Abstract
Purpose
Genotoxic agents (GAs) including cisplatin, doxorubicin, gemcitabine, and topotecan are often used in cancer treat-ment. However, the response to GAs is variable among patients and predictive biomarkers are inadequate to select patients for treatment. Accurate and rapid pharmacodynamics measures of response can, thus, be useful for monitoring therapy and improve clinical outcomes.
Methods
This study focuses on integrating a database of genome-wide response to treatment (The NCI Transcriptional Pharmacodynamics Workbench) with a database of baseline gene expression (GSE32474) for the NCI-60 cell lines to identify mechanisms of response and pharmacodynamic (PD) biomarkers.
Results and conclusions
Our analysis suggests that GA-induced endoplasmic reticulum (ER) stress may signal for GA-induced cell death. Reducing the uptake of GA, activating DNA repair, and blocking ER-stress induction cooperate to prevent GA-induced cell death in the GA-resistant cells. ATF3, DDIT3, CARS, and PPP1R15A appear as possible candidate PD biomarkers for monitoring the progress of GA treatment. Further validation studies on the proposed intrinsic drug-resistant mechanism and candidate genes are needed using in vivo data from either patient-derived xenograft models or clinical chemotherapy trials.
Keywords: Genotoxic agents, Pharmacodynamic biomarkers, Response mechanisms, ER stress
Introduction
Genotoxic agents (GAs) are key members of the chemotherapy repertoire, because cancer cells are often highly proliferative. GAs target DNA by various mechanisms and different targets. For example, cisplatin forms intrastrand crosslinks in DNA, doxorubicin, and topotecan inhibit topoisomerases II and I, respectively, and gemcitabine blocks nucleotide metabolism: all result eventually in failed DNA replication, disturbed DNA integrity, cell cycle inhibition, and induced cell death [1, 2]. However, the response to GAs is inconsistent and varies between tumor types and even within the same tumor type [3, 4]. Insensitivity of some cancer cells towards GAs, thus, remains a major obstacle in cancer treatment. Such insensitivity or resistance can be due to various mechanisms including diminished agent accumulation (efflux) and activation, elevated DNA repair or DNA damage tolerance, enhanced activation of anti-apoptotic proteins, and blocked apoptosis pathways [3, 5, 6]. Resistance mechanisms developed by genetic or epigenetic alterations protect cancer cells from GAs cytotoxic action [4]. For example, mutation and expression of DNA repair and apoptosis genes, such as BRCA1, BRCA2, and TP53, are considered key in predicting a GA response [4, 5]. However, the status of such genes/proteins only modestly correlates with response [7], indicating that the response to GAs is complex and that response after treatment requires careful monitoring.
Early tumor assessment after treatment may enable the adjustment of dose and schedule of the drug, and improve clinical outcomes and ameliorate side effects [8–10]. Conventional monitoring methods to judge response, which are often based on changes in tumor size, can be subjective and time consuming [11]. In addition, early changes in tumor size are often not correlated with biologic changes in cancer cells, causing misleading predictions of drug response [11]. Pharmacodynamic (PD) biomarkers may prove valuable for rapid and accurate evaluation of tumor response in the clinic [9, 10]. Gene expression-based biomarkers have the potential advantage of providing quantitative results with a wide dynamic range [12]. In addition, changes in gene expression after treatment provide information about cellular mechanisms of drug action [12].
The study presented here focuses on identification of genes that may be potential PD biomarkers of GA response or elucidate response mechanisms, by analyzing gene expression data from the NCI Transcriptional Pharmacodynamics Workbench (NCI TPW) (https://tpwb.nci.nih.gov/[13]). The NCI TPW is a publicly available database of drug-induced transcriptional profiles measured in the NCI-60 cell line panel and represents an extensive compilation of directly measured longitudinal transcriptional responses to 15 anticancer agents. The cells were exposed to these agents at a “low” and “high” concentration, approximating the clinical Cmax [14] and the average concentration resulting in 50% inhibition of the cell lines. Human tumor cell lines are traditionally valuable tools to identify genetic variants impacting drug response. Gene expression changes following treatment with four GAs (cisplatin, doxorubicin, gemcitabine, and topotecan) were analyzed. The main hypotheses is that since all four GAs interrupt DNA replication and induce DNA damage, there may be key PD biomarkers and centralized mechanisms of action of GAs. For the PD biomarker study, data available in the NCI TPW were used to collect the lists of genes, whose change in expression after treatment correlated with drug response based on growth inhibition of 50% (log10 GI50, referred as GI50 from this point forward) for each GA. The response and resistance mechanisms of GAs were investigated using differential gene expression profiles between ‘sensitive’ and ‘resistant’ cell lines before and after treatment.
Methods
The 100 genes with the largest absolute correlation between 24-h post-treatment gene expression, and GI50, for the NCI-60 cell lines treated with a lower concentration of each GA were obtained from the NCI TPW [13]. The data were collected from the NCI TPW database (https://tpwb.nci.nih.gov/) using the “Correlation Analysis” option for each of the GA treatments. The cell lines most sensitive and resistant to each GA were classified based on GI50 values provided in the TPW. Baseline gene expression data for the NCI-60 cell lines were obtained from the Gene Expression Omnibus (GSE32474) [15] and were analyzed to find genes differentially expressed between sensitive and resistant cell lines using GEO2R analysis tool (www.ncbi.nlm.nih.gov/geo/geo2r/). Genes which were differentially expressed for multiple GAs were identified using a Venn diagram (Venny 2.1).
For the Receiver Operating Characteristic (ROC) analysis, cell lines were clustered into three groups by dividing the difference between the highest and lowest GI50 values by three for each GA forming high (resistant), intermediate, and low (sensitive) GI50 groups (Fig. 3). The outliers of high GI50 values in the doxorubicin and topotecan treatments were not considered for determining the threshold for dividing the above three groups. The ROC curve was constructed by classifying the resistant and sensitive groups of cell lines based on the changes in RNA expression of the candidate genes after treatment from the TPW. The area under the curve (AUC) values in the ROC curve were calculated using the R with “rms” and “ModelGood” packages.
Fig. 3.
NCI-60 lines GI50 response to cisplatin (a), doxorubicin (b), gemcitabine (c), and topotecan (d). The lines were clustered into three groups (high, intermediate, and low GI50 groups) by dividing the difference between the highest and lowest log10 GI50 value by 3 for each drug. The outliers of high GI50 concentrations in B and D were not considered for this calculation. High log10 GI50 lines (⇑) were represented as resistant cells and low log10 GI50 lines (⇓) were represented as sensitive cells. B breast, C colon, CNS central nervous system, L leukemia, M melanoma, NSL non-small cell lung, O ovarian, P prostate, R renal
Results
Identification of response mechanisms and PD biomarkers of GAs
To investigate cellular response mechanisms after exposure to GAs, 100 genes with the largest absolute correlation between change in gene expression after treatment and sensitivity (measured by GI50) in the NCI-60 cell lines [13] were selected. Gene lists were generated separately for treatment with concentrations of cisplatin (3 μM), doxorubicin (0.1 μM), gemcitabine (0.2 μM), and topotecan (0.01 μM), respectively, for 24 h. To identify centralized cellular mechanisms after exposure to a GA, genes common across multiple agents were included in our study. No genes were identified in common for all 4 drugs. However, 26 negatively correlated genes and 4 positively correlated genes were identified in common for at least two GA drugs, which are shown in the Venn diagram (Fig. 1). Negative correlation is an association between those genes generally up-regulated after GA treatment in cell lines with low GI50 (referred to as ‘sensitive cells’), but down-regulated or unchanged in cell lines with high concentration GI50 (referred to as ‘resistant cells’). Annotation of the 26 up-regulated genes (negative correlation) found that most of them were endoplasmic reticulum (ER) stress-related genes including ATF3, CARS, CEBPG, DDIT3, GADD45A, TXNIP, PPP1R15A, CHAC1, ASNS, CTH, TRIB3, CDKN1A, RNF19B, and WDR45 (Fig. 1a) [16–19]. These results indicate that GAs may induce ER stress contributing to cell arrest or death in sensitive cells. In contrast, GA-induced ER stress may be blocked in resistant cells. Based on published information, the potential relationships between ER-stress genes are summarized in Fig. 2, where the genes highlighted in bold were up-regulated in the sensitive cell line cohort as identified above and in Fig. 1a.
Fig. 1.
Distribution of the 100 genes with the largest absolute correlation between changes in gene expression and GI50 of cisplatin, doxorubicin, gemcitabine, and topotecan, respectively, in the NCI-60 lines treated with a low concentration for 24 h. The negative correlation means that those genes were generally up-regulated after treatment in the low GI50 concentration lines, but down-regulated or not changed in the high GI50 concentration lines. The positive correlation means that those genes were down-regulated in the resistant cell lines after treatment, and up-regulated in the sensitive cell lines. Cisplatin: 3000 nM, doxorubicin: 100 nM, gemcitabine: 200 nM, topotecan: 10 nM. ER-stress-related genes are represented with red color
Fig. 2.
Summary of the functional relationships between the ER-stress-related genes. Genes in bold are the negatively correlated genes in Fig. 1. ERAD ER-associated degradation, ROS reactive oxygen species
The potential of the genes identified above to serve as PD biomarkers was analyzed in cell lines for early clinical assessment after GA treatment. The cell lines in the resistant cell line group (high concentration GI50) and the sensitive cell line group (low concentration GI50) were used for the ROC analysis to identify PD biomarkers. Since the change in RNA expression of each gene in the cell lines varied depending upon GA treatment, the analysis was performed separately for each GA and the mean AUC of each gene was calculated (Table 1). The AUC values for topotecan were, generally, lower, because the range of GI50 values was relatively narrow. Based on the mean AUC values threshold above 80, ATF3, DDIT3, CARS, and PPP1R15A emerged as potential PD biomarker candidates. When the potential PD biomarkers were evaluated as a group, the AUC values increased, indicating that the 4-gene group was more reliable than the single genes as a monitor of response (Table 2).
Table 1.
Area under the curve (AUC) values (%) of ER-stress-related genes in the receiver operator characteristic curve (ROC) constructed in logistic regression for evaluating them as a pharmacodynamic biomarker of genotoxic agents
Cisplatin | Doxorubicin | Gemcitabine | Topotecan | Mean AUC | |
---|---|---|---|---|---|
ATF3 | 82 | 91 | 94 | 78 | 86 |
CARS | 97 | 67 | 91 | 69 | 81 |
CEBPG | 90 | 45 | 91 | 74 | 75 |
DDIT3 | 80 | 95 | 96 | 80 | 88 |
GADD45A | 79 | 79 | 90 | 68 | 79 |
TXNIP | 73 | 80 | 89 | 67 | 77 |
PPP1R15A | 80 | 89 | 89 | 63 | 81 |
CHAC1 | 91 | 74 | 77 | 68 | 78 |
ASNS | 88 | 47 | 82 | 79 | 74 |
CTH | 79 | 58 | 83 | 68 | 72 |
TRIB3 | 74 | 52 | 76 | 70 | 68 |
CDKN1A | 69 | 83 | 67 | 73 | 73 |
RNF19B | 69 | 83 | 76 | 71 | 75 |
WDR45 | 75 | 86 | 60 | 78 | 75 |
Mean AUC values greater than 80 are highlighted in bold
Table 2.
Area under the curve (AUC) values (%) of the combined ER-stress-related genes in the receiver operator characteristic curve (ROC) constructed in logistic regression for evaluating them as pharmacodynamic biomarkers in combination of genotoxic agents
Cisplatin | Doxorubicin | Gemcitabine | Topotecan | Mean AUC | |
---|---|---|---|---|---|
A+B | 82 | 95 | 95 | 83 | 89 |
A+C | 97 | 91 | 95 | 80 | 91 |
A+D | 84 | 98 | 94 | 80 | 89 |
B+C | 97 | 98 | 96 | 80 | 93 |
B+D | 82 | 98 | 95 | 79 | 89 |
C+D | 97 | 88 | 94 | 70 | 87 |
A+B+C | 96 | 98 | 95 | 83 | 93 |
A+B+D | 82 | 98 | 96 | 83 | 90 |
B+C+D | 97 | 99 | 96 | 79 | 93 |
A+C+D | 97 | 97 | 96 | 81 | 93 |
A+B+C+D | 96 | 99 | 97 | 83 | 94 |
A ATF3, B DDIT3, C CARS, D PPP1R15A
Identification of resistance mechanisms of Gas
The causes of drug resistance can be classified as preexisting (intrinsic resistance) or drug-induced (acquired resistance) [20]. The main genetic causes of intrinsic resistance include mutation, deletion, and overexpression of drug response-related genes. Understanding intrinsic resistance mechanisms is important for selecting effective drugs before treatment. To identify intrinsic GA-resistance mechanisms using expression data, the “common” sensitive and resistant cell lines across all four GAs have been redefined. Starting from the sensitive and resistant cell lines for each single GA in Fig. 3, “common” resistant and sensitive cell lines in at least two GAs were identified using a Venn diagram (Fig. 4, Supplementary Table 1). Then, analysis for differentially expressed genes in the “common” resistant lines compared to the “common” sensitive lines (Fig. 4, Supplementary Table 1) was performed using baseline gene expression data obtained using Affymetrix human genome U133 plus 2.0 arrays with the robust multichip analysis (GSE32474) [15]. The top 250 significant probe sets were identified using the GEO2R analysis tool (www.ncbi.nlm.nih.gov/geo/geo2r/). Among the above, there are 46 and 118 protein-encoding genes in the sensitive lines that were expressed higher and lower, respectively, than in the resistant lines (Supplementary Table 2). Gene functional analysis was carried out using DAVID Bioinformatics Resources (https://david.ncifcrf.gov/) with those genes. Among the 118 genes highly expressed in the resistant lines, 21 genes were involved in cell:cell junctions including tight junctions and adherens junctions (Table 3, Supplementary Fig. 1) [21, 22], indicating that cell junction proteins may protect the tumor from GAs. Moreover, ER-stress-, DNA repair-, and PI3K/AKT-related genes were also identified as differentially expressed, and those more highly expressed in the resistant cell lines (Table 3) were investigated for a potential role in GA-resistance mechanisms. Specifically, TNK1, CBLC, and SFN have been reported to activate DNA repair [23–25]. ST14, LCN2, CDH1, PROM1, and EPCAM have been reported to activate PI3K/AKT [26–30] and MST1R (RON) has been reported to activate both DNA repair and PI3K/AKT pathway [31, 32]. ATP2C2 and AGR2 were reported to inhibit ER-stress induction [33, 34], and SPINT1 and 2 were reported to block CHOP expression [35]. Finally, MACC1 and UCA1 were reported to inhibit cell death [36, 37]. These reports suggest that cancer cells harboring active DNA repair and inactive ER-stress induction systems may be protected from GA-induced cell death.
Fig. 4.
Cancer cell lines sensitive or resistant to cisplatin, doxorubicin, gemcitabine, and topotecan, respectively, based on Fig. 3
Table 3.
Cell junction-related genes and ER-stress/DNA damage/CHOP inhibiting genes expressed more highly in the resistant cells than in the sensitive cells
Gene | Probe set ID | Functional pathways in which the gene is involved | Fold change |
---|---|---|---|
EPCAM | 201839_s_at | Cell:cell junctions, PI3K/AKT | 17.76 |
CDH1 | 201131_s_at | Cell:cell junctions, PI3K/AKT | 6.79 |
SFN | 33323_r_at | Cell:cell junctions, DNA repair | 5.71 |
CLDN3 | 203953_s_at | Cell:cell junctions | 4.77 |
KRT18 | 201596_x_at | Cell:cell junctions | 4.36 |
CLDN4 | 201428_at | Cell:cell junctions | 4.35 |
CLDN7 | 202790_at | Cell:cell junctions | 3.03 |
TMC5 | 240304_s_at | Cell:cell junctions | 2.78 |
TMC4 | 226403_at | Cell:cell junctions | 2.57 |
LAD1 | 203287_at | Cell:cell junctions | 2.41 |
PLS1 | 205190_at | Cell:cell junctions | 2.4 |
EPB41L4B | 223427_s_at | Cell:cell junctions | 2.3 |
MARVELD3 | 239148_at | Cell:cell junctions | 2.01 |
DLG3 | 212729_at | Cell:cell junctions | 1.76 |
EPS8L1 | 91826_at | Cell:cell junctions | 1.69 |
MISP | 212925_at | Cell:cell junctions | 1.65 |
TJP3 | 213412_at | Cell:cell junctions | 1.58 |
F11R | 224097_s_at | Cell:cell junctions | 1.55 |
EVPL | 204503_at | Cell:cell junctions | 1.52 |
CRB3 | 232609_at | Cell:cell junctions | 1.39 |
SH3D19 | 1558647_at | Cell:cell junctions | 1.38 |
AGR2 | 209173_at | Inhibit ER stress | 8.86 |
ATP2C2 | 214798_at | Inhibit ER stress | 1.35 |
SPINT2 | 210715_s_at | Block CHOP expression | 6 |
SPINT1 | 202826_at | Block CHOP expression | 4.27 |
LCN2 | 212531_at | PI3K/AKT | 4.06 |
PR0M1 | 204304_s_at | PI3K/AKT | 3.21 |
UCA1 | 227919_at | Inhibit cell death | 2.79 |
MACC1 | 1566764_at | Inhibit cell death | 2.76 |
ST14 | 202005_at | PI3K/AKT | 2.63 |
MST1R | 205455_at | PI3K/AKT, DNA repair | 1.93 |
CBLC | 220638_s_at | DNA repair | 1.8 |
TNK1 | 217149_x_at | DNA repair | 1.22 |
Central nervous system (CNS) cancer cell lines were found to be exclusively included in the sensitive group, while colon cancer cell lines were only in the resistant group (Supplementary Table 1). Neurons are known to be sensitive to ER stress [16]. RNA expression of ER-stress genes between CNS and colon cancer clinical samples were compared using baseline gene expression data available in the cancer genome atlas (TCGA) to begin to validate the findings in tissue samples. All cell junction-, ER-stress-, and DNA repair-related genes, which were more highly expressed in the resistant cell lines, were also more highly expressed in colon cancer samples (Supplementary Figs. 3, 4), suggesting that GA-mediated severe ER stress might be critical to induce GA-mediated cell death in cancer.
Discussion
Understanding resistance mechanisms of cancer cells to GAs continues to be an important focus. Abnormal p53 function and enhanced DNA repair have been broadly accepted as mechanisms of resistance to GAs [7, 38]. Activation of the unfolded protein response (UPR) following ER stress has also been identified as a mechanism of resistance (reviewed in [39]). However, response to GAs varies not only between tumor types, but also between individual tumors of the same type regardless of p53 status and DNA repair systems (Supplementary Table 1) [40–42]. The study presented here sought to identify other broad GA-resistance mechanisms along with potential PD biomarkers for assessing treatment response.
GA response data for the NCI-60 lines indicated that many pro-apoptotic ER-stress-related genes such as ATF3, DDIT3 (CHOP), GADD45A, TXNIP, PPP1R15A (GADD34), CHAC1, and TRIB3 were up-regulated in sensitive compared to resistant cell lines. Severe ER stress induces pro-apoptotic proteins, such as CHOP (DDIT3), and later activates autophagy-induced cell death [43]. Although GA–DNA interactions are considered key leading to cell death, other DNA damage-independent mechanisms are important, and our data suggest that ER stress could be one. ER stress potentially leads to misfolded proteins and is regulated by the Unfolded Protein Response (UPR) which is an adaptive response that can increase cell survival [44, 45]. However, in the case of irreversible ER stress, UPR switches to induction of apoptosis in damaged cells [46, 47]. Therapy-induced ER stress contributes to changes in drug sensitivity, although it is not completely clear what triggers the switch from an adaptive response to apoptosis [39, 48]. A UPR-induced protein, CHOP (DDIT3) initiates apoptosis through regulation of BH3 proteins [49] and also through augmented ROS production, ATP depletion, and reactivated protein synthesis [18]. Sensitive cell lines activated the UPR response to GAs more robustly than the GA-resistant cell lines suggest that the UPR response might be leveraged to increase drug sensitivity in non-responsive tumors. Thus, identifying whether the genotoxic response leads to apoptosis through modulation of the BH3 protein response, change in ROS production, or ATP depletion would provide critical information. However, the UPR activation in sensitive cells may be an attempt by the cell to protect against inevitable cell death. The lack of ER-stress response via UPR in resistant cells may reflect innate resistance that prevents induction of ER stress. To address this, the endogenous gene expression profiles of GA-resistant cells were used to identify genes differentially expressed at baseline compared to sensitive cells. Our analysis showed that many cell junction protein-encoding genes were more highly expressed in the resistant cells (Table 3). Since cell junction proteins regulate drug delivery, they are important for drug efficacy and response [21] and poor drug delivery may be an important factor. Moreover, UPR induced the ER chaperone BiP in an epithelial colon cancer cell, but a cell death protein CHOP in a mesenchymal cell [50]. These reports support our results identifying a common theme for activity and resistance to GA’s involving ER-stress induction and cell junction protein-encoding gene expression. When these potential resistance genes identified in the NCI-60 cell lines were evaluated across a cohort of tumor tissue representing the more resistant cell lines (colon) compared to a more sensitive tumor tissue cohort (CNS), all of the genes were found to be more highly expressed in the resistant cohort, lending further support to the hypothesis.
The previous studies show some cell-surface proteins encoded by the genes identified may support chemo-resistance. EPCAM (epithelial cell adhesion molecule), the gene most differentially expressed (Table 3), was shown to contribute to chemo-resistance in multiple cell types, including prostate, breast, and colon cancers [27, 51, 52]. In addition, several of the cell-surface genes identified have been associated with epithelial-to-mesenchymal transition (EMT), and recent data have identified an intriguing role for EMT in chemo-resistance in cancer [53, 54]. EMT has been linked to increased ER-stress response which enhances tumor cell survival, indicating a possible protective mechanism [55]. Claudin proteins including claudin-3, −4, and −7 (CLDN3, 4, and 7) were reported to be overexpressed in many cancers, and by leading to EMT, were associated with cisplatin resistance in ovarian cancer [56]. The cadherin encoded by CDH4 has also been associated with EMT and resistance to cisplatin [57]. Cytokeratin 18 (KRT18) is an epithelial cell-specific intermediate filament protein that is lost during EMT and its loss is reported to decrease cisplatin sensitivity [58]. However, recent data show KRT18 critically contributes to initiating TGF-β1-induced EMT [59]. Thus, our data support a role for highly expressed genes involved in EMT, and also expression of cell surface and cell:cell junction genes, to potentially contribute to inherent cell resistance.
In addition to cell junction protein-encoding genes, genes blocking DNA damage, ER stress, and apoptosis are also more highly expressed in the resistant cells (Table 3). DNA repair processes in cancer cells are, generally, considered a response to genotoxic damage [7, 38]. DNA repair genes, including TNK1, CBLC, SFN, and MST1R, were more highly expressed in the resistant cells, and all have been reported as participating in resistance. The PARP enzyme TNK1 and an E3 ubiquitin ligase CBLC were reported to be involved in non-homologous end joining (NHEJ) DNA repair and in homologous recombination (HR) DNA repair, respectively [23, 24]. An epithelial cell-surface protein MST1R (RON) was reported to be involved in NHEJ DNA repair [32] and to be related to cisplatin resistance of ovarian cancer cells [60], gemcitabine resistance in pancreatic cancer cells [61], and was considered poor prognostic marker in gastric, bladder, colorectal, breast, and non-small-cell lung cancers [31]. SFN (14–3-3σ) was reported to interact with DNA repair protein human exonuclease 1 (hEXO1) to regulate DNA repair, resulting in resistance to DNA-damaging drugs [25], and was a prognosis marker for poor survival of colorectal cancer patients [62].
GA-induced ER stress may be controlled in the resistant cells; ATP2C2 (SPAC2) was reported to maintain low cytosolic and high luminal free Ca2+ concentration [33] and AGR2 was found to be involved in the UPR to minimize ER stress and cell death [34]. Finally, cell death may be blocked by MACC1 and UCA1. It was reported that MACC1 decreased cisplatin sensitivity in epithelial ovarian cancer cells [36] and UCA1 (a long non-coding RNA) increased cisplatin and gemcitabine resistance of bladder cancer cells [37, 63].
In summary, our analysis suggests that ER-stress induction is a key factor of GA-induced cell death, although the precise mechanisms of GA induction of ER stress remain under investigation and likely vary between drugs. Our results indicate that resistant cells are protected from the ER-stress-induced apoptosis, by inherent resistance mechanisms. One hypothesis is that decreased uptake of GAs, efficient DNA repair, and blocking ER-stress induction may cooperate to reduce GA-induced DNA damage in the GA-resistant lines (Fig. 5). The cooperation between DNA damage and ER stress is supported by the previous studies reporting that DNA damage-induced apoptosis was facilitated by ER protein MG23 [64], DNA repair was inhibited by ER stress [65], and ER chaperone protein BiP was up-regulated by impaired excision repair of DNA [66]. Finally, the results of our analysis suggest that ATF3, DDIT3, CARS, and PPP1R15A are potential candidate PD biomarkers for monitoring response to GA treatment.
Fig. 5.
Suggested resistance mechanisms in the genotoxic agent (GA)-resistant cancer cells. The genes are more highly expressed in the GA-resistant cells than in the GA-sensitive cells. CJP cell junction protein (see Table 3). For additional details and supporting literature reference, please refer to the main text
Supplementary Material
Acknowledgements
The authors would like to thank Dr. Julia Krushkal for reading the manuscript and for providing helpful feedback.
Funding
This project has been funded in whole or in part with federal funds from the National Cancer Institute, National Institutes of Health, under Contract No. HHSN261200800001E. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government.
Footnotes
Compliance with ethical standards
Conflict of interest The authors declare that they have no competing interests
Ethical approval This article does not contain any studies with human participants or animals performed by any of the authors.
Electronic supplementary material The online version of this article (https://doi.org/10.1007/s00280-019-03898-z) contains supplementary material, which is available to authorized users.
References
- 1.Moysan E, Bastiat G, Benoit JP (2013) Gemcitabine versus Modified Gemcitabine: a review of several promising chemical modifications. Mol Pharm 10(2):430–444. 10.1021/mp300370t [DOI] [PubMed] [Google Scholar]
- 2.Swift LH, Golsteyn RM (2014) Genotoxic anti-cancer agents and their relationship to DNA damage, mitosis, and checkpoint adaptation in proliferating cancer cells. Int J Mol Sci 15(3):3403–3431. 10.3390/ijms15033403 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Saunders NA, Simpson F, Thompson EW, Hill MM, Endo-Munoz L, Leggatt G, Minchin RF, Guminski A (2012) Role of intratumoural heterogeneity in cancer drug resistance: molecular and clinical perspectives. EMBO Mol Med 4(8):675–684. 10.1002/emmm.201101131 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Turner NC, Reis-Filho JS (2012) Genetic heterogeneity and cancer drug resistance. Lancet Oncol 13(4):e178–e185. 10.1016/S1470-2045(11)70335-7 [DOI] [PubMed] [Google Scholar]
- 5.Eckstein N (2011) Platinum resistance in breast and ovarian cancer cell lines. J Exp Clin Cancer Res 30:91. 10.1186/1756-9966-30-91 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Cheung-Ong K, Giaever G, Nislow C (2013) DNA-damaging agents in cancer chemotherapy: serendipity and chemical biology. Chem Biol 20(5):648–659. 10.1016/j.chembiol.2013.04.007 [DOI] [PubMed] [Google Scholar]
- 7.Bouwman P, Jonkers J (2012) The effects of deregulated DNA damage signalling on cancer chemotherapy response and resistance. Nat Rev Cancer 12(9):587–598. 10.1038/nrc3342 [DOI] [PubMed] [Google Scholar]
- 8.Wilson MK, Collyar D, Chingos DT, Friedlander M, Ho TW, Karakasis K, Kaye S, Parmar MK, Sydes MR, Tannock IF, Oza AM (2015) Outcomes and endpoints in cancer trials: bridging the divide. Lancet Oncol 16(1):e43–e52. 10.1016/S1470-2045(14)70380-8 [DOI] [PubMed] [Google Scholar]
- 9.Mould DR, Walz AC, Lave T, Gibbs JP, Frame B (2015) Developing exposure/response models for anticancer drug treatment: special considerations. CPT Pharmacometrics Syst Pharmacol 4(1):e00016. 10.1002/psp4.16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Verweij J, de Jonge M, Eskens F, Sleijfer S (2012) Moving molecular targeted drug therapy towards personalized medicine: issues related to clinical trial design. Mol Oncol 6(2):196–203. 10.1016/j.molonc.2012.01.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Yaghmai V, Miller FH, Rezai P, Benson AB 3rd, Salem R (2011) Response to treatment series: part 2, tumor response assessment—using new and conventional criteria. AJR Am J Roentgenol 197(1):18–27. 10.2214/AJR.11.6581 [DOI] [PubMed] [Google Scholar]
- 12.Mizuarai S, Irie H, Kotani H (2010) Gene expression-based pharmacodynamic biomarkers: the beginning of a new era in biomarker-driven anti-tumor drug development. Curr Mol Med 10(6):596–607 [DOI] [PubMed] [Google Scholar]
- 13.Monks A, Zhao Y, Hose C, Hamed H, Krushkal J, Fang J, Sonkin D, Palmisano A, Polley EC, Fogli LK, Konate MM, Miller SB,Simpson MA, Voth AR, Li MC, Harris E, Wu X, Connelly JW, Rapisarda A, Teicher BA, Simon R, Doroshow JH (2018) The NCI transcriptional pharmacodynamics workbench: a tool to examine dynamic expression profiling of therapeutic response in the NCI-60 cell line panel. Cancer Res 10.1158/0008-5472.can-18-0989 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Liston DR, Davis M (2017) Clinically relevant concentrations of anticancer drugs: a guide for nonclinical studies. Clin Cancer Res 23(14):3489–3498. 10.1158/1078-0432.CCR-16-3083 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Pfister TD, Reinhold WC, Agama K, Gupta S, Khin SA, Kinders RJ, Parchment RE, Tomaszewski JE, Doroshow JH, Pommier Y (2009) Topoisomerase I levels in the NCI-60 cancer cell line panel determined by validated ELISA and microarray analysis and correlation with indenoisoquinoline sensitivity. Mol Cancer Ther 8(7):1878–1884. 10.1158/1535-7163.MCT-09-0016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Choy MS, Chen MJ, Manikandan J, Peng ZF, Jenner AM, Melendez AJ, Cheung NS (2011) Up-regulation of endoplasmic reticulum stress-related genes during the early phase of treatment of cultured cortical neurons by the proteasomal inhibitor lactacystin. J Cell Physiol 226(2):494–510. 10.1002/jcp.22359 [DOI] [PubMed] [Google Scholar]
- 17.Dixon SJ, Patel DN, Welsch M, Skouta R, Lee ED, Hayano M, Thomas AG, Gleason CE, Tatonetti NP, Slusher BS, Stockwell BR (2014) Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis. Elife 3:e02523. 10.7554/eLife.02523 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Han J, Back SH, Hur J, Lin YH, Gildersleeve R, Shan J, Yuan CL, Krokowski D, Wang S, Hatzoglou M, Kilberg MS, Sartor MA, Kaufman RJ (2013) ER-stress-induced transcriptional regulation increases protein synthesis leading to cell death. Nat Cell Biol 15(5):481–490. 10.1038/ncb2738 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Huggins CJ, Mayekar MK, Martin N, Saylor KL, Gonit M, Jailwala P, Kasoji M, Haines DC, Quinones OA, Johnson PF (2015) C/EBPgamma is a critical regulator of cellular stress response networks through heterodimerization with ATF4. Mol Cell Biol 36(5):693–713. 10.1128/mcb.00911-15 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lippert TH, Ruoff HJ, Volm M (2011) Current status of methods to assess cancer drug resistance. Int J Med Sci 8(3):245–253 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Deli MA (2009) Potential use of tight junction modulators to reversibly open membranous barriers and improve drug delivery. Biochim Biophys Acta 1788(4):892–910. 10.1016/j.bbamem.2008.09.016 [DOI] [PubMed] [Google Scholar]
- 22.Kohn KW, Zeeberg BM, Reinhold WC, Pommier Y (2014) Gene expression correlations in human cancer cell lines define molecular interaction networks for epithelial phenotype. PLoS One 9(6):e99269. 10.1371/journal.pone.0099269 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bai P (2015) Biology of poly(ADP-Ribose) polymerases: the factotums of cell maintenance. Mol Cell 58(6):947–958. 10.1016/j.molcel.2015.01.034 [DOI] [PubMed] [Google Scholar]
- 24.Frankum J, Moudry P, Brough R, Hodny Z, Ashworth A, Bartek J, Lord CJ (2015) Complementary genetic screens identify the E3 ubiquitin ligase CBLC, as a modifier of PARP inhibitor sensitivity. Oncotarget 6(13):10746–10758. 10.18632/oncotarget.3628 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Andersen SD, Keijzers G, Rampakakis E, Engels K, Luhn P, El-Shemerly M, Nielsen FC, Du Y, May A, Bohr VA, Ferrari S, Zannis-Hadjopoulos M, Fu H, Rasmussen LJ (2012) 14–3-3 checkpoint regulatory proteins interact specifically with DNA repair protein human exonuclease 1 (hEXO1) via a semi-conserved motif. DNA Repair (Amst) 11(3):267–277. 10.1016/j.dnarep.2011.11.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bergin E, Levine JS, Koh JS, Lieberthal W (2000) Mouse proximal tubular cell-cell adhesion inhibits apoptosis by a cadherin-dependent mechanism. Am J Physiol Renal Physiol 278(5):F758–F768. 10.1152/ajprenal.2000.278.5.F758 [DOI] [PubMed] [Google Scholar]
- 27.Ni J, Cozzi P, Hao J, Beretov J, Chang L, Duan W, Shigdar S, Delprado W, Graham P, Bucci J, Kearsley J, Li Y (2013) Epithelial cell adhesion molecule (EpCAM) is associated with prostate cancer metastasis and chemo/radioresistance via the PI3K/Akt/mTOR signaling pathway. Int J Biochem Cell Biol 45(12):2736–2748. 10.1016/j.biocel.2013.09.008 [DOI] [PubMed] [Google Scholar]
- 28.Wang G, Ma N, Meng L, Wei Y, Gui J (2015) Activation of the phosphatidylinositol 3-kinase/Akt pathway is involved in lipocalin-2-promoted human pulmonary artery smooth muscle cell proliferation. Mol Cell Biochem 410(1–2):207–213. 10.1007/s11010-015-2553-5 [DOI] [PubMed] [Google Scholar]
- 29.Weng CC, Kuo KK, Su HT, Hsiao PJ, Chen YW, Wu DC, Hung WC, Cheng KH (2016) Pancreatic tumor progression associated with CD133 overexpression: involvement of increased TERT expression and epidermal growth factor receptor-dependent Akt activation. Pancreas 45(3):443–457. 10.1097/MPA.0000000000000460 [DOI] [PubMed] [Google Scholar]
- 30.Zoratti GL, Tanabe LM, Varela FA, Murray AS, Bergum C, Colombo E, Lang JE, Molinolo AA, Leduc R, Marsault E, Boerner J, List K (2015) Targeting matriptase in breast cancer abrogates tumour progression via impairment of stromal-epithelial growth factor signalling. Nat Commun 6:6776. 10.1038/ncomms7776 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Yao HP, Zhou YQ, Zhang R, Wang MH (2013) MSP-RON signalling in cancer: pathogenesis and therapeutic potential. Nat Rev Cancer 13(7):466–481. 10.1038/nrc3545 [DOI] [PubMed] [Google Scholar]
- 32.Chang HY, Chang TC, Huang WY, Lee CT, Yen CJ, Tsai YS, Tzai TS, Chen SH, Chow NH (2016) RON nuclear translocation under hypoxia potentiates chemoresistance to DNA double-strand break-inducing anticancer drugs. Mol Cancer Ther 15(2):276–286. 10.1158/1535-7163.MCT-15-0311 [DOI] [PubMed] [Google Scholar]
- 33.Vandecaetsbeek I, Vangheluwe P, Raeymaekers L, Wuytack F, Vanoevelen J (2011) The Ca2+ pumps of the endoplasmic reticulum and Golgi apparatus. Cold Spring Harb Perspect Biol 10.1101/cshperspect.a004184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zhao F, Edwards R, Dizon D, Afrasiabi K, Mastroianni JR, Geyfman M, Ouellette AJ, Andersen B, Lipkin SM (2010) Disruption of Paneth and goblet cell homeostasis and increased endoplasmic reticulum stress in Agr2−/− mice. Dev Biol 338(2):270–279. 10.1016/j.ydbio.2009.12.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kawaguchi M, Takeda N, Hoshiko S, Yorita K, Baba T, Sawaguchi A, Nezu Y, Yoshikawa T, Fukushima T, Kataoka H (2011) Membrane-bound serine protease inhibitor HAI-1 is required for maintenance of intestinal epithelial integrity. Am J Pathol 179(4):1815–1826. 10.1016/j.ajpath.2011.06.038 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Zhang R, Shi H, Ren F, Li X, Zhang M, Feng W, Jia Y (2016) Knockdown of MACC1 expression increases cisplatin sensitivity in cisplatin-resistant epithelial ovarian cancer cells. Oncol Rep 35(4):2466–2472. 10.3892/or.2016.4585 [DOI] [PubMed] [Google Scholar]
- 37.Pan J, Li X, Wu W, Xue M, Hou H, Zhai W, Chen W (2016) Long non-coding RNA UCA1 promotes cisplatin/gemcitabine resistance through CREB modulating miR-196a-5p in bladder cancer cells. Cancer Lett 382(1):64–76. 10.1016/j.canlet.2016.08.015 [DOI] [PubMed] [Google Scholar]
- 38.Hosoya N, Miyagawa K (2014) Targeting DNA damage response in cancer therapy. Cancer Sci 105(4):370–388. 10.1111/cas.12366 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Avril T, Vauleon E, Chevet E (2017) Endoplasmic reticulum stress signaling and chemotherapy resistance in solid cancers. Oncogenesis 6(8):e373. 10.1038/oncsis.2017.72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Leroy B, Girard L, Hollestelle A, Minna JD, Gazdar AF, Soussi T (2014) Analysis of TP53 mutation status in human cancer cell lines: a reassessment. Hum Mutat 35(6):756–765. 10.1002/humu.22556 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Rasti M, Azimi T (2015) TP53 Binding to BRCA1 and RAD51 in MCF7 and MDA-MB-468 Breast Cancer Cell Lines In vivo and In vitro. Avicenna J Med Biotechnol 7(2):76–79 [PMC free article] [PubMed] [Google Scholar]
- 42.Sousa FG, Matuo R, Tang SW, Rajapakse VN, Luna A, Sander C, Varma S, Simon PH, Doroshow JH, Reinhold WC, Pommier Y (2015) Alterations of DNA repair genes in the NCI-60 cell lines and their predictive value for anticancer drug activity. DNA Repair (Amst) 28:107–115. 10.1016/j.dnarep.2015.01.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Verfaillie T, Salazar M, Velasco G, Agostinis P (2010) Linking ER stress to autophagy: potential implications for cancer therapy. Int J Cell Biol 2010:930509. 10.1155/2010/930509 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Hetz C (2012) The unfolded protein response: controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Cell Biol 13(2):89–102. 10.1038/nrm3270 [DOI] [PubMed] [Google Scholar]
- 45.Shapiro DJ, Livezey M, Yu L, Zheng X, Andruska N (2016) Anticipatory UPR activation: a protective pathway and target in cancer. Trends Endocrinol Metab 27(10):731–741. 10.1016/j.tem.2016.06.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Tabas I, Ron D (2011) Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress. Nat Cell Biol 13(3):184–190. 10.1038/ncb0311-184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Urra H, Dufey E, Lisbona F, Rojas-Rivera D (1833) Hetz C (2013) When ER stress reaches a dead end. Biochim Biophys Acta 12:3507–3517. 10.1016/j.bbamcr.2013.07.024 [DOI] [PubMed] [Google Scholar]
- 48.Mann MJ, Hendershot LM (2006) UPR activation alters chemosensitivity of tumor cells. Cancer Biol Ther 5(7):736–740 [DOI] [PubMed] [Google Scholar]
- 49.McCullough KD, Martindale JL, Klotz LO, Aw TY, Holbrook NJ (2001) Gadd153 sensitizes cells to endoplasmic reticulum stress by down-regulating Bcl2 and perturbing the cellular redox state. Mol Cell Biol 21(4):1249–1259. 10.1128/MCB.21.4.1249-1259.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Piton N, Wason J, Colasse E, Cornic M, Lemoine F, Le Pessot F, Marguet F, Sabourin JC (2016) Endoplasmic reticulum stress, unfolded protein response and development of colon adenocarcinoma. Virchows Arch 469(2):145–154. 10.1007/s00428-016-1961-6 [DOI] [PubMed] [Google Scholar]
- 51.Gao J, Yan Q, Liu S, Yang X (2014) Knockdown of EpCAM enhances the chemosensitivity of breast cancer cells to 5-fluorouracil by downregulating the antiapoptotic factor Bcl-2. PLoS One 9(7):e102590. 10.1371/journal.pone.0102590 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Liao MY, Lai JK, Kuo MY, Lu RM, Lin CW, Cheng PC, Liang KH, Wu HC (2015) An anti-EpCAM antibody EpAb2–6 for the treatment of colon cancer. Oncotarget 6(28):24947–24968. 10.18632/oncotarget.4453 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong ST, Choi H, El Rayes T, Ryu S, Troeger J, Schwabe RF, Vahdat LT, Altorki NK, Mittal V, Gao D (2015) Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature 527(7579):472–476. 10.1038/nature15748 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zheng X, Carstens JL, Kim J, Scheible M, Kaye J, Sugimoto H, Wu CC, LeBleu VS, Kalluri R (2015) Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 527(7579):525–530. 10.1038/nature16064 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zeindl-Eberhart E, Brandl L, Liebmann S, Ormanns S, Scheel SK, Brabletz T, Kirchner T, Jung A (2014) Epithelial-mesenchymal transition induces endoplasmic-reticulum-stress response in human colorectal tumor cells. PLoS One 9(1):e87386. 10.1371/journal.pone.0087386 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kwon MJ (2013) Emerging roles of claudins in human cancer. Int J Mol Sci 14(9):18148–18180. 10.3390/ijms140918148 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Luo CW, Wu CC, Chang SJ, Chang TM, Chen TY, Chai CY, Chang CL, Hou MF, Pan MR (2018) CHD4-mediated loss of E-cadherin determines metastatic ability in triple-negative breast cancer cells. Exp Cell Res 363(1):65–72. 10.1016/j.yexcr.2017.12.032 [DOI] [PubMed] [Google Scholar]
- 58.Fortier AM, Asselin E, Cadrin M (2013) Keratin 8 and 18 loss in epithelial cancer cells increases collective cell migration and cisplatin sensitivity through claudin1 up-regulation. J Biol Chem 288(16):11555–11571. 10.1074/jbc.M112.428920 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Jung H, Kim B, Moon BI, Oh E-S (2016) Cytokeratin 18 is necessary for initiation of TGF-β1-induced epithelial–mesenchymal transition in breast epithelial cells. Mol Cell Biochem 423(1–2):21–28 [DOI] [PubMed] [Google Scholar]
- 60.Prislei S, Mariani M, Raspaglio G, Mozzetti S, Filippetti F, Ferrandina G, Scambia G, Ferlini C (2010) RON and cisplatin resistance in ovarian cancer cell lines. Oncol Res 19(1):13–22 [DOI] [PubMed] [Google Scholar]
- 61.Logan-Collins J, Thomas RM, Yu P, Jaquish D, Mose E, French R, Stuart W, McClaine R, Aronow B, Hoffman RM, Waltz SE, Lowy AM (2010) Silencing of RON receptor signaling promotes apoptosis and gemcitabine sensitivity in pancreatic cancers. Cancer Res 70(3):1130–1140. 10.1158/0008-5472.CAN-09-0761 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Perathoner A, Pirkebner D, Brandacher G, Spizzo G, Stadlmann S, Obrist P, Margreiter R, Amberger A (2005) 14–3-3sigma expression is an independent prognostic parameter for poor survival in colorectal carcinoma patients. Clin Cancer Res 11(9):3274–3279. 10.1158/1078-0432.CCR-04-2207 [DOI] [PubMed] [Google Scholar]
- 63.Fan Y, Shen B, Tan M, Mu X, Qin Y, Zhang F, Liu Y (2014) Long non-coding RNA UCA1 increases chemoresistance of bladder cancer cells by regulating Wnt signaling. FEBS J 281(7):1750–1758. 10.1111/febs.12737 [DOI] [PubMed] [Google Scholar]
- 64.Yamazaki T, Sasaki N, Nishi M, Takeshima H (2010) Facilitation of DNA damage-induced apoptosis by endoplasmic reticulum protein mitsugumin23. Biochem Biophys Res Commun 392(2):196–200. 10.1016/j.bbrc.2010.01.013 [DOI] [PubMed] [Google Scholar]
- 65.Yamamori T, Meike S, Nagane M, Yasui H, Inanami O (2013) ER stress suppresses DNA double-strand break repair and sensitizes tumor cells to ionizing radiation by stimulating proteasomal degradation of Rad51. FEBS Lett 587(20):3348–3353. 10.1016/j.febslet.2013.08.030 [DOI] [PubMed] [Google Scholar]
- 66.Dicks N, Gutierrez K, Michalak M, Bordignon V, Agellon LB (2015) Endoplasmic reticulum stress, genome damage, and cancer. Front Oncol 5:11. 10.3389/fonc.2015.00011 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.