Skip to main content
Briefings in Bioinformatics logoLink to Briefings in Bioinformatics
. 2015 Jul 27;17(3):468–478. doi: 10.1093/bib/bbv053

A literature mining-based approach for identification of cellular pathways associated with chemoresistance in cancer

Jung Hun Oh 1,, Joseph O Deasy 2
PMCID: PMC6283363  PMID: 26220932

Abstract

Chemoresistance is a major obstacle to the successful treatment of many human cancer types. Increasing evidence has revealed that chemoresistance involves many genes and multiple complex biological mechanisms including cancer stem cells, drug efflux mechanism, autophagy and epithelial–mesenchymal transition. Many studies have been conducted to investigate the possible molecular mechanisms of chemoresistance. However, understanding of the biological mechanisms in chemoresistance still remains limited. We surveyed the literature on chemoresistance-related genes and pathways of multiple cancer types. We then used a curated pathway database to investigate significant chemoresistance-related biological pathways. In addition, to investigate the importance of chemoresistance-related markers in protein–protein interaction networks identified using the curated database, we used a gene-ranking algorithm designed based on a graph-based scoring function in our previous study. Our comprehensive survey and analysis provide a systems biology-based overview of the underlying mechanisms of chemoresistance.

Keywords: chemotherapy, chemoresistance, cancer, systems biology, pathway, gene

Introduction

Resistance to chemotherapy, which can develop over time, is one of the major obstacles to successful cancer treatment in many human cancers [1–7]. It is known that chemoresistance is multifactorial in nature and involves numerous genes and multiple complex signaling pathways [8, 9]. In recent years, growing attention has been paid to cellular systems in cancer stem cells (CSCs) with the capability of self-renewal and differentiation into other tumor cell subtypes [10]. CSCs are considered to be associated with chemoresistance, tumor recurrence and metastasis [11]. Many reports have shown that other biological mechanisms including ATP-binding cassette (ABC) membrane transporter activity, epithelial–mesenchymal transition (EMT) process, autophagy and altered DNA damage response in CSCs are also significantly involved in chemoresistance [12]. However, despite such extensive studies, the underlying biological mechanisms that contribute to chemoresistance have not been entirely understood [13–16]. To better understand the role of genes and pathways involved in chemoresistance, we performed a comprehensive literature review. This review summarizes the associated biological mechanisms in the context of chemoresistance and reports chemoresistance-related genes and pathways of the published chemoresistance-related research. We demonstrate the results of our analysis performed on these genes using a manually curated pathway analysis platform (MetaCore version 6.21; Thomson Reuters Inc., Carlsbad, CA) [17], resulting in the identification of key biological pathways and highly connected protein–protein networks.

Key biological mechanisms associated with chemoresistance

Cancer stem cells

There is increasing evidence that a subpopulation of cells within most tumors called CSCs or tumor initiating cells arises from transformation of normal stem cells [18–22]. CSCs can self-renew and generate other tumor cell types [10]. CSCs are believed to play a significant role in tumorigenesis, progression, metastasis, recurrence and resistance to various kinds of therapies within a heterogeneous tumor mass [10, 11, 23–26]. Therefore, it is important to elucidate the characteristics of CSCs and to identify key biomarkers or pathways of CSCs to develop CSC-related therapies [27]. A number of CSC markers have been identified including ABCG2, ALDH1A1, CD9, CD44, CD133, NANOG, POU5F1 and SOX2, and it has been reported that these markers are associated with chemoresistance [28]. However, the underlying molecular mechanism of chemoresistance associated with CSCs remains still largely unknown [29, 30].

Epithelial mesenchymal transition

EMT is an important biological process by which epithelial cells lose their cell polarity and acquire a migratory mesenchymal phenotype [31]. Accumulated evidence has shown that EMT is implicated in metastasis and chemoresistance in cancer [32, 33]. More specifically, it has been observed that induction of EMT by EMT inducers such as TWIST1, ZEB1 and FOXQ1 promotes tumor invasion and chemoresistance via loss of cell polarity and loss of cell–cell adhesion [33]. EMT-related markers including E-cadherin (CDH1), Snail (SNAI1), ZEB1 and N-cadherin (CDH2) showed significant difference of expression in residual tumors after chemotherapy for esophageal cancer compared with chemo-naive tumors [34]. In particular, a reduced expression of E-cadherin and an increased expression of Snail in residual tumors were associated with poor response to chemotherapy and shortened survival time. This suggests that there is an association between EMT-related markers and chemoresistance. In other studies, in colorectal cancer [35] and ovarian cancer [36], Snail enhanced the effect of EMT and chemoresistance. Cheng et al. showed that EMT induced by hypoxia can contribute to treatment failure and chemoresistance by activation of hypoxia-inducible factor-1α (HIF-1α/HIF1A) and nuclear factor-κB (NF-κB) in pancreatic cancer, suggesting that inactivation of HIF-1α and NF-κB may be a potential therapeutic strategy to overcome hypoxia-induced chemoresistance [37].

Autophagy

Autophagy is a cellular process that involves the degradation and digestion of long-lived or malfunctioning proteins as well as damaged organelles [38, 39]. The role of autophagy in cancer cell death and survival remains controversial [40, 41]. Although deficient autophagy is associated with tumorigenesis, autophagy also contributes to cancer cell survival, protecting cells from stressful conditions such as hypoxia or cytotoxic therapies, thereby resulting in resistance to common anti-cancer therapies [38]. It was found that overexpression of autophagy-related gene 5 (ATG5), which is one of the key regulators of autophagic cell death, was positively correlated with ABCC1 expression. Both ABCC1 and ATG5 were shown to be significantly associated with chemoresistance in gastric cancer [42]. In another study, CDX1 was found to have a protective effect on colon CSCs, leading to chemoresistance through activation of autophagy [43]. Dean et al. summarized each chemoresistance-related ABC transporter gene and its corresponding chemotherapeutic drugs effluxed by the transporter [44].

ABC transporters

ABC transporters play a critical role in the development of chemoresistance in cancer cells [10]. They form a superfamily of membrane proteins, consisting of 48 known proteins, including multidrug resistance protein 1 (P-glycoprotein/MDR1/ABCB1), multidrug resistance-associated protein 1 (MRP1/ABCC1) and breast cancer resistance protein (BCRP/ABCG2) [45]. P-glycoprotein is a key protein involving drug efflux and multidrug resistance in many cancers [46–49]. ABC transporters normally protect cells from excessive extracellular and intracellular concentrations of xenobiotics and toxins [12]. An intriguing property of stem cells is that they express high levels of ABC transporters [44]. Elevated expression levels of ABC transporters in CSCs pump chemotherapy drugs out of the cells and accordingly decrease the intracellular accumulation of drugs, thereby contributing to multidrug resistance [50]. Novel chemotherapeutic strategies are being developed through targeted interventions of ABC transporter-related pathways [10, 51]. Investigators have designed numerous strategies to modulate ABC transporter intrinsic activity [52, 53] or regulate the expression levels of these proteins [54].

Computational systems biology

Computational systems biology is an emerging field that focuses on the modeling of complex biological components for a system-level understanding of molecular and cellular systems using mathematical and computational techniques [55, 56]. However, the development of computational approaches that identify meaningful biological insights from large-scale data is a major challenge [57, 58]. Complex biological components including genes, proteins and transcription factors are often represented as networks in which the connected edges indicate interactions or associations. Beyond simple graph representations, Bayesian modeling in biological networks has been used extensively, as reviewed by Wilkinson [59]. Materi and Wishart summarized computational systems biology approaches that have been used to provide novel insights into tumor genesis, growth, apoptosis, vascularization and therapy [60]. In our previous study, we used a computational systems biology approach based on graph theory to investigate the importance of biomarkers and to identify radiation response-related pathways and hub genes based on literature review [61]. Computational systems biology approaches have also been used to identify potential drug target enzymes in sphingolipid metabolism using metabolic control and pathway analysis [62], and to identify cancer chemoresistance-associated pathways from integrated biological interaction networks collected from public databases [13]. An integrative network inference approach using Bayesian modeling was proposed to infer genetic and metabolic pathways associated with chemoresistance in a way that a gene interaction network and a metabolic network, separately generated using gene expression and metabolite data, are merged into a larger network to predict correlations between genes and metabolizing enzymes [63].

In this study, we mined the scientific literature to identify chemoresistance-related genes, and then used the MetaCore system to construct protein–protein interaction networks and to identify biological pathways. In addition, to investigate the importance of chemoresistance-related markers in protein–protein interaction networks identified using the curated database, we used our gene-ranking algorithm designed in our previous study [61]. Although some microRNAs have been implicated in drug resistance, we limit ourselves to gene products.

Literature mining results

We performed a comprehensive literature review to identify chemoresistance-related genes from published papers. Peer-reviewed papers published until May 2015 were searched by using PubMed (http://www.ncbi.nlm.nih.gov/pubmed/) or GoPubMed (http://www.gopubmed.com/web/gopubmed/) search engine with the following query: (chemo-resistance[Title] OR chemoresistance[Title] OR chemotherapy resistance[Title]) AND (cancer[Title] OR adenocarcinoma[Title] OR carcinoma[Title] OR neoplasms[Title] OR tumor[Title]). Our search resulted in 894 articles, of which 462 were relevant to the aim of study. The inclusion criteria are as follows: (1) studies on chemoresistance-related biomarkers (genes or proteins), and (2) studies on eight cancers (breast, colon, head and neck, lung, ovarian, pancreatic, prostate and stomach). Through the literature review, several types of chemoresistance-related biomarkers including genes, proteins, kinases and protein complexes were identified. To unify the nomenclature of biomarkers differently used across studies, we converted all biomarker terms into their corresponding official gene symbols. As a result, 334 unique genes and four families of proteins were identified. Among them, many biomarkers showed an association between chemoresistance and biological processes including CSCs (CSC markers: ALDH1A1, CD9, CD44, CD133, NANOG, POU5F1 and SOX2), EMT (EMT inducers: TWIST1, FOXQ1, SNAI1 and ZEB1), autophagy (ATG5) and ABC transporters (ABCB1, ABCC1 and ABCG2) as mentioned earlier. Table 1 summarizes chemoresistance-related genes for each cancer. Supplementary data provide the information as well. Ovarian, breast and pancreatic cancers had relatively many biomarkers with 122, 91 and 79, respectively. For these three cancers, 11 biomarkers including ABCB1, AKT1, BCL2, BCL2L1, CD44, ERBB2, NF-κB, NOTCH1, PTGS2, SNAI1 and TP53 were commonly identified. Figure 1 A shows a Venn diagram depicting the number of shared and unique chemoresistance genes among breast, ovarian and pancreatic cancers. Figure 1B shows a Venn diagram for breast, ovarian and lung cancers. Even though in lung cancer, a smaller number of genes was identified with 66 compared with pancreatic cancer, 12 genes were common among the three cancers.

Table 1.

Chemoresistance-related genes identified by literature review

Cancer N Genes
Breast 91 ABCB1, ABCC1, ABCG2, AKT1, ALDH1A1, ANXA2, ATP6V0C, AURKA, AURKB, AXL, BCL2, BCL2L1, BIRC5, BMP6, BRCA1, BSG, CASP3, CAV1, CD44, CSF1, CTBP1, CTBP2, CTNNB1, CXCL1, CXCL2, CXCR7, DEPTOR, DUSP1, EIF4E, EIF5A2, EP300, ERBB2, ESR1, FGF1, FOXQ1, GH1, GHR, GSTP1, HIF1A, HSPB1, HSPG2, IGF1, IL17A, IL8, IMP3, JUN, KIF14, KRT19, KRT8, LAPTM4B, LCMT2, LGALS7, LKB1, MAPK1, MAPK3, MAPT, MMP1, MSH2, MTDH, NDST1, NOTCH1, PDGFRA, PDGFRB, PEA15, PGK1, PIK3CA, PRDX2, PRKCE, PTEN, PTGS2, RARRES1, SIRT1, SLC4A7, SLC9A1, SMAD3, SNAI1, SPHK2, SPP1, SRC, STAT3, TAZ, TGFB1, TNF, TP53, UBE2I, ULK1, YBX1, YWHAE, YWHAZ, NF-κBa, MT proteinsa
Colon 52 ABCB1, ABCC2, ABCC3, ABCF1, ABCG2, AKT1, AKT2, ALDH1A1, AURKA, BCL2, BCL2L1, CA12, CD44, CDX1, CRY2, DKK1, DKK4, DTL, EGFR, HDAC4, HIF1A, IGF1, IGFBP3, IKBKB, KIN, LGR5, LYN, MCL1, MMP7, MMP9, NR1H4, NR1I2, NR4A2, PAK6, POSTN, PPARGC1A, PSAT1, PTEN, PYY, REPS2, SCGB2A1, SDC1, SIRT1, SNAI1, SP1, TFAP2E, TNFSF13, TP53, TYMS, UCP2, VEGFA, NF-κBa
Head and neck 38 ABCB1, ABCC2, ABCG2, ATP7B, BCL2, BDNF, BIRC5, BMI1, BSG, C2orf40, CA9, CASP8, CD44, EGFR, ERCC1, ERCC4, FAM168A, GDF10, GSTP1, HAX1, HECA, HSPA5, HSPB1, HTATIP2, IGF1, NANOG, NEFL, NOS2, NTRK2, POU5F1, PPM1D, PROM1, STAT3, TGFA, TGFBR3, TRIP13, TWIST1, ZEB1
Lung 66 ABCB1, ABCC2, ABCC5, AIFM1, ANXA1, ANXA2, ANXA4, BAX, BCL2, BIRC5, BIRC6, BRCA1, BSG, CASP3, CASP9, CD44, CD9, CDKN1A, CXCR4, DKK3, DUSP1, E2F3, EIF3A, ERBB2, ERCC2, EZH2, FOXM1, GSTP1, HDAC1, HDAC4, HER2, HOXA1, HSP90B1, HSPA5, HSPB1, KDR, KEAP1, KRAS, LCMT2, LGALS1, MAPK14, MCL1, MEOX2, MET, MSH2, NFE2L2, PTGS2, RUNX3, S100A11, SCGN, SKP2, SLCO5A1, SND1, SPHK2, SPP1, STAT3, TLR7, TLR8, TP53, TUBB3, TWIST1, VDAC1, XPA, XRCC1, ZEB1, NF-κBa
Ovarian 122 ABCB1, ABCB3, ABCC1, ABCC2, ABCC3, ACTN4, ACVR1C, AKR1C1, AKT1, AKT2, ANXA11, ANXA4, APAF1, ARID1A, ATP7B, AURKA, AURKB, BAX, BCL2, BCL2L1, CASP3, CASP9, CD44, CDCP1, CDKN1B, CFLAR, CHI3L1, CLEC3B, CLU, CSN1S1, CXCR4, CYCS, DAXX, DICER1, DNMT1, DUSP6, EDNRA, EGFR, ENG, EPB41L1, ERBB2, ERCC1, ERCC4, EZH2, FASN, FOXP1, GLI2, GSC, GSTP1, HDAC1, HK2, HSP90AA1, HTRA2, IGF1, IL6, JAG1, KDM5B, KDR, KIT, KLK4, KLK5, KLK6, KLK7, KRAS, LAPTM4B, LCMT2, LGALS1, MAD2L1, MAPK1, MAPK14, MAPK3, MAPK8, MIEN1, MKI67, MSLN, MSMB, MTDH, MVP, MYD88, NFE2L2, NOTCH1, PAK7, PHB, PML, POSTN, PPARG, PPARGC1A, PPM1D, PRPF4, PRSS8, PTEN, PTGS2, PTK2, RGS10, RGS17, RSF1, SFRP5, SLC28A1, SNAI1, SNAI2, SPHK2, STAT1, STAT3, SULF1, TEAD1, TEAD3, TEAD4, TFAM, TGFBI, TLR4, TNFSF10, TOP2A, TP53, TP73, TRIM27, VEGFB, WNT5A, XIAP, YAP1, ZMYND10, NF-κBa, HSP70a
Pancreatic 79 ABCB1, ABCC2, ABCC5, ABCG2, AKT1, ALCAM, APAF1, BAD, BCL2, BCL2L1, BIRC5, BMI1, BNIP3, BRG1, BTRC, CASP8, CASP9, CBL, CCND1, CCNG2, CD44, CEACAM6, CXCR4, DCK, DLL4, DNMT1, EGFR, ERBB2, ETS1, EZH2, F2R, FASLG, GLI1, HES1, HIF1A, HMGA1, HOXA5, IGF1R, IL1B, IL6, ILK, ISG15, KRT18, L1CAM, LHX2, MACC1, MAP3K7, MAPK8, MDK, MET, MUC1, MUC4, NEU1, NOTCH1, NOTCH2, NRP1, PARK7, PLAU, PROM1, PTGS2, PTK2, RRM1, RRM2, S100A4, SFN, SHH, SIRT1, SLC29A1, SMO, SNAI1, SRC, STAT1, TNFSF10, TP53, TP63, TUBB3, WNT5A, XIAP, NF-κBa
Prostate 28 ABCB1, ABCC1, ANXA3, AURKA, AURKB, BSG, CD44, CDH1, CLU, DAB2IP, EGF, EGR1, ETS1, GATA2, GDF15, HSP27, IGF2, IGFBP2, IL6, KEAP1, MMP9, NFE2L2, NOTCH1, NR2C2, REST, SIRT1, TGFA, TLR4
Stomach 41 ABCB1, ABCC1, ABCG2, AGBL2, AKT1, ATG5, BAX, BCL2, BCL2L1, BMI1, CAV1, CD44, CDH1, DDB2, DPYD, ERCC1, FOXM1, HIF1A, IL-33, IL6, JUN, LGR5, MAPK1, MAPK3, MYC, NOTCH1, NR4A2, PEBP1, PROM1, PTEN, RHOA, SOX2, STAT3, TFAP2E, TP53, TRIM24, TYMP, TYMS, WNT6, NF-κBa, MT proteinsa

aA family of proteins.

Figure 1.

Figure 1.

Venn diagrams depicting the number of shared and unique genes (A) among breast, ovarian, and pancreatic cancers and (B) among breast, ovarian, and lung cancers using the gene list shown in Table 1 (a family of proteins was counted as 1). For both the Venn diagrams, the left-top and right-top circles indicate breast cancer and ovarian cancer, respectively. A colour version of this figure is available at BIB online: http://bib.oxfordjournals.org.

To investigate the most significant biological processes and protein–protein interaction networks based on the list of genes identified through literature review (Table 1), we uploaded the gene symbols into the MetaCore platform. To identify the interaction networks, we used an option of ‘direct interactions’ that allows the platform to search for direct interactions between genes, proteins, transcription factors and compounds from the MetaCore database built by the MetaCore scientists with their data curation process, resulting in directly connected protein–protein interaction networks. Figure 2 A illustrates a directly connected protein–protein interaction network identified using 79 biomarkers of pancreatic cancer. It is worth noting that this network consisted of 72 proteins, showing that the input genes are highly connected to each other. As expected, genes with similar functions are often clustered in the same pathway or biological process [64]. Similar results were found for other cancers (data not shown). As can be seen in Figure 2A, the network is centered around p53 (TP53), with the most interactions with other proteins, implying its important role in chemoresistance. Figure 2B illustrates a simplified network of Figure 2A obtained, using a power graph analysis tool that uses two abstractions: power nodes and power edges shown as thick circles and lines, respectively. A power edge indicates that all nodes inside a power node on one side are connected with all nodes on the other side. For more information for power graph analysis, see [65]. From the directly connected network constructed using the MetaCore software for each cancer, the number of interactions of each gene was counted. Table 2 shows the top 10 genes with the most interactions for ovarian, breast and pancreatic cancers when genes in Table 1 were used for that cancer type. In a directly connected network constructed using all the biomarkers in Table 1, TP53 had the most interactions with other genes with 93 outgoing edges and 64 incoming edges. In ovarian and pancreatic cancers, TP53 also had the most interactions with 64 and 52 edges, respectively, whereas in breast cancer, the JUN gene had the most interactions with 57 edges, demonstrating its key role in chemoresistance. Note that if one gene has interactions with another gene bidirectionally, it was counted for both outgoing and incoming edges.

Figure 2.

Figure 2.

Figure 2.

Connected protein–protein interaction networks constructed using (A) a MetaCore platform tool, and (B) a power graph analysis tool with a list of chemoresistance-related genes of pancreatic cancer.

Table 2.

Ranking of genes based on the number of interactions from directly connected networks constructed by the MetaCore platform when the gene list shown in Table 1 was used

When all genes shown in Table 1 were used
Ranking Gene Number of outgoing edges Number of incoming edges Total
1 TP53 93 64 157
2 SP1 124 31 155
3 JUN 98 33 131
4 ESR1 77 50 127
5 MYC 70 41 111
6 STAT3 62 42 104
7 EP300 78 19 97
8 CTNNB1 59 36 95
9 HIF1A 63 26 89
10 HDAC1 70 16 86
Ovarian cancer
1 TP53 35 29 64
2 STAT3 22 18 40
3 HDAC1 30 5 35
4 HSP90AA1 22 7 29
5 EZH2 21 6 27
6 MAPK14 17 8 25
7 EGFR 11 13 24
8 PPARG 12 12 24
9 CASP3 17 6 23
10 NOTCH1 11 12 23
Breast cancer
1 JUN 43 14 57
2 TP53 33 23 56
3 ESR1 35 19 54
4 STAT3 25 18 43
5 CTNNB1 19 19 38
6 HIF1A 23 15 38
7 AKT1 19 14 33
8 EP300 24 9 33
9 SMAD3 9 15 24
10 BRCA1 10 12 22
Pancreatic cancer
1 TP53 29 23 52
2 NOTCH1 18 13 31
3 HIF1A 24 5 29
4 EGFR 13 13 26
5 STAT1 18 7 25
6 AKT1 13 10 23
7 SRC 13 8 21
8 NF-kBa 18 1 19
9 ETS1 14 4 18
10 EZH2 13 5 18

aA family of proteins.

To investigate the biological importance of genes in the protein–protein interaction networks, we used a graph-based scoring function that we proposed in our previous paper [61]. This approach calculates a global gene score using the Floyd–Warshall algorithm based on a power law and using the number of papers that showed the interaction evidence between two proteins as a weighting value on the interaction. Table 3 shows the top 10 genes for ovarian, breast and pancreatic cancers. Interestingly, TP53 was first ranked in all the three cancers. However, overall there was a difference in genes compared with those in Table 2 that were ranked based on the number of gene interactions. Using the MetaCore software, significant biological processes were identified for ovarian, breast and pancreatic cancers (Table 4 ). In all the three cancers, signal transduction NOTCH signaling and development EMT regulation processes were highly ranked.

Table 3.

Ranking of genes based on our proposed gene ranking algorithm from directly connected networks constructed by the MetaCore platform when the gene list shown in Table 1 was used

When all genes shown in Table 1 were used
Ranking Gene Score
1 EGF 1128.1
2 SP1 1066.0
3 TP53 1043.0
4 VEGFA 1023.6
5 MYC 1000.5
6 ESR1 998.7
7 EGFR 998.7
8 EP300 994.9
9 IGF1 992.3
10 APAF1 986.4
Ovarian cancer
1 TP53 332.4
2 EGFR 319.4
3 APAF1 314.8
4 STAT3 312.6
5 CYCS 312.2
6 BCL2L1 303.8
7 ERBB2 303.7
8 CASP9 301.4
9 XIAP 301.2
10 PTEN 300.9
Breast cancer
1 TP53 276.9
2 EP300 268.6
3 ESR1 264.8
4 STAT3 263.0
5 JUN 260.5
6 SRC 258.0
7 HIF1A 255.6
8 MAPK1 248.3
9 CTNNB1 245.9
10 BRCA1 244.2
Pancreatic cancer
1 TP53 247.8
2 EGFR 236.5
3 HIF1A 232.9
4 CBL 231.6
5 BAD 230.5
6 BCL2L1 227.6
7 STAT1 226.7
8 BCL2 223.7
9 AKT1 223.4
10 SRC 223.3

The score indicates the importance of genes in the network. As the score increases, the importance of the gene increases as well.

Table 4.

Significant biological processes and their corresponding genes

Ovarian cancer
No. Processes/genes FDR N
1 Development_blood vessel morphogenesis 1.08E-13 22
KLK7,CASP3,NF-kB,EGFR,NOTCH1,STAT3,JAG1,EDNRA,MAPK3,ERBB2,NOTCH1,KDR,STAT1,MAPK1; MAPK3,MAPK1,MAPK14,NOTCH1;NOTCH2,AKT2;AKT1,NOTCH1,MAPK8,CXCR4,VEGFB
2 Signal transduction_NOTCH signaling 1.34E-13 24
NF-kB,EGFR,NOTCH1,STAT3,MAPK8,JAG1,MAPK14,MAPK3,WNT5A,ERBB2,NOTCH1,CD44,PTEN,MAPK1; MAPK3,CDKN1B,MAPK1,MAPK14,AKT2;AKT1,NOTCH1,AKT2,TP73,TP53,CFLAR,MAPK8
3 Development_hemopoiesis, erythropoietin pathway 2.51E-10 18
NF-kB,STAT3,KIT,MAPK8,MAPK14,MAPK3,BCL2,KRAS,AKT1,STAT1,MAPK1;MAPK3,CDKN1B,MAPK1, MAPK14,BCL2L1,XIAP,AKT2;AKT1,MAPK8
4 Development_EMT_regulation of epithelial-to-mesenchymal transition 6.94E-10 22
SNAI2,EGFR,NOTCH1,STAT3,MAPK8,JAG1,MAPK14,EDNRA,PTGS2,BCL2,NOTCH1,STAT1,PTEN,MAPK1; MAPK3,SNAI1,CDKN1B,MAPK14,AKT2;AKT1,PTK2,WNT5A,MAPK8,GSC
5 Inflammation_amphoterin signaling 1.18E-09 14
NF-kB,MAPK8,MAPK14,MAPK3,TLR4,AKT1,MAPK1;MAPK3,MYD88,MAPK1,IL6,MAPK14,AKT2;AKT1, AKT2, MAPK8
Breast cancer
1 Signal transduction_NOTCH signaling 1.64E-18 27
PIK3CA,NF-kB,SMAD3,NOTCH1,STAT3,MAPK3,PDGFRB;PDGFRA,ERBB2,JUN,NOTCH1,CD44,TGFB1,PTEN, EP300,EIF4E,MAPK1;MAPK3,PDGFRA,SRC,MAPK1,PDGFRB,TGFB1,AKT2;AKT1,NOTCH1,TP53,PIK3CA, HIF1A,CTNNB1
2 Development_EMT_regulation of epithelial-to-mesenchymal transition 5.34E-15 25
PIK3CA,KRT8,ESR1,SMAD3,NOTCH1,STAT3,JUN,PDGFRB;PDGFRA,PTGS2,BCL2,JUN,NOTCH1,TGFB1, PTEN,MAPK1;MAPK3,SNAI1,PDGFRA,SRC,PDGFRB,TGFB1,AKT2;AKT1,FGF1,TNF,HIF1A,CTNNB1
3 Cell cycle_G1-S growth factor regulation 1.55E-14 22
PIK3CA,NF-kB,SMAD3,STAT3,JUN,MAPK3,ERBB2,JUN,JUN,AKT1,TGFB1,MAPK1;MAPK3,PRKCE,PDGFRA, SRC,MAPK1,PRKCE,TGFB1,AKT2;AKT1,FGF1,IGF1,PIK3CA
4 Inflammation_IL-2 signaling 2.01E-13 16
PIK3CA,NF-kB,ESR1,STAT3,JUN,MAPK3,BCL2,JUN,PTEN,MAPK1;MAPK3,MAPK1,PRKCE,BCL2L1,ESR1, AKT2;AKT1,PIK3CA
5 Inflammation_IL-6 signaling 1.88E-12 16
PIK3CA,CASP3,NF-kB,STAT3,JUN,MAPK3,BCL2,JUN,AKT1,YWHAE,MAPK1;MAPK3,MAPK1,AKT2;AKT1, YWHAZ,PIK3CA,YWHAZ;YWHAE;SFN
Pancreatic cancer
1 Signal transduction_NOTCH signaling 3.23E-16 23
SRC,NF-kB,TP63,EGFR,HES1,NOTCH1,MAPK8,NEU1,NOTCH2,AKT2;AKT1,NOTCH1,WNT5A,SKP2;BTRC, BTRC,ERBB2,DLL4,NOTCH1,TP53,CD44,CCND1,MAPK8,NOTCH2,HIF1A
2 Development_EMT_regulation of epithelial-to-mesenchymal transition 7.48E-15 24
IL1B,SRC,EGFR,NOTCH1,MUC1,MAPK8,ETS1,AKT2;AKT1,PTGS2,BCL2,DLL4,NOTCH1,PTK2,S100A4,CBL, WNT5A,STAT1,MAPK8,MET,MAP3K7,HIF1A,SNAI1,ILK,KRT18
3 Development_blood vessel morphogenesis 8.64E-13 20
IL1B,SRC,NF-kB,EGFR,NOTCH1,NRP1,NOTCH1;NOTCH2,AKT2;AKT1,F2R,NOTCH1,ERBB2,DLL4,L1CAM, NOTCH1,STAT1,MAPK8,ABCG2,MET,HIF1A,CXCR4
4 Apoptosis_anti-apoptosis mediated by external signals via PI3K/AKT 9.79E-11 19
IL1B,BAD,SRC,NF-kB,IL6,EGFR,BCL2L1,AKT2;AKT1,APAF1,BCL2,ERBB2,BIRC5,TP53,IGF1R,PTK2,CASP9, CBL,MET,ILK
5 Development_hedgehog signaling 4.16E-10 19
SRC,BMI1,SHH,TP63,HES1,GLI1,NOTCH1,SIRT1,NOTCH1,WNT5A,SKP2;BTRC,BTRC,SHH,NOTCH1,TP53, AKT1,CCND1,MDK,SNAI1

FDR indicates the significance of biological processes identified.

FDR: false discovery rate.

Numerous studies have identified chemoresistance-related genes or proteins in different cancer types. In bladder cancer, HOXC9, PSEN1, SRSF2, PLAU and HIC2 were found to be associated with chemoresistance [7, 15, 66]. Overexpression of ADAM10 regulated malignant cell growth and invasion, suggesting that ADAM10 could be a candidate therapeutic target [67]. Upregulation of UCA1 activated Wnt signaling and increased cisplatin resistance [68]. In lung cancer, it was reported that upregulation of SCGN [69], BIRC6 [70], EZH2 [71], ABCB1 [72], BCL2 [73], HSPA5/HSP90B1 [74] and TP53/LCMT2 [75] as well as downregulation of HOXA1 [76] were associated with chemoresistance. Numerous studies have reported the interplay between genes and signaling pathways in response to chemoresistance: in breast cancer, (IL17A [77]/BMP6 [78] and ERK), (ABCG2 [79]/BCL2 [80] and PI3K/Akt/NF-κB) and (BIRC5 and PI3K/Akt2 pathway) [81]; in colon cancer, (SDC1 and EGFR) [82], (POSTN and PI3K/Akt/surviving) [83] and (BCL2 and β6-integrin-ERK/MAP kinase pathway) [84]; in ovarian cancer, (XIAP and PI3K/Akt) [85], (MSMB and Lin28b/Let-7) [86] and (SFRP5 and Wnt signaling pathway) [87]; in pancreatic cancer, (IGF1R and PI3K/Akt/NF-κB) [88], (CASP8/CASP9/BCL2 and ERK) [89], (NRP1 and MAPK) [90] and (BNIP3 and PI3K/Akt pathway) [91]; in prostate cancer, (ABCC1 and NOTCH1) [92], (DAB2IP and Egr-1/Clusterin) [93] and (TLR4 and PI3K/Akt pathway) [94]; and in stomach cancer, (PROM1 and PI3K/Akt/p70S6K) [95], (CD44 and Hedgehog) [96], (TP53/MYC and PI3K/Akt and MAPK/ERK) [97] and (ABCB1 and p38-MAPK pathway) [1], where (gene and pathway).

Discussion

We conducted a literature search to find peer-reviewed published studies on chemoresistance with the aim of identifying chemoresistance-related genes, networks showing gene interactions and pathways. To our best knowledge, this is the largest literature review that surveyed papers published on chemoresistance for several cancers.

Unsurprisingly, in our literature review, the PI3K/Akt pathway was commonly found as one of the important pathways associated with chemoresistance in several cancers. The PI3K/Akt pathway is involved in cell growth, proliferation and survival [98, 99]. Alterations to the PI3K/Akt pathway are common in human cancers [100]. The activation of the PI3K/Akt pathway contributes to resistance to specific therapeutic agents [101]. Inhibition of this pathway can induce suppression of cell proliferation and in some circumstances promote cell death in cancer cells [102]. PI3K/Akt pathway inhibitors have been shown to lead to successful chemotherapy by reversing the repression of apoptosis and resistance to therapeutic agents [103, 104]. Hence, the PI3K/Akt pathway has become an attractive target for drug development.

To identify protein–protein interaction networks that can be constructed using a list of genes found via literature review, we used the MetaCore integrated software suite. This analysis resulted in highly condensed networks, implying that there are strong biological interactions among these genes, especially hub genes that interact with many genes. The results support a general assumption that genes with similar functions are often clustered and linked to each other in biological networks. In particular, it is noted that p53 (TP53) has the most interactions with other proteins in several cancers. It has been reported that mutant TP53 is associated with chemoresistance in different cancers. Kandioler-Eckersberger et al. found that fluorouracil, epirubicin and cyclophosphamide (FEC) therapy is dependent on normal p53, whereas paclitaxel responds to p53-deficient breast cancer patients [105]. Fiorini et al. found that mutant p53 stimulates chemoresistance to gemcitabine in pancreatic adenocarcinoma cells, observing a significant expression of CDK1 and CCNB1 genes in mutant p53 cell lines, while a significant decrease for these genes in wild-type p53 cell lines [106]. Bush and Li reviewed the relationship between p53 and multidrug transporters with respect to cancer chemoresistance and concluded that the relationship is conditional, depending on cellular environment, therapeutic agents and the nature of the p53 mutation [107]. To investigate the biological importance of genes, we applied our gene ranking algorithm to protein–protein interaction networks identified using the MetaCore platform. It was found that TP53 was first ranked in ovarian, breast and pancreatic cancers. Consequently, it is obvious that TP53 plays an important role in chemoresistance as a ‘beacon’ gene that helps identify critical gene cascades, although it has not been used as a clinical marker for chemoresistance [108].

In conclusion, these networks identified emphasize the multifactorial dependence of overall treatment response on many genes that are often mutated in individual tumors. For future work, we hypothesize that these network results could help guide the design of multigene predictive signatures with the increased probability of chemotherapy success.

Key Points

  • Chemoresistance is a major obstacle to successful therapy in human cancer and is associated with many genes and multiple complex biological mechanisms.

  • We surveyed chemoresistance-related genes and biological pathways through a comprehensive literature review.

  • We analyze genes found through literature review, and then use a curated database to identify significant pathways and protein–protein interaction networks.

Funding

This work was funded by an internal grant from Memorial Sloan Kettering Cancer Center.

Supplementary Material

Supplementary Data

References

  • 1. Guo X, Ma N, Wang J, et al. Increased p38-MAPK is responsible for chemotherapy resistance in human gastric cancer cells. BMC Cancer 2008;8:375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Rivera E, Gomez H. Chemotherapy resistance in metastatic breast cancer: the evolving role of ixabepilone. Breast Cancer Res 2010;12(Suppl 2):S2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Yang JY, Chang CJ, Xia W, et al. Activation of FOXO3a is sufficient to reverse mitogen-activated protein/extracellular signal-regulated kinase kinase inhibitor chemoresistance in human cancer. Cancer Res 2010;70:4709–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Antoon JW, White MD, Slaughter EM, et al. Targeting NFĸB mediated breast cancer chemoresistance through selective inhibition of sphingosine kinase-2. Cancer Biol Ther 2011;11:678–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. An Y, Ongkeko WM. ABCG2: the key to chemoresistance in cancer stem cells?. Expert Opin Drug Metab Toxicol 2009;5:1529–42. [DOI] [PubMed] [Google Scholar]
  • 6. Gasco M, Crook T. p53 family members and chemoresistance in cancer: what we know and what we need to know. Drug Resist Update 2003;6:323–8. [DOI] [PubMed] [Google Scholar]
  • 7. Lv L, Li Y, Deng H, et al. MiR-193a-3p promotes the multi-chemoresistance of bladder cancer by targeting the HOXC9 gene. Cancer Lett 2015;357:105–13. [DOI] [PubMed] [Google Scholar]
  • 8. Wen X, Zhang HD, Zhao L, et al. Ginsenoside Rh2 differentially mediates microRNA expression to prevent chemoresistance of breast cancer. Asian Pac J Cancer Prev 2015;16:1105–9. [DOI] [PubMed] [Google Scholar]
  • 9. Chuthapisith S, Layfield R, Kerr ID, et al. Proteomic profiling of MCF-7 breast cancer cells with chemoresistance to different types of anti-cancer drugs. Int J Oncol 2007;30:1545–51. [PubMed] [Google Scholar]
  • 10. Abdullah LN, Chow EK. Mechanisms of chemoresistance in cancer stem cells. Clin Transl Med 2013;2:3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Colak S, Zimberlin CD, Fessler E, et al. Decreased mitochondrial priming determines chemoresistance of colon cancer stem cells. Cell Death Differ 2014;21:1170–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Coley HM. Mechanisms and strategies to overcome chemotherapy resistance in metastatic breast cancer. Cancer Treat Rev 2008;34:378–90. [DOI] [PubMed] [Google Scholar]
  • 13. Chao SY, Chiang JH, Huang AM, et al. An integrative approach to identifying cancer chemoresistance-associated pathways. BMC Med Genomics 2011;4:23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Lønning PE. Genes causing inherited cancer as beacons to identify the mechanisms of chemoresistance. Trends Mol Med 2004;10:113–18. [DOI] [PubMed] [Google Scholar]
  • 15. Deng H, Lv L, Li Y, et al. The miR-193a-3p regulated PSEN1 gene suppresses the multi-chemoresistance of bladder cancer. Biochim Biophys Acta 2015;1852:520–8. [DOI] [PubMed] [Google Scholar]
  • 16. Pan B, Yi J, Song H. MicroRNA-mediated autophagic signaling networks and cancer chemoresistance. Cancer Biother Radiopharm 2013;28:573–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. MetaCore™. http://thomsonreuters.com/metacore/
  • 18. Zhang XL, Jia Q, Lv L, et al. Tumorspheres derived from HCC cells are enriched with cancer stem cell-like cells and present high chemoresistance dependent on the Akt pathway. Anticancer Agents Med Chem 2015;15:755–63. [DOI] [PubMed] [Google Scholar]
  • 19. Asuthkar S, Stepanova V, Lebedeva T, et al. Multifunctional roles of urokinase plasminogen activator (uPA) in cancer stemness and chemoresistance of pancreatic cancer. Mol Biol Cell 2013;24:2620–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Paldino E, Tesori V, Casalbore P, et al. Tumor initiating cells and chemoresistance: which is the best strategy to target colon cancer stem cells?. Biomed Res Int 2014;2014:859871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Siddique HR, Saleem M. Role of BMI1, a stem cell factor, in cancer recurrence and chemoresistance: preclinical and clinical evidences. Stem Cells 2012;30:372–8. [DOI] [PubMed] [Google Scholar]
  • 22. Dontu G, Liu S, Wicha MS. Stem cells in mammary development and carcinogenesis: implications for prevention and treatment. Stem Cell Rev 2005;1:207–13. [DOI] [PubMed] [Google Scholar]
  • 23. Li S, Yue D, Chen X, et al. Epigenetic regulation of CD271, a potential cancer stem cell marker associated with chemoresistance and metastatic capacity. Oncol Rep 2015;33:425–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Kang MJ, Kim HP, Lee KS, et al. Proteomic analysis reveals that CD147/EMMPRIN confers chemoresistance in cancer stem cell-like cells. Proteomics 2013;13:1714–25. [DOI] [PubMed] [Google Scholar]
  • 25. Crea F, Danesi R, Farrar WL. Cancer stem cell epigenetics and chemoresistance. Epigenomics 2009;1:63–79. [DOI] [PubMed] [Google Scholar]
  • 26. Vidal SJ, Rodriguez-Bravo V, Galsky M, et al. Targeting cancer stem cells to suppress acquired chemotherapy resistance. Oncogene 2014;33:4451–63. [DOI] [PubMed] [Google Scholar]
  • 27. Misawa A, Katayama R, Koike S, et al. AP-1-Dependent miR-21 expression contributes to chemoresistance in cancer stem cell-like SP cells. Oncol Res 2010;19:23–33. [DOI] [PubMed] [Google Scholar]
  • 28. Okamoto N, Yasukawa M, Nguyen C, et al. Maintenance of tumor initiating cells of defined genetic composition by nucleostemin. Proc Natl Acad Sci USA 2011;108:20388–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Suzuki S, Okada M, Shibuya K, et al. JNK suppression of chemotherapeutic agents-induced ROS confers chemoresistance on pancreatic cancer stem cells. Oncotarget 2015;6:458–470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Chuthapisith S, Eremin J, El-Sheemey M, et al. Breast cancer chemoresistance: emerging importance of cancer stem cells. Surg Oncol 2010;19:27–32. [DOI] [PubMed] [Google Scholar]
  • 31. Larue L, Bellacosa A. Epithelial-mesenchymal transition in development and cancer: role of phosphatidylinositol 3′ kinase/AKT pathways. Oncogene 2005;24:7443–54. [DOI] [PubMed] [Google Scholar]
  • 32. Meng F, Speyer CL, Zhang B, et al. PDGFRα and β play critical roles in mediating Foxq1-driven breast cancer stemness and chemoresistance. Cancer Res 2015;75:584–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Li J, Liu H, Yu J, et al. Chemoresistance to doxorubicin induces epithelial-mesenchymal transition via upregulation of transforming growth factor β signaling in HCT116 colon cancer cells. Mol Med Rep 2015;12:192–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Hara J, Miyata H, Yamasaki M, et al. Mesenchymal phenotype after chemotherapy is associated with chemoresistance and poor clinical outcome in esophageal cancer. Oncol Rep 2014;31:589–96. [DOI] [PubMed] [Google Scholar]
  • 35. Hoshino H, Miyoshi N, Nagai K, et al. Epithelial-mesenchymal transition with expression of SNAI1-induced chemoresistance in colorectal cancer. Biochem Biophys Res Commun 2009;390:1061–5. [DOI] [PubMed] [Google Scholar]
  • 36. Kurrey NK, Jalgaonkar SP, Joglekar AV, et al. Snail and slug mediate radioresistance and chemoresistance by antagonizing p53-mediated apoptosis and acquiring a stem-like phenotype in ovarian cancer cells. Stem Cells 2009;27:2059–68. [DOI] [PubMed] [Google Scholar]
  • 37. Cheng ZX, Wang DW, Liu T, et al. Effects of the HIF-1α and NF-κB loop on epithelial-mesenchymal transition and chemoresistance induced by hypoxia in pancreatic cancer cells. Oncol Rep 2014;31:1891–8. [DOI] [PubMed] [Google Scholar]
  • 38. White EJ, Martin V, Liu JL, et al. Autophagy regulation in cancer development and therapy. Am J Cancer Res 2011;1:362–72. [PMC free article] [PubMed] [Google Scholar]
  • 39. Altman BJ, Rathmell JC. Metabolic stress in autophagy and cell death pathways. Cold Spring Harb Perspect Biol 2012;4:a008763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Zhuang W, Qin Z, Liang Z. The role of autophagy in sensitizing malignant glioma cells to radiation therapy. Acta Biochim Biophys Sin (Shanghai) 2009;41:341–51. [DOI] [PubMed] [Google Scholar]
  • 41. Sui X, Chen R, Wang Z, et al. Autophagy and chemotherapy resistance: a promising therapeutic target for cancer treatment. Cell Death Dis 2013;4:e838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Ge J, Chen Z, Huang J, et al. Upregulation of autophagy-related gene-5 (ATG-5) is associated with chemoresistance in human gastric cancer. PLoS One 2014;9:e110293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Wu S, Wang X, Chen J, et al. Autophagy of cancer stem cells is involved with chemoresistance of colon cancer cells. Biochem Biophys Res Commun 2013;434:898–903. [DOI] [PubMed] [Google Scholar]
  • 44. Dean M, Fojo T, Bates S. Tumour stem cells and drug resistance. Nat Rev Cancer 2005;5:275–84. [DOI] [PubMed] [Google Scholar]
  • 45. Chen K, Huang YH, Chen JL. Understanding and targeting cancer stem cells: therapeutic implications and challenges. Acta Pharmacol Sin 2013;34:732–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Yakirevich E, Sabo E, Naroditsky I, et al. Multidrug resistance-related phenotype and apoptosis-related protein expression in ovarian serous carcinomas. Gynecol Oncol 2006;100:152–9. [DOI] [PubMed] [Google Scholar]
  • 47. Kamazawa S, Kigawa J, Kanamori Y, et al. Multidrug resistance gene-1 is a useful predictor of Paclitaxel-based chemotherapy for patients with ovarian cancer. Gynecol Oncol 2002;86:171–6. [DOI] [PubMed] [Google Scholar]
  • 48. Duan Z, Brakora KA, Seiden MV. Inhibition of ABCB1 (MDR1) and ABCB4 (MDR3) expression by small interfering RNA and reversal of paclitaxel resistance in human ovarian cancer cells. Mol Cancer Ther 2004;3:833–8. [PubMed] [Google Scholar]
  • 49. Fojo AT, Ueda K, Slamon DJ, et al. Expression of a multidrug-resistance gene in human tumors and tissues. Proc Natl Acad Sci USA 1987;84:265–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Ricciardelli C, Ween MP, Lokman NA, et al. Chemotherapy-induced hyaluronan production: a novel chemoresistance mechanism in ovarian cancer. BMC Cancer 2013;13:476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Kuo MT. Roles of multidrug resistance genes in breast cancer chemoresistance. Adv Exp Med Biol 2007;608:23–30. [DOI] [PubMed] [Google Scholar]
  • 52. Patil Y, Sadhukha T, Ma L, et al. Nanoparticle-mediated simultaneous and targeted delivery of paclitaxel and tariquidar overcomes tumor drug resistance. J Control Release 2009;136:21–9. [DOI] [PubMed] [Google Scholar]
  • 53. Ritchie TK, Kwon H, Atkins WM. Conformational analysis of human ATP-binding cassette transporter ABCB1 in lipid nanodiscs and inhibition by the antibodies MRK16 and UIC2. J Biol Chem 2011;286:39489–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Sims-Mourtada J, Izzo JG, Ajani J, et al. Sonic Hedgehog promotes multiple drug resistance by regulation of drug transport. Oncogene 2007;26:5674–9. [DOI] [PubMed] [Google Scholar]
  • 55. Dubitzky W. Understanding the computational methodologies of systems biology. Brief Bioinform 2006;7:315–17. [DOI] [PubMed] [Google Scholar]
  • 56. Kriete A, Lechner M, Clearfield D, et al. Computational systems biology of aging. Wiley Interdiscip Rev Syst Biol Med 2011;3:414–28. [DOI] [PubMed] [Google Scholar]
  • 57. Schadt EE, Linderman MD, Sorenson J, et al. Computational solutions to large-scale data management and analysis. Nat Rev Genet 2010;11:647–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Gomez-Cabrero D, Abugessaisa I, Maier D, et al. Data integration in the era of omics: current and future challenges. BMC Syst Biol 2014;8(Suppl 2):I1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Wilkinson DJ. Bayesian methods in bioinformatics and computational systems biology. Brief Bioinform 2007;8:109–16. [DOI] [PubMed] [Google Scholar]
  • 60. Materi W, Wishart DS. Computational systems biology in cancer: modeling methods and applications. Gene Regul Syst Bio 2007;1:91–110. [PMC free article] [PubMed] [Google Scholar]
  • 61. Oh JH, Wong HP, Wang X, et al. A bioinformatics filtering strategy for identifying radiation response biomarker candidates. PLoS One 2012;7:e38870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Ozbayraktar FB, Ulgen KO. Drug target identification in sphingolipid metabolism by computational systems biology tools: metabolic control analysis and metabolic pathway analysis. J Biomed Inform 2010;43:537–49. [DOI] [PubMed] [Google Scholar]
  • 63. Lecca P. An integrative network inference approach to predict mechanisms of cancer chemoresistance. Integr Biol (Camb) 2013;5:458–73. [DOI] [PubMed] [Google Scholar]
  • 64. Lynn KS, Lu CH, Yang HY, et al. Construction of gene clusters resembling genetic causal mechanisms for common complex disease with an application to young-onset hypertension. BMC Genomics 2013;14:497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Royer L, Reimann M, Andreopoulos B, et al. Unraveling protein networks with power graph analysis. PLoS Comput Biol 2008;4:e1000108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Lv L, Deng H, Li Y, et al. The DNA methylation-regulated miR-193a-3p dictates the multi-chemoresistance of bladder cancer via repression of SRSF2/PLAU/HIC2 expression. Cell Death Dis 2014;5:e1402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Fu L, Liu N, Han Y, et al. ADAM10 regulates proliferation, invasion, and chemoresistance of bladder cancer cells. Tumour Biol 2014;35:9263–8. [DOI] [PubMed] [Google Scholar]
  • 68. Fan Y, Shen B, Tan M, et al. Long non-coding RNA UCA1 increases chemoresistance of bladder cancer cells by regulating Wnt signaling. FEBS J 2014;281:1750–8. [DOI] [PubMed] [Google Scholar]
  • 69. Bai Y, Sun Y, Peng J, et al. Overexpression of secretagogin inhibits cell apoptosis and induces chemoresistance in small cell lung cancer under the regulation of miR-494. Oncotarget 2014;5:7760–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Dong X, Lin D, Low C, et al. Elevated expression of BIRC6 protein in non-small-cell lung cancers is associated with cancer recurrence and chemoresistance. J Thorac Oncol 2013;8:161–70. [DOI] [PubMed] [Google Scholar]
  • 71. Xu C, Hao K, Hu H, et al. Expression of the enhancer of zeste homolog 2 in biopsy specimen predicts chemoresistance and survival in advanced non-small cell lung cancer receiving first-line platinum-based chemotherapy. Lung Cancer 2014;86:268–73. [DOI] [PubMed] [Google Scholar]
  • 72. Han L, Wang YF, Zhang Y, et al. Increased expression and function of P-glycoprotein in peripheral blood CD56+ cells is associated with the chemoresistance of non-small-cell lung cancer. Cancer Chemother Pharmacol 2012;70:365–72. [DOI] [PubMed] [Google Scholar]
  • 73. Sartorius UA, Krammer PH. Upregulation of Bcl-2 is involved in the mediation of chemotherapy resistance in human small cell lung cancer cell lines. Int J Cancer 2002;97:584–92. [DOI] [PubMed] [Google Scholar]
  • 74. Zhang L, Wang S, Wangtao, et al. Upregulation of GRP78 and GRP94 and its function in chemotherapy resistance to VP-16 in human lung cancer cell line SK-MES-1. Cancer Invest 2009;27:453–8. [DOI] [PubMed] [Google Scholar]
  • 75. Shih CM, Chen K, Wang YC, et al. Elevated p53 and p21waf1 mRNA expression in blood lymphocytes from lung cancer patients with chemoresistance. Cancer Detect Prev 2007;31:366–70. [DOI] [PubMed] [Google Scholar]
  • 76. Xiao F, Bai Y, Chen Z, et al. Downregulation of HOXA1 gene affects small cell lung cancer cell survival and chemoresistance under the regulation of miR-100. Eur J Cancer 2014;50:1541–54. [DOI] [PubMed] [Google Scholar]
  • 77. Cochaud S, Giustiniani J, Thomas C, et al. IL-17A is produced by breast cancer TILs and promotes chemoresistance and proliferation through ERK1/2. Sci Rep 2013;3:3456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Lian WJ, Liu G, Liu YJ, et al. Downregulation of BMP6 enhances cell proliferation and chemoresistance via activation of the ERK signaling pathway in breast cancer. Oncol Rep 2013;30:193–200. [DOI] [PubMed] [Google Scholar]
  • 79. Zhang W, Ding W, Chen Y, et al. Up-regulation of breast cancer resistance protein plays a role in HER2-mediated chemoresistance through PI3K/Akt and nuclear factor-kappa B signaling pathways in MCF7 breast cancer cells. Acta Biochim Biophys Sin (Shanghai) 2011;43:647–53. [DOI] [PubMed] [Google Scholar]
  • 80. Simstein R, Burow M, Parker A, et al. Apoptosis, chemoresistance, and breast cancer: insights from the MCF-7 cell model system. Exp Biol Med (Maywood) 2003;228:995–1003. [DOI] [PubMed] [Google Scholar]
  • 81. Xing H, Weng D, Chen G, et al. Activation of fibronectin/PI-3K/Akt2 leads to chemoresistance to docetaxel by regulating survivin protein expression in ovarian and breast cancer cells. Cancer Lett 2008;261:108–19. [DOI] [PubMed] [Google Scholar]
  • 82. Wang X, Zuo D, Chen Y, et al. Shed Syndecan-1 is involved in chemotherapy resistance via the EGFR pathway in colorectal cancer. Br J Cancer 2014;111:1965–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Xiao ZM, Wang XY, Wang AM. Periostin induces chemoresistance in colon cancer cells through activation of the PI3K/Akt/survivin pathway. Biotechnol Appl Biochem 2015;62:401–6. [DOI] [PubMed] [Google Scholar]
  • 84. Liu S, Wang J, Niu W, et al. The β6-integrin-ERK/MAP kinase pathway contributes to chemo resistance in colon cancer. Cancer Lett 2013;328:325–34. [DOI] [PubMed] [Google Scholar]
  • 85. Cheng JQ, Jiang X, Fraser M, et al. Role of X-linked inhibitor of apoptosis protein in chemoresistance in ovarian cancer: possible involvement of the phosphoinositide-3 kinase/Akt pathway. Drug Resist Updat 2002;5:131–46. [DOI] [PubMed] [Google Scholar]
  • 86. Yan BX, Ma JX, Zhang J, et al. PSP94 contributes to chemoresistance and its peptide derivative PCK3145 represses tumor growth in ovarian cancer. Oncogene 2014;33:5288–94. [DOI] [PubMed] [Google Scholar]
  • 87. Su HY, Lai HC, Lin YW, et al. Epigenetic silencing of SFRP5 is related to malignant phenotype and chemoresistance of ovarian cancer through Wnt signaling pathway. Int J Cancer 2010;127:555–67. [DOI] [PubMed] [Google Scholar]
  • 88. Tian X, Hao K, Qin C, et al. Insulin-like growth factor 1 receptor promotes the growth and chemoresistance of pancreatic cancer. Dig Dis Sci 2013;58:2705–12. [DOI] [PubMed] [Google Scholar]
  • 89. Zhao Y, Shen S, Guo J, et al. Mitogen-activated protein kinases and chemoresistance in pancreatic cancer cells. J Surg Res 2006;136:325–35. [DOI] [PubMed] [Google Scholar]
  • 90. Wey JS, Gray MJ, Fan F, et al. Overexpression of neuropilin-1 promotes constitutive MAPK signalling and chemoresistance in pancreatic cancer cells. Br J Cancer 2005;93:233–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Akada M, Crnogorac-Jurcevic T, Lattimore S, et al. Intrinsic chemoresistance to gemcitabine is associated with decreased expression of BNIP3 in pancreatic cancer. Clin Cancer Res 2005;11:3094–101. [DOI] [PubMed] [Google Scholar]
  • 92. Liu C, Li Z, Bi L, et al. NOTCH1 signaling promotes chemoresistance via regulating ABCC1 expression in prostate cancer stem cells. Mol Cell Biochem 2014;393:265–70. [DOI] [PubMed] [Google Scholar]
  • 93. Wu K, Xie D, Zou Y, et al. The mechanism of DAB2IP in chemoresistance of prostate cancer cells. Clin Cancer Res 2013;19:4740–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Zhang Y, Wang Y, Yuan J, et al. Toll-like receptor 4 ligation confers chemoresistance to docetaxel on PC-3 human prostate cancer cells. Cell Biol Toxicol 2012;28:269–77. [DOI] [PubMed] [Google Scholar]
  • 95. Zhu Y, Yu J, Wang S, et al. Overexpression of CD133 enhances chemoresistance to 5-fluorouracil by activating the PI3K/Akt/p70S6K pathway in gastric cancer cells. Oncol Rep 2014;32:2437–44. [DOI] [PubMed] [Google Scholar]
  • 96. Yoon C, Park dJ, Schmidt B, et al. CD44 expression denotes a subpopulation of gastric cancer cells in which Hedgehog signaling promotes chemotherapy resistance. Clin Cancer Res 2014;20:3974–88. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 97. Liu SQ, Yu JP, Yu HG, et al. Activation of Akt and ERK signalling pathways induced by etoposide confer chemoresistance in gastric cancer cells. Dig Liver Dis 2006;38:310–18. [DOI] [PubMed] [Google Scholar]
  • 98. Hua K, Feng W, Cao Q, et al. Estrogen and progestin regulate metastasis through the PI3K/AKT pathway in human ovarian cancer. Int J Oncol 2008;33:959–67. [PubMed] [Google Scholar]
  • 99. Zhao Y, Li Y, Ma H, et al. Modification of sialylation mediates the invasive properties and chemosensitivity of human hepatocellular carcinoma. Mol Cell Proteomics 2014;13:520–36. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 100. Osaki M, Oshimura M, Ito H. PI3K-Akt pathway: its functions and alterations in human cancer. Apoptosis 2004;9:667–76. [DOI] [PubMed] [Google Scholar]
  • 101. Martini M, De Santis MC, Braccini L, et al. PI3K/AKT signaling pathway and cancer: an updated review. Ann Med 2014;46:372–83. [DOI] [PubMed] [Google Scholar]
  • 102. Courtney KD, Corcoran RB, Engelman JA. The PI3K pathway as drug target in human cancer. J Clin Oncol 2010;28:1075–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Fresno Vara JA, Casado E, de Castro J, et al. PI3K/Akt signalling pathway and cancer. Cancer Treat Rev 2004;30:193–204. [DOI] [PubMed] [Google Scholar]
  • 104. Hennessy BT, Smith DL, Ram PT, et al. Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov 2005;4:988–1004. [DOI] [PubMed] [Google Scholar]
  • 105. Kandioler-Eckersberger D, Ludwig C, Rudas M, et al. TP53 mutation and p53 overexpression for prediction of response to neoadjuvant treatment in breast cancer patients. Clin Cancer Res 2000;6:50–6. [PubMed] [Google Scholar]
  • 106. Fiorini C, Cordani M, Padroni C, et al. Mutant p53 stimulates chemoresistance of pancreatic adenocarcinoma cells to gemcitabine. Biochim Biophys Acta 2015;1853:89–100. [DOI] [PubMed] [Google Scholar]
  • 107. Bush JA, Li G. Cancer chemoresistance: the relationship between p53 and multidrug transporters. Int J Cancer 2002;98:323–30. [DOI] [PubMed] [Google Scholar]
  • 108. Knappskog S, Lønning PE. P53 and its molecular basis to chemoresistance in breast cancer. Expert Opin Ther Targets 2012;16(Suppl 1):S23–30. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Data

Articles from Briefings in Bioinformatics are provided here courtesy of Oxford University Press

RESOURCES