Download White Paper - NanoString

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the work of artificial intelligence, which forms the content of this project

Document related concepts

Drosophila melanogaster wikipedia , lookup

Lymphopoiesis wikipedia , lookup

T cell wikipedia , lookup

DNA vaccination wikipedia , lookup

Monoclonal antibody wikipedia , lookup

Immune system wikipedia , lookup

Adaptive immune system wikipedia , lookup

Molecular mimicry wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Psychoneuroimmunology wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Innate immune system wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Immunomics wikipedia , lookup

Transcript
W H I T E PA PE R
Uncovering the TumorImmune System
Interaction
Using the nCounter®
Vantage 3D™ RNA:Protein
Immune Cell Assays
Lucas Dennis, Gokhan Demirkan, Brian Filanoski,
Brian Birditt, Celine Ngouenet, Irena Pekker, and Gary Geiss.
FOR RESEARCH USE ONLY. Not for use in diagnostic procedures.
Contents
Introduction 3
Digital Measurement of Gene and Protein Expression
4
Investigating the Cancer Immunity Cycle with nCounter Vantage 3D RNA:Protein Immune Cell Profiling
5
Correlation of nCounter Protein Measurements with Flow Cytometry
6
Precise, Simultaneous Quantification in Multiplexed Format
7
Cell Profiling Application: Dynamic View of Leukocyte Differentiation
8
Cell Signaling Application: Interferon Gamma Signaling
9
Conclusion
10
Appendix A: Immune Cell Types and Genes Included in
the nCounter Vantage 3D RNA:Protein Immune Cell Profiling Assay
12
Appendix B: Proteins Included in the nCounter Vantage 3D RNA:Protein Immune Cell Profiling Assay
13
Appendix C: Proteins Included in the nCounter Vantage 3D RNA:Protein Immune Cell Signaling Assay
14
WHITE PAPER
Uncovering the Tumor-Immune System
Interaction Using the nCounter® Vantage
3D™ RNA:Protein Immune Cell Assays
Lucas Dennis, Gokhan Demirkan, Brian Filanoski, Brian Birditt, Celine Ngouenet, Irena Pekker, and Gary Geiss.
Introduction
This white paper describes the first RNA:Protein reagents
for profiling the human immune response to tumors and
The ability of mutated cells to give rise to cancer relies upon
for developing new therapeutic approaches and predictive
their capability to interact with cells of the immune system
biomarker
and ultimately evade immune recognition, suppress immune
response. The nCounter® Vantage 3D™ RNA:Protein Immune Cell
signatures
for
immunotherapeutic
treatment
activity, and persist in a chronically inflamed environment .
Assays are highly multiplexed and designed to quantitate mRNA
Many
tumor
and proteins using low levels of sample input. These assays
that
detect 770 mRNAs and up to 30 proteins and are powered by
1,2
immune
cell
microenvironment,
types
are
creating
a
found
complex
in
the
milieu
3-5
affects the growth and evolution of cancerous cells
Robust
immune
monitoring
assays
are
essential
.
for
3D Biology™ technology for simultaneous detection of multiple
analyte types.
characterizing the immune status of cancer patients, and
insights gained from studying the immune response in
treated
patients
will
help
transition
promising
Applications for these products include:
research
1. Identification and quantification of immune cells,
such as those infiltrating a tumor or in a population of
peripheral blood mononuclear cells (PBMCs).
Tremendous growth in the understanding of the immune
2. Assessment of immunological function and response
to immuno-modulation, such as checkpoint inhibitors.
into the clinic.
response to cancer is beginning to generate breakthroughs in
cancer treatment6. However, our understanding of immunooncology is far from complete. There is a need for tools that
better enable researchers to explore the complex relationship
between the immune system and primary tumors—in both the
3. Identification and quantification of tumor-specific
antigens, such as cancer-testis (CT) antigens.
4. Longitudinal monitoring of signaling response to
treatments.
tumor microenvironment and in peripheral blood—as well as to
robustly monitor changes in the immune response associated with
A systems biology approach capable of capturing the functional
potential therapeutic approaches. Transcriptionally active genes
behavior of multiple cell types that interact within the human
and their respective protein products are important molecular
immune system and tumor microenvironment is necessary
indicators of the physiological state of cell populations . As such,
to provide a comprehensive understanding of the complex
expression profiling of genomic and proteomic markers is critical
mechanisms responsible for tumor immune responses and
for identifying relevant immune resistance mechanisms in the
tolerance. Biomarkers will allow for optimization of potential
tumor microenvironment and developing predictive biomarkers
immuno-oncology therapeutics providing mechanistic and
7
8,9
for active immunotherapy .
patient selection information.
3
WHITE PAPER
Digital Measurement of Gene and Protein Expression
Specific hybridization of fluorescent barcodes to nucleic acid targets in solution is the foundation of NanoString’s single-molecule
detection technology10. Over 1000 publications have shown this technology is reliable for gene expression analysis. Recently, this
technology was adapted by Ullal et al. (2014; Sci Transl Med) to enable detection of proteins via oligonucleotide-labeled antibodies
(FIGURE 1)11. NanoString has further refined this technique by combining DNA, RNA, and protein detection into a single assay, referred to
as 3D Biology. Focused RNA:Protein profiling enables specific, multiplexed detection of up to 30 protein targets in conjunction with up
to 770 mRNA targets for quantitative assessments from limited samples (APPENDIX A). Additionally, as both protein and nucleic acid
measurements are provided as digital nCounter® counts, data analysis and comparison of mRNA and protein expression is simplified.
1. Collect cells
3. Stain cells with antibodies
2. Add antibodies with DNA oligo tags
4. Use nCounter to count RNA and protein
FIGURE 1 Illustration of mRNA and protein detection and measurement using nCounter technology. Antibodies are covalently attached to unique DNA tags to enable their
detection by nCounter probes. RNA and protein fractions are combined in a single hybridization reaction for simultaneous RNA:Protein profiling.
4
NanoString® Technologies, Inc.
Investigating the Cancer Immunity Cycle
with nCounter Vantage 3D RNA:Protein Immune Cell Profiling
In their seminal review, Chen and Mellman describe a series of steps (FIGURE 2) that must be initiated and fostered for an anticancer
immune response to effectively kill cancer cells, dubbing these steps the Cancer Immunity Cycle12. This cycle begins with the release
of antigens by cancer cells. Released antigens are processed and then presented, which results in priming and activation of the
adaptive immune response. Once activated, T cells traffic to the tumor, infiltrate, and recognize tumor cells, ultimately leading to their
programmed destruction. When cancer cells are killed by invading T cells, additional antigens are released into the periphery and
start the cycle anew.
Proteins in the nCounter Vantage 3D RNA:Protein
Immune Cell Profiling Assay
Stage of Cancer
Immunity Cycle
Proteins in the nCounter
Vantage 3D RNA:Protein
Immune Cell Profiling
Assay
(2) Antigen Presentation
CD4, CD40, CD154 (CD40L)
(3) Priming and Activation
CD279 (PD-1), CD274 (PD-L1), CD273
(PD-L2), CD25 (IL2R), CD56 (NCAM),
CD357 (GITR), CD137 (OX40), CD27,
CD28, CD127, CD137 (4-1BB)
(4-5) Trafficking and Infiltration CD9
(6-7) Recognition and Killing of CD279 (PD-1), CD274 (PD-L1), CD273
(PD-L2), CD272 (BTLA), HLA-DRA
Cancer Cells
Immune Modulation
CD279 (PD-1), CD278 (ICOS), CD158e1
(KIR3DL1), CD335 (NKp46), CD152
(CTLA-4), CD3 (CD3E), CD8 (CD8A),
CD14, CD19, CD33, CD68, CD163
FIGURE 2 Illustration of the Cancer Immunity Cycle and the associated proteins with each stage that are included in the nCounter Vantage 3D RNA:Protein Immune Cell
Profiling Assay. Illustration used with permission and originally published in: Chen, D.S, Mellman, I. (2013), author update (2015) Oncology Meets Immunology: the CancerImmunity Cycle. Immunity 39(1):1-10.
Movement through this cycle is controlled by a key set of master regulators, collectively known as checkpoint regulators13. The
nCounter Vantage 3D RNA:Protein Immune Cell Profiling Assay allows monitoring of these key regulatory elements via both gene
expression and extracellular proteomic markers (FIGURE 2). Many of the steps of the cancer-immunity cycle are therapeutically
exploitable as shown by the significant clinical benefit observed with checkpoint inhibitors (such as anti-PD-1/PD-L1 and
CTLA-4)14 in patients with melanoma and other solid tumors15.
Identifying and measuring discrete cell populations within heterogeneous samples has been a focus of immunological study for
decades. The classification and enumeration of immune cells via histopathological and flow cytometric analyses within tumor
samples and in the peripheral blood has been shown to be a significant and powerful predictor of patient survival16,17. By including
cell subset-specific surface markers as part of the assay (APPENDIX B), the nCounter Vantage 3D RNA:Protein Immune Cell
Profiling Assay can efficiently measure both cell types and functions as well as follow their changes under therapeutic pressure.
5
WHITE PAPER
Correlation of nCounter Protein Measurements with Flow Cytometry
We compared the digital protein counts to flow cytometry, a commonly used technology for measuring protein expression in cell
suspensions. To characterize and validate the nCounter protein detection technology, three different samples were produced by
transfecting HEK293T cells with ten different expression plasmids that each correspond to proteins recognized by the nCounter
Vantage 3D RNA:Protein Immune Cell Profiling Assay. Each sample was transfected with varying amounts of plasmid DNA, creating
a mixed population of cells expressing the target proteins.
Protein expression was measured in single-plex assays using flow cytometry and in a multiplexed assay using nCounter
(FIGURE 3). The percentage of cells expressing each protein in flow cytometry and the digital counts of protein expression
from the nCounter Analysis System were compared between the samples. Fold-change correlation was high between
the two platforms (R2 > 0.94).
Log2 Fold Change (Flow)
2
R2 = 0.9406
1
-3
-2
-1
0
-1
R2 = 0.9458
0
2
1
3
Sample B vs. A
Sample C vs. A
-2
-3
Log2 Fold Change (nCounter)
FIGURE 3 Correlations in protein signal fold-change between flow cytometry and nCounter assay. Ten proteins in three samples types with varying expression were
assayed. Sample A expressed all proteins at similar levels, while Samples B and C expressed the same proteins at variable levels. Fold-change expression for Sample B vs.
A (green) and Sample C vs. A (orange) are plotted. Flow cytometry data were averaged across 3 replicates; nCounter data were averaged across 6 replicates. CD127 data
were omitted due to expression below the detection level in flow cytometry and low counts using nCounter.
6
NanoString® Technologies, Inc.
Precise, Simultaneous Quantification in Multiplexed Format
In order to assess reproducibility and precision for the nCounter Vantage 3D RNA:Protein Immune Cell Profiling Assay, NanoString
early access customers compared results across multiple users and sites (FIGURE 4). Gene and protein expression levels for 800
measurements were highly correlated in each example and highly reproducible.
A
B
RNA
18
16
14
12
10
8
6
4
2
0
Protein
16
14
12
10
8
6
4
2
0
2
4
6
8
10
12
Replicate 1(Log2 Counts)
14
16
0
18
C
0
2
4
6
8
10
12
Replicate 1(Log2 Counts)
14
16
18
D
Site 1
Inter-user Precision
RNA
18
Protein
16
14
User 2(Log2 Counts)
14
12
10
8
6
4
12
10
8
6
4
2
2
0
Site 2
Inter-user Precision
RNA
18
Protein
16
User 2(Log2 Counts)
Site 2
Intra-user Precision
RNA
18
Protein
Replicate 2(Log2 Counts)
Replicate 2(Log2 Counts)
Site 1
Intra-user Precision
0
0
2
4
6
8
10
12
User 1(Log2 Counts)
14
16
18
0
2
4
6
8
10
12
User 1(Log2 Counts)
14
16
18
FIGURE 4 Precision intra-and inter-user data demonstrated for 800 RNA and protein measurements. Data were generated across two external sites with four different
users to gauge precision and reproducibility for the assay. Site 1 used PBMCs to measure gene and protein expression between a single user (A) and two different users
(C). Site 2 looked at CD3-stimulated T cells between a single user (B) and two different users (D).
7
WHITE PAPER
Cell Profiling Application: Dynamic View of Leukocyte Differentiation
To illustrate the power of simultaneous RNA:Protein detection, time-course experiments for monocyte differentiation into macrophages
were performed using a monocyte cell line THP1, phorbol myristate acetate (PMA) as an inducer of in vitro differentiation, and the
nCounter Vantage 3D RNA:Protein Immune Cell Profiling Assay. When comparing routine gene and protein expression correlation
at a single time-point (FIGURE 5A) with temporal expression profiles (FIGURE 5B), the digital counts vary by analyte type. When
examining a single time-point, protein and gene expression levels appear to be concordant. While this correlation holds true for CD40
across all time point, CD4 and CD9 expression levels bifurcate at 48 hours and beyond, which may be evidence of differences between
transcriptional and translational responses to PMA treatment.
A
∆ Expression 12 hours
after PMA addition
3000
B
RNA
6,000
Protein
2800
Prot-CD40/CD40
600
RNA-CD40/CD40
Digital Protein Counts
2400
2200
Digital Counts
2000
1800
1600
1400
1200
1000
4,500
450
3,000
300
1,500
150
Digital mRNA Counts
2600
800
0
600
400
200
0
12
24
48
72
0
96
Time (hours)
0
CD40
∆ Expression 12 hours
after PMA addition
Prot-CD4/CD4
1,000
RNA-CD4/CD4
Protein
Digital Protein Counts
-100
-200
-300
-400
-500
Digital Counts
2,000
RNA
-600
-700
-800
-900
-1000
1,500
750
1,000
500
500
250
0
CD4
0
12
24
48
72
96
Digital mRNA Counts
0
0
Time (hours)
3000
∆ Expression 12 hours
after PMA addition
RNA
Protein
2800
8,000
Prot-CD9/CD9
2600
1,600
RNA-CD9/CD9
Digital Protein Counts
2200
Digital Counts
2000
1800
1600
1400
1200
1000
800
6,000
1,200
4,000
800
2,000
400
Digital mRNA Counts
2400
600
0
400
200
0
CD9
0
12
24
48
72
96
0
Time (hours)
FIGURE 5 Comparison of static and dynamic expression illustrates relationships between gene and protein expression in monocytes. Monocytes (THP-1 cell lines) were
stimulated with PMA for 12–96 hours to induce differentiation into macrophages. Data normalization was accomplished using ERCC controls and then using the geometric
mean of proteins and genes. Protein background was determined by comparing to normalized data for IgG isotype controls. (A) Bar charts show differential expression of
genes and proteins after 12 hours for each target. (B) Line graphs show time-course data from 0 to 96 hours, giving a deeper view of the changes in target co-expression.
8
NanoString® Technologies, Inc.
Cell Signaling Application: Interferon Gamma Signaling
To examine the regulation of mRNA and protein levels by cytokines, PBMCs were stimulated with varying amounts of interferon
gamma (IFNγ) over 48 hours. Following sampling at 2, 24, and 48 hours post-treatment, the nCounter Vantage 3D RNA:Protein
Immune Cell Assays were used to assess differential expression of both analyte types. Comparison of treated versus untreated
populations at the 2-hour time point with the RNA:Protein Immune Cell Signaling Assay reveals the early upregulation of known IFNγ
targets, including STAT1, STAT2, CCL8, CXCL9, CXCL10, CXCL11, FCGR1A, SERPING1, and IDO1 (FIGURE 6A), at the mRNA level22. As
expected, STAT1 protein expression increases with IFNγ treatment time, whereas the corresponding mRNA plateaus after 24 hours
of exposure (FIGURE 6B). Using the RNA:Protein Immune Cell Profiling Assay, the effects of IFNγ on extracellular proteins were also
measured. STAT1 is known to regulate PD-L1 on the cell surface, and indeed, the protein counts increase considerably between 2
and 24 hours of treatment (FIGURE 6C). In contrast, the changes in surface PD-L2 protein levels, which are not mediated by STAT1
signaling, were less substantial (FIGURE 6D)23.
A
B
miRNA
SAT1 Protein
SAT1 mRNA
4000
Protein
14
3500
12
3000
Digital Counts
16
- Log10 p Value
mRNA
10
8
6
-6
-4
-2
2500
2000
1500
4
1000
2
500
0
0
0
2
6
4
8
10
12
2 hr
Log2 Fold Change
C
D
PD-L1 Protein
48 hr
2 hr
24 hr
48 hr
50ng IFNγ
PD-L2 Protein
CD274 mRNA
PDCD1LG2 mRNA
4000
4000
3500
3500
3000
3000
Digital Counts
Digital Counts
24 hr
Untreated
2500
2000
1500
2500
2000
1500
1000
1000
500
500
0
0
2 hr
24 hr
Untreated
48 hr
2 hr
24 hr
50ng IFNγ
48 hr
2 hr
24 hr
Untreated
48 hr
2 hr
24 hr
48 hr
50ng IFNγ
FIGURE 6 Differential mRNA and protein expression in PBMCs following IFNγ stimulation. (A) PBMCs were treated with 0, 5, 15, or 50 ng of IFNγ, and samples of cells
were taken at 2 hours for RNA:Protein analysis. Data normalization was accomplished using ERCC controls, geometric means (protein), and/or housekeeping genes
(mRNA). Comparing the fold-change in analyte levels between treated and untreated to the significance of those changes reveals the most differentially expressed
targets. Protein expression changes are more apparent at later time points. (B – D) IFNγ treatment was continued to 48 hours, and samples were prepared and analyzed
as described above. Protein data were collected for intracellular (B) and extracellular (C-D) targets with the two nCounter Vantage 3D RNA:Protein Immune Cell Assays.
Both mRNA and protein increase for STAT1 and PD-L1, consistent with published observations.
9
WHITE PAPER
Conclusion
For the first time, researchers can quickly assess the key regulatory molecules and pathways of cancer immunity, measuring gene and
protein expression simultaneously in a single, simple-to-use assay. The nCounter Vantage 3D RNA:Protein Immune Cell Assays permit
sensitive and quantitative investigation of fundamental immunological processes at both the transcriptional and translational level.
Combining measurements onto one platform greatly reduces complexity in data analysis, and as few as cells are required for read out,
permitting analysis on precious, clinically relevant samples.
The data presented here demonstrate the reliability of nCounter technology for measuring protein expression in limited samples.
The precision of RNA:Protein profiling technology and its concordance with the gold standard of flow cytometry is preserved
(FIGURE 3). When applied to biological samples, RNA:Protein profiling offers information on both RNA and protein expression using
the same digital platform and demonstrates precision for replicates generated by a single user and across several users (FIGURE 4).
Expression data from these assays has the potential to promote a better understanding of the interactions between the host immune
system and tumor microenvironment and to identify novel biomarkers allowing for optimization of potential immuno-oncology
therapeutics providing a mechanistic and stratification information.
10
NanoString® Technologies, Inc.
References
1. Hanahan D, Weinberg RA (2011) Hallmarks of cancer:
the next generation. Cell 144:646–674.
13. Pardoll DM (2012) The blockade of immune checkpoints
in cancer immunotherapy. Nat Rev Cancer 12(4):252–264.
2. Cavallo F, De Giovanni C, Nanni P, Forni G, Lollini PL
(2011) 2011: The immune hallmarks of cancer. Cancer
Immunol Immunother 60(3):319–326.
14. Vanneman M, Dranoff G (2012) Combining
immunotherapy and targeted therapies in cancer
treatment. Nat Rev Cancer 12(4):
237–251.
3. Koebel CM, Vermi W, Swann JB, Zerafa N, Rodig SJ,
Old LJ, Smyth MJ, Schreiber RD (2007) Adaptive
immunity maintains occult cancer in an equilibrium state.
Nature 450(7171):903–907.
4. Schreiber RD, Old LJ, Smyth MJ (2011) Cancer
immunoediting: integrating immunity’s roles in cancer
suppression and promotion. Science 331(6024):
1565–1570.
5. Vesely MD, Kershaw MH, Schreiber RD, Smyth MJ (2011)
Natural innate and adaptive immunity to cancer. Annu
Rev Immunol 29:235–271.
6. Couzin-Frankel J (2013) Breakthrough of the year 2013.
Cancer immunotherapy. Science 342(6165):1432–1433.
7. Srinivas PR, Kramer BS, Srivastava S (2001) Trends in
biomarker research for cancer detection. Lancet Oncol
2(11):698–704.
8. Gajewski TF, Fuertes M, Spaapen R, Zheng Y, Kline J
(2011) Molecular profiling to identify relevant immune
resistance mechanisms in the tumor microenvironment.
Curr Opin Immunol 23(2):286–292.
9. Ma Y, Ding Z, Qian Y, Shi X, Castranova V, Harner EJ, Guo
L (2006) Predicting cancer drug response by proteomic
profiling. Clin Cancer Res 12(15):4583–4589.
10.Geiss GK, Bumgarner RE, Birditt B, Dahl T, Dowidar N,
Dunaway DL, Fell HP, Ferree S, George RD, Grogan T,
James JJ, Maysuria M, Mitton JD, Oliveri P, Osborn JL,
Peng T, Ratcliffe AL, Webster PJ, Davidson EH, Hood
L, Dimitrov K (2008) Direct multiplexed measurement
of gene expression with color-coded probe pairs. Nat
Biotechnol 26(3):317–325.
11. Ullal AV, Peterson V, Agasti SS, Tuang S, Juric D, Caster
CM, Weissleder R (2014) Cancer cell profiling by
barcoding allows multiplexed protein analysis in fineneedle aspirates. Sci Transl Med 6(219):219ra9.
12. Chen DS, Mellman I (2013) Oncology meets immunology:
The cancer-immunity cycle. Immunity 39(1):1–10.
15. Ascierto PA, Kalos M, Schaer DA, Callahan MK, Wolchok
JD (2013) Biomarkers for immunostimulatory monoclonal
antibodies in combination strategies for melanoma and
other tumor types. Clin Cancer Res 19(5):1009–1020.
16. Galon J, Angell H, Bedognetti D, Marincola F (2013)
The continuum of cancer immunosurveillance:
prognostic, predictive, and mechanistic signatures.
Immunity 39(1):11–26.
17. Ascierto PA, Capone M, Urba WJ, Bifulco CB, Botti G,
Lugli A, Marincola FM, Ciliberto G, Galon J, Fox BA
(2013) The additional facet of immunoscore:
immunoprofiling as a possible predictive tool for
cancer treatment. |J Transl Med 11:54.
18. Mantovani G, Macciò A, Lai P, Ghiani M, Turnu E, Del
Giacco GS (1995) Membrane-bound/soluble IL-2 receptor
(IL-2R) and levels of IL-1 alpha, IL-2, and IL-6 in the serum
and in the PBMC culture supernatants from 17 patients
with hematological malignancies. Cell Biophys 27(1):1-14.
19. Gonzalo JA, Delaney T, Corcoran J, Goodearl A,
Gutierrez-Ramos JC, Coyle AJ (2001) Cutting edge:
the related molecules CD28 and inducible costimulator
deliver both unique and complementary signals required
for optimal T cell activation. J Immunol 166(1):1-5.
20.Laman JD, Claassen E, Noelle RJ (1996) Functions
of CD40 and its ligand, gp39 (CD40L). 16(1):59-108.
21. Vogel C, Marcotte EM (2012) Insights into the regulation
of protein abundance from proteomic and transcriptomic
analyses. Nat Rev Genet 13(4):227-232.
22.Waddell SJ, Popper SJ, Rubins KH, Griffiths MJ,
Brown PO, Levin M, Relman DA (2010) Dissecting
interferon-induced transcriptional programs in human
blood peripheral blood cells. PLoS ONE 5(3):e9753.
23.Loke P, Allison JP (2003) PD-L1 and PD-L2 are
differentially regulated by Th1 and Th2 cells.
PNAS 100(9):5336-5341.
11
WHITE PAPER
Appendix A: Immune Cell Types and Genes Included in the nCounter Vantage 3D RNA:Protein
Immune Cell Profiling Assay
Cell Type
Description
Panel Genes
B Cells
Perform several roles, including generating and presenting antibodies, cytokine
production, and lymphoid tissue organization.
BLK, CD19 (CD19), CR2 (CD21), HLA-DOB,
MS4A1 (CD20), TNFRSF17 (CD269)
T Cells
Play a central role in immunity and distinguished from other lymphocytes (e.g., B cells) by
the presence of a T cell receptor (TCR) on the cell surface.
CD2, CD3E, CD3G, CD6
A subset of CD3+CD4+ effector T cells that secrete cytokines with different activities.
ANP32B (APRIL), BATF, NUP107, CD28
(CD28), ICOS (CD278)
TH1
Produce IL-2 and IFNγ and promote cellular immunity by acting on CD8+ cytotoxic T cells,
NK cells and macrophages.
CD38, CSF2 (GM-CSF), IFNG, IL12RB2,
LTA, CTLA4 (CD152), TXB21, STAT4
TH2
Produce IL-4, IL-5 and IL-13 and promote humoral immunity by acting on B cells.
CXCR6 (CD186), GATA3, IL26, LAIR2
(CD306), PMCH, SMAD2, STAT6
TH17
Produce IL-17A, IL-17F, IL-21 and IL-22 and promote anti-microbial inflammation.
IL17A, IL17RA (CD217), RORC
TFH
Trigger the formation and maintenance of germinal centers through the expression of
CD40L and the secretion of IL-21 and IL-4, thereby playing a critical role in mediating the
selection and survival of B cells.
CXCL13, MAF, PDCD1 (CD279), BCL6
CD3+CD4+ T cells that inhibit effector B and T cells and play a central role in
suppression of autoimmune responses.
FOXP3
Tcm (central
memory)
Educated T cells that rapidly respond to antigen. Central memory T cells express
L-selectin and CCR7; they secrete IL-2, but not IFNγ or IL-4.
ATM, DOCK9, NEFL, REPS1, USP9Y
Tem (effector
memory)
Educated T cells that rapidly respond to antigen. Effector memory T cells do not
express L-selectin or CCR7; they secrete effector cytokines like IFNγ and IL-4.
AKT3, CCR2 (CD192), EWSR1 (EWS),
LTK, NFATC4
Cytotoxic (CD8) T Cells
Effector T cells with cytotoxic granules that interact with target cells expressing cognate
antigen and promote apoptosis of target cells.
CD8A (CD8), CD8B (CD8B), FLT3LG,
GZMM (MET1), PRF1
Gamma Delta T Cells
(Tγδ)
Express surface antigen recognition complex type 2 and represent a small percentage of
the peripheral T cell population. Functions span innate and adaptive immune responses,
including direct cytolysis and establishment of memory phenotypes.
CD160, FEZ1, TARP (TCRG)
Natural Killer (NK) Cells
Provide a rapid cytotoxic response to virally infected cells and tumors. These cells also
play a role in the adaptive immune response by readily adjusting to the immediate
environment and formulating antigen-specific immunological memory.
BCL2, FUT5, NCR1 (CD335), ZNF205
CD56bright
Constitute the majority of NK cells in secondary lymphoid tissues. Abundant cytokine
producers and weakly cytotoxic before activation.
FOXJ1, MPPED1, PLA2G6, RRAD
CD56dim
Constitute the majority of NK cells in the periphery and are more cytotoxic than
CD56bright cells.
GTF3C1, GZMB, IL21R (CD360)
Cells that process antigen material and present it on the cell surface to T cells, thereby
acting as messengers between the innate and adaptive immune systems.
CCL13, CCL17, CCL22 (MDC), CD209
(CD209), HSD11B1
iDC
(immature)
Play a critical role in initiating tumor immunity. Tumor cells can exploit the functional roles
of iDCs for tumor progression via release of soluble factors such as VEGF.
CD1A, CD1B, CD1E, F13A1, SYT17
aDC
(activated)
Promote the induction of the adaptive immune response by presenting captured
antigen to naive T cells.
CCL1, EBI3, IDO1 (INDO), LAMP3
(CD208), OAS3
Similar in appearance to plasma cells and share many characteristics with myeloid dendritic cells. These cells can produce high levels of IFNα.
IL3RA (CD123)
Scavengers of dead or dying cells and cellular debris. Macrophages have roles in innate
immunity by secreting pro-inflammatory and anti-inflammatory cytokines.
APOE, CCL7 (FIC), CD68, CHIT1,
CXCL5, MARCO, MSR1 (CD204)
Mast Cells
Granulocytes that can influence tumor cell proliferation and invasion and promote
organization of the tumor microenvironment by modulating the immune response.
CMA1, CTSG, KIT (CD117), MS4A2,
PRG2, TPSAB1
Neutrophils
Phagocytic granulocytes that act as first-responders and migrate towards a site of
inflammation. Typically a hallmark of acute inflammation.
CSF3R (CD114), FPR2, MME (CD10)
Eosinophils
White blood cells responsible for combating multicellular parasites and some types of
infections.
CCR3 (CD193), IL5RA (CD125), PTGDR2
(CD294; GPR44), SMPD3, THBS1
Adaptive Immmune Response
Helper T Cells
Regulatory T Cells (Treg)
Cytotoxic Cells
Conventional (Myeloid)
Dendritic Cells (DC)
Dendridic Cells
Innate Immmune Response
Memory
T Cells
Plasmacytoid Dendritic
Cells (pDC)
Granulocytes
Macrophages
12
NanoString® Technologies, Inc.
Appendix B: Proteins Included in the nCounter Vantage 3D RNA:Protein Immune Cell Profiling Assay
Protein
Name
Immunological Activity
CD3 (CD3E)
Member of the T cell receptor-CD3 complex, which plays an important role in coupling antigen recognition to several intracellular signal
transduction pathways. The CD3 complex is found on the surface of all mature T cells.
CD4
Glycoprotein found on the surface of T helper cells, monocytes, macrophages, and dendritic cells. Acts as a co-receptor that amplifies TCR
signaling via recruitment of Lck. It can also interact directly with MHC class II molecules on the surface of antigen-presenting cells.
CD8 (CD8A)
Cytotoxic T lymphocyte cell surface glycoprotein that mediates cell-cell interactions between MHC class I antigen-presenting cells and T
cells.
CD9
Marker of naïve lymphocyte populations with roles in cell adhesion and migration.
CD14
Co-receptor expressed by macrophages, neutrophils, and dendritic cells that — along with TLR4 and MD-2 — is responsible for detection
of pathogen-associated molecules such as lipopolysaccharide and lipoteichoic acid.
CD19
Expressed on the surface of dendritic cells and B cells, this surface molecule assembles with antigen receptors in order to decrease the
threshold for antigen receptor-dependent stimulation.
CD25 (IL2R)
Expressed by lymphocytes, this receptor acts along with its ligand, IL2, to promote the differentiation of T cells into effector T cells and
memory T cells after an antigen-mediated initial T cell response.
CD27
Member of TNF-receptor superfamily and required for the generation and long-term maintenance of T cell immunity via binding of its
ligand, CD70. Known to play a role in regulating B cell activation and immunoglobulin synthesis.
CD28
Receptor for CD80 and CD86 proteins. Expressed on T cells that provide co-stimulatory signals required for T cell activation and survival.
CD33
Transmembrane receptor that binds sialic acids and a myeloid lineage cell marker.
CD40
Broadly expressed co-stimulatory protein found on antigen-presenting cells that is required for their activation, typically via binding of
CD40L. This member of the TNF receptor superfamily is essential in mediating a variety of immune and inflammatory responses including
T cell-dependent immunoglobulin class switching, memory B cell development, and germinal center formation.
CD45RO
Protein tyrosine phosphatase receptor specifically expressed in hematopoietic cells and shown to be an essential regulator of T cell and B
cell antigen receptor signaling.
CD56 (NCAM)
Homophilic binding glycoprotein expressed on the surface of natural killer cells.
CD68
Glycoprotein expressed on monocytes and macrophages that binds to low density lipoprotein and serves as a marker of these lineages.
CD73 (NT5E)
Frequently used marker of lymphocyte differentiation. This protein catalyzes the conversion of AMP to adenosine.
CD127 (IL-7Ra)
Expressed on many immune cell types including B cells, T cells, monocytes, and dendritic cells, this receptor and its ligand, IL7, play a
critical role in lymphocyte development.
CD134 (OX40)
Secondary costimulatory molecule expressed on activated antigen-presenting cells that plays a critical role in maintaining an immune
response by enhancing the survival of T cells.
CD137 (4-1BB)
Expressed by activated T cells, dendritic cells, follicular dendritic cells, natural killer cells, granulocytes, and cells of blood vessel walls at
sites of inflammation. Known to possess a co-stimulatory role in activated T cells, enhancing T cell proliferation and cytolytic activity.
CD152
(CTLA-4)
Regulatory protein expressed on the surface of helper T cells. Downregulates the immune response by sending an inhibitory signal to T
cells.
CD154 (CD40L) TNF superfamily protein primarily expressed on activated T cells. Binds to CD40 on antigen-presenting cells, often in a co-stimulatory role.
CD158e1
(KIR3DL1)
Transmembrane glycoprotein expressed by natural killer cells and subsets of T cells.
CD163
Receptor for the hemoglobin-haptoglobin complex that has been shown to mark cells of the monocyte and macrophage lineages.
CD272 (BTLA)
Induced during T cell activation, this regulatory protein acts to negatively regulate T cell immune responses. Ligand for TNFRSF14 (HVEM).
CD273 (PD-L2)
Co-stimulatory molecule essential for T cell proliferation and interferon-gamma production in a PDCD1-independent manner. Interaction
with PDCD1 inhibits T cell proliferation by blocking cell cycle progression and cytokine production.
CD274 (PD-L1)
A regulatory protein ligand that modulates activation or inhibition of T cells, B cells, and myeloid cells. Reduces the proliferation of these
CD8+ T cells via interaction with PD-1 and B7-1.
CD278 (ICOS)
Co-stimulatory molecule expressed on activated T cells that is thought to be particularly important for Th2 cell function.
CD279 (PD-1)
Promotes self-tolerance by stimulating apoptosis in antigen-specific T cells and reducing apoptosis in regulatory T cells.
CD335
(NKp46)
Triggering receptor expressed on the surface of activated natural killer cells.
13
WHITE PAPER
Appendix C: Proteins Included in the nCounter Vantage 3D RNA:Protein Immune Cell Signaling Assay
Protein
Name
Immunological Activity
Aiolos (IKZF3) Transcription factor that controls B cell proliferation and development.
BatF
Transcription factor that controls Th17, Tfh and CD8+ dendritic cell differentiation. Also controls class switch recombination in B
cells.
CCL5
Chemoattractant for monocytes, eosinophils, and CD4+ memory cells.
CXCL5
Chemoattractant for neutrophils.
CXCL8 (IL8)
Chemoattractant for neutrophils and T cells.
FoxP3
Transcription factor that controls regulatory T cell development and activity.
GATA3
Transcription factor required for Th2 differentiation.
GM-CSF
Cytokine growth factor for granulocytes and monocytes.
Helios (IKZF2) Transcription factor expressed during T cells development and in regulatory T cells.
IDO
Tryptophan catabolizing enzyme that limits T cell proliferation and promotes differentiation of regulatory T cells.
IFNγ
Pleotropic cytokine with activities including macrophage activation, development and function of Th1 T cells, antigen processing
and presentation, inhibition of proliferation, B cell class switching.
Ikaros (IKZF1)
Transcription factor with role in lymphocyte development.
IL-1β
Proinflammatory cytokine that promotes lymphocyte activation, especially Th17 T cells, neutrophil influx, fibroblast proliferation.
IL-10
Anti-inflammatory cytokine that inhibits T cell activity and promotes regulatory T cells and M2 macrophages.
IL-12p40
Cytokine required for IFNg production by Th1 T cells and NK cells.
IL-15RA
(CD215)
Receptor for IL-15 that promotes proliferation and activation of T cells (especially memory CD8+ T cells) and NK cells.
IL-2
Cytokine produced by T cells in response to stimulation and promotes T cell proliferation.
IL-6
Pleotropic cytokine with activities including lymphocyte and monocyte activation and differentiation.
LAP(TGF-β)
Pleotropic cytokine with regulatory effects on osteoblast activity, regulatory T cell and Th17 T cell differentiation and activity.
Perforin
(PRF1)
Pore forming component of cytolytic granules secreted by CD8+ T cells and NK cells.
STAT1 (total)
Transcription factor activated by interferon signaling.
STAT3 (total)
Transcription factor activated by proinflammatory cytokines.
T-bet
Transcription factor that controls Th1 CD4+ T cell differentiation.
Th-Pok
(ZBTB7B)
Transcription factor that promotes CD4+ T cell linage commitment.
PTEN
Intracellular phosphatase that inhibits PI3K/AKT signaling.
IRF4
Transcription factor that represses proinflammatory gene expression.
Bcl-6
Transcription repressor required for B cell germinal center formation.
14
NanoString® Technologies, Inc.
NanoString® Technologies, Inc.
WHITE PAPER
NanoString Technologies, Inc.
LEARN MORE
SALES CONTACTS
530 Fairview Ave N
Seattle, Washington 98109 USA
Visit www.http://www.nanostring.com/products/
vantage-rna-protein to learn more about
nCounter Vantage 3D Immune Cell Assays.
United States:
[email protected]
EMEA:
[email protected]
Tel: (888) 358-6266 | Fax: (206) 378-6288
www.nanostring.com | [email protected]
Other Regions:
Asia Pacific & Japan: [email protected]
[email protected]
© 2016 NanoString® Technologies, Inc. All rights reserved. NanoString, NanoString Technologies, nCounter, 3D Biology, Vantage 3D, and nSolver are registered trademarks or
trademarks of their respective companies in the United States and/or other countries.
FOR RESEARCH USE ONLY. Not for use in diagnostic procedures.
NOVEMBER 2016 USSC PM0014-04