identification of key prognostic biomarker and its correlation...

12
Research Article Identification of Key Prognostic Biomarker and Its Correlation with Immune Infiltrates in Pancreatic Ductal Adenocarcinoma Hong Luan, 1 Chuang Zhang, 2,3 Tuo Zhang, 1 Ye He, 1 Yanna Su, 1 and Liping Zhou 4 1 Department of Laboratory Medicine, The First Aliated Hospital of China Medical University, Shenyang, Liaoning 110001, China 2 Key Laboratory of Anticancer Drugs and Biotherapy, The First Aliated Hospital of China Medical University, Shenyang, Liaoning 110001, China 3 Department of Medical Oncology, The First Aliated Hospital of China Medical University, Shenyang, 110001 Liaoning, China 4 Department of Graduate Faculty, The First Aliated Hospital of China Medical University, Shenyang, Liaoning Province 110001, China Correspondence should be addressed to Liping Zhou; [email protected] Received 30 May 2020; Accepted 20 August 2020; Published 31 August 2020 Academic Editor: Carlo Chiarla Copyright © 2020 Hong Luan et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Pancreatic ductal adenocarcinoma (PDAC) is an extremely malignant tumor. The immune prole of PDAC and the immunologic milieu of its tumor microenvironment (TME) are unique; however, the mechanism of how the TME engineers the carcinogenesis of PDAC is not fully understood. This study is aimed at better understanding the relationship between the immune inltration of the TME and gene expression and identifying potential prognostic and immunotherapeutic biomarkers for PDAC. Analysis of data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases identied dierentially expressed genes (DEGs), including 159 upregulated and 53 downregulated genes. Gene Ontology analysis and Kyoto Encyclopedia of Genes and Genomes enrichment were performed and showed that the DEGs were mainly enriched for the PI3K-Akt signaling pathway and extracellular matrix organization. We used the cytoHubba plugin of Cytoscape to screen out the most signicant ten hub genes by four dierent models (Degree, MCC, DMNC, and MNC). The expression and clinical relevance of these ten hub genes were validated using Gene Expression Proling Interactive Analysis (GEPIA) and the Human Protein Atlas, respectively. High expression of nine of the hub genes was positively correlated with poor prognosis. Finally, the relationship between these hub genes and tumor immunity was analyzed using the Tumor Immune Estimation Resource. We found that the expression of SPARC, COL6A3, and FBN1 correlated positively with inltration levels of six immune cells in the tumors. In addition, these three genes had a strong coexpression relationship with the immune checkpoints. In conclusion, our results suggest that nine upregulated biomarkers are related to poor prognosis in PDAC and may serve as potential prognostic biomarkers for PDAC therapy. Furthermore, SPARC, COL6A3, and FBN1 play an important role in tumor-related immune inltration and may be ideal targets for immune therapy against PDAC. 1. Introduction Pancreatic ductal adenocarcinoma (PDAC) is an extremely malignant tumor of the digestive system, with a 5-year sur- vival rate of only 8% [1]. GLOBOCAN 2018 estimated that pancreatic cancer ranked as the seventh leading cause of cancer death worldwide, with approximately 458,918 new incidence cases and 432,242 deaths [2]. Because of its poor prognosis, newly diagnosed cases and deaths from pancreatic cancer have signicantly increased at the same pace over the last few decades [2]. Currently, surgical resection is still the most eective treatment, which signicantly improves the 5-year survival rate to 2030%. However, less than 20% of all patients are eligible for resection because the majority of Hindawi Disease Markers Volume 2020, Article ID 8825997, 12 pages https://doi.org/10.1155/2020/8825997

Upload: others

Post on 11-Oct-2020

7 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Identification of Key Prognostic Biomarker and Its Correlation ...downloads.hindawi.com/journals/dm/2020/8825997.pdfResearch Article Identification of Key Prognostic Biomarker and

Research ArticleIdentification of Key Prognostic Biomarker and ItsCorrelation with Immune Infiltrates in PancreaticDuctal Adenocarcinoma

Hong Luan,1 Chuang Zhang,2,3 Tuo Zhang,1 Ye He,1 Yanna Su,1 and Liping Zhou 4

1Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China2Key Laboratory of Anticancer Drugs and Biotherapy, The First Affiliated Hospital of China Medical University, Shenyang,Liaoning 110001, China3Department of Medical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, 110001 Liaoning, China4Department of Graduate Faculty, The First Affiliated Hospital of China Medical University, Shenyang,Liaoning Province 110001, China

Correspondence should be addressed to Liping Zhou; [email protected]

Received 30 May 2020; Accepted 20 August 2020; Published 31 August 2020

Academic Editor: Carlo Chiarla

Copyright © 2020 Hong Luan et al. This is an open access article distributed under the Creative Commons Attribution License,which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Pancreatic ductal adenocarcinoma (PDAC) is an extremely malignant tumor. The immune profile of PDAC and the immunologicmilieu of its tumor microenvironment (TME) are unique; however, the mechanism of how the TME engineers the carcinogenesis ofPDAC is not fully understood. This study is aimed at better understanding the relationship between the immune infiltration of theTME and gene expression and identifying potential prognostic and immunotherapeutic biomarkers for PDAC. Analysis of datafrom The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases identified differentially expressedgenes (DEGs), including 159 upregulated and 53 downregulated genes. Gene Ontology analysis and Kyoto Encyclopedia ofGenes and Genomes enrichment were performed and showed that the DEGs were mainly enriched for the PI3K-Akt signalingpathway and extracellular matrix organization. We used the cytoHubba plugin of Cytoscape to screen out the most significantten hub genes by four different models (Degree, MCC, DMNC, and MNC). The expression and clinical relevance of these tenhub genes were validated using Gene Expression Profiling Interactive Analysis (GEPIA) and the Human Protein Atlas,respectively. High expression of nine of the hub genes was positively correlated with poor prognosis. Finally, the relationshipbetween these hub genes and tumor immunity was analyzed using the Tumor Immune Estimation Resource. We found that theexpression of SPARC, COL6A3, and FBN1 correlated positively with infiltration levels of six immune cells in the tumors. Inaddition, these three genes had a strong coexpression relationship with the immune checkpoints. In conclusion, our resultssuggest that nine upregulated biomarkers are related to poor prognosis in PDAC and may serve as potential prognosticbiomarkers for PDAC therapy. Furthermore, SPARC, COL6A3, and FBN1 play an important role in tumor-related immuneinfiltration and may be ideal targets for immune therapy against PDAC.

1. Introduction

Pancreatic ductal adenocarcinoma (PDAC) is an extremelymalignant tumor of the digestive system, with a 5-year sur-vival rate of only 8% [1]. GLOBOCAN 2018 estimated thatpancreatic cancer ranked as the seventh leading cause ofcancer death worldwide, with approximately 458,918 new

incidence cases and 432,242 deaths [2]. Because of its poorprognosis, newly diagnosed cases and deaths from pancreaticcancer have significantly increased at the same pace over thelast few decades [2]. Currently, surgical resection is still themost effective treatment, which significantly improves the5-year survival rate to 20–30%. However, less than 20% ofall patients are eligible for resection because the majority of

HindawiDisease MarkersVolume 2020, Article ID 8825997, 12 pageshttps://doi.org/10.1155/2020/8825997

Page 2: Identification of Key Prognostic Biomarker and Its Correlation ...downloads.hindawi.com/journals/dm/2020/8825997.pdfResearch Article Identification of Key Prognostic Biomarker and

patients have locally advanced or metastatic disease at thetime of diagnosis [3]. Therefore, there is an urgent need formore effective therapeutic strategies.

In recent years, cancer immunotherapy has become anactive area of cancer research. This approach is designed toimprove the immunogenicity of tumor cells, stimulate andenhance the antitumor immune response, and inhibit tumorgrowth and progression. Since the FDA approval of the firsttargeted drug ipilimumab (humanized anti-CTLA-4 IgG1monoclonal antibody) in 2011, six more immune checkpointinhibitors have been approved for cancer therapy [4, 5]. Atpresent, immune checkpoint inhibitors have achieved signif-icant therapeutic effects in melanoma [6], squamous non-small-cell lung cancer [7], renal cell carcinoma [8, 9], andurothelial cancer [10, 11]. However, the immune profile ofPDAC and the immunologic milieu of its tumor microenvi-ronment (TME) are unique relative to other malignanttumors, and the mechanism of how the TME engineers thecarcinogenesis of PDAC is not entirely clear. Therefore, it isnecessary to find new possible prognostic and immunothera-peutic biomarkers for this disease.

In the current study, we analyzed pancreatic cancer geneexpression data from The Cancer Genome Atlas (TCGA)and Gene Expression Omnibus (GEO) databases to identifydifferentially expressed genes (DEGs). Gene Ontology (GO)and Kyoto Encyclopedia of Genes and Genomes (KEGG)pathway enrichment and protein-protein interaction (PPI)network analyses were performed to reveal the interactiverelationships between the DEGs to explore the underlyingmolecular mechanisms involved in the carcinogenesis andprogression of PDAC. Subsequently, we used the cyto-Hubba plugin of Cytoscape to search for hub genes andidentify the most significant hub genes using four differentmodels (Degree, Maximal Clique Centrality (MCC), Den-sity of Maximum Neighborhood Component (DMNC),and Maximum Neighborhood Component (MNC)). Wefurther validated the expression levels of the hub genesand their clinical relevance using Gene Expression ProfilingInteractive Analysis (GEPIA) and the Human Protein Atlas,respectively. Finally, we evaluated the associations betweenthe prognosis-related genes and several immune cell typesbased on the Tumor Immune Estimation Resource

�e differentially expressed genes on chromosomes1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 X Y

Over-expressed genesUnder-expressed genes

�e gene positions are based on GRCh38.p2(NCBI). 9222 genes.

(a)

Volcano plot20

16

12

–lo

g10(P

-val

ue)

–6 –4 –2 0 2 4 6log2(foldchange)

8

4

0

(b)

16610

18

159

1234

7313

2

GSE62452 GSE15471

TCGA

(c)

25 5 102

73

53

329

23

GSE62452 GSE15471

TCGA

(d)

Figure 1: Identification of DEGs in pancreatic cancer using three databases. (a) The differentially expressed genes from the TCGA databasewere determined by GEPIA. (b) The volcano map for the GSE15471 dataset. (c) A Venn diagram showing 159 upregulated genes in PDAC.(d) A Venn diagram showing 53 downregulated genes in PDAC.

2 Disease Markers

Page 3: Identification of Key Prognostic Biomarker and Its Correlation ...downloads.hindawi.com/journals/dm/2020/8825997.pdfResearch Article Identification of Key Prognostic Biomarker and

(TIMER). In summary, our study identified potentialprognostic biomarkers and immunotherapeutic targetsfor PDAC.

2. Materials and Methods

2.1. Data Collection and Processing. The mRNA expressionprofiles of PDAC and adjacent normal tissues were obtainedfrom GEPIA (TCGA Data Online Analysis Tool; http://gepia2.cancer-pku.cn/#index). PDAC microarray profileswere retrieved from the GEO database (http://www.ncbi.nlm.nih.gov/geo/) using the keywords “pancreatic cancer”OR “PDAC”OR “pancreatic adenocarcinoma.” “Homo sapi-

ens” and “Expression profiling by array” were included in thenext round of screening. Inclusion criteria consisted of (1)human pancreatic tissue samples, (2) tumor and adjacentnoncancerous tissue samples, and (3) more than 30 samplesin the tumor and adjacent noncancerous groups. Based onthese criteria, the gene expression microarray datasetsGSE15471 [12] and GSE62452 [13] were downloaded foranalysis. GSE15471 is comprised of 39 tumors and pairedadjacent normal tissues, and GSE62452 contains 69 tumorsand 61 adjacent normal tissue samples. We used the GEO2Ranalysis tool (http://www.ncbi.nlm.nih.gov/geo/geo2r/) toscreen for differentially expressed mRNAs between thenormal tissue and tumor samples from the GSE15471 and

Enrichment GO_BP_dotExtracellular matrix organization

p.adjust

2.50e-07

Count(10)(20)(30)

5.00e-07

7.50e-07

1.25e-06

1.00e-06

Extracellular structure organization

Positive regulation of cell migration

Skeletal system development

Cell-substrate adhesion

Collagen metabolic process

Collagen catabolic process

Extracellular matrix disassembly

Formation of primary germ layer

Endodermal cell differentiation

0.05 0.10 0.15 0.20 0.25Gene ratio

(a)

PI3K-Akt signaling pathway

Focal adhesion

ECM-receptor interaction

Human papillomavirus infection

Protein digestion and absorption

Amoebiasis

p.adjust

Count(5.0)

0.05

0.10

0.15

0.20

(7.5)

(10.0)

(12.5)

Small cell lung cancer

Arrhythmogenic rightventricular cardiomyopathy (ARVC)

Rheumatoid arthritis

Malaria

0.04 0.08 0.12 0.16Gene ratio

Enrichment KEGG_dot

(b)

COL1A

1

COL3A1

COL10A1LAMC2

FN1

COL11A1

LAMA3

COL5A2

ITGB4

COL12A1

AREG

COL6A3

ITGA

2

ITGA

3

MET

SERP

INB3

THBS2

COMP

LAM

B3

KEGG termsECM-receptor interaction

Focal adhesion

Protein digestion and absorptionPI3K-Akt signaling pathway

Amoebiasis

(c)

logFC

1 3

KEGG termsECM-receptor interaction

Focal adhesion

Protein digestion and absorptionPI3K-Akt signaling pathway

Amoebiasis

(d)

Figure 2: Functional enrichment analysis of DEGs. (a) Top ten enriched biological processes for the DEGs. (b) Top ten enriched KEGGpathways for the DEGs. (c) Hierarchical clustering of gene expression profiles in each KEGG pathway. (d) Chord plots showing therelationship between the hub genes and the KEGG pathway.

3Disease Markers

Page 4: Identification of Key Prognostic Biomarker and Its Correlation ...downloads.hindawi.com/journals/dm/2020/8825997.pdfResearch Article Identification of Key Prognostic Biomarker and

GSE62452 datasets. P < 0:05 and ∣log 2FC ∣ >1 were set as thecut-off criteria for identifying DEGs.

2.2. Functional Enrichment Analysis. Gene Ontology (GO)analysis and Kyoto Encyclopedia of Genes and Genomes(KEGG) enrichment were performed on the DEGs, and theresults were visualized using the R package. A P value <0.05 was set as the cut-off criterion.

2.3. PPI Network Construction and Module Analysis. TheSTRING database (https://string-db.org/) was used to drawthe PPI network diagram of the DEGs. The hub genes wereidentified and visualized using Cytoscape [14]. Specifically,the hub genes were identified using the Degree, MCC,

DMNC, and MNC models with the Cytoscape plugincytoHubba [15].

2.4. Validation of Hub Genes. We analyzed the expression ofthe hub gene using GEPIA (http://gepia.cancer-pku.cn/index.html) [16]. Because there were few normal pancreatic tissuesamples in the TCGA database, the expression level of spe-cific DEGs was validated using the TCGA PDAC tumor dataand matched data for normal tissue in the TCGA andGenotype-Tissue Expression (GTEx) databases. ∣log 2FC ∣ >1 and P < 0:05 were considered statistically significant. Pro-tein expression of the DEGs was evaluated in the pancreatictumor and nontumor tissues using the Human Protein Atlastool (https://www.proteinatlas.org/) [17]. Mutation data were

ASPNCOL12A1 ITGA3

COL6A3

COL3A1 THBS2

COL11A1COL5A2

FBN1

COL1A1

ITGA2FN1

POSTN

VCAN

EGF

ALBMP1MMP14

SPARC

BGN

(a)

ALB

MMP14

VCANIGFBP5

MMP1

LTBP1 SP COL1A1

BGNCOL5A2

EBN1

HBS2COL11A1

COL3A1FN1

POSTN

COL12A1 OL6A3

CDH11 ASPN

(b)

LTBP1 IGFBP5

MFAP5

ACTA2

COL5A2

FAP

COL8A1COL12A1

COL5A3COL11A1

CDH11THBS2

TGFBI

POSTN

FBN1

BGNVCAN

SPARCMATN3

APOL1

(c)

MFAP5

ACTA2

IGGBP5LTBP1

APOL1

MATN3SPARC

CDH11

THBS2

BGNVCAN

FBN1

COL11A1

COL6A3

COL5A2POSTN

TGFBI FAP

COL8A1

COL12A1

(d)

Degree

DMNC MCC

MNC

0

00

1

9

0

0

0

1

10

9

0

0

0

0

(e)

Figure 3: Identification of hub genes. (a–d) The hub genes were identified using four models (Degree, MCC, DMNC, and MNC) with theCytoscape plugin cytoHubba. (e) A Venn diagram was used to identify the ten hub genes in PDAC.

4 Disease Markers

Page 5: Identification of Key Prognostic Biomarker and Its Correlation ...downloads.hindawi.com/journals/dm/2020/8825997.pdfResearch Article Identification of Key Prognostic Biomarker and

obtained from the cBioPortal for Cancer Genomics (https://www.cbioportal.org/) [18].

2.5. Association of Hub Gene Expression with the Survival ofPatients with PDAC. We analyzed the association of patientprognosis with the hub genes using the human protein atlaswebsite. Based on the fragments per kilobase million (FPKM)value of each gene, pancreatic cancer patients were classifiedinto two expression groups (high expression and low expres-sion), and the correlation between the expression level andpatient survival was examined. The prognosis of each groupof patients was examined using Kaplan-Meier survivalestimators, and the survival outcomes of the two groups werecompared by the log-rank test. P < 0:05 was consideredstatistically significant.

2.6. Associations between Hub Genes and Immune CellInfiltration and Immune Checkpoints. TIMER is an onlinewebsite that uses RNA-Seq expression profiling data to detectimmune cell infiltration in more than 30 cancer types [19].We used this approach to analyze the correlation betweenthe expression levels of the hub genes and the abundance ofimmune infiltrates (B cells, CD4+ T cells, CD8+ T cells,neutrophils, macrophages, and dendritic cells) and four

immunological checkpoints (CTLA4, CD274, PDCD1, andPDCD1LG2).

3. Results

3.1. Screening for DEGs in PDAC.We identified the differen-tially expressed genes in PDAC in the TCGA databaseusing GEPIA (Figure 1(a)). Based on the thresholds of P< 0:05 and |log 2FC ∣ >1, a total of 9202 DEGs (8724upregulated and 478 downregulated) were identified.DEGs between normal and cancer tissues in the GEOdatasets GSE15471 and GSE62452 were screened usingGEO2R. A total of 295 DEGs (189 upregulated and 106downregulated genes) were identified in profileGSE62452, and 1794 DEGs (1561 upregulated and 233downregulated genes) were found in profile GSE15471(Figure 1(b)). The overlapping DEGs within the three datasetsconsisted of 159 upregulated genes (Figure 1(c)) and 53 down-regulated genes (Figure 1(d)).

3.2. Functional Analysis of the DEGs. To analyze the underly-ing interplay of the DEGs, GO analysis and KEGG pathwayenrichment were performed using the R package. Based onthe GO analysis, the upregulated DEGs mainly participatedin the extracellular matrix organization, extracellular

BGN

10

8

6

4

2

10

8

6

4

20

10

8

6

4

2

0

10

8

6

4

2

0

10

12

14

8

6

4

2

0

10

8

6

4

2

0

8

6

4

2

0

10

8

6

4

2

0

8

6

4

2

0

8

6

4

2

0

COL5A2 COL6A3 COL11A1 COL12A1

VCANTHBS2SPARCPOSTNFBN1

PAAD(num(T)=179; num(N)=171)

PAAD(num(T)=179; num(N)=171)

PAAD(num(T)=179; num(N)=171)

PAAD(num(T)=179; num(N)=171)

PAAD(num(T)=179; num(N)=171)

PAAD(num(T)=179; num(N)=171)

PAAD(num(T)=179; num(N)=171)

PAAD(num(T)=179; num(N)=171)

PAAD(num(T)=179; num(N)=171)

PAAD(num(T)=179; num(N)=171)

⁎ ⁎ ⁎ ⁎ ⁎

⁎⁎⁎⁎⁎

Figure 4: Expression analysis of ten hub genes in PDAC based on GEPIA. The mRNA expression levels in TCGA pancreatic tumors (n = 179)and matching normal tissue (n = 171) from the TCGA and GTEx databases. P < 0:05 was considered statistically significant.

5Disease Markers

Page 6: Identification of Key Prognostic Biomarker and Its Correlation ...downloads.hindawi.com/journals/dm/2020/8825997.pdfResearch Article Identification of Key Prognostic Biomarker and

structure organization, positive regulation of cell migra-tion, skeletal system development, and cell-substrate adhe-sion (Figure 2(a)). KEGG pathway analysis revealed thatthe DEGs were mainly enriched in the PI3K-Akt signalingpathway, focal adhesion, and ECM-receptor interaction

(Figure 2(b)). Genes specifically enriched in each KEGGterm were visualized (Figures 2(c) and 2(d)).

3.3. PPI Network Analysis and Screening of Hub Genes. Toexplore the potential interactions between the DEGs, we used

Normal

BGN

COL1

2A1

POST

NTB

HS2

VCA

NSP

ARC

FBN

1CO

L6A

3

Negative

Weak Weak Weak Moderate

ModerateModerate

Moderate StrongWeakModerate

Negative

NegativeNegative

NegativeModerate

Tumor Normal Tumor

(a)

POSTN

BGN

FBN1

COL5A2

VCAN

THBS2

SPARC

COL11A1

COL6A3

COL12A1

1.1%

2.9%

2.9%

1.7%

2.9%

1.1%

1.7%

2.9%

1.7%

1.1%

Missense mutation (unknown significance)

Truncating mutation (unknown significance)

Genetic alteration

Amplification

Deep deletion

No alterations

(b)

Figure 5: Validation of protein expression and genetic alterations of ten hub genes in pancreatic cancer. (a) Representative protein expressionof eight genes in pancreatic cancer tumors and normal tissue. Data was obtained from the Human Protein Atlas database. (b) Geneticalterations of ten genes in pancreatic cancer. Data was obtained from the cBioPortal.

6 Disease Markers

Page 7: Identification of Key Prognostic Biomarker and Its Correlation ...downloads.hindawi.com/journals/dm/2020/8825997.pdfResearch Article Identification of Key Prognostic Biomarker and

Surv

ival

pro

vabi

lity

Low expression (n = 117)

1.00.90.80.70.60.50.40.30.20.1

0.00 1 2 3 4 5 6 7

High expression (n = 59)

P = 0.26

Time (years)

(a)

Surv

ival

pro

vabi

lity

1.00.90.80.70.60.50.40.30.20.10.0

P = 0.044

0 1 2 3 4 5 6 7Time (years)

Low expression (n = 36)High expression (n = 140)

P = 0.044P

(b)

Surv

ival

pro

vabi

lity

1.00.90.80.70.60.50.40.30.20.1

0.0

P = 0.013

0 1 2 3 4 5 6 7Time (years)

Low expression (n = 38)High expression (n = 138)

(c)

Surv

ival

pro

vabi

lity

1.00.90.80.70.60.50.40.30.20.1

0.0

P = 0.011

0 1 2 3 4 5 6 7Time (years)

Low expression (n = 77)High expression (n = 99)

P = 0.011P

(d)

Surv

ival

pro

vabi

lity

1.00.90.80.70.60.50.40.30.20.10.0

P = 0.0012

0 1 2 3 4 5 6 7Time (years)

Low expression (n = 64)High expression (n = 112)

P = 0.0012P

(e)

Surv

ival

pro

vabi

lity

1.00.90.80.70.60.50.40.30.20.1

0.0

P = 0.028

0 1 2 3 4 5 6 7Time (years)

Low expression (n = 36)High expression (n = 140)

P = 0.028P

(f)

Surv

ival

pro

vabi

lity

1.00.90.80.70.60.50.40.30.20.10.0

P = 0.013

0 1 2 3 4 5 6 7Time (years)

Low expression (n = 38)High expression (n = 138)

P = 0.013P

(g)

Surv

ival

pro

vabi

lity

1.00.90.80.70.60.50.40.30.20.1

0.0

P = 0.041

0 1 2 3 4 5 6 7Time (years)

Low expression (n = 40)High expression (n = 136)

(h)

Figure 6: Continued.

7Disease Markers

Page 8: Identification of Key Prognostic Biomarker and Its Correlation ...downloads.hindawi.com/journals/dm/2020/8825997.pdfResearch Article Identification of Key Prognostic Biomarker and

the STRING database to study the relationship between thevarious DEGs. We identified ten hub genes based on theDegree (Figure 3(a)), MCC (Figure 3(b)), DMNC(Figure 3(c)), and MNC (Figure 3(d)) Cytoscape models.The ten hub genes were BGN, COL5A2, COL6A3, COL11A1,COL12A1, FBN1, POSTN, SPARC, THBS2, and VCAN(Figure 3(e)).

3.4. Validation of Expression and Alteration of Hub Genes inPDAC. To verify the differential expression of the hub genesbetween PDAC and normal pancreatic tissue, we analyzedthe ten hub genes using the GEPIA-based TCGA database.We found that the mRNA expression levels of the hub geneswere significantly increased in PDAC compared to normalpancreatic tissue (Figure 4). Protein expression levels weresimilarly compared using the Human Protein Atlas database.

Typical immunohistochemistry images (https://www.proteinatlas.org/) for the protein expression of eight of theten genes (COL5A2 and COL11A1 were not included in thedatabase) in tumor and normal pancreatic tissue are shownin Figure 5(a). Genetic alterations found in the ten genes inpancreatic cancer are shown in Figure 5(b). Missensemutations were the most common type of mutation in theupregulated genes.

3.5. Identification of the Clinical Prognostic Value of HubGenes in PDAC. To estimate the influence of hub geneexpression on the prognosis of PDAC, we performedsurvival analysis for the hub genes using the HumanProtein Atlas online tool for differential analysis. Highexpression of nine of the hub genes was positively corre-lated with poor prognosis (COL5A2 P = 0:044; COL6A3

Surv

ival

pro

vabi

lity

1.00.90.80.70.60.50.40.30.20.1

0.0

P = 0.040

0 1 2 3 4 5 6 7Time (years)

Low expression (n = 122)High expression (n = 54)

P = 0.040P

(i)

Surv

ival

pro

vabi

lity

1.00.90.80.70.60.50.40.30.20.1

0.0

P = 0.0027

0 1 2 3 4 5 6 7Time (years)

Low expression (n = 40)High expression (n = 136)

P = 0.0027P

(j)

Figure 6: Survival analysis of the ten hub genes in PDAC based on the Human Protein Atlas. (a) BGN; (b) COL5A2; (c) COL6A; (d)COL11A1; (e) COL12A1; (f) FBN1; (g) POSTN; (h) SPARC; (i) THBS2; (j) VCAN. P < 0:05 was considered statistically significant.

SPA

RC ex

pres

sion

leve

l (lo

g2 (T

PM) Purity

PAA

DPA

AD

PAA

D

B cell CD8+ T cell CD4+ T cell Macrophage Neutrophil Dendritic cell

Macrophage Neutrophil Dendritic cell

Macrophage Neutrophil Dendritic cellPurity B cell CD8+ T cell CD8+ T cell

Purity B cell CD8+ T cell CD8+ T cell

COL6

A3

expr

essio

n le

vel (

log2

(TPM

)FB

N1

expr

essio

n le

vel (

log2

(TPM

)

0.0

2.5

5.0

7.5

3

6

0.25 0.50 0.75 1.00 0.0 0.1 0.2 0.3 0.4 0.1 0.2 0.3 0.4 0.1 0.2 0.3 0.1 0.2 0.3 0.05 0.10 0.15 0.20 0.2 0.4 0.6 0.80.4 0.5 0.0

0.25 0.50 0.75 1.00 0.0 0.1 0.2 0.3 0.4 0.1 0.2 0.3 0.4 0.1 0.2 0.3 0.1 0.2 0.3 0.05 0.10 0.15 0.20 0.2 0.4 0.6 0.80.4 0.5 0.0

0.25 0.50 0.75 1.00 0.0 0.1 0.2 0.3 0.4 0.1 0.2 0.3 0.4 0.1 0.2 0.3 0.1 0.2 0.3 0.05 0.10 0.15 0.20 0.2 0.4 0.6 0.80.4 0.5 0.0

9

12

12

14

10

8

6

4

Infiltration level

Infiltration level

Infiltration level

cor = –0.223p = 3.34e–03

partial. cor = 0.276p = 2.54e–04

partial. cor = 0.481p = 2.66e–11

partial. cor = 0.198p = 1.00e–02 partial. cor = 0.7

p = 1.55e–26partial. cor = 0.584

p = 4.98e–17partial. cor = 0.629

p = 3.03e–20

partial. cor = 0.613p = 5.11e–19

partial. cor = 0.577p = 1.49e–16partial. cor = 0.671

p = 1.06e–23partial. cor = 0.222

p = 3.67e–03partial. cor = 0.442

p = 1.47e–09partial. cor = 0.241

p = 1.47e–03cor = 0.159p = 3.78e–02

cor = 0.222p = 3.49e–03

partial. cor = 0.358p = 1.54e–06

partial. cor = 0.598p = 5.49e–18

partial. cor = 0.167p = 3.04e–02 partial. cor = 0.766

p = 3.39e–34partial. cor = 0.643

p = 2.52e–21partial. cor = 0.689

p = 2.13e–25

Figure 7: Correlation analysis of prognosis-related genes with tumor-infiltrating immune cell types using TIMER.

8 Disease Markers

Page 9: Identification of Key Prognostic Biomarker and Its Correlation ...downloads.hindawi.com/journals/dm/2020/8825997.pdfResearch Article Identification of Key Prognostic Biomarker and

P = 0:013; COL11A1 P = 0:011; COL12A1 P = 0:0012;FBN1 P = 0:028; POSTN P = 0:013; SPARC P = 0:041;THBS2 P = 0:040; VCAN P = 0:0027) (Figure 6).

3.6. Immune Infiltration Analysis. To explore whether therewas a correlation between immune cell infiltration into thetumors and hub gene expression, we analyzed the relation-ships between the ten hub gene signatures and tumor purityand six important immune cell types (CD4+ T cells, CD8+ Tcells, B cells, neutrophils, macrophages, and dendritic cells)using TIMER. We observed that most of these prognosis-related genes were positively correlated with the infiltratinglevels of the different immune cell types but negatively relatedto tumor purity (Supplementary Figure 1, SupplementaryTable 1). In particular, SPARC, COL6A3, and FBN1showed remarkable positive correlations with theinfiltrating levels of the six immune cell types (Figure 7).

Since immunotherapy is currently focused on immuno-logical checkpoint inhibitors (e.g., CTLA4, PDCD1,PDCD1LG2, and CD274), we chose SPARC, COL6A3, andFBN1 as potential target genes and further analyzed the coex-pression relationship of these three genes with these immunecheckpoint-related genes by TIMER. We found that all threeof these potential target genes had strong coexpression rela-tionships with CTLA4, PDCD1, PDCD1LG2, and CD274(Figure 8).

4. Discussion

Pancreatic cancer is one of the most malignant and aggres-sive tumors. Traditional treatment methods (e.g., surgery,chemotherapy, radiotherapy, and other locoregional thera-pies) provide low survival rates. Currently, several clinicalstudies have focused on immunotherapeutic strategies inpancreatic cancer [20]. Therefore, a better understanding ofthe immune infiltration into pancreatic tumors and the iden-tification of novel PDAC immune-related biomarkers mayprove useful for immunotherapy.

In the current study, we found 212 reliable DEGs inPDAC by the comprehensive analysis of three datasets. Thesedifferentially expressed genes were then subjected to GO andKEGG pathway enrichment analysis, which indicated thatthese DEGs were mainly enriched in pathways involved inthe extracellular matrix (ECM) and extracellular structureorganization and the PI3K-Akt signaling pathway. TheECM, a major component of the tumor microenvironment,is known to undergo significant changes during angiogenesisand tumor progression [21]. Activation of the PI3K-AKT sig-naling pathway can enhance pancreatic cancer cell prolifera-tion and invasion [22, 23]. Our results can help elucidate theunderlying mechanisms of TME in pancreatic cancer prolif-eration and invasion. Furthermore, we identified ten hubgenes through PPI network analysis. According to the results

cor = –0.223p = 3.34e–03

cor = –0.186p = 1.44e–02

partial.cor = 0.38p = 2..91e-07

cor = –0.223p = 3.34e–03

cor = –0.185p = 1.51e-02

partial.cor = 0.292p = 1.09e–04

cor = –0.223p = 3.34e–03

cor = –0.308p = 3.84e–05

partial.cor = 0.71p = 1.48e–27

cor = –0.223p = 3.34e–03

cor = –0.152p = 4.68e–02

partial.cor = 0.455p = 4.05e–10

Expr

essio

n le

vel (

log2

(TPM

)

Purity1.00

0.75

0.50

0.25

4

3

2

1

0

0.25 0.50 0.75 1.00 6 8 10 12 0.25 0.50 0.75 1.00 6 8 10 12 0.25 0.50 0.75 1.00 6 8 10 12 0.25 0.50 0.75 1.00 6 8 10 12-1

1.00

0.75

0.50

0.25

4

3

2

1

0

-1

1.00

0.75

0.50

0.25

4

3

2

1

0

-1

1.00

0.75

0.50

0.25

4

3

2

1

0

-1

SPARC Purity SPARC Purity SPARC Purity SPARCPu

rity

CTLA

4

Purit

yPD

CD1

Purit

yPD

CD1L

G2

Purit

yCD

274

Expr

essio

n le

vel (

log2

(TPM

)

Expr

essio

n le

vel (

log2

(TPM

)

Expr

essio

n le

vel (

log2

(TPM

)

Expression level (log2 (TPM) Expression level (log2 (TPM) Expression level (log2 (TPM) Expression level (log2 (TPM)

cor = –0.159p = 3.78e-02

cor = –0.186p = 1.44e–02

partial.cor = 0.39p = 1.36e–07

cor = –0.159p = 3.78e–02

cor = –0.185p = 1.51e–02

partial.cor = 0.297p = 7.91e–05

cor = –0.159p = 3.78e–02

cor = –0.308p = 3.84e–05

partial.cor = 0.675p = 4.02e–24

cor = –0.159p = 3.78e–02

cor = –0.152p = 4.68e–02

partial.cor = 0.478p = 4.01e-11

0.25 0.50 0.75 1.002 4 6 8 100.25 0.50 0.75 1.002 4 6 8 100.25 0.50 0.75 1.002 4 6 8 100.25 0.50 0.75 1.002 4 6 8 10

1.00

0.75

0.50

0.25

0.004

3

2

1

0

1.00

0.75

0.50

0.25

0.004

3

2

1

0

1.00

0.75

0.50

0.25

0.004

3

2

1

0

1.00

0.75

0.50

0.25

0.004

3

2

1

0

Purity COL6A3Purity COL6A3Purity COL6A3Purity

Purit

yCT

LA4

Purit

yPD

CD1

Purit

yPD

CD1L

G2

Purit

yCD

274

COL6A3

Expr

essio

n le

vel (

log2

(TPM

)

Expr

essio

n le

vel (

log2

(TPM

)

Expr

essio

n le

vel (

log2

(TPM

)

Expr

essio

n le

vel (

log2

(TPM

)

Expression level (log2 (TPM) Expression level (log2 (TPM) Expression level (log2 (TPM) Expression level (log2 (TPM)

cor = –0.222p = 3.49e–03

cor = –0.152p = 4.68e–02

partial.cor = 0.577p = 1.54e–16

cor = –0.222p = 3.49e–03

cor = –0.308p = 3.84e–05

partial.cor = 0.782p = 1.67e–36

cor = –0.222p = 3.49e–03

cor = –0.185p = 1.51e–02

partial.cor = 0.338p = 6.06e–06

cor = –0.222p = 3.49e–03

cor = –0.186p = 1.44e–02

partial.cor = 0.451p = 5.80e–10

0.25 0.50 0.75 1.00 42 6 8 0.25 0.50 0.75 1.00 42 6 8 0.25 0.50 0.75 1.00 42 6 8 0.25 0.50 0.75 1.00 42 6 8

1.00

0.75

0.50

0.25

0.004

3

2

1

0

1.00

0.75

0.50

0.25

0.004

3

2

1

0

1.00

0.75

0.50

0.25

0.00

4

3

2

1

0

1.00

0.75

0.50

0.25

0.004

3

2

1

0

Purit

yPD

CD1L

G2

Purit

yCD

274

Purit

yPD

CD1

Purit

yCT

LA4

Purity FBN1 Purity FBN1 Purity FBN1 Purity FBN1

Expr

essio

n le

vel (

log2

(TPM

)

Expr

essio

n le

vel (

log2

(TPM

)

Expr

essio

n le

vel (

log2

(TPM

)

Expr

essio

n le

vel (

log2

(TPM

)

Expression level (log2 (TPM) Expression level (log2 (TPM) Expression level (log2 (TPM) Expression level (log2 (TPM)

Figure 8: The association between the expression levels of SPARC (a), COL6A3 (b), and FBN1 (c) with immune checkpoints using TIMER.

9Disease Markers

Page 10: Identification of Key Prognostic Biomarker and Its Correlation ...downloads.hindawi.com/journals/dm/2020/8825997.pdfResearch Article Identification of Key Prognostic Biomarker and

of our prognostic analysis, high expression of COL5A2,COL6A3, COL11A1, COL12A1, FBN1, POSTN, THBS2,SPARC, or VCAN was associated with poor prognosis forpatients with pancreatic cancer. These results suggest thatthese genes could serve as potential prognostic biomarkersfor PDAC.

Immune cell infiltration in the tumor microenvironmenthas received much attention and has become a promisingtherapeutic target. To further investigate the relationshipbetween these prognosis-related genes and different immunecell types, we predicted gene-immune cell interactions usingTIMER. We found that the expression levels of SPARC,COL6A3, and FBN1 were significantly correlated with sixinfiltrating immune cell types (CD4+ T cells, CD8+ T cells,B cells, neutrophils, macrophages, and dendritic cells).

SPARC is a multifunctional calcium-binding glycopro-tein that is usually secreted into the extracellular matrix andplays a key role in proliferation, migration, adhesion, and dif-ferentiation [24, 25]. Previous studies have shown thatSPARC is localized in the tumor stroma and overexpressedin various cancers, such as breast [26], lung [27, 28],and melanoma [29]. Moreover, SPARC expression is dra-matically increased in gastric tumors and associated withpoor outcome [30]. Stromal SPARC expression is observedin almost 40% of pancreatic adenocarcinoma patients whohave undergone curative resection, and this expression isan independent prognostic factor [31]. Consistent withour research, high SPARC expression in PDAC is associ-ated with poor prognosis.

COL6A3 is an extracellular matrix protein that is typi-cally found in most connective tissues, including muscle,skin, tendon, and vessels. Many recent studies have demon-strated the important role of COL6A3 in the diagnosis andprognosis of colorectal, lung, and prostate cancer [32, 33].A high level of expression of COL6A3 has been observed inpancreatic tumors, where it was correlated with negativeprognostic factors [34]. This finding is consistent with theresults obtained in our study.

FBN1 encodes fibrillin, which is the primary componentof microfibrils in the extracellular matrix. A previous studyreported that FBN1 overexpression plays a key role in thedevelopment of germ cell tumors [35]. FBN1 is also a targetgene for microRNA- (miR-) 133b. miR-133b inhibits theproliferation, migration, and invasion of gastric cancer cellsby increasing FBN1 expression [36]. Moreover, hypermethy-lated FBN1 is found in tissue samples from colorectal cancerpatients but not in healthy controls, suggesting that hyper-methylated FBN1 may be a sensitive biomarker for this dis-ease [37]. The role of FBN1 in the development ofpancreatic cancer and its correlation with the prognosis ofpatients has not been reported previously. In summary, wespeculate that SPARC, COL6A3, and FBN1 might beinvolved in the development of PDAC by regulating thefunctions of the TME.

Previous studies have suggested that the tumor microen-vironment is one of the significant factors determining theprognosis of PDAC and may even play a role in the resistanceto treatment [38, 39]. However, the tumor microenviron-ment is complex and determined by many factors. We com-

prehensively analyzed the correlation between immunecheckpoints and identified three potential prognosis-relatedgenes (SPARC, COL6A3, and FBN1). We were surprised tofind that these genes had a significant coexpression relation-ship with CTLA4, PDCD1, PDCD1LG2, and CD274. Theseresults suggest that the three prognosis-related genes mayaffect the development of pancreatic cancer by affectingimmune checkpoints. Future studies should identify the spe-cific roles of these genes in the regulation of PDAC develop-ment and progression.

5. Conclusion

Taken together, our results suggest that the nine high expres-sion hub genes (COL5A2, COL6A3, COL11A1, COL12A1,FBN1, POSTN, THBS2, SPARC, and VCAN) may be associ-ated with the development and prognosis of PDAC. All thesegenes may be potential prognostic biomarkers for PDAC. Inaddition, three hub genes (SPARC, COL6A3, and FBN1)may also play a vital role in the microenvironment of pancre-atic cancer through the regulation of tumor-infiltratingimmune cells, suggesting they could serve as potential thera-peutic targets for the modulation of the antitumor immuneresponse. Nevertheless, further investigation is needed toconfirm the mechanisms underlying the potential roles ofthese biomarkers in the immune microenvironment.

Data Availability

The data used to support the findings of this study are avail-able from the corresponding author upon request.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Authors’ Contributions

LPZ designed and supervised the study. HL and CZ analyzedthe data, manufactured the figures, and wrote the manu-script. TZ, YH, YNS, and LPZ reviewed the manuscript. Allauthors have read and approved the final manuscript.

Supplementary Materials

Supplementary 1. Supplementary Figure 1: correlation analy-sis of the expression levels of BGN, COL5A2, COL11A1,COL12A1, POSTN, THBS2, and VCAN with tumor-infiltrating immune cell types using TIMER.

Supplementary 2. Supplementary Table 1: relationshipbetween 10 hub gene expression and immune infiltrationlevel in PDAC.

References

[1] R. L. Siegel, K. D. Miller, and A. Jemal, “Cancer statistics,2017,” CA: a Cancer Journal for Clinicians, vol. 67, no. 1,pp. 7–30, 2017.

[2] F. Bray, J. Ferlay, I. Soerjomataram, R. L. Siegel, L. A. Torre,and A. Jemal, “Global cancer statistics 2018: GLOBOCAN

10 Disease Markers

Page 11: Identification of Key Prognostic Biomarker and Its Correlation ...downloads.hindawi.com/journals/dm/2020/8825997.pdfResearch Article Identification of Key Prognostic Biomarker and

estimates of incidence and mortality worldwide for 36 cancersin 185 countries,” CA: a Cancer Journal for Clinicians, vol. 68,no. 6, pp. 394–424, 2018.

[3] T. Kamisawa, L. D. Wood, T. Itoi, and K. Takaori, “Pancreaticcancer,” Lancet, vol. 388, no. 10039, pp. 73–85, 2016.

[4] R. K. Vaddepally, P. Kharel, R. Pandey, R. Garje, and A. B.Chandra, “Review of indications of FDA-approved immunecheckpoint inhibitors per NCCN guidelines with the level ofevidence,” Cancers, vol. 12, no. 3, p. 738, 2020.

[5] D. McDermott, J. Haanen, T. T. Chen, P. Lorigan, S. O'Day,and MDX010-20 Investigators, “Efficacy and safety of ipilimu-mab in metastatic melanoma patients surviving more than 2years following treatment in a phase III trial (MDX010-20),”Annals of Oncology, vol. 24, no. 10, pp. 2694–2698, 2013.

[6] V. Brower, “Checkpoint blockade immunotherapy for cancercomes of age,” JNCI Journal of the National Cancer Institute,vol. 107, no. 3, article djv069, 2015.

[7] J. Brahmer, K. L. Reckamp, P. Baas et al., “Nivolumab versusdocetaxel in advanced squamous-cell non-small-cell lung can-cer,” New England Journal of Medicine, vol. 373, no. 2,pp. 123–135, 2015.

[8] H. J. Hammers, E. R. Plimack, J. R. Infante et al., “Safety andefficacy of nivolumab in combination with ipilimumab inmetastatic renal cell carcinoma: the CheckMate 016 study,”Journal of Clinical Oncology, vol. 35, no. 34, pp. 3851–3858, 2017.

[9] Y. Tomita, S. Fukasawa, N. Shinohara et al., “Nivolumab ver-sus everolimus in advanced renal cell carcinoma: Japanese sub-group 3-year follow-up analysis from the phase III CheckMate025 study,” Japanese Journal of Clinical Oncology, vol. 49,no. 6, pp. 506–514, 2019.

[10] P. Sharma, M. Retz, A. Siefker-Radtke et al., “Nivolumab inmetastatic urothelial carcinoma after platinum therapy(CheckMate 275): a multicentre, single-arm, phase 2 trial,”The Lancet Oncology, vol. 18, no. 3, pp. 312–322, 2017.

[11] P. Sharma, A. Siefker-Radtke, F. de Braud et al., “Nivolumabalone and with ipilimumab in previously treated metastaticurothelial carcinoma: CheckMate 032 nivolumab 1 mg/kg plusipilimumab 3 mg/kg expansion cohort results,” Journal ofClinical Oncology, vol. 37, no. 19, pp. 1608–1616, 2019.

[12] S. Yang, P. He, J. Wang et al., “A novel MIF signaling pathwaydrives the malignant character of pancreatic cancer by targetingNR3C2,” Cancer Research, vol. 76, no. 13, pp. 3838–3850, 2016.

[13] L. Badea, V. Herlea, S. O. Dima, T. Dumitrascu, andI. Popescu, “Combined gene expression analysis of whole-tissue and microdissected pancreatic ductal adenocarcinomaidentifies genes specifically overexpressed in tumor epithe-lia,” Hepato-Gastroenterology, vol. 55, no. 88, pp. 2016–2027, 2008.

[14] P. Shannon, A. Markiel, O. Ozier et al., “Cytoscape: a softwareenvironment for integrated models of biomolecular interac-tion networks,” Genome Research, vol. 13, no. 11, pp. 2498–2504, 2003.

[15] C. H. Chin, S. H. Chen, H. H. Wu, C. W. Ho, M. T. Ko, andC. Y. Lin, “cytoHubba: identifying hub objects and sub-networks from complex interactome,” BMC Systems Biology,vol. 8, Supplement 4, p. S11, 2014.

[16] Z. Tang, C. Li, B. Kang, G. Gao, C. Li, and Z. Zhang, “GEPIA: aweb server for cancer and normal gene expression profilingand interactive analyses,” Nucleic Acids Research, vol. 45,no. W1, pp. W98–W102, 2017.

[17] M. Uhlen, C. Zhang, S. Lee et al., “A pathology atlas of thehuman cancer transcriptome,” Science, vol. 357, no. 6352, arti-cle eaan2507, 2017.

[18] E. Cerami, J. Gao, U. Dogrusoz et al., “The cBio cancer geno-mics portal: an open platform for exploring multidimensionalcancer genomics data,” Cancer Discovery, vol. 2, no. 5, pp. 401–404, 2012.

[19] T. Li, J. Fan, B. Wang et al., “TIMER: a web server for compre-hensive analysis of tumor-infiltrating immune cells,” CancerResearch, vol. 77, no. 21, pp. e108–e110, 2017.

[20] D. Schizas, N. Charalampakis, C. Kole et al., “Immunotherapyfor pancreatic cancer: a 2020 update,” Cancer TreatmentReviews, vol. 86, article 102016, 2020.

[21] A. Naba, K. R. Clauser, D. R. Mani, S. A. Carr, and R. O.Hynes, “Quantitative proteomic profiling of the extracellularmatrix of pancreatic islets during the angiogenic switch andinsulinoma progression,” Scientific Reports, vol. 7, no. 1, article40495, 2017.

[22] G. Hassan, J. Du, S. M. Afify, A. Seno, and M. Seno, “Cancerstem cell generation by silenced MAPK enhancingPI3K/AKT signaling,” Medical Hypotheses, vol. 141, article109742, 2020.

[23] J. Jiang, J. Bai, T. Qin, Z. Wang, and L. Han, “NGF from pan-creatic stellate cells induces pancreatic cancer proliferation andinvasion by PI3K/AKT/GSK signal pathway,” Journal of Cellu-lar and Molecular Medicine, vol. 24, no. 10, pp. 5901–5910,2020.

[24] J. Feng and L. Tang, “SPARC in tumor pathophysiology and asa potential therapeutic target,” Current Pharmaceutical Design,vol. 20, no. 39, pp. 6182–6190, 2014.

[25] J. Vaz, D. Ansari, A. Sasor, and R. Andersson, “SPARC,” Pan-creas, vol. 44, no. 7, pp. 1024–1035, 2015.

[26] A. Zhu, P. Yuan, F. du et al., “SPARC overexpression in pri-mary tumors correlates with disease recurrence and overallsurvival in patients with triple negative breast cancer,” Onco-target, vol. 7, no. 47, pp. 76628–76634, 2016.

[27] Y. Huang, J. Zhang, Y. Y. Zhao et al., “SPARC expression andprognostic value in non-small cell lung cancer,” Chinese Jour-nal of Cancer, vol. 31, no. 11, pp. 541–548, 2012.

[28] K. Komiya, T. Nakamura, C. Nakashima et al., “SPARC is apossible predictive marker for albumin-bound paclitaxel innon-small-cell lung cancer,” OncoTargets and Therapy,vol. 9, pp. 6663–6668, 2016.

[29] M. Bellenghi, R. Puglisi, F. Pedini et al., “SCD5-induced oleicacid production reduces melanoma malignancy by intracellu-lar retention of SPARC and cathepsin B,” The Journal ofPathology, vol. 236, no. 3, pp. 315–325, 2015.

[30] P. Liao, W. Li, R. Liu et al., “Genome-scale analysis identifiesSERPINE1 and SPARC as diagnostic and prognostic bio-markers in gastric cancer,” OncoTargets and Therapy, vol. 11,pp. 6969–6980, 2018.

[31] M. Murakawa, T. Aoyama, Y. Miyagi et al., “The impact ofSPARC expression on the survival of pancreatic ductal adeno-carcinoma patients after curative resection,” Journal of Cancer,vol. 10, no. 3, pp. 627–633, 2019.

[32] W. Liu, L. Li, H. Ye, H. Tao, and H. He, “Role of COL6A3 incolorectal cancer,” Oncology Reports, vol. 39, no. 6, pp. 2527–2536, 2018.

[33] Y. Duan, G. Liu, Y. Sun et al., “COL6A3 polymorphisms wereassociated with lung cancer risk in a Chinese population,”Respiratory Research, vol. 20, no. 1, p. 143, 2019.

11Disease Markers

Page 12: Identification of Key Prognostic Biomarker and Its Correlation ...downloads.hindawi.com/journals/dm/2020/8825997.pdfResearch Article Identification of Key Prognostic Biomarker and

[34] C. Svoronos, G. Tsoulfas, M. Souvatzi, and E. Chatzitheoklitos,“Prognostic value of COL6A3 in pancreatic adenocarcinoma,”Annals of Hepato-Biliary-Pancreatic Surgery, vol. 24, no. 1,pp. 52–56, 2020.

[35] Z. Cierna, M.Mego, I. Jurisica et al., “Fibrillin-1 (FBN-1) a newmarker of germ cell neoplasia in situ,” BMC Cancer, vol. 16,no. 1, p. 597, 2016.

[36] D. Yang, D. Zhao, and X. Chen, “MiR-133b inhibits prolifera-tion and invasion of gastric cancer cells by up-regulating FBN1expression,” Cancer Biomarkers, vol. 19, no. 4, pp. 425–436,2017.

[37] W. H. Li, H. Zhang, Q. Guo et al., “Detection of SNCA andFBN1 methylation in the stool as a biomarker for colorectalcancer,” Disease Markers, vol. 2015, Article ID 657570, 6pages, 2015.

[38] M. Ligorio, S. Sil, J. Malagon-Lopez et al., “Stromal microenvi-ronment shapes the intratumoral architecture of pancreaticcancer,” Cell, vol. 178, no. 1, pp. 160–175.e27, 2019.

[39] D. F. Quail and J. A. Joyce, “Microenvironmental regulation oftumor progression and metastasis,” Nature Medicine, vol. 19,no. 11, pp. 1423–1437, 2013.

12 Disease Markers