When studying cellular heterogeneity and immune composition within the tumor microenvironment, traditional cell profiling methods have been limited by the number of markers they can interrogate.
Now, digital software suites can be used to profile the cellular composition of tissues from bulk RNA-sequencing (RNA-seq) data. In particular, complex tissue types such as fresh/frozen (FF) or formalin-fixed paraffin-embedded (FFPE) samples require specialized methods to remove noise and technical variation.
This application note explores the performance characteristics of a NGS targeted enrichment panel for profiling both FF and FFPE tissue samples using a state-of-the-art iSort software solution for digital cytometry.
Download this application note to learn more about:
- Efficient and reliable enumeration of cell types from FF or FFPE tissues
- Target enrichment for targeted RNA sequencing
- Software to quantify cellular composition from bulk tissue expression data
Application Note
Genomics
Author
Aki Nakao1
Aaron M. Newman1,2
Ash A. Alizadeh1,2
Maximilian Diehn1,2
Manjula Aliminati3
Mistuni Ghosh3
Jayati Ghosh3
Kristi Stephenson3
Ashutosh Ashutosh3
1. CiberMed, Inc.,
California, USA
2. Stanford University, Stanford,
California, USA
3. Agilent Technologies Inc.,
California, USA
Abstract
CiberMed’s iSort software suite is a widely used digital cytometry solution for
profiling the cellular composition of complex tissues from bulk RNA-sequencing
(RNA-seq) data. iSort Fractions ‘tissue mode’ quantifies the relative fractions
of 22 functionally defined hematopoietic subsets along with three non-immune
cell types—fibroblasts, endothelial cells, and epithelial cells—from bulk tissue
expression data. To enable accurate enumeration of these 25 cell types from
fresh/frozen (FF) or formalin-fixed paraffin-embedded (FFPE) samples, iSort
Fractions includes specialized methods to remove noise and technical variation
across diverse platforms and sample preservation conditions. In this study, we
assess the accuracy, robustness, and reproducibility of the Agilent SureSelect CD
CiberMed Tissue panel using iSort Fractions. This panel was developed to enable
highly efficient and reliable enumeration of 25 cell types from FF or FFPE tissues.
Total RNA from paired FF and FFPE tumor specimens was extracted and
prepared for sequencing using Agilent SureSelect XT HS2 RNA reagents and for
target enrichment using the SureSelect CD CiberMed Tissue panel. The resulting
sequencing data were analyzed using iSort Fractions tissue mode. Across all
evaluable phenotypes (n = 25), cell type fractions determined by iSort were
highly concordant between paired FF and FFPE tumor samples (rho = 0.93).
This substantially outperformed whole-transcriptome profiling (rho = 0.78) while
requiring approximately 20-fold less sequencing. Additionally, strong reproducibility
was observed between technical replicates profiled by targeted sequencing (rho ≥
0.97 for FF and rho ≥ 0.96 for FFPE). The SureSelect CD CiberMed Tissue panel is
available through the Agilent Community Design program to enable highly accurate
cellular profiling of fresh/frozen and fixed tissue specimens with iSort.
Agilent SureSelect Targeted RNA-Seq
Paired With iSort Enables Robust
Enumeration of Cell Type Composition
in Solid Tissue Samples
2
Introduction
Cellular heterogeneity and complex intercellular interactions
underlie diverse physiological and pathological states,
including various malignancies. Therefore, it is critically
important to study the phenotypic and genotypic composition
of cell subsets within the diseased milieu. It is also
essential to monitor changes in their relative abundances
during disease progression and in response to therapy.
The importance of studying cellular heterogeneity and
composition within the tumor microenvironment (TME) is
well established.1-3 Enumerating cell type composition has
prognostic value and holds great promise as a potential
predictive biomarker for therapy response.4,5 Therefore,
traditional methods such as flow/mass cytometry,6,7
immunohistochemistry (IHC),8
and immunofluorescence (IF)9
are routinely employed for quantifying and characterizing
tissue heterogeneity (Table 1). However, these methods can
only interrogate a modest number of markers and there is
often a trade-off between the number of markers that can
be measured and the throughput of the assay. Cytometry
by time-of-flight (CyTOF)10 is destructive to the sample and
does not enable co-interrogation of cell type fractions and
cell type expression profiles across thousands of genes.
In more recent years, single-cell RNA sequencing (scRNAseq) approaches have been embraced as a means of
characterizing cell type composition and gene expression
at the single-cell level. However, scRNA-seq is limited by
sample preparation artifacts, including dissociation-induced
distortions in cellular composition, and remains cost
prohibitive for large-scale cellular profiling.11 Similarly, FFPE
tissue specimens, which are collected as part of routine
clinical care, cannot be dissociated into a cell suspension
without disruption of cell type composition.
Given these limitations for characterizing FF and FFPE
samples, deconvolution algorithms for determining cell
type abundances from bulk tissue expression profiles have
gained traction.1,2,12-16 These methods enable dissection
of cell-type-specific signals from bulk sequencing data.
Comparative analyses of scRNA-seq, deconvolution of bulk
expression data, and IHC revealed that deconvolution is
free from artifacts arising from cell separation and tissue
dissociation.17 Of these deconvolution methods, CIBERSORT
and CIBERSORTx, have emerged as robust and accurate tools
for determining cell type proportions from blood and tissue
samples.3,17-28 In fact, CIBERSORTx was recently identified
as one of the five fastest-growing software tools in the
biosciences.29
CiberMed further optimized and standardized CIBERSORTx
with proprietary enhancements and validated the improved
algorithms with different sequencing platforms and sample
types, including FF and FFPE preservation states. CiberMed
currently offers two algorithms by way of two flagship
products within the iSort digital cytometry suite (Figure 1).
iSort Fractions reliably determines cell subset abundance
from bulk tissue or blood expression data and iSort HiRes
infers cell-type-specific gene expression profiles from bulk
tissue or blood expression data.
Table 1. Comparison of cell profiling methods.
Software-Based Deconvolution Single-Cell RNA-Seq Flow Cytometry/CyTOF IHC
Throughput +++++ + ++ +++
Requires Tissue Dissociation No Yes Yes No
Artifacts Introduced by Dissociation No Yes Yes No
Workflow Simplicity +++++ + ++ ++++
Manual Data Analysis Required No Yes Yes Yes
Number of Cell Types/Cell Type Resolutions +++++ +++++ +++ +++
3
Table 2. Human cell types quantified by blood mode (22 cell types) and
tissue mode (25 cell types) of iSort Fractions.
Parent Subsets Cell Type Description
B cells
B cells naïve
B cells memory
Plasma cells Plasma cells
CD8 T cells T cells CD8
CD4 T cells
T cells CD4 naïve
T cells CD4 memory resting
T cells CD4 memory activated
T cells follicular helper
T cells regulatory (Tregs)
Gama delta T cells T cells gamma delta
NK cells NK cells resting
NK cells activated
Monocytes and
Macrophages
Monocytes
Macrophages M0
Macrophages M1
Macrophages M2
Dendritic cells Dendritic cells resting
Dendritic cells activated
Mast cells Mast cells resting
Mast cells activated
Eosinophils Eosinophils
PMNs Neutrophils
Fibroblasts
Endothelial cells
Epithelial cells
iSort Fractions ‘blood mode’ quantifies cell type abundances
from bulk RNA-seq data by applying the well-established
LM22 leukocyte gene signature matrix to distinguish
22 human hematopoietic cell subsets. LM22 has been
validated in pure leukocyte subset titrations, blood samples,
and tumors from multiple cancer types. CiberMed’s iSort
Fractions tissue mode uses the LM22 signature matrix plus
tissue-specific signature profiles to quantify 25 cell types,
including 22 immune subsets, fibroblasts, endothelial cells,
and epithelial cells, from bulk tissue RNA-seq profiles of FF or
FFPE biospecimens (see Table 2 for the complete list of cell
subsets).3,27,30,47,48,49
In this application note, we demonstrate robust, accurate,
and reproducible enumeration of cell subsets in solid tumor
samples using iSort Fractions combined with the Agilent
SureSelect target enrichment solution (Figure 2). The
SureSelect CD CiberMed Tissue panel targets genes from
the LM22 signature matrix along with additional genes for
discriminating fibroblasts, endothelial cells, and epithelial
cells with iSort Fractions tissue mode (see Table 3 for panel
details). Four pairs of matched FF and FFPE samples from
non-small cell lung cancer (NSCLC) tumor biopsies were
included in the assessment. SureSelect XT HS2 RNA libraries
were prepared in triplicate from total RNA extracted from
each sample and enriched with the SureSelect CD CiberMed
Tissue panel.
Blood
Tissue
SureSelect CD CiberMed Tissue SureSelect CD CiberMed Heme†
Intended Application Enumeration of 25 cell subsets (Table 2) with iSort Fractions Enumeration of 22 immune cell subsets (Table 2) with iSort Fractions
Sample Type FF, FFPE Whole blood, PBMCs
Signature Matrix LM22 (immune subsets) and TR4 (non-immune cell types) LM22 (immune cell types)
Number of Targets 1,423 genes 547 genes
Total Capture Size 5.7 Mb 1.8 Mb
Recommended Min Reads/Sample 2 M (1 M × 2, 150 bp) 500 k (250 k × 2, 150 bp)
†
An application note detailing the Agilent SureSelect CD CiberMed Heme panel is available at www.agilent.com/cs/library/applications/ap-isort-sureselect-5994-6964en-agilent.pdf.
Table 3. Comparison of panel designs and the intended application for each.
4
Across all 25 evaluable cell types, high concordance was
observed between matched FF and FFPE samples. The
concordance was markedly stronger for targeted RNA-seq
with the SureSelect CD CiberMed Tissue panel than for wholetranscriptome sequencing applied to the same samples
(Figure 3). There was also high reproducibility between
technical replicates (Figure 4). Because of several proprietary
enhancements, we found that iSort Fractions outperformed
CIBERSORTx for the enumeration of cell type composition
(Figure 3).
Notably, targeted enrichment with the SureSelect CD
CiberMed Tissue panel reduced the sequencing requirement
to only 2M (1M × 2) input reads per sample, as opposed
to 40M (20M × 2) input reads per sample for wholetranscriptome sequencing, a reduction of 20-fold. Together,
these results underscore the accuracy, reliability, and costeffectiveness of the combined SureSelect/iSort assay for cell
type abundance profiling from solid tissue samples.
Figure 2. End-to-end workflow supported by kitted reagents and instrumentation from Agilent and an integrated analysis solution from CiberMed.
Figure 1. Schematic of iSort algorithms for digital cytometry. iSort Fractions determines cell type proportions and iSort HiRes determines cell-type-specific gene
expression profiles from bulk tissue expression data.
Sample
Extraction
cDNA
Conversion Library Prep Target Enrichment Sequencing iSort
Analysis
(Detailed
workflow)
Kitted Reagents
Instrumentation
SureSelect HS2
RNA reagents
SureSelect CD
CiberMed panels
TapeStation
systems
Bravo Automation system Magnis system
Pre-designed + Optimized panels and kitted SureSelect reagents simplify the wet lab workflow.
Sample QC supported at multiple steps in the workflow.
Automated sample processing increases reproducibility and walk away time.
Compatible with both Bravo and Magnis systems.
Fragment Analyzer
systems
1
Digital
Cytometry
iSortTM Fractions iSortTM HiRes
BULK RNA
PROFILE
5
Materials and methods
Samples
Paired FF and FFPE tumor samples from four patients with
NSCLC were procured from Proteogenex (Inglewood, CA,
USA). Total RNA of the FF samples was extracted using the
AllPrep DNA/RNA Micro Kit (Qiagen). The AllPrep DNA/RNA
FFPE Kit (Qiagen) was used to extract the RNA of one FFPE
sample (FFPE-1). The RNAstorm FFPE RNA extraction kit
(Biotium, formerly Cell Data Sciences) was used to extract
RNA from the remaining three FFPE samples. RNA quality
was assessed using the Agilent RNA 6000 Pico kit (Agilent
p/n 5067-1513) and RNA concentrations were determined
using the Qubit RNA HS Assay kit (Thermo Fisher Scientific
p/n Q32855). All FF samples had RIN > 6 whereas FFPE
samples had DV200 values from 52 to 82.
Library preparation and target enrichment for targeted
RNA sequencing
Library preparation and targeted enrichment were performed
using an Agilent Bravo NGS Workstation Option B following
the Agilent SureSelect XT HS2 RNA system user guide
G9993-90010.31 Agilent SureSelect XT HS2 RNA reagent
kits G9991A and G9991B were employed to generate three
technical replicate libraries per sample, each starting with
an input of 30 ng total RNA. Agilent SureSelect XT HS2 RNA
target enrichment kit part number G9994A was employed
to enrich sample libraries with the SureSelect CD CiberMed
Tissue panel.
Agilent SureDesign software was used to create the
SureSelect CD CiberMed Tissue panel. Manual curation was
performed to ensure full coverage of the coding region for
every included gene.
Enriched libraries were sequenced as 150 bp × 2 paired-end
reads on Illumina NovaSeq 6000 instrument. Each sample
was sequenced to approximately 50M (25M × 2) total reads
to allow thorough performance assessment at varying
depths of coverage.
Data processing
As input to iSort, RNA sequencing reads (in FASTQ format)
were first summarized to gene expression values in
transcripts per million (TPM). While this process can be
accomplished using any standard mapping/alignment
approach, for the data presented here, we used Salmon v1.932
to map and quantify RNA-seq reads. The TPM values were
then used as input to iSort Fractions v1.4, which determined
the relative fractions of 25 immune subsets (Table 2) in each
tissue sample.
CiberMed software tools, iSort Fractions and iSort HiRes, are
currently available as docker-containerized tools that can be
run locally. iSort Fractions is offered also by way of a userfriendly web interface run securely through AWS (Amazon
Web Services) at https://isort.cibermed.com. For more details
about the iSort digital cytometry suite, see the brochure at
https://isort.cibermed.com/iSort_ProductsAndServices.pdf or
contact CiberMed directly for questions at https://cibermed.
com/contact.
Performance evaluation
The accuracy of iSort Fractions was evaluated using
Spearman rho, Pearson r, and root mean squared error
(RMSE) to quantify the concordance of estimated cell type
proportions, both within and between paired FF and FFPE
samples.
Read titration analysis
To determine the impact of the number of reads per sample
on deconvolution accuracy and reproducibility, paired reads
were randomly sampled to 5M, 2M, 1M, 500k, 100k, and 10k
effective reads per sample. Effective reads are defined as the
number of reads mapped “on-target” to the genes included
in the SureSelect CD CiberMed Tissue panel. Input reads are
calculated as [minimum effective reads]/[on-target map rate].
6
Results
All RNA-seq data generated in this study passed internal
quality control requirements.
Accuracy
Four pairs of matched FF and FFPE samples, derived
from NSCLC tumor biopsies, with three replicates each,
were analyzed using iSort Fractions tissue mode, which
quantifies 25 cell types, including 22 hematopoietic subsets
and three non-immune cell types: fibroblast, endothelial,
and epithelial cells.
Figure 3 summarizes the concordance of iSort Fractions
results between FF and FFPE samples for 25 cell types (top)
and 23 cell types (bottom) with the latter excluding fibroblasts
and epithelial cells to better visualize the 0 to 10% fractional
abundance range. While performance gains can be seen for
iSort over CIBERSORTx (Figure 3b versus Figure 3a), targeted
sequencing with the SureSelect CD CiberMed Tissue panel
substantially outperformed whole transcriptome sequencing
applied to the same samples (Figure 3c versus Figure 3b).
These data indicate that targeted digital cytometry with
SureSelect and iSort can overcome FFPE-related distortions
to enable reliable cell profiling in fixed tissue specimens
0 20 40 60 80
0 20 40 60 80
iSort (%) FFPE
iSort (%) FF
rho = 0.93
P = 1 × 10−45
RMSE = 4.2
25 cell types
0 20 40 60 80
0 20 40 60 80
CIBERSORTx (%) FFPE
CIBERSO
RTx (%) FF
rho = 0.76
P = 7 × 10−20
RMSE = 4.8
25 cell types
0 20 40 60 80
0 20 40 60 80
iSort (%) FFPE
iSort (%) FF
rho = 0.78
P = 1 × 10−21
RMSE = 4.7
25 cell types
0 2 4 6 8 10 12
0 2 4 6 8 10
CIBERSORTx (%) FFPE
CIBERSO
RTx (%) FF
r = 0.67
P = 2 × 10−13
rho = 0.70
P = 1 × 10−14
RMSE = 2.2
23 cell types
Whole Transcriptome RNA-Seq
Whole transcriptome RNA-Seq
How
0 2 4 6 8 10 12
0 2 4 6 8 10
iSort (%) FFPE
iSort (%) FF
r = 0.74
P = 8 × 10−17
rho = 0.72
P = 5 × 10−16
RMSE = 1.8
23 cell types
0 2 4 6 8 10 12
0 2 4 6 8 10
iSort (%) FFPE
iSort (%) FF
r = 0.91
P = 4 × 10−35
rho = 0.92
P = 1 × 10−37
RMSE = 1
23 cell types
SureSelect CD CiberMed
Tissue Panel
iSort Fractions
Whole Transcriptome RNA-Seq
iSort Fractions
Whole Transcriptome RNA-Seq
CIBERSORTx
B cells naive
B cells memory
Plasma cells
T cells CD8
T CD4 naive
T CD4 memory resting
T CD4 memory activated
T cells follicular helper
T cells regulatory (Tregs)
T cells gamma delta
NK cells resting
NK cells activated
Monocytes
Macrophages M0
Macrophages M1
Macrophages M2
Dendritic cells resting
Dendritic cells activated
Mast cells resting
Mast cells activated
Eosinophils
Neutrophils
Fibroblasts
Endothelial cells
Epithelial cells
a b c
Source:
Method:
Figure 3. Scatterplots of cell type fractions determined by deconvolution of paired FF (y-axis) and FFPE (x-axis) bulk tumor expression profiles (n = 4 pairs);
comparing (a) whole-transcriptome sequencing data analyzed by CIBERSORTx, (b) whole-transcriptome sequencing data analyzed by iSort Fractions, and (c)
Agilent SureSelect CD CiberMed Tissue panel data analyzed by iSort Fractions. Results are shown for all 25 evaluable cell types (top) and 23 cell types (bottom) to
expand the lower range of fractional abundances (0 to 10%). Each point represents a sample colored by cell type.
7
Figure 4 summarizes the sample-level concordance between
FF and FFPE cell type fractions. The SureSelect CD CiberMed
Tissue panel outperformed whole-transcriptome sequencing
for all four samples.
Reproducibility
To assess reproducibility, RNA from all samples was
processed and sequenced in triplicate and the concordance
of iSort Fractions results across technical replicates was
evaluated. All pairwise comparisons across the three
replicates demonstrated strong reproducibility, with nearly
perfect correlations obtained for all samples (Figure 5).
0 20 40 60 80
0 20 40 60 80
iSort (%) FFPE
iSort (%) FF
rho = 0.93
P = 1 × 10−45
RMSE = 4.2
25 cell types
0 20 40 60 80
0 20 40 60 80
CIBERSORTx (%) FFPE
CIBERSO
RTx (%) FF
rho = 0.76
P = 7 × 10−20
RMSE = 4.8
25 cell types
0 20 40 60 80
0 20 40 60 80
iSort (%) FFPE
iSort (%) FF
rho = 0.78
P = 1 × 10−21
RMSE = 4.7
25 cell types
0 2 4 6 8 10 12
0 2 4 6 8 10
CIBERSORTx (%) FFPE
CIBERSO
RTx (%) FF
r = 0.67
P = 2 × 10−13
rho = 0.70
P = 1 × 10−14
RMSE = 2.2
23 cell types
Whole Transcriptome RNA-Seq
Whole transcriptome RNA-Seq
How
0 2 4 6 8 10 12
0 2 4 6 8 10
iSort (%) FFPE
iSort (%) FF
r = 0.74
P = 8 × 10−17
rho = 0.72
P = 5 × 10−16
RMSE = 1.8
23 cell types
0 2 4 6 8 10 12
0 2 4 6 8 10
iSort (%) FFPE
iSort (%) FF
r = 0.91
P = 4 × 10−35
rho = 0.92
P = 1 × 10−37
RMSE = 1
23 cell types
SureSelect CD CiberMed
Tissue Panel
iSort Fractions
Whole Transcriptome RNA-Seq
iSort Fractions
Whole Transcriptome RNA-Seq
CIBERSORTx
B cells naive
B cells memory
Plasma cells
T cells CD8
T CD4 naive
T CD4 memory resting
T CD4 memory activated
T cells follicular helper
T cells regulatory (Tregs)
T cells gamma delta
NK cells resting
NK cells activated
Monocytes
Macrophages M0
Macrophages M1
Macrophages M2
Dendritic cells resting
Dendritic cells activated
Mast cells resting
Mast cells activated
Eosinophils
Neutrophils
Fibroblasts
Endothelial cells
Epithelial cells
a b c
Source:
Method:
0 20 40 60 80
0 20 40 60 80
iSort (%) FFPE
iSort (%) FF
rho = 0.93
P = 1 × 10−45
RMSE = 4.2
25 cell types
0 20 40 60 80
0 20 40 60 80
CIBERSORTx (%) FFPE
CIBERSO
RTx (%) FF
rho = 0.76
P = 7 × 10−20
RMSE = 4.8
25 cell types
0 20 40 60 80
0 20 40 60 80
iSort (%) FFPE
iSort (%) FF
rho = 0.78
P = 1 × 10−21
RMSE = 4.7
25 cell types
0 2 4 6 8 10 12
0 2 4 6 8 10
CIBERSORTx (%) FFPE
CIBERSO
RTx (%) FF
r = 0.67
P = 2 × 10−13
rho = 0.70
P = 1 × 10−14
RMSE = 2.2
23 cell types
Whole Transcriptome RNA-Seq
Whole transcriptome RNA-Seq
How
0 2 4 6 8 10 12
0 2 4 6 8 10
iSort (%) FFPE
iSort (%) FF
r = 0.74
P = 8 × 10−17
rho = 0.72
P = 5 × 10−16
RMSE = 1.8
23 cell types
0 2 4 6 8 10 12
0 2 4 6 8 10
iSort (%) FFPE
iSort (%) FF
r = 0.91
P = 4 × 10−35
rho = 0.92
P = 1 × 10−37
RMSE = 1
23 cell types
SureSelect CD CiberMed
Tissue Panel
iSort Fractions
Whole Transcriptome RNA-Seq
iSort Fractions
Whole Transcriptome RNA-Seq
CIBERSORTx
B cells naive
B cells memory
Plasma cells
T cells CD8
T CD4 naive
T CD4 memory resting
T CD4 memory activated
T cells follicular helper
T cells regulatory (Tregs)
T cells gamma delta
NK cells resting
NK cells activated
Monocytes
Macrophages M0
Macrophages M1
Macrophages M2
Dendritic cells resting
Dendritic cells activated
Mast cells resting
Mast cells activated
Eosinophils
Neutrophils
Fibroblasts
Endothelial cells
Epithelial cells
a b c
Source:
Method:
0 20 40 60 80
0 20 40 60 80
iSort (%) FFPE
iSort (%) FF
rho = 0.93
P = 1 × 10−45
RMSE = 4.2
25 cell types
0 20 40 60 80
0 20 40 60 80
CIBERSORTx (%) FFPE
CIBERSO
RTx (%) FF
rho = 0.76
P = 7 × 10−20
RMSE = 4.8
25 cell types
0 20 40 60 80
0 20 40 60 80
iSort (%) FFPE
iSort (%) FF
rho = 0.78
P = 1 × 10−21
RMSE = 4.7
25 cell types
0 2 4 6 8 10 12
0 2 4 6 8 10
CIBERSORTx (%) FFPE
CIBERSO
RTx (%) FF
r = 0.67
P = 2 × 10−13
rho = 0.70
P = 1 × 10−14
RMSE = 2.2
23 cell types
Whole Transcriptome RNA-Seq
Whole transcriptome RNA-Seq
How
0 2 4 6 8 10 12
0 2 4 6 8 10
iSort (%) FFPE
iSort (%) FF
r = 0.74
P = 8 × 10−17
rho = 0.72
P = 5 × 10−16
RMSE = 1.8
23 cell types
0 2 4 6 8 10 12
0 2 4 6 8 10
iSort (%) FFPE
iSort (%) FF
r = 0.91
P = 4 × 10−35
rho = 0.92
P = 1 × 10−37
RMSE = 1
23 cell types
SureSelect CD CiberMed
Tissue Panel
iSort Fractions
Whole Transcriptome RNA-Seq
iSort Fractions
Whole Transcriptome RNA-Seq
CIBERSORTx
B cells naive
B cells memory
Plasma cells
T cells CD8
T CD4 naive
T CD4 memory resting
T CD4 memory activated
T cells follicular helper
T cells regulatory (Tregs)
T cells gamma delta
NK cells resting
NK cells activated
Monocytes
Macrophages M0
Macrophages M1
Macrophages M2
Dendritic cells resting
Dendritic cells activated
Mast cells resting
Mast cells activated
Eosinophils
Neutrophils
Fibroblasts
Endothelial cells
Epithelial cells
a b c
Source:
Method:
Figure 4. Box plots summarizing sample-level concordance of cell type fractions from paired FF and FFPE NSCLC specimens, analyzed by sample pair
across 23 cell types and stratified by sequencing assay (Agilent SureSelect CD CiberMed Tissue panel versus whole-transcriptome sequencing). Fibroblasts
and epithelial cells were excluded to focus on cell types in the range of 0 to 10% fractional abundance. Concordance was evaluated by (a) root mean squared
error (RMSE), (b) Spearman rho, and (c) Pearson r. Statistical significance was determined with a two-sided paired t-test. All results were obtained using iSort
Fractions tissue mode.
0.0
0.5
1.0
1.5
2.0
2.5
3.0
RMSE
SureSelect Whole
Transcriptome
Median 1 1.7
P = 0.04
0.70
0.75
0.80
0.85
0.90
0.95
1.00
Spearman rho
SureSelect Whole
Transcriptome
Median 0.93 0.78
P = 0.06
0.70
0.75
0.80
0.85
0.90
0.95
1.00
Pearson r
SureSelect Whole
Transcriptome
Median 0.93 0.78
P = 0.04
Sample pair 1
Sample pair 2
Sample pair 3
Sample pair 4
0 10 30 50
0 10 30 50
FF
iSort (%) (replicate 1)
iSort (%) (replicate 2)
r = 1.0
P < 1 × 10-10
rho = 0.98
P < 1 × 10−10
0 10 30 50
0 10 30 50
FF
iSort (%) (replicate 2)
iSort (%) (replicate 3)
r = 1.0
P < 1 × 10−10
rho = 0.98
P < 1 × 10−10
0 10 30 50
0 10 30 50
FF
iSort (%) (replicate 1)
iSort (%) (replicate 3)
r = 1.0
P < 1 × 10−10
rho = 0.97
P < 1 × 10−10
0 10 30 50
0 10 30 50
FFPE
iSort (%) (replicate 1)
iSort (%) (replicate 2)
r = 1.0
P < 1 × 10−10
rho = 0.98
P < 1 × 10−10
0 10 30 50
0 10 30 50
FFPE
iSort (%) (replicate 2)
iSort (%) (replicate 3)
r = 1.0
P < 1 × 10−10
rho = 0.96
P < 1 × 10−10
0 10 30 50
0 10 30 50
FFPE
iSort (%) (replicate 1)
iSort (%) (replicate 3)
r = 1.0
P < 1 × 10−10
rho = 0.96
P < 1 × 10−10
B cells naive
B cells memory
Plasma cells
T cells CD8
T CD4 naive
T CD4 memory resting
T CD4 memory activated
T cells follicular helper
T cells regulatory (Tregs)
T cells gamma delta
NK cells resting
NK cells activated
Monocytes
Macrophages M0
Macrophages M1
Macrophages M2
Dendritic cells resting
Dendritic cells activated
Mast cells resting
Mast cells activated
Eosinophils
Neutrophils
Fibroblasts
Endothelial cells
Epithelial cells
a
b
Figure 5. Scatterplots comparing iSort Fractions results between sequencing replicates, shown for (a) four FF samples and (b) four FFPE tumor samples. Each
data point represents a sample colored by cell type. The Agilent SureSelect CD CiberMed Tissue panel was applied to all samples.
8
Sequencing requirement
Samples were sequenced to an average of 50M (25M × 2)
reads for the SureSelect CD CiberMed Tissue panel.
To determine the minimum number of reads needed per
sample without compromising performance, we performed a
titration experiment. In this experiment, the number of effective
reads per sample was down-sampled to predefined quantities
before running iSort Fractions (where “effective reads” denotes
on-target reads only). Figure 6 summarizes the concordance
between FF and FFPE cell type fractions for all three replicates
using pre-determined numbers of effective reads. All three
replicates maintained accurate and stable performance down
to 1M (500k × 2) effective reads (Figure 6). Figure 7 shows all
data points from Replicate 1 before and after down-sampling
to 1M (500k × 2) effective reads.
Based on these data, when using the SureSelect CD CiberMed
Tissue panel, the total number of required input reads is
expected to range from approximately 1.3M per sample
(assuming a 75% on-target mapping rate) to approximately
2M per sample (assuming a 50% on-target rate). Therefore, a
conservative projection of 2M (1M × 2) total input reads per
sample should be sufficient. With the recommended input of
at least 40M (20M × 2) reads for iSort Fractions from wholetranscriptome sequencing, the SureSelect CD CiberMed
Tissue panel offers a 20-fold reduction in sequencing costs
while achieving superior performance.
Figure 6. Impact of reads per sample on the concordance between FF and FFPE cell type proportions determined by iSort Fractions. “All” denotes all evaluable
reads per sample. The Agilent SureSelect CD CiberMed Tissue panel maintained stable performance down to 1M (500k × 2) effective (“on-target”) reads per
sample for all three replicates. Because Pearson r and RMSE are heavily driven by abundant fibroblasts and epithelial cells, they are shown without these two cell
types. RMSE, root mean squared error.
0.3
0.4
0.5
0.6
0.7
0.8
0.9
1.0
23 cell types
Pearson r
All
5M
2M
1M
500k
100k
10k
0.0
0.5
1.0
1.5
2.0
2.5
3.0
3.5
23 cell types
RMSE
All
5M
2M
1M
500k
100k
10k
Replicate 1
Replicate 2
Replicate 3
0.3
0.4
0.5
0.6
0.7
0.8
0.9
1.0
25 cell types
Spearman rho
All
5M
2M
1M
500k
100k
10k
Number of effective reads per sample
9
Figure 7. Impact of reads per sample on the concordance between cell type fractions in paired FF and FFPE samples, comparing (a) all evaluable reads per
sample (no down-sampling) with (b) 1M (500k × 2) effective reads per sample (on-target reads per sample). The results are shown for all 25 cell types (top) and 23
cell types (bottom), with the latter expanding the fractional abundance range between 0 and 15%. Each data point denotes a sample colored by cell type. Results
were calculated using the Agilent SureSelect CD CiberMed Tissue panel applied to four matching pairs of FF (y-axis) and FFPE (x-axis) tumor specimens.
0 5 10 15
0 5 10 15
23 cell types
iSort (%) FFPE
iSort (%) FF
r = 0.90
P = 1 × 10−35
rho = 0.92
P = 1 × 10−37
RMSE = 1.0
Lorem ipsum
0 5 10 15
0 5 10 15
23 cell types
iSort (%) FFPE
iSort (%) FF
Lorem ipsum
r = 0.89
P = 3 × 10−33
rho = 0.89
P = 4 × 10-32
RMSE = 1.0
Lorem ipsum
a b All Reads 1M (500 k × 2) Effective Reads
0 10 30 50
0 10 30 50
25 cell types
iSort (%) FFPE
iSort (%) FF
r = 0.95
P = 1 x 10−51
rho = 0.93
P = 1 × 10−45
RMSE = 4.2
0 10 30 50
0 10 30 50
25 cell types
iSort (%) FFPE
iSort (%) FF
r = 0.96
P = 9 x 10−55
rho = 0.91
P = 1 × 10−39
RMSE = 4
B cells naive
B cells memory
Plasma cells
T cells CD8
T CD4 naive
T CD4 memory resting
T CD4 memory activated
T cells follicular helper
T cells regulatory (Tregs)
T cells gamma delta
NK cells resting
NK cells activated
Monocytes
Macrophages M0
Macrophages M1
Macrophages M2
Dendritic cells resting
Dendritic cells activated
Mast cells resting
Mast cells activated
Eosinophils
Neutrophils
Fibroblasts
Endothelial cells
Epithelial cells
10
Conclusion
In this application note, we describe the performance
characteristics of the SureSelect CD Tissue panel for profiling
both FF and FFPE tissue samples with iSort, a state-of-the-art
software solution for digital cytometry offered by CiberMed.
There are several applications that would benefit from the
enhanced robustness, accuracy, and cost-effectiveness
of this new joint assay for cytometry by sequencing.
These include routine and translational tissue analysis,35
retrospective characterization of bulk tissue expression data
to derive new insights into cellular composition,1
and largescale validation of sequencing analyses.36 Other applications
include the general assessment of cell type composition
under diverse physiological and pathological conditions,
all without the need for antibodies, fresh specimens, viable
material, or millions of cells.
As shown previously, by comparing FFPE samples to a
matched FF reference, the methodology is robust to FFPE
artifacts and recovers the cell type abundance profile of
matched references. Moreover, the Agilent SureSelect CD
Tissue panel substantially outperforms whole-transcriptome
profiling with potential to greatly reduce sequencing cost.
The utility of the core iSort methodology has been
demonstrated in different tissues and various cancer
types.27,30,47,48 It has also been demonstrated in multiple
other contexts, including immuno-oncology,37,38 organ
transplantation,39 cardiology,40 Crohn’s disease,30 and neonatal
sepsis.41 The ability to deconvolve alternative genomic data
types, including methylation and proteomic profiles, has
also been demonstrated.19,42 Furthermore, iSort has been
analytically validated for whole blood samples and fresh,
frozen, and fixed tumor specimens.27,30 The core algorithm
underlying iSort has become the standard methodology for
deconvolution in large cancer studies and datasets, such as
The Cancer Genome Atlas (TCGA), and is being applied in
clinical trials.43-46 Given the performance gains demonstrated
here, the SureSelect/iSort assay for digital cytometry
promises to facilitate many exciting and impactful future
applications.
References
1. Thorsson, V.; Gibbs, D. L.; Brown, S. D.; Wolf, D.; Bortone,
D. S.; Yang, T. H. O.; Porta-Pardo, E.; Gao, G. F.; Plaisier,
C. L.; Eddy, J. A.; et al. The Immune Landscape of
Cancer. Immunity. 2018, 48 (4), P812-830. https://doi.
org/10.1016/j.immuni.2018.03.023.
2. Farc, O.; Cristea, V. An Overview of the Tumor
Microenvironment, from Cells to Complex Networks
(Review). Exp. Ther. Med. 2020, 21 (1). https://doi.
org/10.3892/etm.2020.9528
3. Newman, A. M.; Liu, C. L.; Green, M. R.; Gentles, A. J.;
Feng, W.; Xu, Y.; Hoang, C. D.; Diehn, M.; Alizadeh, A.
A. Robust Enumeration of Cell Subsets from Tissue
Expression Profiles. Nat. Methods 2015, 12 (5), 453–
457. https://doi.org/10.1038/nmeth.3337.
4. Gentles, A. J.; Newman, A. M.; Liu, C. L.; Bratman, S. V.;
Feng, W.; Kim, D.; Nair, V. S.; Xu, Y.; Khuong, A.; Hoang,
C. D.; et al. The Prognostic Landscape of Genes and
Infiltrating Immune Cells across Human Cancers. Nat.
Med. 2015, 21 (8), 938–945. https://doi.org/10.1038/
nm.3909.
5. Bentham, R.; Litchfield, K.; Watkins, T. B. K.; Lim, E. L.;
Rosenthal, R.; Martínez-Ruiz, C.; Hiley, C. T.; Bakir, M. A.;
Salgado, R.; Moore, D. A.; et al. TRACERx Consortium.
Using DNA Sequencing Data to Quantify T Cell Fraction
and Therapy Response. Nature 2021, 597 (7877),
555–560. https://doi.org/10.1038/s41586-021-03894-
5.
6. Herold, N. C.; Mitra, P. Immunophenotyping; StatPearls
Publishing, 2023.
7. Alfonso, B.-F.; Al-Rubeai, M. Flow Cytometry. In
Comprehensive Biotechnology; Elsevier, 2011; pp
559–578.
8. Dixon, A. R.; Bathany, C.; Tsuei, M.; White, J.; Barald, K.
F.; Takayama, S. Recent Developments in Multiplexing
Techniques for Immunohistochemistry. Expert Rev.
Mol. Diagn. 2015, 15 (9), 1171–1186. https://doi.org/10
.1586/14737159.2015.1069182
9. Rashid, R.; Gaglia, G.; Chen, Y.-A.; Lin, J.-R.; Du, Z.;
Maliga, Z.; Schapiro, D.; Yapp, C.; Muhlich, J.; Sokolov,
A.; et al. Highly Multiplexed Immunofluorescence
Images and Single-Cell Data of Immune Markers in
Tonsil and Lung Cancer. Sci. Data 2019, 6 (1), 1–10.
https://doi.org/10.1038/s41597-019-0332-y.
11
10. Palit, S.; Heuser, C.; de Almeida, G. P.; Theis, F. J.;
Zielinski, C. E. Meeting the Challenges of HighDimensional Single-Cell Data Analysis in Immunology.
Front. Immunol. 2019, 10. https://doi.org/10.3389/
fimmu.2019.01515.
11. Lähnemann, D.; Köster, J.; Szczurek, E.; McCarthy, D. J.;
Hicks, S. C.; Robinson, M. D.; Vallejos, C. A.; Campbell,
K. R.; Beerenwinkel, N.; Mahfouz, A.; et al. Eleven Grand
Challenges in Single-Cell Data Science. Genome Biol.
2020, 21 (1). https://doi.org/10.1186/s13059-020-
1926-6.
12. Aran, D.; Hu, Z.; Butte, A. J. xCell: Digitally Portraying
the Tissue Cellular Heterogeneity Landscape. Genome
Biol. 2017, 18 (1). https://doi.org/10.1186/s13059-017-
1349-1.
13. Racle, J.; Gfeller, D. EPIC: A Tool to Estimate the
Proportions of Different Cell Types from Bulk Gene
Expression Data. In Bioinformatics for Cancer
Immunotherapy; Springer US: New York, NY, 2020; Vol.
2120, pp 233–248.
14. Li, B.; Li, T.; Liu, J. S.; Liu, X. S. Computational
Deconvolution of Tumor-Infiltrating Immune
Components with Bulk Tumor Gene Expression Data. In
Bioinformatics for Cancer Immunotherapy; Springer US:
New York, NY, 2020; Vol. 2120, pp 249–262.
15. Becht, E.; Giraldo, N. A.; Lacroix, L.; Buttard, B.; Elarouci,
N.; Petitprez, F.; Selves, J.; Laurent-Puig, P.; SautèsFridman, C.; Fridman, W. H.; de Reyniès, A. Estimating
the Population Abundance of Tissue-Infiltrating
Immune and Stromal Cell Populations Using Gene
Expression. Genome Biol. 2016, 17 (1). https://doi.
org/10.1186/s13059-016-1070-5.
16. Shen-Orr, S. S.; Tibshirani, R.; Butte, A. J. Gene
Expression Deconvolution in Linear Space. Nat.
Methods 2012, 9 (1), 9–9. https://doi.org/10.1038/
nmeth.1831.
17. Newman, A. M.; Steen, C. B.; Liu, C. L.; Gentles, A.
J.; Chaudhuri, A. A.; Scherer, F.; Khodadoust, M. S.;
Esfahani, M. S.; Luca, B. A.; Steiner, D.; et al. Determining
Cell Type Abundance and Expression from Bulk Tissues
with Digital Cytometry. Nat. Biotechnol. 2019, 37 (7),
773–782. https://doi.org/10.1038/s41587-019-0114-2.
18. Bionetworks, S. Synapse. Synapse.org. https://www.
synapse.org/ (accessed 2023-10-30).
19. Nadel, B. B.; Oliva, M.; Shou, B. L.; Mitchell, K.; Ma, F.;
Montoya, D. J.; Mouton, A.; Kim-Hellmuth, S.; Stranger,
B. E.; Pellegrini, M.; Mangul, S. Systematic Evaluation
of Transcriptomics-Based Deconvolution Methods and
References Using Thousands of Clinical Samples. Brief.
Bioinform. 2021, 22 (6). https://doi.org/10.1093/bib/
bbab265.
20. Avila Cobos, F.; Alquicira-Hernandez, J.; Powell, J. E.;
Mestdagh, P.; De Preter, K. Benchmarking of Cell Type
Deconvolution Pipelines for Transcriptomics Data.
Nat. Commun. 2020, 11 (1). https://doi.org/10.1038/
s41467-020-19015-1.
21. Le, T.; Aronow, R. A.; Kirshtein, A.; Shahriyari, L. A Review
of Digital Cytometry Methods: Estimating the Relative
Abundance of Cell Types in a Bulk of Cells. Brief.
Bioinform. 2021, 22 (4). https://doi.org/10.1093/bib/
bbaa219.
22. Chen, B.; Khodadoust, M. S.; Liu, C. L.; Newman, A.
M.; Alizadeh, A. A. Profiling Tumor Infiltrating Immune
Cells with CIBERSORT. In Methods in Molecular Biology;
Springer New York: New York, NY, 2018; Vol. 1711, pp
243–259.
23. Sutton, G. J.; Poppe, D.; Simmons, R. K.; Walsh, K.;
Nawaz, U.; Lister, R.; Gagnon-Bartsch, J. A.; Voineagu, I.
Comprehensive Evaluation of Deconvolution Methods
for Human Brain Gene Expression. Nat. Commun. 2022,
13 (1), 1–18. https://doi.org/10.1038/s41467-022-
28655-4.
24. Breen, M. S.; Ozcan, S.; Ramsey, J. M.; Wang, Z.;
Ma’ayan, A.; Rustogi, N.; Gottschalk, M. G.; Webster, M.
J.; Weickert, C. S.; Buxbaum, J. D.; Bahn, S. Temporal
Proteomic Profiling of Postnatal Human Cortical
Development. Transl. Psychiatry 2018, 8 (1), 1–14.
https://doi.org/10.1038/s41398-018-0306-4.
25. Li, L.; Shen, L.; Ma, J.; Zhou, Q.; Li, M.; Wu, H.; Wei,
M.; Zhang, D.; Wang, T.; Qin, S.; Xing, T. Evaluating
Distribution and Prognostic Value of New TumorInfiltrating Lymphocytes in HCC Based on a scRNA-Seq
Study with CIBERSORTx. Front. Med. (Lausanne) 2020,
7. https://doi.org/10.3389/fmed.2020.00451.
26. Newman, A.M.; Gulati, S.G.; Clarke, M.F.; Sikandar,
S.S. Methods Utilizing Single Cell Genetic Data for
Cell Population Analysis and Applications Thereof.
US202003700112 A1, November 26, 2020.
12
27. Newman, A. M.; Nakao, A.; Li, K.; Liu, C.-L.; Mathi,
K.; Sigal, N.; Maecker, H.; Diehn, M.; Alizadeh, A. A.
Analytical Validation of Digital Cytometry (iSort) for
Leukocyte Enumeration Using Stored Blood. J. Clin.
Oncol. 2020, 38 (15_suppl), 3542–3542. https://doi.
org/10.1200/jco.2020.38.15_suppl.3542.
28. Chakravarthy, A.; Furness, A.; Joshi, K.; Ghorani, E.;
Ford, K.; Ward, M. J.; King, E. V.; Lechner, M.; Marafioti,
T.; Quezada, S. A.; Thomas, G. J.; Feber, A.; Fenton, T.
R. Pan-Cancer Deconvolution of Tumour Composition
Using DNA Methylation. Nat. Commun. 2018, 9 (1).
https://doi.org/10.1038/s41467-018-05570-1.
29. Hutson, M. Hunting for the Best Bioscience Software
Tool? Check This Database. Nature 2023. https://doi.
org/10.1038/d41586-023-00053-w.
30. Chen, H.; Chen, C.; Yuan, X.; Xu, W.; Yang, M.-Q.; Li,
Q.; Shen, Z.; Yin, L. Identification of Immune Cell
Landscape and Construction of a Novel Diagnostic
Nomogram for Crohn’s Disease. Front. Genet. 2020, 11.
https://doi.org/10.3389/fgene.2020.00423.
31. A, V. Agilent.com. https://www.agilent.com/cs/library/
usermanuals/public/G9993-90010.pdf (accessed
2023-10-30).
32. Patro, R.; Duggal, G.; Love, M. I.; Irizarry, R. A.; Kingsford,
C. Salmon Provides Fast and Bias-Aware Quantification
of Transcript Expression. Nat. Methods 2017, 14 (4),
417–419. https://doi.org/10.1038/nmeth.4197.
33. Dobin, A.; Davis, C. A.; Schlesinger, F.; Drenkow, J.;
Zaleski, C.; Jha, S.; Batut, P.; Chaisson, M.; Gingeras,
T. R. STAR: Ultrafast Universal RNA-Seq Aligner.
Bioinformatics 2013, 29 (1), 15–21. https://doi.
org/10.1093/bioinformatics/bts635.
34. Li, B.; Dewey, C. N. RSEM: Accurate Transcript
Quantification from RNA-Seq Data with or without a
Reference Genome. BMC Bioinformatics 2011, 12 (1).
https://doi.org/10.1186/1471-2105-12-323.
35. Orange, D. E.; Yao, V.; Sawicka, K.; Fak, J.; Frank, M.
O.; Parveen, S.; Blachere, N. E.; Hale, C.; Zhang, F.;
Raychaudhuri, S.; et al. RNA Identification of PRIME
Cells Predicting Rheumatoid Arthritis Flares. N. Engl. J.
Med. 2020, 383 (3), 218–228. https://doi.org/10.1056/
nejmoa2004114.
36. Gohil, S. H.; Iorgulescu, J. B.; Braun, D. A.; Keskin,
D. B.; Livak, K. J. Applying High-Dimensional
Single-Cell Technologies to the Analysis of Cancer
Immunotherapy. Nat. Rev. Clin. Oncol. 2021, 18 (4),
244–256. https://doi.org/10.1038/s41571-020-
00449-x.
37. Nabet, B. Y.; Esfahani, M. S.; Moding, E. J.; Hamilton,
E. G.; Chabon, J. J.; Rizvi, H.; Steen, C. B.; Chaudhuri,
A. A.; Liu, C. L.; Hui, A. B.; et al. Noninvasive Early
Identification of Therapeutic Benefit from Immune
Checkpoint Inhibition. Cell 2020, 183 (2), 363-376.e13.
https://doi.org/10.1016/j.cell.2020.09.001.
38. Lozano, A. X.; Chaudhuri, A. A.; Nene, A.; Bacchiocchi,
A.; Earland, N.; Vesely, M. D.; Usmani, A.; Turner, B. E.;
Steen, C. B.; Luca, B. A.; et al. T Cell Characteristics
Associated with Toxicity to Immune Checkpoint
Blockade in Patients with Melanoma. Nat. Med. 2022,
28 (2), 353–362. https://doi.org/10.1038/s41591-021-
01623-z.
39. Huang, S.; Chen, H.; Guo, Z.; He, X. A Comprehensive
Cibersort Study and Gene-Expression Based
Transcriptomic Analysis on Patterns of Immune
Infiltration in Ischemia-Reperfusion Injury Livers
Post Liver Transplantation. Transplantation 2020,
104 (S3), S195–S195. https://doi.org/10.1097/01.
tp.0000699368.56252.17.
40. Journal-of-cardiology.com. https://www.journal-ofcardiology.com/article/S0914-5087(20)30289-6/
fulltext (accessed 2023-10-30).
41. Jiang, Z.; Luo, Y.; Wei, L.; Gu, R.; Zhang, X.; Zhou,
Y.; Zhang, S. Bioinformatic Analysis and Machine
Learning Methods in Neonatal Sepsis: Identification
of Biomarkers and Immune Infiltration. Biomedicines
2023, 11 (7), 1853. https://doi.org/10.3390/
biomedicines11071853.
42. Voss, M. H.; Buros Novik, J.; Hellmann, M. D.; Ball, M.;
Hakimi, A. A.; Miao, D.; Margolis, C.; Horak, C.; WindRotolo, M.; De Velasco, G.; et al. Correlation of Degree
of Tumor Immune Infiltration and Insertion-andDeletion (Indel) Burden with Outcome on Programmed
Death 1 (PD1) Therapy in Advanced Renal Cell Cancer
(RCC). J. Clin. Oncol. 2018, 36 (15_suppl), 4518–4518.
https://doi.org/10.1200/jco.2018.36.15_suppl.4518.
43. Metzger Filho, O.; Stover, D. G.; Asad, S.; Ansell, P. J.;
Watson, M.; Loibl, S.; Geyer, C. E.; O’Shaughnessy, J.;
Untch, M.; Rugo, H. S.; et al. Immunophenotype and
Proliferation to Predict for Response to Neoadjuvant
Chemotherapy in TNBC: Results from BrighTNess
Phase III Study. J. Clin. Oncol. 2019, 37 (15_suppl),
510–510. https://doi.org/10.1200/jco.2019.37.15_
suppl.510.
44. CTG labs - NCBI. Clinicaltrials.gov. https://clinicaltrials.
gov/study/NCT03979508 (accessed 2023-10-30).
45. Clinicaltrials.gov. https://classic.clinicaltrials.gov/
ProvidedDocs/10/NCT03955510/Prot_000.pdf
(accessed 2023-10-30).
46. Medjebar, S.; Richard, C.; Fumet, J.-D.; Malo, J.; Elkrief,
A.; Blais, N.; Tehfe, M.; Florescu, M.; Boidot, R.; Truntzer,
C.; et al. Angiotensin-Converting Enzyme Inhibitor
Prescription Is Associated with Decreased ProgressionFree Survival (PFS) and Overall Survival (OS) in
Patients with Lung Cancers Treated with PD-1/PD-L1
Immune Checkpoint Blockers. J. Clin. Oncol. 2019, 37
(15_suppl), e20512–e20512. https://doi.org/10.1200/
jco.2019.37.15_suppl.e20512
www.agilent.com
For Research Use Only. Not for use in diagnostic procedures.
PR7001-2303
This information is subject to change without notice.
© Agilent Technologies, Inc. 2024
Published in the USA, March 15, 2024
5994-7248EN
47. Steen, C. B.; Liu, C. L.; Alizadeh, A. A.; Newman, A. M.
Profiling Cell Type Abundance and Expression in Bulk
Tissues with CIBERSORTx. Methods in Molecular
Biology 2020, 135–157. https://doi.org/10.1007/978-1-
0716-0301-7_7
48. Craven, K. E.; Gökmen-Polar, Y.; Badve, S. S. CIBERSORT
Analysis of TCGA and METABRIC Identifies Subgroups
with Better Outcomes in Triple Negative Breast Cancer.
Scientific Reports 2021, 11 (1). https://doi.org/10.1038/
s41598-021-83913-7
49. Newman, A.M. et al, J. Clinical Oncology, 38, Supp.
e15243 (2020) https://ascopubs.org/doi/abs/10.1200/
JCO.2020.38.15_suppl.e15243