it too
can be used as a method control.
3.5.3 Isotype Controls Isotype controls are basically antibodies of the same immunoglob-
ulin class as the primary antibody including both the heavy and
light chain molecules but lacking the specific component of that
antibody to react with the target antigen or are directed against an
irrelevant target. Isotype controls can serve as experiment controls
and many labs use these to demonstrate the background staining or
nonspecific staining present when using the test antibody. Setting a
statistical gate on the positive population was based on where the
negative signal ended for the isotype control. But there are some
inherent limitations to using isotype controls for this purpose
[19]. In some cases, the isotype control may not exactly match
the target antibody and may react with an unexpected antigen,
what is known as nonspecific binding. This may introduce uncer-
tainty about what is positive versus what could be nonspecific
staining. Another difference in using an isotype control is that it
may have a different fluorescence to protein (F/P) ratio versus the
target antibody. If the F/P ratios are different then the background
Quality Control of Immunophenotyping 265
3.5.4 Postanalytic Quality Internal/intraassay Controls are method controls that occur natu-
Controls Internal/Intraassay rally in a sample and demonstrate what positive versus negative
Controls looks like for each same cell types being studied. For instance,
lysed whole blood WBC’s include lymphocyte, monocyte, and
granulocyte populations. An antibody like CD33 reacts with
monocytes and granulocytes but is negative on lymphocytes.
CD19 is positive on B cells but is negative on T and NK cells as
well as other myeloid populations present. In each of these exam-
ples you have one or more positive populations and one or more
negative populations. In most bone marrow specimens other exam-
ples can be used within the tube to confirm different antibody
reactivities, both positive and negative, using the same approach.
See Fig. 23: Using intraassay controls.
An advantage of using internal/intraassay controls is that now
the test antibody’s reactivity can be described because the condi-
tions, F/P ratio, and isotype are all in one place compared to a using
a separate isotype control tube.
Each lab should establish their own criteria for describing the
use of internal controls as a method and cocktail quality control
check. Abnormal populations typically found in malignancy may
aberrantly express antigens or be missing antigens that are expected
266 Bruce Greig
3.5.5 Postanalytic Quality Flow cytometer list mode data is stored in a specific format that
Control Methods Using includes not only a record of every signal or event but also the time
Time as a Parameter point at which that event occurred. This additional measurement is
known as the Time parameter. The Time parameter can be a useful
quality control indicator to alert you to problems with the sample
acquisition, especially the fluidics [20]. For example, when an
acquisition is either interrupted by something briefly blocking the
fluidics or if the sample runs dry, these incidents can result in a loss
of signal or the inclusion of bubble events in the list mode
file results. Bubbles will look like collected events especially on
light scatter. Looking at a histogram of time versus another param-
eter should normally show a steady sequence of events, but if there
is an interruption during the acquisition or bubbles present, the
histogram will demonstrate a noticeable change in the pattern or
shift of a parameter. An interruption in the acquisition can be
identified using the Time dot plot. Results before the interruption
should look continuous; however, the results after an interruption
may produce a false signal that could change the appearance of your
data. The good news is that you can still salvage an otherwise faulty
Quality Control of Immunophenotyping 267
Fig. 24 Using Time as a gating parameter. The FSC/SSC histogram on the left shows what happens when a
sample is run until empty. This introduces air bubbles, which appear as the multiple black dots in the upper
area of the histogram. The middle histogram, Time versus CD45 shows a clear break where the bubbles
started being acquired. A gate is then drawn around the “clean” events to omit the bubble events. The bubble-
free-gated FSC/SSC histogram on the right no longer shows any bubble events
3.5.6 Postanalytic Quality Another issue that can impact the quality of your results is when the
Control Methods Using sample contains doublets or cellular aggregates. Doublets are when
Singlet Gates two or more cells for whatever reason are bound together as they
pass through the flow cell chamber and are counted as one event.
Using a singlet gate is an important consideration especially in
experiments that measure DNA ploidy. If two cells with a 2N
DNA content (resting, G1/G0 phase) are measured as one cell
they could be mistaken as one cell having twice as much (prolifer-
ating, G2 + M) content. Samples with numerous doublets would
then falsely skew the number of proliferating cells versus resting
cells. Doublets can also be seen when specimens undergo several
centrifugation steps such as preparing bulk lysed samples. Bulk
lysing is often used in experiments that look for rare events, espe-
cially minimal residual disease (MRD) leukemia cases. To capture
these rare events more specimen is required than that used under
routine circumstances. In a bulk lysing procedure, using 1 ml of
sample instead of 100 μl increases the number of cells tenfold. Now
you have significantly more WBCs than before, and here is where
the problem of doublet aggregate combinations is likely to occur
268 Bruce Greig
(x 1,000)
200 250
5
10
CD19 PerCP-Cy5-5-A
Singlets
4
150
10
FSC-H
100
3
10
50
2
10
2 3 4 5
50 100 150 200 250
10 10 10 10 FSC-A (x 1,000)
CD7 FITC-A
Doublets
Fig. 25 Using a singlet gate to omit doublet events. In this example, the blue population before singlet gating
appear to be positive for both CD19, a pan-B cell marker and CD7, a T cell marker. Normal B cells should be
negative for CD7 and normal T cells should be negative for CD19. Using a Singlet gate that excludes doublets
shows the red populations to be as expected for both CD7 and CD19. Singlet gating is a good practice
regardless of the sample type since this phenomenon may happen quite frequently
Fig. 26 Using 7-AAD to identify and gate out nonviable cells. Events in dot plot A have a high number of
nonviable cells which take up the 7-AAD dye, demonstrated by the very bright expression of the 7-AAD on the
Y-axis. The dead cells are seen in dot plot B, FSC/SSC as the secondary population on the SSC axis above the
lymphocytes. Dot plot C, SSC/CD19, includes the dead cells which dilute the actual percentage of B cells
present in the sample. Gating on just the cells that are 7-AAD negative in dot plot D cleans up the analysis by
now omitting the dead cells. The SSC/CD19 dot plot F () shows only the viable cells with a more accurate CD19
result
3.6 External Quality Another regulatory requirement for clinical labs performing immu-
Assessment (EQA) nophenotyping is to participate in an external quality assessment
program (EQA) also known as proficiency surveys. Laboratory
regulatory agencies such as the College of American Pathologists
require that a laboratory reporting clinical results of any assay
participate in an EQA that is designed to test each aspect of the
testing and reporting procedure. The laboratory’s performance is
then compared and graded against other laboratories performing
the same testing. Proficiency surveys typically occur 2–3 times per
270 Bruce Greig
NK Lymph CD16+/56+/CD3
% FL-A 2017
FL-C 2016
FL-B 2016
-100 -80 -60 -40 -20 0 20 40 60 80 100
Fig. 27 Proficiency results over time. The CD19% over three different surveys shows results that fall both
above and below the mean of the acceptable results. Conversely, the results of the NK Lymphocytes
consistently run above the mean during the same period. This may be indicative of an analysis gating issue
or antibody that might need to be further investigated for suitability
year and will usually include two or more samples with each mail-
ing. Proficiency testing must follow the laboratory’s written proce-
dures for all the steps used to test and report patient samples. This
not only provides information about the validated procedures but
also measures the performance of the instruments used, acceptabil-
ity of the reagents, and should over time even demonstrate poten-
tial issues that may not be as obvious on a day to day basis. For
instance, if the returned results for CD19 in an immunodeficiency
panel are acceptable but consistently trend higher than the mean of
the other participants it may indicate either a reagent, gating issue,
or instrument problem that might otherwise have gone unnoticed
(see Fig. 27: EQA results over time).
3.7 Quality Control All testing in Immunophenotyping requires that records of quality
Records control performance be maintained and retained for a certain
period of time[27]. There is a saying that goes, “If you did not
document what happened then it did not happen.” Quality assur-
ance and quality control records are the proof of that purpose. Each
step in the process of preanalytic, analytic, and postanalytic perfor-
mance must be documented for acceptability. Best practices in
quality control management include having an overall quality assur-
ance plan along with the records of procedure and instrument
validations, reagents used in testing, daily instrument performance
and control results, assay records of specimen analysis that include
the gating diagrams, and any proficiency testing results. For a
summary of records that should be readily accessible and up to
date see the Notes section of this chapter.
3.8 Conclusion Quality control (QC) and quality assurance (QA) in immunophe-
notyping spans all three phases of laboratory testing: preanalytic,
analytic, and postanalytic procedures. As part of an overall quality
Quality Control of Immunophenotyping 271
4 Notes
4.1 Reagent Recipes 1. PBS buffer, pH 7.2, with sodium azide. Some laboratories
Used in Immuno- include bovine calf serum in their PBS buffer as an additional
phenotyping stabilization agent for the cells. This recipe does not include
that additive.
Caution: Sodium azide is an irritant and a poison. Avoid all
direct skin contact, wear gloves, and wash hands thoroughly
after handling.
Materials:
(a) PBS, 1, Mediatech, Inc., Cat. # 21-040-CM.
(b) Sodium azide, Fisher Scientific, #S227-500.
Working solution:
(a) Add 1 g sodium azide to 1 l of plain PBS.
(b) Mix until dissolved.
(c) Check that the pH is between 7.2 and 7.4.
(d) Label and date (date made and date expired).
(e) Store at 2–8 C when not in use. Expiration ¼ 3 months.
2. Ammonium chloride (NH4Cl) RBC Lysing Agent:
Materials:
(a) Ammonium chloride, Fisher Scientific, #A661-500.
(b) Potassium bicarbonate, Fisher Scientific, P184-500.
(c) Disodium EDTA, Fisher Scientific, #S311-500.
Working solution:
(a) Place the following into a 1-l volumetric flask:
l 8.26 g ammonium chloride.
l 1.0 g potassium bicarbonate.
l 0.037 g (37 mg) disodium EDTA.
(b) Add deionized water to 1-l line on flask. Mix until all
ingredients are dissolved.
(c) Check pH: 7.2–7.4. Discard, if >7.4.
(d) Dispense into 100 ml bottles. Fill bottles to near the top
so that there is no air space∗.
272 Bruce Greig
(e) Cap tightly. Put Parafilm around the cap to prevent air
exposure.
(f) Label and Date (date made, and date expired). Expira-
tion ¼ 1 week. Store at 4 C until ready to use.
∗NOTE: There should be as little air on top of the
lysing solution as possible. Air will cause a decrease in the
pH over time and lower the effectiveness of the lyse
solution.
3. 7-AAD Viability Dye.
Materials:
(a) 7-Aminoactinomycin D, Molecular Probes, Cat.#A-
1310, 1 mg.
(b) Dimethyl sulfoxide (DMSO), Fisher Scientific, Cat.
#D128-500.
(c) 500 ml Flow PBS, stock solution.
7-AAD Stock Solution:
(a) Add 1 ml DMSO to 1.0 mg 7-AAD.
(b) Store at room temperature or in the dark for up to
1 month.
7-AAD Working Dilution
(a) Use 5 μl of 7-AAD stock solution/1–two million cells.
4.4 Instrument Set up single tubes and stain with one antibody-fluorochrome for
Optimization each detector on the instrument and add an extra tube for an
Procedure Using unstained sample. Follow your normal surface staining procedure.
Single-Stained Cells A typical 8-color Optimization Panel would include the following
tubes:
1. Start with the PMT settings from the bead QC. Be sure to turn
off the Compensation for this exercise. Compensation will be
set up after the Optimization procedure.
2. Run the unstained tube-1 and adjust forward and side scatter
light signals to display all three WBC populations clearly.
Adjust each PMT setting so that the leading edge of the nega-
tive signal extends just beyond the background or noise, as
performed in the previous method.
276 Bruce Greig
Fig. 28 Optimized Gating setup that demonstrates CD4 gated on Lymphocytes and Monocytes (R1). The bright
population are CD4+ lymphocytes, the intermediate population are monocytes, the negative population are
CD4- lymphocytes and granulocytes. Using this setup is a way to optimize PMT settings to clearly show all
three populations.
3. After running the unstained sample run the first tube of the
stained sample. If using CD4 draw a gate that includes both the
lymphocytes and monocytes. See Fig. 28 : Optimized Gating
setup that includes lymphocytes and monocytes.
4. Next, draw either a single-parameter histogram of the conju-
gate being acquired, that is, CD4 FITC ¼ FL1, etc. or a
two-parameter dot plot of SSC/FL1. Adjust the PMT voltages
for each detector until the three separate populations can be
clearly seen like that of the CD4 histogram and dot plot of the
figure.
5. Continue acquiring each single-color tube adjusting the PMT
voltages to achieve the best separation between the three popu-
lations until each PMT is optimized.
6. Save the now optimized PMT settings. Once the PMT set-
tings are established the instrument is ready to set up the
compensation.
3. Analytic Records.
Instrument Maintenance—records of all maintenance pro-
cedures as well as any maintenance performed by a service
engineer including scheduled maintenance.
Instrument Performance including all daily QC results and
signed monthly summary should be kept for at least 2 years.
4. Postanalytic Records.
(a) Gated dot plots and histograms should be saved either
electronically or as a hard copy. Clinical laboratories are
required to keep these records for at least 10 years.
References
1. CAP all common checklist and CAP flow cyto- 10. Greig B, Stetler-Stevenson M, Lea J (2014)
metry checklist, College of American Patholo- Stabilization media increases recovery in pauci-
gists, 325 Waukegan Rd. Northfield, IL cellular cerebrospinal fluid specimens submit-
60053, www.cap.org ted for flow cytometry testing. Cytometry B
2. Banfi G, Salvagno GL, Lippi G (2007) The role Clin Cytom 86(2):135–138
of ethylenediamine tetraacetic acid (EDTA) as 11. Hulspas R (2010) Titration of fluorochrome-
in vitro anticoagulant for diagnostic purposes. conjugated antibodies for labeling cell surface
Clin Chem Lab Med 45(5):565–576 markers on live cells. Curr Protoc Cytom.
3. Bowen RAR, Remaley AT (2014) Interferences Chapter 6:Unit 6.29. doi: https://doi.org/
from blood collection tube components on 10.1002/0471142956.cy0629s54
clinical chemistry assays. Biochem Med. Pub- 12. Wood BL, Arroz M, Barnett D et al (2007)
lished online 2014 Feb 15. https://doi.org/ 2006 Bethesda international consensus recom-
10.11613/BM.2014.006 mendations on the immunophenotypic analysis
4. Davis BH, Dasgupta A, Kussick S, on behalf of of hematolymphoid neoplasia by flow cytome-
ICSH/ICCS working group et al (2013) Vali- try: optimal reagents and reporting for the flow
dation of cell-based fluorescence assays: prac- cytometric diagnosis of hematopoietic neopla-
tice guidelines from the ICSH and ICCS- part sia. Cytometry B Clin Cytom 72B:S12–S22
II- preanalytical issues. Cytometry B Clin 13. Department of Health and Human Services,
Cytom 84B:286–290 Centers for Medicare and Medicaid Services
5. Heinlein C, Deppert W, Braithwaite A et al (1992) Clinical laboratory improvement
(2010) A rapid and optimization-free proce- amendments of 1988; final rule. Fed Register.
dure allows the in vivo detection of subtle cell 7146 [42CFR493.1256]
cycle and ploidy alterations in tissues by flow 14. Department of Health and Human Services,
cytometry. Cell Cycle 9. https://doi.org/10. Centers for Medicare and Medicaid Services
4161/cc.9.17.12831 (2003) Clinical laboratory improvement
6. Novelli P, Savoia I, Cambieri R et al (2000) amendments of 1988; final rule. Fed Register.
Collagenase digestion and mechanical disag- 7166 [42CFR493.1256(f)]
gregation as a method to extract and immuno- 15. Bagwell CB, Adams EG (1993) Fluorescence
phenotype tumour lymphocytes in cutaneous spectral overlap compensation for any number
T cell lymphomas. Clin Exp Dermatol of flow cytometry parameters. Ann N Y Acad
25:423–431 Sci 677:167–184
7. Becton Dickinson MedimachineTM brochure. 16. D Biosciences, technical bulletin: an introduc-
Becton Dickinson Biosciences, 2350 Qume tion to compensation for multicolor assays on
Dr., San Jose, CA, 95131 digital flow cytometers (2009) https://www.
8. Moriarty AT, Wiersema L, Snyder W et al bdbiosciences.com/documents/Compensa
(1993) Immunophenotyping of cytologic spe- tion_Multicolor_TechBulletin.pdf
cimens by flow cytometry. Diagn Cytopathol 9 17. Maecker HT, Trotter J (2008) Flow cytometry
(3):252–258 controls, instrument setup, and the determina-
9. Caldwell CW (1998) Analyte-specific reagents tion of positivity. Cytometry A 69A
in the flow cytometry laboratory. Arch Pathol (9):1037–1042
Lab Med 122:861–864
Quality Control of Immunophenotyping 279
18. Landay A et al (1992) Clinical applications of Laboratory Standards Institute, 940 West Val-
flow cytometry: quality assurance and immuno- ley Road, Suite 1400, Wayne, Pennsylvania
phenotyping of peripheral blood lymphocytes, 19087–1898 USA
Document H42-T, Vol. 12, No. 6, pp. 6–8 26. Schmid I, Krall WJ, Uittenbogaart CH et al
19. Keeney M, Gratama JW, Chin-Yee IH et al (1992) Dead cell discrimination with
(1998) Isotype controls in the analysis of lym- 7-aminoactinomycin D in combination with
phocytes and CD34+ stem and progenitor cells dual color immunofluorescence in single laser
by flow cytometry – time to let go! Cytometry flow cytometry. Cytometry 13(2):204–208
343:280–283 27. CAP Policy PP Retention of laboratory records
20. Lindmo T, Fundingsrud K (1981) Measure- and materials. College of American Patholo-
ment of the distribution of time intervals gists, 325 Waukegan Rd. Northfield, IL
between cell passages in flow cytometry as a 60053, www.cap.org
method for the evaluation of sample prepara- 28. CD-Chex Plus™ Immunology Control for
tion procedures. Cytometry 2:151–154 Cluster Designation package insert. Streck.
21. Sphero TM Rainbow calibration particles. 7002 S. 109 Street, La Vista, NE 68128
Spherotech, Inc. 27845 Irma Lee Circle, Lake 29. All Common Checklist, pp 26-29. College of
Forest, IL 60045 American Pathologists, 325 Waukegan Road,
22. BD Biosciences (2014) Multicolor flow cyto- Northfield, IL 60093-2750. Aug 22, 2018
metry: beyond the basics, course workbook. 30. Flow Cytometry Checklist. College of Ameri-
BD Biosciences, San Jose, CA can Pathologists, 325 Waukegan Rd., North-
23. Darzynkiewicz Z, Halicka HD, Zhao H (2010) field, IL, 60093-2750. Aug 22, 2018.
Analysis of cellular DNA content by flow and 31. Sutherland DR, Anderson L, Keeney M, et al.
laser scanning cytometry. Adv Exp Med Biol (1996) The ISHAGE Guidelines for CD34+
676:137–147 cell determination by flow cytometry. Interna-
24. Nunez R (2001) DNA measurement and cell tional Society of Hematotherapy and Graft
cycle analysis by flow cytometry. Curr Issues Engineering. J Hematother. 5(3):213–26
Mol Biol 3(2):67–70 32. Davis
25. Clinical and Laboratory Standards Institute 33. Biolegend Blog. The Stain Index: What is it
(CLSI) (2007) Clinical flow cytometric analysis and what does it tell you. http://www.biole-
of neoplastic hematolymphoid cells; approved gend.com/newsdetail/1245/
guideline—Second Edition. CLSI document
H43-A2 (ISBN 1-56238-635-2). Clinical and
Chapter 15
Abstract
Immunophenotyping by multiparameter flow cytometry is a rapid and efficient technique to simultaneously
assess and correlate multiple individual cell properties like size and internal complexity along with antigen
expression in a population of cells. This method is utilized for rapid characterization of the blasts and
classification of acute myeloid leukemia (AML), in both the peripheral blood (PB) and bone marrow (BM).
This technique is not only useful in the initial diagnosis but also in monitoring and determining prognosis
of the disease through minimal residual disease (MRD) testing. This chapter provides an overview of
procedures for specimen processing, staining, and immunophenotyping of AML and describes the princi-
ples of data analysis for AML classification and MRD testing.
Key words Immunophenotype, Acute myeloid leukemia, Myelodysplastic syndrome, Minimal resid-
ual disease, Flow cytometry
1 Introduction
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_15, © Springer Science+Business Media, LLC, part of Springer Nature 2019
281
282 Pallavi Kanwar Galera et al.
2 Materials
2.2 Monoclonal Table 1 describes the antibodies utilized in our laboratory for
Antibody Panels detection, characterization, and classification of AML with appro-
priate corresponding fluorochromes (see Note 1).
Table 1
Antibodies and fluorochromes utilized in our laboratory*
3 Methods
3.2 Labeling This method allows the detection of cytoplasmic antigens, along
with Monoclonal with the assessment of expression of surface antigens. It utilizes a
Antibodies Against cross-linking fixative to stabilize cellular contents prior to labeling
Combined Surface with antibodies against cytoplasmic antigens. The procedure outlined
and Intracellular here utilizes Erythrocyte Lysis Buffer (Qiagen) and Fix Perm Reagent
Antigens A and B (Caltag), but other products may be used. It is also critical
to appropriately titrate the amount of antibody to get an optimal
antibody dilution for maximum signal-to-noise ratio [21, 22].
1. Lyse RBCs from sample using Erythrocyte Lysis Buffer: To lyse
the RBCs add 2 mL of the Erythrocyte Lysis Buffer to 200 μL
of cell suspension in a 12 75 mm disposable polystyrene
tube. Allow to lyse for approximately 10 min. Spin at 216 g
for 5 min. Decant supernatant.
Immunophenotyping of Acute Myeloid Leukemia 285
3.3 Instrument A variety of instruments are available and different laboratories may
Settings use different instruments to perform flow cytometric analysis. Set-
tings and calibrations are instrument-dependent, and manufac-
turer’s guidelines should be employed. The number of cells
acquired for analysis per tube may vary depending on the level of
detection sensitivity desired. In our laboratory we acquire at least
100,000 cells/tube for routine analysis; however, higher numbers
are necessary for MRD testing.
3.4 Data Analysis The initial evaluation of results consists of an assessment of the
quality of data and viability of the specimen. One strategy to
3.4.1 Data Quality
evaluate quality and consistency of the acquired data is to observe
and Viability
data over time to identify air aspiration artifacts due to sample
exhaustion and fluidic instabilities (see Note 5) (Fig. 1). The second
step in ensuring the quality of data collected is to make certain that
the data analyzed is an actual measure of a single cell at a time.
Frequently, high cell concentration, high flow rates or nonspecific
antibody bindings can induce cell aggregates and clump forma-
tions. To analyze exclusively single cells, a combination of forward
light scatter area (FSC-A) and forward light scatter width (FSC-W)
and/or height (FSC-H) is utilized to exclude cell doublets and
aggregates from the data (Fig. 1).
Finally, to evaluate the viability of the specimen and to exclude
debris and degenerated cells, viability dyes (propidium iodide,
7AAD) can be utilized. Alternatively, for practical purposes, nonvi-
able cells may be excluded utilizing light scatter characteristics
[19, 23, 24].
286 Pallavi Kanwar Galera et al.
Fig. 1 Normal bone marrow. Data quality: Data is observed over time to identify and exclude fluidic instabilities
such as air aspiration artifacts (upper left graph). A combination of forward light scatter area (FSC-A) and
forward light scatter width (FSC-W) (lower left graph), and/or forward light scatter height (FSC-H) (lower
middle graph) is utilized to exclude doublets and to ensure that the data analyzed is a measure of a single cell
at a time. Initial gating (right graph): Based on the side light scatter (SSC-A) and CD45 expression the marrow
populations can be broadly selected as red blood cells (brown), granulocytes (green), lymphocytes (blue),
monocytes (aqua), and cells in the blast gate (red)
3.4.2 Initial Cell The initial gating, after excluding all elements that are not single
Population Gating viable cells, is performed to define broad categories of cell popula-
tions. This is done by examining side light scatter (SSC-A) and
CD45 signals (see Note 6) [25]. Using this combination, the
following populations can be broadly identified: erythroid (low
SSC-A and negative CD45), granulocytes (moderate to high
SSC-A and moderate CD45), lymphocytes (low SSC-A and bright
CD45), monocytes (intermediate SSC-A between lymphocytes and
granulocytes and bright CD45) and blasts (low SSC-A and dim
CD45) (Fig. 1) [26]. Though an effective initial gate, the “blast
gate” based on SSC-A and CD45 may be inaccurate [27] since
there may be other populations that fall in this gate, namely baso-
phils, hypogranular myeloid cells, plasmacytoid dendritic cells, and
immature monocytes. Also, there would be instances where the
blasts may have a brighter or dimmer CD45 and a varied SSC-A.
Other populations may also, to a certain extent, be relatively
ill-defined using this initial gating approach. Therefore, a panel of
additional markers is required to further delineate the blasts as well
as other distinct cell populations.
Immunophenotyping of Acute Myeloid Leukemia 287
Fig. 2 Bone marrow with increased, phenotypically abnormal myeloblasts, comprising 23% of total cells. The
blasts (red dots) express CD45, CD34, CD13, CD117, CD33 (bright), CD38 (dim/negative), HLA-DR, CD64
(dim/negative), and CD123 (dim). Blasts are negative for CD14, CD15, CD16, CD11b, CD7, and CD2
3.4.3 Strategies for Blast To assess the presence of myeloblasts, commonly used antigens
Identification include CD34 (Fig. 2) and CD117 since blasts usually express
these markers. However, some myeloblasts may be negative for
CD34 and/or CD117 and not even fall within the typical location
in the CD45 and SSC-A graphs, making their recognition difficult.
In these instances, other reagents and strategies are necessary for
their detection. For example, blasts in acute promyelocytic leuke-
mia are CD34 dim or negative and may mimic normal granulocytes
but they usually have distinct phenotypic features [28]. Also,
monoblasts usually express no or dim CD34, requiring the use of
reagents for monocyte-associated markers for their recognition.
3.4.4 Strategies Myeloid lineage markers commonly used include CD13, CD15,
for Lineage Assignment CD33, MPO, and CD16 along with markers expressed during
monocytic differentiation such as CD11b, CD64, CD14, and
CD4 (Fig. 2). Erythroid precursors express CD71, CD105,
CD117, Glycophorin A (CD 235a), and CD36, and megakaryo-
cytic precursors express platelet-associated glycoproteins such as
CD61 and CD42b. Assessment of lymphoid markers (CD3,
CD2, CD4, CD5, CD7, CD19, CD56, CD22, and CD79a)
should be included at the time of initial diagnosis, to assign lineage
or to look for aberrant antigen expression. Non-lineage-specific
markers (CD38, CD56, CD123, and HLA-DR) are also helpful
to further delineate blasts and aberrancies.
288 Pallavi Kanwar Galera et al.
Fig. 3 Bone marrow involved by AML with FLT3 mutation and phenotypically abnormal myeloblasts comprising
95% of total cells. The blasts (red dots) express CD45, CD34 (dim to negative), CD13, CD117 (variable), CD64
(variable), CD38 (partial), HLA-DR, CD123, CD33, CD7 (partial aberrant). Blasts are negative for CD16, CD14,
and other T-cell and B-cell markers (not shown)
3.4.5 Background In acute myeloid leukemias, it may be useful to evaluate the back-
Myelopoiesis ground myelopoiesis to determine the presence of possible dysplas-
tic changes. For this purpose, knowledge of normal maturation
patterns utilizing various combinations of antigens is essential.
Patterns generated by CD45 and SSC-A (SSC-A reflecting granu-
larity), CD13 and CD16, CD11b and CD64, and CD11b and
CD16 have been well studied for assessment of myeloid maturation
and are reproducible [4, 5] (Fig. 5). Alterations of these patterns
and other aberrancies like loss of CD10 on mature granulocytes, or
expression of CD56 on myeloid and monocytic cells are helpful
clues into probable underlying myelodysplasia [7, 19, 30].
Fig. 4 A patient with history of autoimmune lymphoproliferative syndrome with FAS mutation. Bone marrow
aspirate shows two populations of myeloblasts (red dots) based on CD34 expression with the minority of the
blasts (approximately 25%) expressing CD34 and the majority of the blasts are negative for CD34. The blasts
also express CD45, CD117, CD13, HLA-DR (bright), CD56 (aberrant), and CD33. A small subset with dim
expression of CD19 is noted. The blasts are negative for CD7, CD14, and CD64. They were also negative for
CD10, CD20, CD3, and CD5 (not shown)
Fig. 5 A patient with history of BRCA1 and BRCA2 mutations and therapy for breast cancer 7 years prior to
presentation. Bone marrow aspirate shows abnormal myeloblasts (red dots), comprising 13% of total cells.
The blasts express CD45, CD34, CD13, CD33, CD117 (dim to negative), HLA-DR, CD7 (partial & aberrant) and
are negative for CD38 (aberrant), CD2, CD16, CD14, CD64, and CD123. Granulocytes (green dots) reveal
disrupted maturation pattern based on the expression of CD13 and CD16
Fig. 6 AML with t(9;11)(p21.3;q23.3); KMT2A-MLLT3. Bone marrow aspirate reveals over 90% blasts, with
two different immunophenotypically abnormal populations: The first population is of immature myeloblasts
(red dots), representing approximately 81% of total cells and expressing CD45(dim), CD34 (partial, 72% of
total cells), CD117, CD13, HLA-DR, CD33 (bright), CD123, and CD64 (dim to negative). These blasts are
negative for CD14, CD16, and CD11b. They are also negative for CD15, CD61/CD42b, T-cell and B-cell
markers (not shown). The second population is of monoblasts (aqua dots), representing approximately 10% of
total cells with intermediate SSC-A and expressing CD45 (brighter than first population), CD13, HLA-DR
(bright), CD38, CD33 (bright), CD64 (bright), CD11b (variable), CD36 (not shown). These are negative for CD34,
CD117, CD16, and CD14
3.4.7 Minimal Residual The detection limit of leukemic blasts by flow cytometry ranges
Disease from 0.1% to 0.01% of leukocytes. The detection limit is dependent
on various factors, including the number of cells collected, anti-
body panel, flow cytometer, and distinct immunophenotypic prop-
erties of the leukemic blasts. For a detection sensitivity of 0.01%, at
least 1,000,000 cells should be analyzed.
Two strategies are commonly employed in MRD testing for
AML by flow cytometry. The first strategy is identifying a combina-
tion of aberrant markers and their expression levels on leukemic
blasts that are usually absent in normal hematopoietic precursors.
This strategy is widely known as “leukemia-associated immunophe-
notype” or LAIP. At diagnosis a broad, extensive antigen panel is
utilized to define the LAIP. Subsequently after treatment, the
selected antibody panel is used to identify blasts with pretreatment
LAIP. The advantages of this approach are that the data analysis is
relatively simple, and sensitivity for each LAIP (defined based on the
background expression in nonleukemic specimens) is known. The
disadvantages include the requirement to know the complete phe-
notype of the pretreatment leukemic sample, the need for custo-
mized antibody panels for subsequent evaluations after the
evaluation of the initial diagnostic specimen and, finally, the immu-
nophenotype of the leukemic blasts might change after treatment
[3, 9, 11, 13, 20, 23].
The second strategy known as “difference from normal” iden-
tifies leukemic blasts based on immunophenotypic differences
between the leukemic blasts and normal hematopoietic precursors.
There is no requirement of defining or having prior knowledge of
LAIPs at diagnosis (which is a hindrance in many tertiary or referral
centers that do not have this information available); the same
standardized panel can be used for all evaluations and since this
method is based on differences between blasts and normal myeloid
precursors, immunophenotypic shifts and change of LAIPs after
treatment do not interfere with the assessment. However, extensive
knowledge of antigen expression patterns of normal hematopoietic
precursors and their maturation patterns is essential for an accurate
evaluation [3, 9, 11, 20, 23]. In practice, a combination of the two
strategies is often utilized.
Immunophenotyping of Acute Myeloid Leukemia 293
4 Notes
Acknowledgments
References
1. Swerdlow SH, Campo E, Harris NL et al (eds) 12. Dohner H, Estey E, Grimwade D et al (2017)
(2017) WHO classification of tumours of hae- Diagnosis and management of AML in adults:
matopoietic and lymphoid tissues, 4th edn. 2017 ELN recommendations from an interna-
IARC, Lyon tional expert panel. Blood 129(4):424–447
2. Craig FE, Foon KA (2008) Flow cytometric 13. Terwijn M, van Putten WL, Kelder A et al
immunophenotyping for hematologic neo- (2013) High prognostic impact of flow cyto-
plasms. Blood 111:3941–3967 metric minimal residual disease detection in
3. Chen X, Cherian S (2017) Acute myeloid leu- acute myeloid leukemia: data from the
kemia immunophenotyping by flow cytometric HOVON/SAKK AML 42A study. J Clin
analysis. Clin Lab Med 37:753–769 Oncol 31(31):3889–3897
4. van Lochem EG, van der Velden VH, Wind HK 14. Venditti A, Buccisano F, Del Poeta G et al
et al (2004) Immunophenotypic differentia- (2000) Level of minimal residual disease after
tion patterns of normal hematopoiesis in consolidation therapy predicts outcome in
human bone marrow: reference patterns for acute myeloid leukemia. Blood 96
age-related changes and disease-induced shifts. (12):3948–3952
Cytometry B Clin Cytom 60(1):1–13 15. Zhu HH, Zhang XH, Qin YZ et al (2013)
5. Gorczyca W, Sun ZY, Cronin W et al (2011) MRD-directed risk stratification treatment
Immunophenotypic pattern of myeloid popu- may improve outcomes of t(8;21) AML in the
lations by flow cytometry analysis. Methods first complete remission: results from the
Cell Biol 103:221–266 AML05 multicenter trial. Blood 121
6. Loken MR, Chu SC, Fritschle W (2009) Nor- (20):4056–4062
malization of bone marrow aspirates for hemo- 16. Freeman SD, Virgo P, Couzens S et al (2013)
dilution in flow cytometric analyses. Cytometry Prognostic relevance of treatment response
B Clin Cytom 76(1):27–36 measured by flow cytometric residual disease
7. Wood BL (2007) Myeloid malignancies: mye- detection in older patients with acute myeloid
lodysplastic syndromes, myeloproliferative dis- leukemia. J Clin Oncol 31(32):4123–4131
orders, and acute myeloid leukemia. Clin Lab 17. Walter RB, Gooley TA, Wood BL et al (2011)
Med 27(3):551–575. vii Impact of pretransplantation minimal residual
8. Wood BL, Arroz M, Barnett D et al (2007) disease, as detected by multiparametric flow
2006 Bethesda international consensus recom- cytometry, on outcome of myeloablative hema-
mendations on the immunophenotypic analysis topoietic cell transplantation for acute myeloid
of hematolymphoid neoplasia by flow cytome- leukemia. J Clin Oncol 29(9):1190–1197
try: optimal reagents and reporting for the flow 18. Getta BM, Devlin SM, Levine RL et al (2017)
cytometric diagnosis of hematopoietic neopla- Multicolor flow cytometry and multigene next-
sia. Cytometry B Clin Cytom 72(Suppl 1): generation sequencing are complementary and
S14–S22 highly predictive for relapse in acute myeloid
9. Wood BL (2016) Principles of minimal residual leukemia after allogeneic transplantation. Biol
disease detection for hematopoietic neoplasms Blood Marrow Transplant 23(7):1064–1071
by flow cytometry. Cytometry B Clin Cytom 19. van Dongen JJ, Lhermitte L, Böttcher S, Euro-
90(1):47–53 Flow Consortium (EU-FP6, LSHB-CT-2006-
10. Xu J, Jorgensen JL, Wang SA (2017) How do 018708) et al (2012) EuroFlow antibody
we use multicolor flow cytometry to detect panels for standardized n-dimensional flow
minimal residual disease in acute myeloid leu- cytometric immunophenotyping of normal,
kemia? Clin Lab Med 37:787–802 reactive and malignant leukocytes. Leukemia
26(9):1908–1975
11. Zhou Y, Wood BL (2017) Methods of detec-
tion of measurable residual disease in AML. 20. Schuurhuis GJ, Heuser M, Freeman S et al
Curr Hematol Malig Rep 12:557–567 (2018) Minimal/measurable residual disease
in AML: a consensus document from the
Immunophenotyping of Acute Myeloid Leukemia 295
European LeukemiaNet MRD working party. expression of immature B-cell antigen CD19
Blood 131(12):1275–1291 together with stem cell antigen CD34. Blood
21. Reilly JT (1996) Use and evaluation of leuko- 80:470–477
cyte monoclonal antibodies in the diagnostic 33. Baer MR, Stewart CC, Lawrence D et al
laboratory: a review. Clin Lab Haematol (1997) Expression of the neural cell adhesion
18:1–5 molecule CD56 is associated with short remis-
22. Owens MA, Loken MR (1995) Quality control sion duration and survival in acute myeloid
of flow cytometer and reagents. In: Owens leukemia with t(8;21)(q22;q22). Blood
MA, Loken MR (eds) Flow cytometry: princi- 90:1643–1648
ples for clinical laboratory practice. Quality 34. Iriyama N, Hatta Y, Takeuchi J et al (2013)
assurance for quantitative immunophenotyp- CD56 expression is an independent prognostic
ing. Wiley, New York, pp 45–72 factor for relapse in acute myeloid leukemia
23. Wood BL (2013) Flow cytometric monitoring with t(8;21). Leuk Res 37:1021–1026
of residual disease in acute leukemia. Methods 35. De J, Zanjani R, Hibbard M et al (2007)
Mol Biol 999:123–136 Immunophenotypic profile predictive of KIT
24. Chen W, Luu HS (2017) Immunophenotyping activating mutations in AML1-ETO leukemia.
by multiparameter flow cytometry. Methods Am J Clin Pathol 128:550–557
Mol Biol 1633:51–73 36. Ouyang J, Goswami M, Peng J et al (2016)
25. Stelzer GT, Shults KE, Loken MR (1993) Comparison of multiparameter flow cytometry
CD45 gating for routine flow cytometric anal- immunophenotypic analysis and quantitative
ysis of human bone marrow specimens. Ann N RT-PCR for the detection of minimal residual
Y Acad Sci 677:265–280 disease of core binding factor acute myeloid
26. Borowitz MJ, Guenther KL, Shults KE et al leukemia. Am J Clin Pathol 145(6):769–777
(1993) Immunophenotyping of acute leuke- 37. Exner M, Thalhammer R, Kapiotis S et al
mia by flow cytometric analysis. Use of CD45 (2000) The “typical” immunophenotype of
and right-angle light scatter to gate on leuke- acute promyelocytic leukemia (APL-M3):
mic blasts in three-color analysis. Am J Clin does it prove true for the M3-variant? Cytome-
Pathol 100:534–540 try 42:106–109
27. Harrington AM, Olteanu H, Kroft SH (2012) 38. Takenokuchi M, Kawano S, Nakamachi Y et al
A dissection of the CD45/side scatter “blast (2012) FLT3/ITD associated with an imma-
gate”. Am J Clin Pathol 137:800–804 ture immunophenotype in PML-RARα leuke-
28. Orfao A, Chillón MC, Bortoluci AM et al mia. Hematol Rep 4(4):e22
(1999) The flow cytometric pattern of CD34, 39. Montesinos P, Rayón C, Vellenga E et al
CD15 and CD13 expression in acute myelo- (2011) Clinical significance of CD56 expres-
blastic leukemia is highly characteristic of the sion in patients with acute promyelocytic leu-
presence of PML-RARalpha gene rearrange- kemia treated with all-trans retinoic acid and
ments. Haematologica 84(5):405–412 anthracycline-based regimens. Blood
29. Kussick SJ, Wood BL (2003) Using 4-color 117:1799–1805
flow cytometry to identify abnormal myeloid 40. Muñoz L, Nomdedéu JF, Villamor N et al
populations. Arch Pathol Lab Med (2003) Acute myeloid leukemia with MLL
127:1140–1147 rearrangements: clinicobiological features,
30. van de Loosdrecht AA, Alhan C, Béné MC et al prognostic impact and value of flow cytometry
(2009) Standardization of flow cytometry in in the detection of residual leukemic cells. Leu-
myelodysplastic syndromes: report from the kemia 17:76–82
first European LeukemiaNet working confer- 41. Creutzig U, Harbott J, Sperling C et al (1995)
ence on flow cytometry in myelodysplastic syn- Clinical significance of surface antigen expres-
dromes. Haematologica 94(8):1124–1134 sion in children with acute myeloid leukemia:
31. Hurwitz CA, Raimondi SC, Head D et al results of study AML-BFM-87. Blood
(1992) Distinctive immunophenotypic fea- 86:3097–3108
tures of t(8;21)(q22;q22) acute myeloblastic 42. Alsabeh R, Brynes RK, Slovak ML et al (1997)
leukemia in children. Blood 80:3182–3188 Acute myeloid leukemia with t(6;9) (p23;q34):
32. Kita K, Nakase K, Miwa H et al (1992) Pheno- association with myelodysplasia, basophilia,
typical characteristics of acute myelocytic leu- and initial CD34 negative immunophenotype.
kemia associated with the t(8;21)(q22;q22) Am J Clin Pathol 107:430–437
chromosomal abnormality: frequent
296 Pallavi Kanwar Galera et al.
43. Lai YY, Li Y, Shi Y et al (2012) Characteristics 52. Liu YR, Zhu HH, Ruan GR et al (2013)
of 11 patients with acute myeloid leukemia NPM1-mutated acute myeloid leukemia of
accompanied with karyotype aberration t monocytic or myeloid origin exhibit distinct
(6;9). Zhongguo Shi Yan Xue Ye Xue Za Zhi immunophenotypes. Leuk Res 37:737–741
20:1293–1296 53. Angelini DF, Ottone T, Guerrera G et al
44. Oyarzo MP, Lin P, Glassman A et al (2004) (2015) A leukemia-associated CD34/
Acute myeloid leukemia with t(6;9)(p23;q34) CD123/CD25/CD99+ immunophenotype
is associated with dysplasia and a high fre- identifies FLT3-mutated clones in acute mye-
quency of flt3 gene mutations. Am J Clin loid leukemia. Clin Cancer Res 21:3977–3985
Pathol 122:348–358 54. Lin LI, Chen CY, Lin DT et al (2005) Charac-
45. Slovak ML, Gundacker H, Bloomfield CD et al terization of CEBPA mutations in acute mye-
(2006) A retrospective study of 69 patients loid leukemia: most patients with CEBPA
with t(6;9)(p23;q34) AML emphasizes the mutations have biallelic mutations and show a
need for a prospective, multicenter initiative distinct immunophenotype of the leukemic
for rare ‘poor prognosis’ myeloid malignancies. cells. Clin Cancer Res 11:1372–1379
Leukemia 20:1295–1297 55. Hou HA, Lin LI, Chen CY et al (2009) Reply
46. Medeiros BC, Kohrt HE, Arber DA et al to ‘heterogeneity within AML with CEBPA
(2010) Immunophenotypic features of acute mutations; only CEBPA double mutations,
myeloid leukemia with inv(3)(q21q26.2)/t but not single CEBPA mutations are associated
(3;3)(q21;q26.2). Leuk Res 34:594–597 with favorable prognosis’. Br J Cancer
47. Raya JM, Martı́n-Santos T, Luño E et al (2015) 101:738–740
Acute myeloid leukemia with inv(3)(q21q26.2) 56. Tang JL, Hou HA, Chen CY et al (2009)
or t(3;3)(q21;q26.2): clinical and biological fea- AML1/RUNX1 mutations in 470 adult
tures and comparison with other acute myeloid patients with de novo acute myeloid leukemia:
leukemias with cytogenetic aberrations involving prognostic implication and interaction with
long arm of chromosome 3. Hematology other gene alterations. Blood 114:5352–5361
13:1607845415Y0000000003 57. Schnittger S, Dicker F, Kern W et al (2011)
48. Bernstein J, Dastugue N, Haas OA et al (2000) RUNX1 mutations are frequent in de novo
Nineteen cases of the t(1;22)(p13;q13) acute AML with noncomplex karyotype and confer an
megakaryblastic leukaemia of infants/ children unfavorable prognosis. Blood 117:2348–2357
and a review of 39 cases: report from a t(1;22) 58. Haferlach T, Kohlmann A, Klein HU et al
study group. Leukemia 14:216–218 (2009) AML with translocation t(8;16)(p11;
49. Carroll A, Civin C, Schneider N et al (1991) p13) demonstrates unique cytomorphological,
The t(1;22) (p13;q13) is nonrandom and cytogenetic, molecular and prognostic features.
restricted to infants with acute megakaryoblas- Leukemia 23:934–943
tic leukemia: a pediatric oncology group study. 59. Dang H, Chen Y, Kamel-Reid S et al (2013)
Blood 78:748–752 CD34 expression predicts an adverse outcome
50. Inaba H, Zhou Y, Abla O et al (2015) Hetero- in patients with NPM1-positive acute myeloid
geneous cytogenetic subgroups and outcomes leukemia. Hum Pathol 44:2038–2046
in childhood acute megakaryoblastic leukemia: 60. Chen CY, Chou WC, Tsay W et al (2013)
a retrospective international study. Blood Hierarchical cluster analysis of immunopheno-
126:1575–1584 typed classify AML patients with NPM1 gene
51. Nomdedéu J, Bussaglia E, Villamor N et al mutation into two groups with distinct prog-
(2011) Immunophenotype of acute myeloid nosis. BMC Cancer 13:107
leukemia with NPM mutations: prognostic
impact of the leukemic compartment size.
Leuk Res 35:163–168
Chapter 16
Abstract
Immunophenotyping by flow cytometry is an important component in the diagnostic evaluation of patients
with acute lymphoblastic leukemia. This technique further permits the detection of minimal residual disease
after therapy, a robust prognostic factor that may guide individualized treatment. We describe here
laboratory methods for both the initial characterization of lymphoblasts at diagnosis, and the detection
of rare leukemic lymphoblasts after treatment. In addition to antibody combinations suitable for diagnosis
and detection of minimal residual disease, we describe procedures for peripheral blood and bone marrow
sample preparation, procedures for labeling of cell-surface and intracellular proteins with fluorochrome-
conjugated antibodies, and approaches to analysis of immunophenotypic data.
Key words Immunophenotyping, Flow cytometry, Acute lymphoblastic leukemia, Minimal residual
disease, Antibodies
1 Introduction
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_16, © Springer Science+Business Media, LLC, part of Springer Nature 2019
297
298 Joseph A. DiGiuseppe and Jolene L. Cardinali
2 Materials
2.2 Antibody Antibody combinations suitable for the diagnosis and detection of
Combinations minimal residual disease (MRD) in acute lymphoblastic leukemia
(ALL) are presented in Table 1. These combinations were devel-
oped for use on flow cytometers equipped with three lasers (excita-
tions at 405 nm, 488 nm, and 633 nm), and capable of detecting at
Immunophenotyping of Acute Lymphoblastic Leukemia 299
Table 1
Antibody combinations suitable for diagnosis and detection of minimal residual disease in acute
lymphoblastic leukemia
PB, V450,
Tube FITC PE PerCP-Cy5.5 PE-Cy7 APC APC-AF750 or BV421 V500
1 Kappa Lambda CD20 CD19 CD10 CD38 CD5 CD45
2 CD20 CD22 CD34 CD19 CD13 + CD33 CD38 CD10 CD45
3 CD20 CD49f CD34 CD19 CD58 CD38 CD10 CD45
4 CD24 CD304 CD34 CD19 CD86 CD38 CD10 CD45
5 CD16 CD56 CD5 CD3 CD7 CD8 CD4 CD45
6 CD7 CD1a CD3 CD2 CD5 CD8 CD4 CD45
7 cyMPO cyCD3 CD34 – CD7 – HLA-DR CD45
8 cyMPO cyCD22 cyCD79a CD19 CD34 – HLA-DR CD45
9 nTdt cyCD3 cyCD79a CD19 CD34 – HLA-DR CD45
FITC fluorescein isothiocyanate, PE phycoerythrin, PerCP-Cy5.5 peridinin-chlorophyll-Cy5.5, APC allophycocyanin,
APC-AF750 APC-Alexa Fluor 750, PB Pacific Blue, V450 BD Horizon™ V450, BV421 Brilliant Violet™ 421, V500 BD
Horizon™ V500
3 Methods
3.3 Labeling 1. Label 12 75-mm polystyrene tubes with two patient identi-
Procedure for Cell- fiers and the antibody combination to be added.
Surface Antigens 2. Deliver 100 μL of the appropriate antibody combination (cock-
tail) to each tube (see Note 4).
3. Deliver 100 μL of the cell suspension (or preincubated cell
suspension in the case of immunoglobulin light chain studies)
as prepared in Subheadings 3.1 and 3.2 to each tube and vortex
(see Note 5).
4. Incubate the tubes for 15 min at room temperature in the dark.
5. Pipet 2 mL of FACS™ Lysing Solution into each tube and
vortex.
6. Incubate for 10 min at room temperature in the dark.
7. Centrifuge for 5 min at 800 RCF, and remove the supernatant.
8. Dilute to a volume of 2 mL with 0.5% PBS/BSA/azide and
vortex.
9. Centrifuge for 5 min at 800 RCF and remove the supernatant.
10. Fix with 500 μL of 0.5% PBS/BSA/azide and 500 μL of 2%
formaldehyde.
11. Acquire the labeled cells on the flow cytometer (see Note 6).
Acquisition of 100,000 viable cells is sufficient for initial diag-
nosis; for MRD detection, acquisition of 1,000,000 cells is
attempted (see Note 7). To mitigate carryover during acquisi-
tion for MRD detection, tubes containing distilled water
should be positioned between sample tubes.
3.5 Data Analysis At both diagnosis and assessment of MRD after therapy, the goal of
data analysis is the identification of normal and abnormal cell
populations on the basis of their immunophenotypic and light-
scatter properties. Data analysis is an iterative process, which
requires a thorough understanding of normal and abnormal matu-
ration patterns [6, 9], and familiarity with the technical artifacts
that may produce spurious immunophenotypic patterns [10]. The
magnitude of this challenge is underscored by a recent study of
MRD detection in B lymphoblastic leukemia, in which analytic
precision among a cohort of experienced practitioners using iden-
tical data sets was disappointing [11]. Although the development of
algorithms capable of analyzing complex multiparametric data sets
holds the promise of improving analytic precision [12], such
approaches have yet to be widely adopted in routine practice.
Fig. 1 Gating strategies to exclude overt data artifacts. (Left) Assessment of acquisition using event number to
detect potential fluidic perturbations. (Center) Exclusion of coincident events (cellular doublets or higher-
ordered aggregates) by gating on singlets defined by FSC-A and FSC-H. (Right) Exclusion of debris and
nonviable cells by gating on cells with the FSC and SSC properties of viable cells
Fig. 2 Identification of blasts at diagnosis in B lymphoblastic leukemia (peripheral blood). The B-lymphoblasts
(blue) have dim CD45 relative to mature B cells (red), and low SSC. In addition to B-lineage antigens (CD19,
CD22), the blasts express CD10 and CD34 (All viable singlets are shown)
3.5.2 Identification The next step in data analysis is identification of the abnormal
of the Abnormal population. A display of CD45 vs. SSC is very useful in this regard,
Population: Diagnosis as lymphoblasts commonly occupy a region defined by dim CD45
and low SSC (Figs. 2 and 3). In the case of B lymphoblastic
leukemia, CD45 expression may be virtually undetectable
(Fig. 1), while in the case of T lymphoblastic leukemia, overlap of
CD45 expression between the blasts and mature T cells is more
common (Fig. 2). Bivariate plots of antigen vs. light scatter, or
antigen vs. antigen are then used to refine phenotypic
304 Joseph A. DiGiuseppe and Jolene L. Cardinali
Fig. 4 Identification of MRD in B lymphoblastic leukemia (bone marrow). The initial CD19+ gate includes a
preponderance of normal B-cell precursors (blue), with dim CD45, little or no CD20, and substantial CD34; in
contrast, the MRD (red, highlighted) differs from the normal B-cell precursors in its expression one or more
myeloid antigens (CD13 + CD33), lack of CD10 and CD38, and overexpression of CD49f and CD58 (All viable
singlets are shown in the upper left and center panels; only B cells are shown in the remaining panels)
306 Joseph A. DiGiuseppe and Jolene L. Cardinali
However, the MRD in this case differs from the normal B-cell
precursors in its expression of one or more myeloid antigens
(CD13 + CD33), lack of CD10 and CD38 expression, and over-
expression of CD49f (see Note 11) and CD58 (Fig. 4). In the
example of MRD detected in a patient treated for T lymphoblastic
leukemia (Fig. 5), iterative gating discloses an abnormal T-cell
population (blue, highlighted) with dim CD5 and CD45, absent
surface CD3, and coexpression (albeit weak) of CD4 and CD8.
These phenotypic features differ from those of the normal CD4+
and CD8+ T cells (red and green, respectively) but resemble those
of a subset of blasts seen in the patient’s diagnostic specimen
(Fig. 5, lower right panel).
Immunophenotyping of Acute Lymphoblastic Leukemia 307
4 Notes
Acknowledgments
References
1. Swerdlow SH, Campo E, Harris NL et al Darzynkiewicz Z, Roederer M, Tanke HJ (eds)
(2017) WHO classification of tumours of hae- Cytometry. New developments, Methods in
matopoietic and lymphoid tissues, 4th edn. cell biology, vol 75, 4th edn. Elsevier, San
IARC, Lyon Diego, CA, pp 559–576
2. Borowitz MJ, DiGiuseppe JA (2014) Acute 10. Wood B (2006) 9-color and 10-color flow
lymphoblastic leukemia/lymphoblastic lym- cytometry in the clinical laboratory. Arch
phoma. In: Orazi A, Weiss LM, Foucar K et al Pathol Lab Med 130:680–690
(eds) Knowles’ neoplastic hematopathology. 11. Keeney M, Wood BL, Hedley BD et al (2017)
Lippincott Williams & Wilkins, Philadelphia, A QA program for MRD testing demonstrates
pp 1019–1029 that systematic education can reduce discor-
3. DiGiuseppe JA (2016) Acute lymphoblastic dance among experienced interpreters. Cyto-
leukemia/lymphoma: diagnosis and minimal metry B Clin Cytom 94:239–249
residual disease detection by flow cytometric 12. DiGiuseppe J, Tadmor MD, Pe’er D (2015)
immunophenotyping. In: Detrick B, Detection of minimal residual disease in B lym-
Hamilton R, Schmitz J (eds) Manual of molec- phoblastic leukemia using viSNE. Cytometry B
ular and clinical laboratory immunology, 8th Clin Cytom 88B:294–304
edn. ASM Press, Washington, D.C., pp 13. Gaipa G, Basso G, Aliprandi S et al (2008)
207–216 Prednisone induces immunophenotypic mod-
4. Wood B, Soma L (2016) Acute myeloid leuke- ulation of CD10 and CD34 in nonapoptotic
mia: diagnosis and minimal residual disease B-cell precursor acute lymphoblastic leukemia
detection by flow cytometry. In: Detrick B, cells. Cytometry B Clin Cytom 74:150–155
Schmitz J, Hamilton RG (eds) Manual of 14. Roshal M, Fromm JR, Winter S et al (2010)
molecular and clinical laboratory immunology, Immaturity associated antigens are lost during
8th edn. ASM Press, Washington, D.C. induction for T cell lymphoblastic leukemia:
5. Borowitz MJ, Bene M-C, Harris NL et al implications for minimal residual disease detec-
(2017) Acute leukaemias of ambiguous line- tion. Cytometery B Clin Cytom 78:139–146
age. In: Swerdlow SH, Campo E, Harris NL 15. McKenna RW, Washington LT, Aquino DB
et al (eds) WHO classification of tumours of et al (2001) Immunophenotypic analysis of
haematopoietic and lymphoid tissues, 4th edn. hematogones (B-lymphocyte precursors) in
IARC, Lyon, pp 180–187 662 consecutive bone marrow specimens by
6. Wood BL (2016) Principles of minimal residual 4-color flow cytometry. Blood 98:2498–2507
disease detection for hematopoietic neoplasms 16. Kantarjian H, Stein A, Gokbuget N et al
by flow cytometry. Cytometry B Clin Cytom (2017) Blinatumomab versus chemotherapy
90:47–53 for advanced acute lymphoblastic leukemia. N
7. Gaipa G, Basso G, Biondi A et al (2013) Detec- Engl J Med 376:836–847
tion of minimal residual disease in pediatric 17. Park JH, Riviere I, Gonen M et al (2018)
acute lymphoblastic leukemia. Cytometry B Long-term follow-up of CD19 CAR therapy
Clin Cytom 84:359–369 in acute lymphoblastic leukemia. N Engl J
8. Borowitz MJ, Devidas M, Hunger SP et al Med 378:449–459
(2008) Clinical significance of minimal residual 18. Cherian S, Miller V, McCullouch V et al (2018)
disease in childhood acute lymphoblastic leu- A novel flow cytometric assay for detection of
kemia and its relationship to other prognostic residual disease in patients with
factors: a Children’s oncology group study. B-lymphoblastic leukemia/lymphoma post
Blood 111:5477–5485 anti-CD19 therapy. Cytometery B Clin
9. Wood B (2004) Multicolor immunophenotyp- Cytom 94B:112–120
ing: human immune system hematopoiesis. In:
310 Joseph A. DiGiuseppe and Jolene L. Cardinali
19. DiGiuseppe JA, Fuller SG, Borowitz MJ acute lymphoblastic leukemia: efficiency of
(2009) Overexpression of CD49f in precursor minimal residual disease (MRD) detection by
B-cell acute lymphoblastic leukemia: potential flow cytometric immunophenotyping com-
usefulness in minimal residual disease detec- pared with commonly used markers. Mod
tion. Cytometry B Clin Cytom 76:150–155 Pathol 89:388A
20. Shinoda-Matsuoka H, DiGiuseppe JA (2009)
Overexpression of CD49f in precursor B-cell
Chapter 17
Abstract
Flow-cytometric demonstration of the typical chronic lymphocytic leukemia (CLL) immunophenotype is
vital for diagnosis. CLL has a characteristic immunophenotype, expressing CD5, CD19, dim CD20, dim
CD22, CD23, bright CD43, dim CD45, dim to negative CD79b, dim CD81, CD200, and dim monoclo-
nal surface immunoglobulin. This characteristic immunophenotype allows a definitive diagnosis and the
ruling out of another leukemia or lymphoma. Flow cytometry also provides important prognostic informa-
tion and accurate assessment of response to therapy. Here we describe optimal specimen collection, red cell
lysis, appropriate panel, cell staining, acquisition on a flow cytometer, and analysis for CLL specimens.
Key words Flow cytometry, Immunophenotype, Chronic lymphocytic leukemia, Minimal residual
disease
1 Introduction
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_17, © Springer Science+Business Media, LLC, part of Springer Nature 2019
311
312 Dalia A. Salem and Maryalice Stetler-Stevenson
a
5 5 5
10 10 10
4 4 4
10 10 10
CD5 V450
CD5 V450
CD5 V450
3 3 3
10 10 10
2 2 2
10 10 10
1 1 1
-10 -10 -10
-101 102 103 104 105 -101 102 103 104 105 -100 102 103 104 105
CD79b PE CD20 AH7 CD22 PerCP Cy55
5
10 105 105
Lambda-m PE
4 4
10 10
4
10
CD38 V450
CD81 FITC
3
10 3 10
3
10
2
10 2
10
102
2
10
1 2 -10
-10
2 3 4 5 2 3 4 5 2
-10 10 10 10 -10 10 10 10 -10 0 102 10
3
10
4
10
5
104 104
CD200 PC7
CD38 V450
104
CD49d PE
103 10
3
3
10
2
10 102
102
2
-10 -102 0.00% 94.08% 2
-10
1 2 3 4 5 2 2 3 4 5 1 2 3 4 5
10 10 10 10 10 -10 10 10 10 10 10 10 10 10 10
CD19 PerCP Cy55 CD5 V450 CD5 APC
Fig. 1 (a) Gated on CD19 positive lymphocytes. Abnormal CLL cells (black) with normal B-cells (gray) in
background. CLL cells are positive for CD5, have bright CD43, and have dim CD79b, CD20, CD22, and CD81.
CD38 is negative. The CLL cells are monoclonal, dim positive for lambda surface light chain and negative for
kappa surface light chain. (b) Gated on abnormal CLL cells. Cells are CD200 positive. Prognostic markers
CD49d (less than 30% positive) and CD38 are negative
cases with a circulating CLL cell count <5 103/μl and recog-
nized nodal, splenic, or other extramedullary involvement. The
differential diagnosis of CLL/SLL primarily includes monoclonal
B-cell lymphocytosis (MBL) and mantle cell lymphoma (MCL).
Flow Cytometry in CLL 313
2 Materials
2.1 Lysing Solution 1. Weigh 8.29 g ammonium chloride (NH4Cl) and 1 g potassium
(1) bicarbonate (KHCO3) and place them into 1000 ml glass or
clear plastic bottle.
2. Add 4 ml 0.5 M EDTA solution, and fill to a volume of
1000 ml with distilled water (H2O). Cap and mix well.
3. Store at room temperature for up to 6 months.
2.2 Wash Solution: 1. Weigh 4 g BSA and add to 4 l of 1 PBS with 0.1% sodium
Phosphate Buffered azide. Cap and mix well by inverting several times. Allow wash
Saline (PBS) with 10% solution to sit at room temperature for 30 min, mix again, and
Bovine Serum Albumin check to be sure the BSA has dissolved.
(BSA) and Sodium 2. Perform pH testing after BSA is in solution and while the
Azide solution is at room temperature. Adjust pH to 7.3–7.5 by
adding small amounts of concentrated HCL (if the pH is
>7.5) or concentrated NaOH (if the pH is <7.3) (see
Notes 2 and 3).
3. Store at 4 C for 2 months from preparation.
314 Dalia A. Salem and Maryalice Stetler-Stevenson
2.3 Fixative: 0.5% 1. Mix 50 ml of 10% buffered formaldehyde with 950 ml of PBS
Formalin Fixative in a bottle, cap and mix well (see Note 4).
Solution 2. Wrap bottle with foil and tape to protect from light. Check PH
and adjust to 7.3–3.5 (see Subheading 2.2).
3. Store at room temperature for up to 1 year.
3 Method
3.1 Cocktails The panel in Table 1 is an example of an 8 color panel that can be
and Panels used to diagnose CLL and monitor disease post therapy. Each
cocktail is made by addition of the appropriate volumes of different
Antibody (Ab) as recommended by the vendor and/or titration.
The amount of the cocktail to be added during staining tube
(Subheading 3.6, step one below) is based on the sum of the
different Ab volumes in this cocktail. The cocktail should be stored
at 4 C and light exposure minimalized. The stability of these
cocktails should be determined by each clinical laboratory.
Table 1
Antibody panel for CLL diagnosis/follow-up
3.3 Pre-lysis, Cell Minimizing sample treatment before staining and timely sample
Count, and Viability processing greatly reduces the risk of losing cells of interest (espe-
Assessment [18, 19] cially in case of CLL MRD) and maintains cell viability and
integrity.
1. Transfer 5 ml of either PB or BM into labelled 50 ml conical
tube(s).
2. Add ammonium chloride lysing Solution (see Subheading 2.1)
to fill each conical tube. Cap and invert several times to mix.
Incubate 10 min room temperature, inverting to mix occasion-
ally and at end.
3. Centrifuge tubes at 500 g for 5 min, check for pellet and
aspirate or decant supernatant into waste container (see Note 5).
4. Cap and vortex tube(s) gently for less than 5 s to dislodge pellet
(s). Fill the tube with PBS, cap, and invert to mix. Repeat
step 3 above (see Note 6).
5. Cap and vortex tube gently to dislodge the entire cell pellet.
Add PBS to the cell pellet according to the pellet size (0.2–2 ml
for small pellets and 3–5 ml for larger pellets), and flick or
vortex gently to mix. Record the final cell suspension volume
to be used later to adjust the cell count.
6. Assess viability and perform a cell count by using a viability dye
that is excluded from viable cells and automated cell counters
or a hemocytometer [20]. Samples with less than 75% viability
should be rejected unless the sample is irreplaceable.
3.4 Cell 1. Divide the total WBCs count (obtained by multiplying the cell
Concentration count in the “count” tube by the dilution factor multiplied by
Adjustment the preparation volume in Subheading 3.3, step 5) by the
target WBCs count to get the amount of the required final
volume. Example: if the total WBCs count ¼ 15.0 106 and
the target count is 20 106/ml, then the final volume should
be 0.75 ml.
2. Adjust the final volume of the cell suspension to reach the
target cell concentration of 20 106 cells/ml (see Note 7).
3.5 Staining Cells 1. Deliver the appropriate volume of the premade cocktail (Sub-
with Antibodies heading 3.1) to the bottom of the labeled 12 75 mm tubes
[18, 19 ] using automatic pipette.
2. Add 100 μl patient cell preparation to each tube and vortex
briefly to mix. Incubate 30 min at room temperature protected
from the light.
3. Wash stained tubes by adding 3–4 ml wash solution to each
tube; vortex to mix then centrifuge at 500 g for 5 min, check
for pellet and aspirate or decant tube into waste container, flick
or vortex tube to dislodge entire cell pellet.
316 Dalia A. Salem and Maryalice Stetler-Stevenson
3.7 Analysis Sequential gating of the list mode data is performed to distinguish
abnormal cells of interest from normal cells based on light scatter
and antigen profile. An example of a typical sequence of hierarchical
analysis gating follows:
1. The FSC-A vs. time plot of ungated cells is evaluated for
discontinuity indicating fluidic disturbances (e.g. bubbles,
clogs, or in the case of low cell number the tube being acquired
dry). A time gate is drawn to exclude such events (Fig. 2).
2. A viable gate is drawn on the time gated SSC-A vs. FSC-A plot
to exclude debris with very low scatter properties (Fig. 2) and is
placed under the time gate.
3. A singlets gate is drawn on the viable gated FSC-H vs. FSC-A
plots to exclude doublets (Fig. 2) and is placed under the
viable gate.
4. Antigen vs. SSC gates are drawn to define T-cells (CD3+) and
B-cells (CD19+) (Fig. 2) and placed under the singlet gate.
5. The antigen based gates are displayed on a FSC-A vs. SSC-A
plot to create a scatter gate that includes all lymphocytes
(Fig. 2). This lymphocyte gate is placed under the singlet gate.
6. The antigen profile of the lymphoid cells is evaluated under the
lymphocyte and B cell antigen gates to identify the CLL immu-
nophenotype (Fig. 1).
7. For MRD assessment, low level CLL involvement must be
detected among a predominately polyclonal B-cell back-
ground. There are two general analysis approaches.
(a) Method 1: Based upon European Research Initiative in
CLL (ERIC) method [2, 16, 21]. The ERIC method
utilizes cells stained with cocktail 2 in Table 1 and a
Boolean gating strategy in which CD19 positive B-cells
are evaluated and a series of analysis gates are created in
areas where normal B-cell populations are not observed
(Fig. 3): A lymphocyte scatter gate that includes all CD19
Flow Cytometry in CLL 317
Fig. 2 Basic analysis for CLL: (a) Time gate. (b) Time gate displayed. Viable gate to exclude debris. (c) Time
gate displayed. Singlets gated to exclude doublets. (d) Gated on Time, Viable, and Singlets gates.
Antigen vs. SSC gates to define T-cells (CD3+) (e) Antigen vs. SSC gates to define B-cells. (f) Gated on
time, viable, and singlets gates. Antigen-based gates are displayed on a FSC-A vs. SSC-A plot to ensure
lymphocyte gate includes all lymphocytes. (g) Final analysis gate: Gated on time, viable, singlets, and
lymphocyte gates
Fig. 3 CLL MRD Method 1: (a) Viable and Lymphocyte gates. (b) CD19+ gate. (c) CD19+CD3– gate. (d–g)
Gated on viable, singlet, lymphocyte and CD19+CD3– gates. The four gates of this method. (h) Minimal
residual CLL. Gated on Viable, Singlet, Lymphocyte, CD19+CD3– gates and gates 1–4
Fig. 4 CLL MRD analysis method 2: (a) Viable and lymphocyte gates. (b) CD19+ gate. (c) Gated on viable,
singlet, lymphocyte, and CD19+ gates. Abnormal CLL cells CD20 dim and CD5+ in ellipse. (d) Gated on viable,
singlet, lymphocyte and CD19+ gates. Abnormal CLL cells CD5+ and homogeneously CD38 negative in
ellipse. (e) Gated on viable, singlet, lymphocyte and CD19+ gates. Abnormal CLL cells in ellipse masked by
polyclonal B cells. (f) Gated on viable, singlet, lymphocyte, CD19+, and abnormal CD5, CD20, and CD38 gates.
Abnormal CLL cells in ellipse are monoclonal, dim positive for kappa and negative for lambda
Flow Cytometry in CLL 319
4 Notes
References
1. Campo E, Ghia P, Montserrat E et al (2017) mature B cell neoplasms. Cytometry B Clin
Chronic lymphocytic leukemia/small lympho- Cytom 50:243–248
cytic lymphoma. In: Swerdlow SH, Campo E, 8. Kampalath B, Barcos MP, Stewart C (2003)
Harris NL, Pileri SA, Jaffe ES, Stein H, Thiele J Phenotypic heterogeneity of B cells in patients
et al (eds) WHO classification of tumours of with chronic lymphocytic leukemia/small lym-
hematopoietic and lymphoid tissues, 4th edn. phocytic lymphoma. Am J Clin Pathol 119
IRAC, Lyon, pp 216–221 (6):824–832. https://doi.org/10.1309/
2. Rawstron AC, Villamor N, Ritgen M et al 4agut3lkeurd7t7k
(2007) International standardized approach 9. Damle R, Wasil T, Fais F et al (1999) Ig V gene
for flow cytometric residual disease monitoring mutation status and CD38 expression as novel
in chronic lymphocytic leukaemia. Leukemia prognostic indicators in chronic lymphocytic
21(5):956–964. https://doi.org/10.1038/sj. leukemia. Blood 94:1840–1847
leu.2404584 10. Durig J, Naschar M, Schmucker U, Renzing-
3. Muller-Hermelink HKME, Catovsky D, Kohler K, Holter A, Huttmann T, Duhrsen U
Campo E, Harris NL, Stien H (2007) Chronic (2002) CD38 expression is an important prog-
lymphocytic leukemia/small lymphocytic lym- nostic marker in chronic lymphocytic leukae-
phoma. WHO classification of hematopoietic mia. Leukemia 16:30–35
and lymphoid tissues. IARC, Lyon 11. Hamblin TJ, Orchard JA, Ibotson RE et al
4. Alapat D, Coviello-Malle J, Owens R et al (2002) CD38 expression and immunoglobulin
(2012) Diagnostic usefulness and prognostic variable region mutations are independent
impact of CD200 expression in lymphoid prognostic variables in chronic lymphocytic
malignancies and plasma cell myeloma. Am J leukemia but CD38 expression may vary dur-
Clin Pathol 137(1):93–100. https://doi.org/ ing the course of the disease. Blood
10.1309/ajcp59uorcyzevqo 99:1023–1029
5. Marti G, Rawstron AC, Ghia P, Hillmen P, 12. Bulian P, Shanafelt TD, Fegan C et al (2014)
Houlston RS, Kay N, Aurran Schleinitz T, CD49d is the strongest flow cytometry-based
Caporaso N, on behalf of The International predictor of overall survival in chronic lympho-
Familial CLL Consortium (2005) Diagnostic cytic leukemia. J Clin Oncol 32(9):897–904.
criteria for monoclonal B-cell lymphocytosis https://doi.org/10.1200/jco.2013.50.8515
(MBL). Br J Haematol 130:325–332 13. Chiorazzi N (2012) Implications of new prog-
6. Ahmad E, Steinberg SM, Goldin L, Hess CJ, nostic markers in chronic lymphocytic leuke-
Caporaso N, Kreitman RJ, Wiestner A, mia. Hematology Am Soc Hematol Educ
Wilson W, White T, Marti G, Stetler-Stevenson Program 2012:76–87. https://doi.org/10.
M (2008) Immunophenotypic features distin- 1182/asheducation-2012.1.76
guishing familial chronic lymphocytic leukemia 14. Gattei V, Bulian P, Del Principe MI et al (2008)
from sporadic chronic lymphocytic leukemia. Relevance of CD49d protein expression as
Cytometry B Clin Cytom 74:221–226 overall survival and progressive disease prog-
7. Kingma DW, Imus P, Xie XY, Jasper G, nosticator in chronic lymphocytic leukemia.
Sorbara L, Stewart C, Stetler-Stevenson M Blood 111(2):865–873. https://doi.org/10.
(2002) CD2 is expressed by a sub-population 1182/blood-2007-05-092486
of normal B cells and is frequently present in
Flow Cytometry in CLL 321
15. Gooden CE, Jones P, Bates R et al (2018) H43-A2, 2nd edn. Clinical and Laboratory
CD49d shows superior performance character- Standards Institute, Wayne
istics for flow cytometric prognostic testing in 19. Stelzer GT, Marti G, Hurley A et al (1997) U.
chronic lymphocytic leukemia/small lympho- S.-Canadian consensus recommendations on
cytic lymphoma. Cytometry B Clin Cytom 94 the immunophenotypic analysis of hematologic
(1):129–135. https://doi.org/10.1002/cyto. neoplasia by flow cytometry: standardization
b.21384 and validation of laboratory procedures. Cyto-
16. Rawstron AC, Kennedy B, Evans PA et al metry 30(5):214–230
(2001) Quantitation of minimal disease levels 20. Cadena-Herrera D, Esparza-De Lara JE,
in chronic lymphocytic leukemia using a sensi- Ramirez-Ibanez ND et al (2015) Validation of
tive flow cytometric assay improves the predic- three viable-cell counting methods: manual,
tion of outcome and can be used to optimize semi-automated, and automated. Biotechnol
therapy. Blood 98:29–35 Rep (Amst) 7:9–16. https://doi.org/10.
17. Moreton P, Kennedy B, Lucas G et al (2005) 1016/j.btre.2015.04.004
Eradication of minimal residual disease in 21. Rawstron AC, Bottcher S, Letestu R et al
B-cell chronic lymphocytic leukemia after (2013) Improving efficiency and sensitivity:
alemtuzumab therapy is associated with pro- European research initiative in CLL (ERIC)
longed survival. J Clin Oncol 23 update on the international harmonised
(13):2971–2979. https://doi.org/10.1200/ approach for flow cytometric residual disease
jco.2005.04.021 monitoring in CLL. Leukemia 27
18. Wayne P (2007) Clinical flow Cytometric anal- (1):142–149. https://doi.org/10.1038/leu.
ysis of neoplastic Hematolymphoid cells; 2012.216
approved guideline, 2nd ed. CLSI document
Chapter 18
Abstract
Paroxysmal nocturnal hemoglobinuria (PNH) is a rare but often debilitating disease which may lead to
death in up to 35% of patients within 5 years if unrecognized and untreated. Detection of PNH and
assessment of PNH clone size in RBC and WBC lineages by flow cytometric analysis has increased in
importance due to the availability of novel therapies. These therapies typically block the hemolysis of red
blood cells and thus significantly lower the morbidities and mortality associated with this disease. This
chapter describes validated, state-of-the-art, high-sensitivity flow cytometric methodologies based on latest
published testing guidelines for PNH.
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6_18, © Springer Science+Business Media, LLC, part of Springer Nature 2019
323
324 Andrea J. Illingworth et al.
2 Materials
Table 1
Recommended CD235a-FITC and CD59-PE conjugates for high-sensitivity PNH RBC assay
3 Methods
3.1 Instrument 1. Standard setup instructions from both major manufacturers are
Setup; Key Points optimized for specific applications and do not transfer directly
to PNH testing in either red or white cell assays.
328 Andrea J. Illingworth et al.
Table 2
Recommended clones/conjugates for high-sensitivity detection of PNH WBC on Beckman Coulter
cytometers
Table 3
Recommended clones/conjugates for high-sensitivity detection of PNH WBC on Becton Dickinson
cytometers
Table 4
Recommended clones/conjugates for cross-platform three-laser B-C Navios/BD Canto II High
sensitivity PNH WBC 5-, 6-, and 7-color assays
3.2 Sample For instruments that do not automatically save “TIME” as a param-
Preparation eter, this parameter must be collected so that fluidics can be moni-
and Instrument Setup tored if needed. Instrument setup for the RBC assay is performed
for RBC Analysis using a fresh normal blood sample, diluted 1:100 with clean Phos-
phate buffered saline (PBS). Light scatter voltages, photomultiplier
tube voltages (PMTv) and compensation are optimized as detailed
in several previous publications [18, 23, 24, 28], and recent exam-
ples can be viewed in Figs. 1 and 2 of [24]).
3.3 Recommended Anti-coagulated (EDTA is preferred, but heparin can also be used)
Staining Protocol peripheral blood samples should be less than 48 h old. If possible,
for RBC these samples should be kept refrigerated for this may extend the
stability of these samples.
330 Andrea J. Illingworth et al.
3.4 Sample For instruments that do not automatically save “TIME” as a param-
Preparation eter, this parameter must be collected so that fluidics can be moni-
and Instrument Setup tored if needed. To analyze white blood cells, 100 μl of
for WBC Analysis anticoagulated peripheral blood is stained with the reagent cocktail
of choice and subject to an RBC lysis step. Regardless of lysing agent
used, the cells need to be washed thereafter with phosphate buffered
saline supplemented with 1% bovine serum albumin before acquisi-
tion. A multistep Boolean gating approach has been adopted to
efficiently gate neutrophils and monocytes that begins with setting
light scatter voltages in linear mode to ensure that all cells of interest
(neutrophils and monocytes for PNH “clone” detection and lym-
phocytes used as internal staining controls) are properly on-scale and
above the threshold/discriminator, used to remove subcellular
debris. Light scatter voltages and PMTv are optimized as detailed
in several previous publications [18, 23, 24, 28].
Immunophenotyping of PNH 331
3.5 Establishing Light scatter voltages, photomultiplier tube voltages (PMTv) and
the Compensation compensation for the PNH WBC assay are optimized as detailed in
Matrix several previous publications [18, 23, 24, 28] and examples are
shown in Figs. 6 and 7 for a FLAER-based 6-color assay on a
10-color Navios cytometer.
3.6 Recommended All individual antibodies were verified for appropriate reactivity
Staining Protocol with target cells and titrated to optimize specific staining perfor-
for Five- and Six-Color mance prior to being cocktailed for use in the single tube five- or
WBC Assays six-color assays. It is recommended that samples be stained fresh or
(Neutrophils after a maximum storage time of 48 h at 4 C [19, 23, 24].
and Monocytes) 1. Using reverse-pipetting, pipet 100 μl of fresh peripheral blood
carefully into the bottom of a test tube without touching the
side of the tube.
2. Add an appropriate volume of premixed and validated cocktail
directly to the blood aliquot in the bottom of the tube and mix
very gently but thoroughly as described above for the RBC
assay.
3. Incubate for 20 min in the dark (incubation times for up to
60 min have been validated).
4. Lyse the RBCs using an appropriate lysing agent. Immuno-
Prep, VersaLyse (BC), FACSLyse (BD) or ammonium
chloride-based lysing agents following the manufacturer’s
recommendations. While all are acceptable, those containing
fixatives may help retain cellular integrity better than those that
do not.
5. After lysing, wash cells once with PBS, resuspend in 1 ml of
PBS and acquire on the cytometer. Note: Samples should be
acquired immediately, as delays can cause light-scatter changes,
especially when fixative-free lysing agents are used.
6. Acquire a minimum of 50,000 neutrophils and 5000 mono-
cytes in listmode for clinical test samples.
7. If small numbers of GPI-deficient cells are observed, acquisi-
tion times should be increased until a statistically reliable num-
ber of “PNH phenotypes” are acquired (20–50 cells).
4 Analysis
4.1 RBC Analysis For high-sensitivity RBC analysis, most previously published
Guidelines recommend the use of CD235a-FITC for lineage-
specific RBC gating and CD59-PE to detect GPI-deficient cells
[18, 24]. RBCs are analyzed by a series of gating dot plots and
diagnostic dot plots:
1. TIME (x axis) versus SS log (y axis) dot plot. TIME is collected
as a parameter (as is recommended for all clinical flow assays)
and monitored during acquisition so that if fluidics problems
are encountered, the sample can be reacquired if possible, or if
not, data acquired prior to the fluidics hiatus can be “gated”
and only that portion of the data file subsequently analyzed
(Fig. 1, top row left).
2. FS log (x axis) versus SS log (y axis) dot plot to gate out debris
(Fig. 1, top row middle). It is important to adjust the threshold
(discriminator) for the forward scatter so that no RBCs are
excluded from acquisition. Make sure that gate includes the
RBC cluster and excludes any cellular debris on the left.
3. CD235a-FITC (x axis) versus FS (y axis) dot plot to gate singlet
RBCs and to quantify and exclude any remaining RBC aggre-
gates (Fig. 1, top row right).
4. CD59-PE (x axis) versus CD235a-FITC (y axis) dot plot (Fig. 1,
bottom row left). Check the Type III area for cells, which are
CD235a brightly positive/negative for CD59 and record the
percentage. Check the Type II area for cells, which are also
brightly positive for CD235a with intermediate CD59 expres-
sion and record the percentage.
5. CD59-PE (x axis) versus CD235a-FITC (y axis) density plot
(Fig. 1, bottom row middle). Use this plot if the separation is
not clear based on the previous dot plot.
6. CD59-PE single parameter histogram (Fig. 1, bottom row
right). Bivariate dot plots and/or density plots are typically
recommended over single-parameter histograms, especially
Immunophenotyping of PNH 333
Fig. 1 Normal patient with no PNH RBC clone. Sample stained with CD235a-FITC/CD59-PE
4.2 Separation In most cases the PNH clusters are evident and the initial region
Between Normal Type settings can be used for the quantification of the Type II and Type
I RBC, PNH Type II III PNH RBC based on a combination of bivariate dot plots,
and PNH Type III RBCs density plots and the single parameter CD59 histogram (Fig. 2).
However, in some patients the regions may need to be adjusted
(dynamic region setting) based on slight patient-specific differences
in the CD59 expression rather than strictly adhering to the initial
region setting. Occasionally there is no clear distinction (clustering)
between PNH Type II and PNH Type III or between PNH Type II
and normal Type I cells which renders the interpretation
334 Andrea J. Illingworth et al.
Fig. 2 Patient with PNH RBC Type II and PNH RBC Type III clone. Sample stained with CD235a-FITC/CD59-PE
Fig. 3 PNH sample containing 94.2% PNH neutrophils and 94.8% PNH monocytes (data not shown) stained
with CD235aFITC/CD59-PE/CD71-APC
Fig. 4 PNH sample containing 97.3% PNH neutrophils and 99.6% PNH monocytes (data not shown) stained
with CD235aFITC/CD59-PE/CD71-APC
large-clone PNH cases, Type III PNH iRBCs are the predominant
PNH population although striking examples of predominantly
Type II PNH iRBCs can also be observed (Fig. 5).
This study has also shown that if analysis of the red cell lineage
is to be clinically useful, it needs to be accurate. For large-clone
PNH cases, accurate data cannot be derived from mature RBCs
(due to transfusion and hemolysis). For large-clone PNH cases,
accurate data can be derived only from immature RBCs (iRBCs).
For small clone PNH cases (clone sizes <1%) and BMFS cases
containing small numbers of PNH RBC phenotypes, it is difficult
to collect enough iRBCs for statistically precise data. However, in
the latter, sufficient mature PNH RBCs can usually be identified to
generate reliable and precise data.
338 Andrea J. Illingworth et al.
Fig. 5 PNH sample containing 99.25% PNH neutrophils (top right plot) and 99% PNH monocytes (top left plot)
stained with CD235aFITC/CD59-PE/CD71-APC700
Fig. 6 FLAER/CD24/CD14-based WBC assay with no PNH clone in neutrophils and monocytes. Sample stained
with FLAER-Alexa 488/CD24-PE/CD15-PC5/CD64-PC7/CD14-APC700/CD45-KrO
Fig. 7 FLAER/CD24/CD14-based WBC assay with PNH clone present in neutrophils and monocytes. Sample
stained with FLAER-Alexa 488/CD24-PE/CD15-PC5/CD64-PC7/CD14-APC700/CD45-KrO
Fig. 8 CD157 and FLAER-based cross-platform assay with PNH clone present in neutrophils and monocytes.
Sample stained with FLAER-Alexa 488/CD157-PE/CD15PerCPeFluor710/CD64-PECy7/CD45-eFluor450
Fig. 9 Minor clone in all three lineages showing a quantifiable PNH clone (>50 PNH cells)
346 Andrea J. Illingworth et al.
6 Reporting
7 Assay Validation
8.1 Reagent Since publication of the 2010 Guidelines [16] a large number of
Selection specific clones/conjugates have been assessed for lineage-specific
gating and detection of GPI-deficient neutrophils and monocytes.
1. Optimal titration ranges must be established for selected con-
jugates and validated both individually and in combination
with other reagents on both normal and PNH samples.
2. Using this information, high-sensitivity assays were developed
using four-color cocktails based on FLAER, CD24, CD15 and
CD45 (for neutrophils) and FLAER, CD14, CD64 and CD45
(for monocytes) and (Supplementary Data, 23, 24).
3. Platform-specific versions of these robust four-color assays
were developed for clinical cytometers with four or more
PMTs [18, 23] and were capable of successful deployment
beyond the laboratories of original developers [39]. These
assays and reagent sets were found to be highly reliable across
FC500 and FACSCalibur instrument platforms in Interna-
tional studies performed under the auspices of the UK
NEQAS organization using whole blood stabilized
samples [40].
350 Andrea J. Illingworth et al.
8.2 Instrument- 1. Note that cocktailing of reagents is very important for WBC
Specific Reagent assays, just as it is for the RBC assay. If reagents are added one
Cocktails for Four-, at a time, there is increased risk of failing to add individual
Five-, and Six-Color reagents, or adding too much or too little, especially where
WBC Assays very small volumes of some individual reagents need to be
accurately pipetted. Such errors run the risk of generating
“laboratory-induced PNH” if the GPI-specific reagents are
inadvertently omitted. For laboratories that run the assay infre-
quently, there is an increased requirement to ensure that the
reagents used in the assay are working as specified. In this
setting, it is recommended that the laboratory stain a normal
sample at least once per month to validate the performance of
all reagents used in the assay [18, 22].
2. As detailed elsewhere, the Practical Guidelines [18] defined
two 4-color reagent cocktails for the high sensitivity detection
of PNH neutrophils and PNH monocytes for both Beckman
FC500 and BD FACSCalibur platforms. Subsequent studies
using these assays/reagent sets have shown them to be repro-
ducible and robust using fresh samples across multiple labora-
tories [39] and in International studies performed with
stabilized whole blood samples under the auspices of the UK
NEQAS organization [40].
Immunophenotyping of PNH 351
References
15. Sugimori C, Chuhjo T, Feng X et al (2006) assay in the primary screening of PNH clones.
Minor population of CD55-CD59- blood cells Am J Clin Pathol 132:564–572
predicts response to immunosuppressive ther- 26. Dalal BI, Khare NS (2013) Flow cytometric
apy and prognosis in patients with aplastic ane- testing for paroxysmal nocturnal hemoglobin-
mia. Blood 107:1308–1314 uria: CD64 is better for gating monocytes than
16. Borowitz MJ, Craig FE, DiGiuseppe JA et al CD33. Cytometry B Clin Cytom 84:33–36
(2010) Guidelines for the diagnosis and moni- 27. Sutherland DR, Ortiz F, Quest G et al (2018)
toring of paroxysmal nocturnal hemoglobin- High-sensitivity 5-, 6-, and 7-color PNH WBC
uria and related disorders by flow cytometry. Assays for both Canto II and Navios Platforms.
Cytometry B Clin Cytom 78B:211–230 Cytometry B Clin Cytom 94:637–651
17. Killick SB, Bown N, Cavenagh J et al (2016) 28. Keeney M, Illingworth A, Sutherland DR
Guidelines for the diagnosis and management (2017) Paroxysmal Nocturnal Hemoglobin-
of adult aplastic anaemia. Br J Haematol uria assessment by flow cytometric analysis.
172:187–207 Clin Lab Med 37:855–867
18. Sutherland DR, Keeney M, Illingworth A 29. Sutherland DR, Richards SJ, Ortiz, F et al.
(2012) Practical guidelines for the high- Addition of CD71 to high-sensitivity
sensitivity detection and monitoring of parox- CD235a/CD59 PNH RBC assay allows the
ysmal nocturnal hemoglobinuria clones by flow additional accurate delineation of PNH Type
cytometry. Cytometry B Clin Cytom III, PNH Type II and normal (Type I) nucle-
82:195–208. and Supplementary Data ated RBCs: Cytometry B 2019, in press
19. Illingworth A, Marinov I, Sutherland DR et al 30. Hernández-Campo PM, Almeida J, Sánchez
(2018) ICCS/ESCCA consensus guidelines to ML et al (2006) Normal patterns of expression
detect GPI-deficient cells in PNH and related of glycosylphosphatidylinositol-anchored pro-
disorders part 3—data analysis, reporting and teins on different subsets of peripheral blood
case studies. Cytometry B Clin Cytom cells: A frame of reference for the diagnosis of
94:49–66. and Supplementary Data paroxysmal nocturnal hemoglobinuria. Cyto-
20. Oldaker T, Whitby L, Saber M et al (2018) metry B Clin Cytom 70:71–81
ICCS/ESCCA Consensus Guidelines for the 31. Sutherland DR, Acton E, Keeney M et al
Flow Cytometry Testing of Patients with sus- (2014) Use of CD157 in FLAER-based assays
pected Paroxysmal Hemoglobinuria (PNH) for high-sensitivity PNH neutrophil and PNH
and related Disorders Part 4 - Assay Validation monocyte detection. Cytometry B Clin Cytom
and quality assurance. Cytometry B Clin 86:44–55
Cytom 94:67–81 32. Owens MA, Vall HG, Hurley AA, Wormsley SB
21. Illingworth A, Oldaker T, Sutherland DR et al.: (2000) Validation and quality control of immu-
Verification of PNH assay sensitivity through nophenotyping in clinical flow cytometry. J
spiking experiment. (2018). https://www. Immunol Methods 243:33–50
cytometry.org/web/modules/Module10.pdf 33. International Organization for Standardization
22. Davis BH, Keeney M, Brown R et al (2014) (ISO) Statistics-Vocabulary and Symbols. ISO
CLSI H52-A2 red blood cell diagnostic testing 3534-1. Geneva; 1993
using flow cytometry; approved guideline, 2nd 34. https://www.iso.org
edn. Clinical and Laboratory Standards Insti-
tute, Wayne, PA. ISBN Number: 1-56238- 35. Libeer J-C (2001) Role of external quality
957-2 assurance schemes in assessing and improving
quality in medical laboratories. Clin Chim Acta
23. Sutherland DR, Richards SJ, Keeney M et al 309:173–177
(2015) High-sensitivity detection of PNH red
blood cells, red cell precursors and white blood 36. CLSI EP12-A2 (2008) User Protocol for Eval-
cells. Curr Protoc Cytom 72(Unit 6.37):1–29 uation of Qualitative Test Performance- Sec-
ond Edition – Approved Guideline. National
24. Sutherland DR, Illingworth A, Marinov I et al Committee for Clinical Laboratory Standards,
(2018) ICCS/ESCCA Consensus Guidelines Wayne, PA
to detect GPI-deficient cells in Paroxysmal
Nocturnal Hemoglobinuria (PNH) and related 37. Clinical and Laboratory Standards Institute
disorders part 2—reagent selection and assay (2004) Protocols for Determination of Limits
optimization for high-sensitivity testing. Cyto- of Detection and Limits of Quantitation
metry B Clin Cytom 94:23–48 Approved Guideline. CLSI document EP17.
CLSI, Wayne, PA
25. Sutherland DR, Kuek N, Azcona-Olivera J et al
(2009) Use of FLAER-based white blood cell 38. New York State Department of Health (2011)
Clinical Laboratory Evaluation Program, Assay
354 Andrea J. Illingworth et al.
A E
Aberrant marker expression................................. 287, 289 Effector Tregs (eTreg) ........................142, 146, 152, 156
Acoustic cell enrichment............................................... 214 Endothelial ........................................................... 203–210
Acute leukemia ....................................293, 297, 301, 308 Expansion ............................................................ 116, 183,
Acute lymphoblastic leukemia (ALL) ................. 297–308 186, 324, 339
Acute myeloid leukemia (AML)..................................254,
281–293, 297, 324 F
Adipose tissue (AT).............................................. 115–125 Fetal calf serum (FCS) ............................ 71, 77, 182, 298
Analysis ................................................................ 2, 31, 53, FLAER................................................................. 327–329,
69, 81, 106, 122, 130, 143, 185, 194, 204, 213,
331, 336, 339–345, 349–351
228, 282, 298, 316, 324 Flow cytometric analysis ....................................... 53, 106,
Antibodies ........................................................... 2, 3, 6, 7, 130, 193–200, 285
16, 19, 20, 26, 31, 53, 69, 89, 105, 116, 131, 144,
Flow cytometry (FC) .................................................1, 31,
181, 195, 203, 215, 229, 282, 298, 317, 325 53, 69, 81, 106, 123, 130, 142, 185, 194, 203,
Aplastic anemia (AA) .................................. 324, 325, 339 215, 228, 282, 297, 311, 324
Fluorescence ........................................................ 1–27, 53,
B
64, 71, 82, 85, 87, 88, 91, 93, 95, 97, 98, 106,
Body mass index (BMI)...............................115, 123–125 107, 122–124, 143, 144, 152, 154, 156–158,
160–164, 166, 169, 172, 173, 182, 185, 190,
C 200, 209, 210, 223, 235, 240, 244, 249, 259,
Cardiovascular (CV) ..................................................... 115 260, 264, 267, 274, 293, 307, 316
Fluorescence minus one (FMO)
CD59 ................................................................... 325, 326,
330, 332–334, 336–338, 346 controls ........................................... 16, 19, 21, 23,
CD127...............................................................5, 90, 142, 24, 26, 97, 154–157, 200, 224, 240, 259
144–148, 152, 154, 156, 157, 160–162, 165, Fluorescent cell barcoding (FCB) ............................53–68
FoxP3.................................................... 90, 142, 144–148,
168, 181, 186, 191
Chronic lymphocytic leukemia (CLL) ................ 311–320 152, 154–156, 159–163, 165, 168, 182
Circulating tumor cells (CTCs) .......................... 213–225 Free fatty acids (FFAs) .................................................. 116
Color compensation.....................................................247,
G
252–258, 262, 276
Compensation .............................................. 3, 32, 59, 71, Gating .................................................................. 2, 45, 55,
93, 107, 112, 122, 133, 143, 198, 205, 229, 329 77, 81, 107, 122, 136, 144, 185, 197, 204, 218,
Cytokines ..............................................39, 84, 91, 94, 97, 265, 282, 303, 316, 332
112, 116, 141, 180, 182–183, 186, 188, 191, 324 Germinal center (GC).......................................... 118, 120
Cytometry by time of flight (CyTOF)......................1, 44, Glycophosphatidyl inositol-anchored protein
107, 110 (GPI-AP) ........................................................... 323
D H
Dendritic cells (DCs) .............................................. 70, 91, Hanks’ balanced salt solution (HBSS)................ 117, 118
98, 193–200, 286, 342 Harmonization ....................................................... 70, 146
Dried antibody cocktails ...........................................70, 77 High-dimensional ..........................................1–27, 44, 97
Dulbecco’s modified Eagle’s medium Human................................................................. 4, 39, 70,
(DMEM).......................................... 117, 118, 120 81, 106, 116, 130, 142, 180, 205
J. Philip McCoy, Jr (ed.), Immunophenotyping: Methods and Protocols, Methods in Molecular Biology, vol. 2032,
https://doi.org/10.1007/978-1-4939-9650-6, © Springer Science+Business Media, LLC, part of Springer Nature 2019
355
IMMUNOPHENOTYPING: METHODS AND PROTOCOLS
356 Index
I Phenotyping ............................................................ 53, 56,
70, 105–113, 120, 213–225
Immunofluorescence .................................................... 110 Plasmacytoid dendritic cells (pDCs) ...........193–200, 286
Immunology.............................................. 1, 12, 101, 130 Polychromatic................................. 19, 20, 130, 242, 244
Immunophenotype ................................................. 32, 39, Proficiency testing ..........................................83, 264, 270
194, 281, 288–291, 297, 304, 311, 313, 316
Immunophenotyping............................................. v, 1–27, Q
31–50, 53–78, 89, 107, 115–125, 141–173,
179–191, 203–210, 227–278, 281–293, Quality assurance (QA).......................228, 259, 270, 277
297–308, 311, 323–351 Quality control (QC)............................................... v, 2, 3,
Immunoprofiling........................................................... 106 6, 20, 85, 87, 101, 154, 221–223, 227–278
Induced regulatory T cells (iTregs).............................. 142
R
Innate lymphoid cells (ILCs) .............................. 179–191
Insulin resistance (IR).......................................... 115, 116 Reactive oxygen species (ROS) .................................... 116
Isolation ................................................................. 55, 111, Red blood cells (RBCs) .......................................... 40, 55,
117–121, 181, 183–184, 189, 205, 207, 213, 214 111, 130, 133, 147, 149, 150, 159, 190, 195,
208, 214, 215, 217, 218, 224, 229, 230, 232,
L 233, 241, 271, 277, 282, 284, 286, 325–337,
Levey–Jennings (L-J) ........................................... 246, 247 339, 340, 342, 343, 346, 347, 350, 351
Lyophilization ........................................v, 70–73, 75, 146 Regulatory T cells (Tregs) .......................................70–72,
90, 141–147, 151, 152, 154–158, 160–162, 165
M Room temperature (RT) .................................... 6, 18, 20,
39, 40, 42, 43, 55–60, 120, 121, 124, 130, 133,
Magnetic depletion .............................................. 214, 218 181, 184–186, 190, 191, 194–196, 204–208,
Mass cytometry (MC)...................................................v, 1, 216–223, 230, 231, 272, 283, 284, 298, 301,
31–50, 81, 106, 107, 110 302, 313–315, 317, 330
Memory T cells..................................................... 131, 137
Microparticles (MPs) .....................................55, 203–210 S
Minimal residual disease (MRD)........................ 267, 282,
285, 289, 297–299, 301, 302, 304–308, 313, Spillover spreading error matrix (SSM) .......................... 6,
315, 316, 318 12–17, 26
Molecular indexing ....................................................... 223 Staining ................................................................ 4, 33, 53,
Monoclonal antibodies (mAb) ................................ v, 132, 69, 82, 108, 118, 131, 143, 182, 197, 206, 214,
134, 147, 149–151, 155, 160–164, 167–169, 231, 285, 307, 314, 327
173, 181–183, 195–198, 229, 233, 283–285 Stromal vascular fraction (SVF) .......................... 117–123
Multicolor flow cytometry.......................... 153, 198, 262 Subcutaneous adipose tissue (SAT) .................... 116–124
Multiplexing .................................................................. 110 Suppressor T cells.......................................................... 251
Myelodysplastic syndrome (MDS)............. 324, 325, 339
T
N T cells4, 67, 70, 83, 117, 129, 142, 179, 193, 251, 303,
Naive T cells ................................................ 129, 131, 137 311, 323
Natural killer (NK) cells...................................... 4, 70, 93, Toll-like receptor (TLR) ............................. 118, 122, 123
98, 105–113, 116, 117, 120, 137, 180, 193, 251, Transcription factors (TF) .................................. 117, 125,
265, 317, 342 142, 163, 180, 182, 185, 186, 189, 191
Natural Tregs (nTregs) ........................................ 141, 153 Type-2 diabetes (T2D) ................................................. 115
P W