Shi 2021

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Genomics 113 (2021) 1988–1998

Contents lists available at ScienceDirect

Genomics
journal homepage: www.elsevier.com/locate/ygeno

Original Article

MIN score predicts primary response to infliximab/adalimumab and


vedolizumab therapy in patients with inflammatory bowel diseases☆
Yuan Shi a, 1, Wei He b, 1, Ming Zhong a, *, Minhao Yu a, *
a
Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
b
Department of Pathology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China

A R T I C L E I N F O A B S T R A C T

Keywords: Infliximab/adalimumab (IFX/ADA) and vedolizumab (VDZ) are the most widely used biologics in inflammatory
Inflammatory bowel disease bowel diseases. Current models used to predict their efficacies are restricted to either Crohn’s disease or ul­
Infliximab cerative colitis or to only one type of biologic, which makes them limited in external validation. We therefore
Response predictors
designed a comprehensive comparison among these models to identify the most meaningful predictors for patient
responses. Several biomarkers and models were compared for their abilities to predict both IFX/ADA and VDZ
responses by receiver operating characteristic curves. Least absolute shrinkage and selection operator regression
was adopted to determine a simplified gene signature. Verification was performed in biopsy samples by
immunohistochemical staining. The GIMATS module (based on counts of IgG plasma cells, inflammatory
monocytes, activated T cells, and stromal cells) had the best overall performance for response prediction in both
biologics (IFX/ADA, AUC = 0.720–0.853; VDZ, AUC = 0.661–0.728). Based on this module, patients were
equally divided into 3 groups: M type (GIMATS-low, metabolism), with a preference for IFX/ADA; I type
(GIMATS-high, immune), with a preference for VDZ; and N type (GIMATS-medium, normal), with no preference
for either treatment. Furthermore, to improve clinical utility, a simplified 6-gene model, MIN score, was
established to determine the baseline expression of G0S2, S100A9, SELE, CHI3L1, MMP1 and CXCL13 and
function as a substitute for GIMATS module. Our study suggested that the classification of metabolic or immune
type by MIN score was valuable for IBD diagnosis to assist with selection of IFX/ADA and VDZ.

1. Introduction injuries [2]. As a result, therapies for IBD have been developed to block
these processes.
Inflammatory bowel diseases (IBD), Crohn’s disease (CD) and ul­ The anti-TNF-α antibodies, including infliximab or adalimumab
cerative colitis (UC), are progressive inflammatory disorders in the (IFX/ADA), and the gut-selective therapy, such as vedolizumab (VDZ),
gastrointestinal tract. The aetiology of IBD remains unclear, but agree­ are two of the most widely used biologics for treatment in both CD and
ment has been reached on the crucial role of immune dysfunction as well UC, but approximately one-third of patients receiving IFX/ADA treat­
as other factors, such as environmental exposure, genetic susceptibility ment will show primary non-response, while another one-third will
and intestinal flora disturbance. undergo secondary loss of response [3]. The efficacies of these therapies
Currently, the mainstream view is that the imbalance between pro- are hard to predict. Moreover, given that both IFX/ADA and VDZ have
inflammatory Th (T helper) 17 and anti-inflammatory Treg (regulato­ significant therapeutic effects compared with placebo [4,5], physicians’
ry) cell differentiation plays a significant part in the pathogenesis of prescriptions can influence the treatment outcome. For patients with
abnormal intestinal immunity in IBD [1] and promotes the action of IFX/ADA treatment failure, an equal dosage of VDZ shows reduced ef­
molecules such as tumour necrosis factor α (TNF-α), interleukin-1 beta ficacy compared to that in those naïve to IFX/ADA [6,7], indicating the
(IL-1β), IL-6, and IL-23 to accelerate the development of mucosal need for prospective guidelines for first-line decision-making.


This work was supported by the National Natural Science Foundation of China (81702300, 81873555). The authors declared no financial disclosures or conflicts
of interest.
* Corresponding authors at: 160 Pujian Rd, Shanghai, China.
E-mail addresses: drzhongming@sjtu.edu.cn (M. Zhong), fishmeangood@163.com (M. Yu).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.ygeno.2021.04.011
Received 8 October 2020; Received in revised form 8 February 2021; Accepted 5 April 2021
Available online 16 April 2021
0888-7543/© 2021 Elsevier Inc. All rights reserved.
Y. Shi et al. Genomics 113 (2021) 1988–1998

Though controversial, many studies have reported predictors of 2.3. Calculation of immune signatures and models
biological efficacy from the perspective of immunologic features, patient
characteristics (e.g., age, gender, body mass index, etc.) and disease Previously, several models have been established to facilitate quan­
progression. Both the innate and adaptive immune systems have been tified evaluations of immune infiltration according to distinct genetic
studied. Genes related to bacterial component recognition and cytokine features of immune cells.
signalling are important for the IFX/ADA response [8]. Several genes, The ESTIMATE algorithm was introduced to determine fractions of
including TNF receptor superfamily 11B (TNFRSF11B, osteoprotegerin), stromal and immune cells using gene expression signatures by R package
stanniocalcin-1 (STC1), prostaglandin-endoperoxide synthase 2 (version 1.0.13) [18]. Immune signatures were constituted with genes
(PTGS2), interleukin 13 receptor alpha 2 (IL13RA2), interleukin 11 (IL- involved in both infiltration of immune cells and the process of
11) [9] and oncostatin M (OSM) [10], were identified as potential bio­ haematopoiesis.
markers of IFX/ADA resistance. In addition, a few studies have shown Specific tumour-infiltrating lymphocytes (TILs) were evaluated
that the expression of integrin α4β7 [11], the level of IL-6 in circulating through two approaches in this study, gene set enrichment analysis
blood [12], and a 4-gene signature of regulator of G protein signalling 13 (GSEA) and deconvolution. For the first mentioned of two, the 28
(RGS13), Dachsous cadherin-related 2 (DCHS2), cilia and flagella asso­ immune-related signatures, as described by Charoentong et al. [19],
ciated protein 91 (MAATS1) and Piwi like RNA-mediated gene silencing expressed in neither cancer cell lines nor normal tissues. The single
1 (PIWIL1) at baseline [13] could predict the response to VDZ. sample GSEA (ssGSEA) was used to decompose cellular profiles and
However, most of these models have been restricted to either a single generate 28 subpopulations of TILs via GSVA (version 1.20.0) R pack­
disease (CD or UC) or only one type of biologic, presenting challenges for ages [20]. The expression level of each gene was normalized across all
application to clinical practices. Therefore, we explored public cohort patients using z-score. In addition, one of the most popular deconvolu­
data with a number of methodologies, including ESTIMATE (Estimation tion methods, CIBERSORT (https://cibersort.stanford.edu/), were
of stromal and immune cells in malignant tumour tissues using expres­ employed to calculate the score of 22 cell types using characteristics of
sion data) and the GIMATS (IgG plasma cells, inflammatory mono­ 547 marker genes [21]. Since these two methods produced similar re­
nuclear phagocytes, activated T cells, and stromal cells) module, to sults, we only compared the abilities of the 28 TILs subpopulations with
discover the decisive predictors for biologic selection and to provide other biomarkers to predict patients’ response towards IFX/ADA and
clues regarding the underlying mechanisms of the treatment response. VDZ. Scores of 22 cell types were used for further explanations of
mechanisms underlying GIMATS -high and -low subgroups.
2. Methods The GIMATS module score was calculated based on IgG plasma cells,
inflammatory mononuclear phagocytes, activated T cells, and stromal
2.1. Study population cells. It was derived by single-cell analyses of inflamed tissues from anti-
TNF-α-treated ileum CD patients. Genes listed as Subtype Set-high and
The flowchart of this study was shown in Fig. S1. All data for bio­ -low group were displayed in Table S3. Datasets were transformed to z-
informatic analyses were obtained from the GEO (GSE12251, GSE16879 scores. Transformed genes that belonged to Subtype Set-high and -low
and GSE73661, www.ncbi.nlm.nih.gov/geo) and SRA databases were averaged and projected onto the GIMATS module and “No module”
(ERP115036, SRP077046, SRP100787, ERP114636, and ERP113396) as described by Martin et al. [22].
according to the following inclusion criteria: organism: Homo sapiens;
expression profiling by array; samples with IBD; samples obtained from 2.4. DEGs analysis
mucosal biopsy (Table S1). Sample exclusion schemes were shown in
Fig. S2. DEGs analysis was performed with the DESeq2 (version 1.38.0) R
Besides, mucosal biopsies of patients with (n = 4) or with no (n = 4) package through the tximport (version 1.0.3) R package [23,24], using
response to IFX therapy were obtained from Renji Hospital, School of the raw count produced by Kallisto. P-values were adjusted (adj.p) with
medicine, Shanghai Jiao Tong University, for immunohistochemical the Benjamini and Hochberg method. DEGs with a fold change >1 and
staining (IHC). Baseline clinical characteristics of these patients were an adj.p < 0.05 between two groups were considered to have statistical
summarized in Table S2. Ethics approvals of collection and preservation significance.
of these samples were obtained from Institute Research Ethics Com­ The clusterProfiler (version 3.14.3) R package was used to match
mittee of Renji Hospital, and informed consents were signed following biological pathways for genes that found to be up- or down-regulated in
the Declaration of Helsinki [14]. Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway enrich­
ment analyses with a cut-off value of an adj.p < 0.05 [25].
The weighted gene co-expression network analysis (WGCNA) was
2.2. The expression profiles of RNA sequencing (RNA-seq) and performed to construct a co-expression network of specific genes in IBD
microarray samples via the WGCNA (version 1.69) R package [26]. The minimum
module size was 30, and the module detection sensitivity of deepSplit
Quality assessments of ERP115036, SRP077046, SRP100787, was 2, while the cut-off height for the merging of the modules was 0.2,
ERP114636 and ERP113396 were performed using FastQC software meaning that modules whose eigengenes had correlation coefficients
(version 0.11.7) (http://www.bioinformatics.babraham.ac.uk/project above 0.8 were merged.
s/fastqc/). To quantify gene expression, reads of all RNA-seq samples
were mapped to filtered assembled transcriptomes with the Kallisto 2.5. Definition of outcomes
(version 0.43.0) R package [15]. Gene expression values were presented
either as transcript per million (TPM) for model/marker calculation or Treatment efficacies were evaluated under endoscopy by either
as raw counts for differentially expressed genes (DEGs) analysis. Batch Simple Endoscopic Score for CD (SES-CD) [27] or Mayo Endoscopic
effects were moved by the sva (version 3.34.0) R package [16]. Subscore (MES) [28] with a histological activity system [29] at an in­
Raw CEL data were imported into R through the Affy (version 1.54.0) terval of 4–6 weeks. SES-CD represented the sum of scores for ulcer size,
R package, and robust multi-array (RMA)-based correction was per­ ulcerated surface, affected surface, and luminal narrowing in each in­
formed to determine the comparability of expression values among all testinal segment. In CD patients, the response was defined as the
samples [17]. For merged microarray dataset (GSE12251, GSE16879, achievement of a SES-CD ≤ 2. Meanwhile, MES was one of the most
GSE73661), batch effects were moved by the sva (version 3.34.0) R widely used indexes for UC, in which patients were given 0 (normal or
package. inactive disease) to 3 (spontaneous bleeding or ulceration) according to

1989
Y. Shi et al. Genomics 113 (2021) 1988–1998

Fig. 1. The top 5 biomarkers for IFX/ADA treatment response prediction in microarray (A) and RNA-seq datasets (B). The AUCs of different biomarkers for IFX/ADA
treatment response prediction (C). The top 5 biomarkers for VDZ treatment response prediction in microarray (D) and RNA-seq datasets (E). The AUCs of different
biomarkers for VDZ treatment response prediction (F). The logistic regression of the low, median and high GIMATS module score for IFX/ADA or VDZ treatment
response (G). In (C) (F), reference lines represented boundaries between top 5 predictors and the rest in corresponding datasets.
Abbreviations: ADA, adalimumab; AUC, area under the curve; CI, confident interval; GIMATS, IgG plasma cells, inflammatory mononuclear phagocytes, activated T
cells, and stromal cells; IFX, infliximab; OR, odds ratio; RNA-seq, RNA sequencing; ROC, receiver operating characteristic curve; VDZ, vedolizumab

1990
Y. Shi et al. Genomics 113 (2021) 1988–1998

Fig. 2. Estimated abundances of immune cells (A) and accumulations of monocyte/macrophage subgroups (B) among different GIMATS module groups. Abbrevi­
ations: NK, natural killer; GIMATS, IgG plasma cells, inflammatory mononuclear phagocytes, activated T cells, and stromal cells.

their clinical presentation and mucosal appearance. For UC patients, the considered statistically significant. Other R packages adopted in this
response was considered to be complete mucosal healing when the MES study included ggplot2 (version 3.3.2) [35], pheatmap (version 1.0.12)
was ≤1 and the histological score was Grade 0–1, indicating only [36], corrplot (version 0.84) [37], survminer (version 0.4.6) [38], pROC
structural change or chronic inflammation presented in haematoxylin- (version 1.16.1) [39], forestplot (version 1.9) [40] and rms (version
eosin sections. 6.0–1) [41].

2.6. Protein-protein interaction (PPI) network hub genes 3. Results

To explore gene interactions, the search tool STRING 10.5 3.1. The comparison between markers to predict the response of IFX/ADA
(http://string-db.org) [30] was employed to establish a DEGs PPI or VDZ
network, which was drawn by Cytoscape (version 3.6.1) [31]. A com­
bined score > 0.7 was set as the cut-off point. The maximal clique The enrichment levels of 28 immune signatures, 2 related immune
centrality (MCC) algorithm of CytoHubba (version 0.1) was used to models (the ESTIMATE score and GIMATS module), and the expression
explore PPI network hub genes [32]. ClueGO (version 2.5.4) facilitated of several genes, TNFRSF11B, STC1, PTGS2, IL13RA2, IL11, and OSM,
the visualization of the results [33]. were previously identified as predictive biomarkers in IFX/ADA
response. They were assessed for prediction abilities by ROC in merged
2.7. Immunohistochemistry microarray cohort (GSE12251, GSE16879 and GSE73661) and the RNA-
seq cohort (ERP113396) (Table S4).
Biopsy samples from IBD patients were fixed in 4% para­ The top 5 immune signatures with the highest area under the curve
formaldehyde followed by paraffin embedding. After deparaffinization (AUC) values in microarray data were shown in Fig. 1A: IL13RA2 (AUC
of the tissue sections, antigen unmasking was performed with heat- = 0.927), immature dendritic cells (AUC = 0.895), TNFRSF11B (AUC =
induced epitope retrieval in 10 mM citrate buffer for 10 min. Next, the 0.873), GIMATS module (AUC = 0.853) and STC1 (AUC = 0.850).
sections were incubated with 3% hydrogen peroxide for 30 min and Furthermore, in the RNA-seq dataset (Fig. 1B), OSM exhibited the
blocked with 5% BSA for 1 h at room temperature. Serial tissue sections highest AUC of 0.752, followed by IL11 (AUC = 0.726), GIMATS (AUC
were stained with primary antibodies against G0S2 (12091–1-AP, Pro­ = 0.720), PTGS2 (AUC = 0.720) and neutrophil cells (AUC = 0.718). In
teintech), CXCL13 (10927–1-AP, Proteintech), MMP1 (10371–2-AP, general, only the GIMATS module showed satisfactory robustness and
Proteintech), CHI3L1 (12036–1-AP, Proteintech), E-selectin (20894–1- validity in both datasets (Fig. 1C).
AP, Proteintech) or S100A9 (26992–1-AP, Proteintech) and a goat anti- To evaluate the capabilities to predict VDZ response, immune models
rabbit IgG H&L (HRP) secondary antibody (ab6721, Abcam) and and some other predictive markers, RGS13, DCHS2, MAATS1, and
counterstained with haematoxylin. Sections were visualized by a Nikon PIWIL1, were utilized in the microarray (GSE73661) and RNA-seq
microscope equipped with a Nikon camera and Vision 4.1 software in cohort (ERP114636). Exact AUC values of all the features were pre­
three different fields at 20× and 40× magnification for each sample. sented in Table S5. For the microarray data (Fig. 1D), the GIMATS
Patients’ response status to IFX was blind to both experimenters and module and natural killer cells showed the best performance, followed
researchers who interpreted staining results. by activated dendritic cells (AUC = 0.634), mast cells (AUC = 0.634)
and gamma delta T cells (AUC = 0.629). Besides, in the RNA-seq cohort
2.8. Statistics and analysis (Fig. 1E), the top 5 signatures were type 2 T helper cells (AUC = 0.754),
the GIMATS module (AUC = 0.728), activated CD4+ T cells (AUC =
To extract the core subset of data to determine the GIMATS module, a 0.726), the Stromal score (AUC = 0.716), and natural killer T cells (AUC
linear regression technique was used based on the least absolute = 0.716).
shrinkage and selection operator (LASSO) algorithm as implemented in The data above demonstrated that the GIMATS module had the best
the glmnet (version 2.0–16) R package [34]. The 500-bootstrap resam­ overall performance in both IFX/ADA and VDZ treatment response
pling procedure was used in the validation set to build confidence in­ prediction (Fig. 1F).
tervals for receiver operating characteristic curves (ROC). Logistic
regression was performed using the stats (version 3.4.1) R packages. All
statistical tests were two-sided, and p-values less than 0.05 were

1991
Y. Shi et al. Genomics 113 (2021) 1988–1998

Fig. 3. Biologics response correlated with distinct functional pathways. The KEGG analysis of differential gene expressions between GIMATS-high and -low groups
(A). The dendrogram of nine distinct modules defined by the WGCNA (B). Module-trait relationship revealed both positive and negative correlation with clinical
traits and modules. Each row in the table corresponded to a module and each column to a specific clinical trait. Values listed in the heatmap showed the correlation
coefficient and P-value within parentheses. The colour of the cell indicated the correlation coefficient between the traits: red indicates a high positive correlation and
blue a high negative correlation (C).
Abbreviations: KEGG, Kyoto Encyclopaedia of Genes and Genomes; WGCNA, weighted gene co-expression network analysis; GIMATS, IgG plasma cells, inflammatory
mononuclear phagocytes, activated T cells, and stromal cells. (For interpretation of the references to colour in this figure legend, the reader is referred to the web
version of this article.)

3.2. The GIMATS module provides assistance for treatment selection macrophages decreased (p < 0.001). This analysis confirmed the
different activation patterns of the innate immune system among
Patients from each dataset were divided into three equal groups GIMATS groups.
according to their GIMATS module values and assessed by the logistic
regression model (Fig. 1G, Tables S6–7). Of note, compared to patients 3.4. Differentially expressed genes between the high and low GIMATS
with low GIMATS module scores, those with high GIMATS module groups
scores showed a significantly worse response to IFX/ADA therapy
[microarray: odds ratio (OR) = 0.01, 95% confidence interval (CI) = To further identify the underlying mechanisms of these features,
0.00–0.06, p < 0.001; RNA-seq: OR = 0.17, 95%CI = 0.03–0.79, p = DEGs between the GIMATS -high and -low patients were determined
0.032) but tended to have better clinical outcomes when receiving VDZ based on the thresholds of |log2(fold-change)| > 1 and p < 0.05,
(microarray: OR = 6.00, 95%CI = 0.72–129.52, p = 0.138; RNA-seq: OR revealing 2006 upregulated and 1332 downregulated genes (Table S9).
= 6.60, 95%CI = 1.50–34.76, p = 0.017). Moreover, KEGG analysis was conducted, and Fig. 3A summarized
On the other hand, the response rate to IFX/ADA or VDZ seemed to the results for identified signatures. The GIMATS-high group was char­
show no significant difference in patients with a median GIMATS acterized by the enrichment of immune-associated pathways, including
module score, who may therefore benefit from either biologic. cytokine-cytokine receptor interaction, IL-17, chemokine and TNF sig­
nalling, whereas in the GIMATS-low group, metabolic pathways were
3.3. Estimated abundance of immune cells among the different GIMATS predominantly enriched, such as retinol, drug and xenobiotic meta­
groups bolism. In addition, 36.9%, 43.4%, and 19.7% of CD patients were
divided into the GIMATS-low, − median, and -high groups, respectively,
The gene expression profile obtained from the merged RNA-seq co­ while 28.1%, 25.0%, and 46.9% of the UC patients belonged to the
horts (ERP115036, SRP077046, SRP100787, ERP114636, and GIMATS-low, − median, and -high groups, respectively. UC patients
ERP113396, n = 216) was used to calculate the estimated abundance of were more likely to be assigned to the GIMATS-high group (Fig. S3,
22 types of tumour-infiltrating immune cells in the 3 GIMATS groups by Fisher’s test p < 0.001).
the CIBERSORT algorithm (Fig. 2A). Apart from the cell populations in
the GIMATS module (inflammatory macrophages, activated dendritic 3.5. Weighted gene coexpression network analysis (WGCNA) identified
cells and highly activated T cells), monocytes and M0 macrophages were the hub genes of each GIMATS group
also observed to be highly accumulated in the GIMATS-high group
compared with those in the GIMATS-low group (p = 0.010, <0.001, Biological components and processes are expected to be regulated in
respectively, Fig. 2B; Table S8), while the abundance of M2 a coordinated manner, resulting in the coexpression of genes that are

1992
Y. Shi et al. Genomics 113 (2021) 1988–1998

Fig. 4. Protein-protein interaction (PPI) networks established with 50 hub genes in each GIMATS module. The venn diagram of the intersection of GIMATS-related
DEGs and genes from the GIMATS-related WGCNA module (A, D and G). The top 50 hub genes from the PPI network ranked by maximal clique centrality (MCC)
algorithm (B, E and H). Edges represented protein-protein associations. The red nodes represented genes with high MCC sores, while the yellow node represented
genes with low MCC sores. The relationship between DEGs and GO terms with a p-value <0.05 in ClueGO network diagrams (C, F and I). Nodes represented pathways
that were significantly enriched. Size of coloured nodes indicated the enrichment significance. The edges were kappa scores that measured the interrelatedness of
nodes. A thick edge that represented annotated genes had previously been experimentally verified to be part of that term/pathway. The thin one represented that
most genes were inferred by electronic annotation (IEA). Some edges were bended for a better visualization.
Abbreviations: DEGs, differentially expressed genes; GO, gene ontology; GIMATS, IgG plasma cells, inflammatory mononuclear phagocytes, activated T cells, and
stromal cells. (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

1993
Y. Shi et al. Genomics 113 (2021) 1988–1998

(caption on next page)

1994
Y. Shi et al. Genomics 113 (2021) 1988–1998

Fig. 5. A 6-gene response prediction model, MIN score, based on LASSO regression (A). Satisfactory linear dependences of MIN score in both training (B, microarray,
n = 95) and validation (C, RNA-seq, n = 216) cohorts. The logistic regression of the M, N, I type for IFX/ADA or VDZ treatment response (D). Abbreviations: CD,
Crohn’s disease; UC, ulcerative colitis; GIMATS, IgG plasma cells, inflammatory mononuclear phagocytes, activated T cells, and stromal cells; Inf.Mac, inflammatory
macrophage; AC.DC, activated dendritic cell; pDC, plasmacytoid dendritic cell; ACKR1.Ecs, ACKR1 + endothelial cell; AC.Fib, activated fibroblast; res.Macs, resident
macrophage; ILCs, innate lymphoid cells; T1cyTRM, type 1 cytokines tissue-resident memory T cell; enteric_neu, enteric neurons; CD36EC, CD36+ endothelial cell;
ADA, adalimumab; IFX, infliximab; VDZ, vedolizumab; AUC, area under the curve; LASSO, least absolute shrinkage and selection operator; RNA-seq, RNA sequencing

associated with the same protein complex or biological pathway. (AUC = 0.958, 95%CI = 0.927–0.982).
Therefore, WGCNA was applied to classify similarly expressed genes into To evaluate the predictive performance of MIN score, ROC and
modules and identify the protein-based coexpression networks in our precision-recall curves were shown in Fig. S5 and Table S11. Moreover,
study. Nine distinct modules were generated through a hierarchical a logistic regression model was used to examine the predictive value of
clustering tree, as shown in the dendrogram (Fig. 3B). A total number of MIN score (Fig. 5D). IBD patients were divided into three equal cohorts.
718 genes in the turquoise module were found to be related to the Notably, compared with patients with low MIN score (GIMATS M type),
GIMATS-low signature (r = 0.60, p < 0.001, Fig. 3C, Fig. 4A-C), among those with a high MIN score, which corresponded to the GIMATS I type,
which 451 genes were significantly downregulated. had poorer rates of response to IFX/ADA therapy (microarray: OR =
To further explore the interactions among these genes, we selected 0.01, 95%CI = 0.00–0.06, p < 0.001; RNA-seq: OR = 0.06, 95%CI =
the top 50 genes (hub genes) (Table S10), ranked by the connectivity 0.01–0.31, p = 0.003), but were more likely to respond to VDZ
within the corresponding module, to draw the PPI network. It could be (microarray: OR = 6.00, 95%CI = 0.72–129.52, p = 0.138; RNA-seq: OR
seen that metabolic processes associated with the PPAR and adipocy­ = 9.53, 95%CI = 2.04–57.78, p = 0.007). Patients with a median MIN
tokine signalling pathways as well as fatty acid beta-oxidation were the score (N type) displayed no remarkable preference for either of the two
core of the relationships within the network (Fig. 4A). medications.
In addition, the green and brown modules were found to be associ­ This 6-gene panel was further confirmed in our IBD patient cohort
ated with the GIMATS-high module (r = 0.69 and 0.61, p < 0.001 and < using immunohistochemistry (IHC). As expected, the results showed
0.001, respectively, Fig. 3C). Within them, 122 and 166 genes were that G0S2, CXCL13, MMP1, CHI3L1, SELE and S100A9 staining were
significantly upregulated. The top 50 hub genes were identified in each much stronger in biopsies from patients with poor response to IFX
group. Genes associated with the IL-17 signalling pathway, cytokine- (Fig. 6). We found that CHI3L1 and CXCL13 were widely detected in
cytokine receptor interaction and leukocyte migration showed remark­ biopsies from patients with poor response, but were barely seen in those
able enrichment in the green module (Fig. 4D-F), while genes involved with good response. Of note, SELE was expressed solely in endothelial
in extracellular matrix organization, ECM-receptor interaction, blood cells of specimens from poorly responded patients and was relatively
vessel development and endodermal cell differentiation were enriched weak in well responded patients’ samples. Likewise, MMP1 and G0S2
in the brown module (Fig. 4G-I). expression were found in endothelial cells. In contrast, S100A9 was
The data above demonstrated that the GIMATS-low group was predominantly expressed in infiltrating immune cells, which was much
typically associated with active metabolic processes, and therefore, we more obvious in specimens from patients with a good response.
named it the metabolic type (M type); the GIMATS-high group was Collectively, these results suggested that the MIN score could be used to
identified as the immune type (I type) due to its close association with predict the response of IBD patients to IFX therapy.
immune activation. The remaining GIMATS-median group was consid­
ered to be the normal type (N type). 4. Discussion

In this study, we applied various immune-related models to predict


3.6. Establishment of a simplified 6-gene model by LASSO regression the IBD biological response. Among the validated signatures, GIMATS
module showed satisfactory prediction precision for both IFX/ADA and
The GIMATS module exhibited an ability to classify IBD patients into VDZ datasets. Therefore, further analyses were focused on GIMATS
M, I and N type, who can probably benefit from IFX/ADA or VDZ or both module and its related features of gene expression. Since GIMATS
therapies, respectively. To make it more convenient for practical utili­ module was originally generated in CD patients, we proved that it can be
zation, we performed a sparse regression analysis according to a unified extended to general scenarios that also include UC patients. In addition,
score based on the expression of the associated genes. the LASSO algorithm was used to reduce the data dimensionality and
Gene expressions in the training cohort of 95 patients (GSE12251, select the best weighting coefficients, resulting in a simple but practical
GSE16879 and part of GSE73661) were analysed with a statistical 6-gene signature, MIN score, which optimizes the clinical feasibility for
regression algorithm based on LASSO. To prevent over-fitting and data pre-treatment testing.
leakage during the process of model establishment, we used 5-fold cross As a scoring system based on the inflammatory status, a low MIN
validation. The training data (microarray, n = 95) was shuffled score reveals the presence of relatively weak inflammation in the bio­
randomly and split into 5 groups. For each unique group, we defined psied mucosa. Intriguingly, functional analyses found that the corre­
itself as a test set and the remaining groups as one training set. The sponding group of patients (M type) usually showed more active
training set was used to establish LASSO model, which was then un­ metabolic activities, highlighting the significant role of metabolism,
derwent evaluations in the test set. The average accuracy was 0.863. especially fatty acid catabolic processes, in the pathogenesis of IBD and
Finally, an optimized 6-gene signature (MIN score) was yielded as a the function of TNF-α inhibitors.
weighted sum of expression levels of 6 genes for each patient (Fig. 5A, Intense intestinal inflammation is associated with elevated serum
Tables S6–7). The MIN score was calculated according to the following levels of free fatty acids, which provide additional rich energy supplies
formula: MIN score = G0S2 × 0.157 + S100A9 × 0.610 + SELE×0.268 to immune cells at the sites of inflammation despite the presence of
+ CHI3L1 × 0.896 + MMP1 × 0.500 + CXCL13 × 0.564. glucose [42], while the increases in the pro-inflammatory cytokine TNF-
The Spearman correlation between the MIN score and GIMATS α are also able to induce a shift towards abnormal lipid profiles in in­
module score was 0.974 (Fig. 5B). The merged RNA-seq cohort flammatory conditions, the process of which can be partially reversible
mentioned above was adopted as a validation cohort. As shown in through TNF-α blockade [43]. These alterations in energy homeostasis
Fig. 5C, the validation cohort also demonstrated good linear dependence were found more in CD than in UC [44], which is consistent with the
(Spearman correlation = 0.869, p < 0.001). The ROC of the MIN score to differential expression of peroxisome proliferator-activated receptors
match the GIMATS module in validation cohort was illustrated in Fig. S4

1995
Y. Shi et al. Genomics 113 (2021) 1988–1998

Fig. 6. Represented IHC staining results of biopsy derived from IBD patients well (A) or poor (B) response to IFX. Scale bar, 100 um. Abbreviations: IBD, inflam­
matory bowel disease; IFX, infliximab; IHC, immunohistochemistry.

(PPARs) [45], members of the steroid receptor superfamily that play probably cause the innate immune system to assume an IFX/ADA-
vital roles in both inflammation and fibrosis formation [46]. Although sensitive status and provide clues to physicians regarding the selection
the 3 subtypes, PPARα, PPARγ, and PPARβ/δ, are different in terms of of biologics.
their distribution, all of them participate in the regulation of fatty acid In addition, researchers have long identified the function of intesti­
metabolism [47]. Therefore, the activation of the PPAR pathway can nal microbiota as a bridge between immunity and lipid metabolism, and

1996
Y. Shi et al. Genomics 113 (2021) 1988–1998

the abundance of intestinal flora that produce short-chain fatty acids expression) in daily practices for community hospitals with limited
(SCFAs) was also closely related to treatment outcome. SCFAs are the medical resources. Besides, given that different genetic assays would
main source of energy for intestinal epithelial cells and participate in the produce different information on gene mutations and fusions, expres­
regulation of intestinal haemostasis. SCFA-producing microflora act as sion levels of genes seem less stable to guide clinical procedures. Hence,
an epithelial barrier of metabolic and immune function, the imbalance in this study, we established the MIN score and validated it using
of which can lead to the onset of IBD and the existence of persistent platform-varied data to ascertain the robustness. We believe that, with
inflammation [48]. Studies have shown that, in both CD and UC, a the further reduction of sequencing cost and the emerging of next-
sustained response to IFX/ADA or likelihood of disease recurrence was generation technologies, genetics biomarkers would be found highly
significantly correlated with the high abundance of intestinal SCFA- valuable in predicting treatment outcomes. Compared to GIMATS
producing bacteria (e.g., Clostridiales and F. prausnitzii) at baseline module, this 6-gene scoring system developed for therapy decision is
[49–51]. simple, efficient and affordable with alternative laboratory techniques
On the other hand, patients who had higher MIN scores (I type) of RNA-seq, microarray, real-time polymerase chain reaction and IHC.
tended to respond better to VDZ than IFX/ADA, and the functional In conclusion, our findings regarding the predictive value of the MIN
analysis was mainly focused on immunologic changes. Although the score provided new insights to assist with treatment choices in the
pharmacologic target of VDZ, integrin α4β7, is located on both innate future. IBD patients were divided into 3 groups based on their baseline
and adaptive immune cells, a limited impact on intestinal T cells has MIN score, the M type, I type and N type groups, reflecting the disease
been observed during VDZ treatment [52], highlighting the significance characteristics. The M and I types may favour treatment with IFX/ADA
of the innate immune system in the action of medication. and VDZ, respectively, while the N type can probably benefit from either
The baseline accumulation of polarized macrophages was highly treatment. This grouping emphasized the significant role of metabolism
correlated with the I-type response in our findings, as validated in other and immune response analysis at the time of IBD diagnosis instead of the
studies that observed a switch from M1-type to M2-type macrophages in differentiation between CD and UC.
patients who achieved VDZ-induced clinical remission [53]. This re­ Supplementary data to this article can be found online at https://doi.
flected a shift in homeostasis towards an anti-inflammatory state rather org/10.1016/j.ygeno.2021.04.011.
than overactivation of the immune system. Additionally, cytokine sig­
nalling pathways, including the IL-17 pathway that was ranked among Funding
the top 3 most remarkable pathways in our results, are known for their
function in the promotion of the development of IBD. IL-17 is produced This work was supported by the National Natural Science Foundation
by Th17 cells, which, under normal circumstances, are located in the of China (81702300, 81873555).
mucosa to protect the host from invasive microorganisms. When
induced by cytokines such as IL-1β and IL-6, the imbalance in Th17 Ethics and data availability statements
signalling as well as Th or Treg differentiation leads to disease pro­
gression [2]. Although there are already some models of cytokines used Ethical approval was obtained from Institute Research Ethics Com­
for IFX/ADA prediction [54], little evidence has been found to link the mittee of Renji Hospital. Informed consent was signed for experimen­
IL-17 pathway to the VDZ response. However, in these complicated tation with human subjects. The data underlying this article are
processes, IL-6 also serves as a pro-inflammatory and anti-apoptosis available in the article and in its online supplementary material.
factor through activation of signal transducer and activator of tran­ Supplementary tables have been uploaded to the following link:
scription (STAT) 3 in T cells [55]. An increase in the baseline IL-6 level https://www.dropbox.com/s/tgnz52c0uq71u23/Revised%
and its tendency to decrease over the first 6 weeks had significant power 20Supplementary%20tables%20for%20MIN%20score.xlsx?dl=0.
to predict VDZ-induced mucosal healing [12].
In our study, certain approaches were used to prevent over-fitting Brief summary
and data leakage issues. Primarily, we performed LASSO regression to
optimize gene selections, and adopted one internal and two external The 6-gene MIN score is able to classify the metabolic or immune
validation sets. During the process of model establishment, 5-fold cross type of inflammatory bowel disease patients at diagnosis and assists with
validation was employed to enhance robustness, after which, MIN score the first-line selection of infliximab/adalimumab or vedolizumab
was also tested in external RNA-seq validation as well as an independent therapy.
IHC cohort. The outcomes supported our previous conclusion.
However, there were still limitations. First, this study was based on Declaration of Competing Interest
public datasets and bioinformatic tools, and the correlations would
require further experiments to verify if the exact causal relationships The authors declared no conflict interest.
exist. LASSO only works based on several assumptions. Considering its
limitations in usage, variable sparsity and irrepresentable condition References
should be checked before processing the data [56]. In this study, the
conduction of LASSO regression was followed by multiple validations to [1] C.T. Weaver, R.D. Hatton, Interplay between the TH17 and TReg cell lineages: a
ensure its proper usage. Although we used the IHC experiment in an (co-)evolutionary perspective, Nat. Rev. Immunol. 9 (2009) 883–889.
[2] H.S.P. de Souza, C. Fiocchi, Immunopathogenesis of IBD: current state of the art,
attempt to utilize our model in the real clinical practice, it is undeniable
Nat. Rev. Gastroenterol. Hepatol. 13 (2016) 13–27.
that the sample size was too small to draw a convincing conclusion. We [3] K. Papamichael, A. Gils, P. Rutgeerts, B.G. Levesque, S. Vermeire, W.J. Sandborn,
called for a new independent validation set in the future to confirm our N. Vande Casteele, Role for therapeutic drug monitoring during induction therapy
with TNF antagonists in IBD: evolution in the definition and management of
findings. Second, prescriptions of ADA and VDZ were unavailable in our
primary nonresponse, Inflamm. Bowel Dis. 21 (2015) 182–197.
centre, therefore no biopsy sample was obtained for validation experi­ [4] B.G. Feagan, P. Rutgeerts, B.E. Sands, S. Hanauer, J.F. Colombel, W.J. Sandborn,
ments in response prediction of ADA/VDZ. Third, most clinical trials G. Van Assche, J. Axler, H.J. Kim, S. Danese, I. Fox, C. Milch, S. Sankoh, T. Wyant,
included in the data sources in our research had an active follow-up J. Xu, A. Parikh, Vedolizumab as induction and maintenance therapy for ulcerative
colitis, N. Engl. J. Med. 369 (2013) 699–710.
interval of 6 weeks, which was much shorter than the typical internal [5] W.J. Sandborn, B.G. Feagan, P. Rutgeerts, S. Hanauer, J.F. Colombel, B.E. Sands,
(3–6 months if no emergencies happened) [57]. Future prospective M. Lukas, R.N. Fedorak, S. Lee, B. Bressler, I. Fox, M. Rosario, S. Sankoh, J. Xu,
studies should have a more reasonable timetable for clinical observation K. Stephens, C. Milch, A. Parikh, Vedolizumab as induction and maintenance
therapy for Crohn’s disease, N. Engl. J. Med. 369 (2013) 711–721.
or used other sensitive but non-invasive tests for response assessment. [6] B.G. Feagan, D.T. Rubin, S. Danese, S. Vermeire, B. Abhyankar, S. Sankoh,
Fourth, it is a challenge to apply prognosis biomarkers (e.g. gene A. James, M. Smyth, Efficacy of Vedolizumab induction and maintenance therapy

1997
Y. Shi et al. Genomics 113 (2021) 1988–1998

in patients with ulcerative colitis, regardless of prior exposure to tumor necrosis [31] M. Kohl, S. Wiese, B. Warscheid, Cytoscape: software for visualization and analysis
factor antagonists, Clin. Gastroenterol. Hepatol. 15 (2017) 229–239 (e225). of biological networks, Methods Mol. Biol. (Clifton, N.J.) 696 (2011) 291–303.
[7] B.E. Sands, W.J. Sandborn, G. Van Assche, M. Lukas, J. Xu, A. James, [32] C.H. Chin, S.H. Chen, H.H. Wu, C.W. Ho, M.T. Ko, C.Y. Lin, cytoHubba: identifying
B. Abhyankar, K. Lasch, Vedolizumab as induction and maintenance therapy for hub objects and sub-networks from complex interactome, BMC Syst. Biol. 8 (Suppl.
Crohn’s disease in patients Naïve to or who have failed tumor necrosis factor 4) (2014) S11.
antagonist therapy, Inflamm. Bowel Dis. 23 (2017) 97–106. [33] G. Bindea, B. Mlecnik, H. Hackl, P. Charoentong, M. Tosolini, A. Kirilovsky, W.
[8] S. Bek, J.V. Nielsen, A.B. Bojesen, A. Franke, S. Bank, U. Vogel, V. Andersen, H. Fridman, F. Pagès, Z. Trajanoski, J. Galon, ClueGO: a Cytoscape plug-in to
Systematic review: genetic biomarkers associated with anti-TNF treatment decipher functionally grouped gene ontology and pathway annotation networks,
response in inflammatory bowel diseases, Aliment. Pharmacol. Ther. 44 (2016) Bioinformatics (Oxford, England) 25 (2009) 1091–1093.
554–567. [34] J. Friedman, T. Hastie, R. Tibshirani, Regularization paths for generalized linear
[9] I. Arijs, K. Li, G. Toedter, R. Quintens, L. Van Lommel, K. Van Steen, P. Leemans, models via coordinate descent, J. Stat. Softw. 33 (2010) 1–22.
G. De Hertogh, K. Lemaire, M. Ferrante, F. Schnitzler, L. Thorrez, K. Ma, X.Y. [35] H. Wickham, ggplot2: Elegant Graphics for Data Analysis, Springer-Verlag, New
R. Song, C. Marano, G. Van Assche, S. Vermeire, K. Geboes, F. Schuit, F. Baribaud, York, 2016.
P. Rutgeerts, Mucosal gene signatures to predict response to infliximab in patients [36] R. Kolde, pheatmap: Pretty Heatmaps, 2015.
with ulcerative colitis, Gut 58 (2009) 1612–1619. [37] V.S. Taiyun Wei, R Package “Corrplot”: Visualization of a Correlation Matrix, 2017.
[10] N.R. West, A.N. Hegazy, B.M.J. Owens, S.J. Bullers, B. Linggi, S. Buonocore, [38] M.K. Alboukadel Kassambara, Przemyslaw Biecek, Scheipl Fabian, Survminer:
M. Coccia, D. Görtz, S. This, K. Stockenhuber, J. Pott, M. Friedrich, G. Ryzhakov, Drawing Survival Curves Using ‘ggplot2’, 2020.
F. Baribaud, C. Brodmerkel, C. Cieluch, N. Rahman, G. Müller-Newen, R.J. Owens, [39] X. Robin, N. Turck, A. Hainard, N. Tiberti, F. Lisacek, J.C. Sanchez, M. Müller,
A.A. Kühl, K.J. Maloy, S.E. Plevy, S. Keshav, S.P.L. Travis, F. Powrie, Oncostatin M pROC: an open-source package for R and S+ to analyze and compare ROC curves,
drives intestinal inflammation and predicts response to tumor necrosis factor- BMC Bioinformatics 12 (2011) 77.
neutralizing therapy in patients with inflammatory bowel disease, Nat. Med. 23 [40] T.L. Max Gordon, forestplot: Advanced Forest Plot Using ’grid’ Graphics, 2020.
(2017) 579–589. [41] E.H. Frank Jr., rms: Regression Modeling Strategies, 2020.
[11] E.K. Boden, D.M. Shows, M.V. Chiorean, J.D. Lord, Identification of candidate [42] T. Karrasch, F. Obermeier, R.H. Straub, Systemic metabolic signaling in acute and
biomarkers associated with response to Vedolizumab in inflammatory bowel chronic gastrointestinal inflammation of inflammatory bowel diseases, Hormone
disease, Dig. Dis. Sci. 63 (2018) 2419–2429. Metab. Res. 46 (2014) 445–451.
[12] C. Soendergaard, J.B. Seidelin, C. Steenholdt, O.H. Nielsen, Putative biomarkers of [43] C. Popa, M.G. Netea, P.L. van Riel, J.W. van der Meer, A.F. Stalenhoef, The role of
vedolizumab resistance and underlying inflammatory pathways involved in IBD, TNF-alpha in chronic inflammatory conditions, intermediary metabolism, and
BMJ Open Gastroenterol. 5 (2018), e000208. cardiovascular risk, J. Lipid Res. 48 (2007) 751–762.
[13] B. Verstockt, S. Verstockt, M. Veny, J. Dehairs, K. Arnauts, G. Van Assche, G. De [44] E.A. Scoville, M.M. Allaman, C.T. Brown, A.K. Motley, S.N. Horst, C.S. Williams,
Hertogh, S. Vermeire, A. Salas, M. Ferrante, Expression levels of 4 genes in colon T. Koyama, Z. Zhao, D.W. Adams, D.B. Beaulieu, D.A. Schwartz, K.T. Wilson, L.
tissue might be used to predict which patients will enter endoscopic remission after A. Coburn, Alterations in lipid, amino acid, and energy metabolism distinguish
vedolizumab therapy for inflammatory bowel diseases, Clin. Gastroenterol. Crohn’s disease from ulcerative colitis and control subjects by serum metabolomic
Hepatol. 18 (2019) 1142–1151. profiling, Metabolomics 14 (2018) 17.
[14] World Medical Association Declaration of Helsinki: ethical principles for medical [45] J. Decara, P. Rivera, A.J. López-Gambero, A. Serrano, F.J. Pavón, E. Baixeras,
research involving human subjects, Jama 310 (2013) 2191–2194. F. Rodríguez de Fonseca, J. Suárez, Peroxisome proliferator-activated receptors:
[15] N.L. Bray, H. Pimentel, P. Melsted, L. Pachter, Near-optimal probabilistic RNA-seq experimental targeting for the treatment of inflammatory bowel diseases, Front.
quantification, Nat. Biotechnol. 34 (2016) 525–527. Pharmacol. 11 (2020) 730.
[16] J.T. Leek, W.E. Johnson, H.S. Parker, A.E. Jaffe, J.D. Storey, The sva package for [46] H.E. Xu, M.H. Lambert, V.G. Montana, D.J. Parks, S.G. Blanchard, P.J. Brown, D.
removing batch effects and other unwanted variation in high-throughput D. Sternbach, J.M. Lehmann, G.B. Wisely, T.M. Willson, S.A. Kliewer, M.
experiments, Bioinformatics (Oxford, England) 28 (2012) 882–883. V. Milburn, Molecular recognition of fatty acids by peroxisome proliferator-
[17] L. Gautier, L. Cope, B.M. Bolstad, R.A. Irizarry, affy—analysis of Affymetrix activated receptors, Mol. Cell 3 (1999) 397–403.
GeneChip data at the probe level, Bioinformatics (Oxford, England) 20 (2004) [47] R.B. Clark, The role of PPARs in inflammation and immunity, J. Leukoc. Biol. 71
307–315. (2002) 388–400.
[18] K. Yoshihara, M. Shahmoradgoli, E. Martínez, R. Vegesna, H. Kim, W. Torres- [48] E.C. Martens, M. Neumann, M.S. Desai, Interactions of commensal and pathogenic
Garcia, V. Treviño, H. Shen, P.W. Laird, D.A. Levine, S.L. Carter, G. Getz, microorganisms with the intestinal mucosal barrier, Nat. Rev. Microbiol. 16 (2018)
K. Stemke-Hale, G.B. Mills, R.G. Verhaak, Inferring tumour purity and stromal and 457–470.
immune cell admixture from expression data, Nat. Commun. 4 (2013) 2612. [49] M. Sun, W. Wu, Z. Liu, Y. Cong, Microbiota metabolite short chain fatty acids,
[19] P. Charoentong, F. Finotello, M. Angelova, C. Mayer, M. Efremova, D. Rieder, GPCR, and inflammatory bowel diseases, J. Gastroenterol. 52 (2017) 1–8.
H. Hackl, Z. Trajanoski, Pan-cancer immunogenomic analyses reveal genotype- [50] Y. Zhou, Z.Z. Xu, Y. He, Y. Yang, L. Liu, Q. Lin, Y. Nie, M. Li, F. Zhi, S. Liu, A. Amir,
immunophenotype relationships and predictors of response to checkpoint A. González, A. Tripathi, M. Chen, G.D. Wu, R. Knight, H. Zhou, Y. Chen, Gut
blockade, Cell Rep. 18 (2017) 248–262. microbiota offers universal biomarkers across ethnicity in inflammatory bowel
[20] S. Hänzelmann, R. Castelo, J. Guinney, GSVA: gene set variation analysis for disease diagnosis and infliximab response prediction, mSystems 3 (2018).
microarray and RNA-seq data, BMC Bioinformatics 14 (2013) 7. [51] S. Rajca, V. Grondin, E. Louis, G. Vernier-Massouille, J.-C. Grimaud, Y. Bouhnik,
[21] A.M. Newman, C.L. Liu, M.R. Green, A.J. Gentles, W. Feng, Y. Xu, C.D. Hoang, D. Laharie, J.-L. Dupas, H. Pillant, L. Picon, M. Veyrac, M. Flamant, G. Savoye,
M. Diehn, A.A. Alizadeh, Robust enumeration of cell subsets from tissue expression R. Jian, M. Devos, G. Paintaud, E. Piver, M. Allez, J.Y. Mary, H. Sokol, J.-
profiles, Nat. Methods 12 (2015) 453–457. F. Colombel, P. Seksik, Alterations in the intestinal microbiome (dysbiosis) as a
[22] J.C. Martin, C. Chang, G. Boschetti, R. Ungaro, M. Giri, J.A. Grout, K. Gettler, L.- predictor of relapse after infliximab withdrawal in Crohn’s disease, Inflamm.
S. Chuang, S. Nayar, A.J. Greenstein, M. Dubinsky, L. Walker, A. Leader, J.S. Fine, Bowel Dis. 20 (2014) 978–986.
C.E. Whitehurst, M.L. Mbow, S. Kugathasan, L.A. Denson, J.S. Hyams, J. [52] S. Zeissig, E. Rosati, C.M. Dowds, K. Aden, J. Bethge, B. Schulte, W.H. Pan,
R. Friedman, P.T. Desai, H.M. Ko, I. Laface, G. Akturk, E.E. Schadt, H. Salmon, N. Mishra, M. Zuhayra, M. Marx, M. Paulsen, A. Strigli, C. Conrad, D. Schuldt,
S. Gnjatic, A.H. Rahman, M. Merad, J.H. Cho, E. Kenigsberg, Single-cell analysis of A. Sinha, H. Ebsen, S.C. Kornell, S. Nikolaus, A. Arlt, D. Kabelitz, M. Ellrichmann,
crohn’s disease lesions identifies a pathogenic cellular module associated with U. Lützen, P.C. Rosenstiel, A. Franke, S. Schreiber, Vedolizumab is associated with
resistance to anti-TNF therapy, Cell 178 (2019). changes in innate rather than adaptive immunity in patients with inflammatory
[23] S. Anders, W. Huber, Differential expression analysis for sequence count data, bowel disease, Gut 68 (2019) 25–39.
Genome Biol. 11 (2010) R106. [53] L. Bertani, L. Baglietto, L. Antonioli, M. Fornai, G. Tapete, E. Albano, L. Ceccarelli,
[24] C. Soneson, M.I. Love, M.D. Robinson, Differential analyses for RNA-seq: M.G. Mumolo, C. Pellegrini, E. Lucenteforte, N. de Bortoli, M. Bellini, S. Marchi,
transcript-level estimates improve gene-level inferences, F1000Research 4 (2015) C. Blandizzi, F. Costa, Assessment of serum cytokines predicts clinical and
1521. endoscopic outcomes to vedolizumab in ulcerative colitis patients, Br. J. Clin.
[25] G. Yu, L.G. Wang, Y. Han, Q.Y. He, clusterProfiler: an R package for comparing Pharmacol. 86 (2020) 1296–1305.
biological themes among gene clusters, Omics 16 (2012) 284–287. [54] I.V. Obraztsov, K.E. Shirokikh, O.I. Obraztsova, M.V. Shapina, M.-H. Wang, I.
[26] P. Langfelder, S. Horvath, WGCNA: an R package for weighted correlation network L. Khalif, Multiple cytokine profiling: a new model to predict response to tumor
analysis, BMC Bioinformatics 9 (2008) 559. necrosis factor antagonists in ulcerative colitis patients, Inflamm. Bowel Dis. 25
[27] M. Daperno, G. D’Haens, G. Van Assche, F. Baert, P. Bulois, V. Maunoury, (2019) 524–531.
R. Sostegni, R. Rocca, A. Pera, A. Gevers, J.Y. Mary, J.F. Colombel, P. Rutgeerts, [55] J. Mudter, M.F. Neurath, Apoptosis of T cells and the control of inflammatory
Development and validation of a new, simplified endoscopic activity score for bowel disease: therapeutic implications, Gut 56 (2007) 293–303.
Crohn’s disease: the SES-CD, Gastrointest. Endosc. 60 (2004) 505–512. [56] G. Vasconcelos, When the LASSO Fails???, 2017.
[28] K.W. Schroeder, W.J. Tremaine, D.M. Ilstrup, Coated oral 5-aminosalicylic acid [57] C. Maaser, A. Sturm, S.R. Vavricka, T. Kucharzik, G. Fiorino, V. Annese,
therapy for mildly to moderately active ulcerative colitis. A randomized study, E. Calabrese, D.C. Baumgart, D. Bettenworth, P. Borralho Nunes, J. Burisch,
N. Engl. J. Med. 317 (1987) 1625–1629. F. Castiglione, R. Eliakim, P. Ellul, Y. González-Lama, H. Gordon, S. Halligan,
[29] K. Geboes, R. Riddell, A. Ost, B. Jensfelt, T. Persson, R. Löfberg, A reproducible K. Katsanos, U. Kopylov, P.G. Kotze, E. Krustinš, A. Laghi, J.K. Limdi, F. Rieder,
grading scale for histological assessment of inflammation in ulcerative colitis, Gut J. Rimola, S.A. Taylor, D. Tolan, P. van Rheenen, B. Verstockt, J. Stoker, ECCO-
47 (2000) 404–409. ESGAR Guideline for Diagnostic Assessment in IBD Part 1: Initial diagnosis,
[30] D. Szklarczyk, A. Franceschini, S. Wyder, K. Forslund, D. Heller, J. Huerta-Cepas, monitoring of known IBD, detection of complications, J. Crohns Colitis 13 (2019)
M. Simonovic, A. Roth, A. Santos, K.P. Tsafou, M. Kuhn, P. Bork, L.J. Jensen, C. von 144–164.
Mering, STRING v10: protein-protein interaction networks, integrated over the tree
of life, Nucleic Acids Res. 43 (2015) D447–D452.

1998

You might also like